Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Anal Bioanal Chem ; 411(12): 2493-2509, 2019 May.
Article in English | MEDLINE | ID: mdl-30911800

ABSTRACT

Inhalation of Bacillus anthracis spores can cause a rapidly progressing fatal infection. B. anthracis secretes three protein toxins: lethal factor (LF), edema factor (EF), and protective antigen (PA). EF and LF may circulate as free or PA-bound forms. Both free EF (EF) and PA-bound-EF (ETx) have adenylyl cyclase activity converting ATP to cAMP. We developed an adenylyl cyclase activity-based method for detecting and quantifying total EF (EF+ETx) in plasma. The three-step method includes magnetic immunocapture with monoclonal antibodies, reaction with ATP generating cAMP, and quantification of cAMP by isotope-dilution HPLC-MS/MS. Total EF was quantified from 5PL regression of cAMP vs ETx concentration. The detection limit was 20 fg/mL (225 zeptomoles/mL for the 89 kDa protein). Relative standard deviations for controls with 0.3, 6.0, and 90 pg/mL were 11.7-16.6% with 91.2-99.5% accuracy. The method demonstrated 100% specificity in 238 human serum/plasma samples collected from unexposed healthy individuals, and 100% sensitivity in samples from 3 human and 5 rhesus macaques with inhalation anthrax. Analysis of EF in the rhesus macaques showed that it was detected earlier post-exposure than B. anthracis by culture and PCR. Similar to LF, the kinetics of EF over the course of infection were triphasic, with an initial rise (phase-1), decline (phase-2), and final rapid rise (phase-3). EF levels were ~ 2-4 orders of magnitude lower than LF during phase-1 and phase-2 and only ~ 6-fold lower at death/euthanasia. Analysis of EF improves early diagnosis and adds to our understanding of anthrax toxemia throughout infection. The LF/EF ratio may also indicate the stage of infection and need for advanced treatments.


Subject(s)
Anthrax/pathology , Antigens, Bacterial/blood , Bacillus anthracis/pathogenicity , Bacterial Toxins/blood , Chromatography, High Pressure Liquid/methods , Respiratory Tract Infections/pathology , Tandem Mass Spectrometry/methods , Toxemia/pathology , Adenosine Triphosphate/metabolism , Animals , Anthrax/blood , Case-Control Studies , Cyclic AMP/biosynthesis , Disease Progression , Enzyme-Linked Immunosorbent Assay , Humans , Limit of Detection , Macaca mulatta , Polymerase Chain Reaction , Respiratory Tract Infections/blood , Toxemia/blood , Toxemia/microbiology
2.
J Biol Chem ; 291(42): 22021-22029, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27555325

ABSTRACT

The protective antigen (PA) moiety of anthrax toxin binds to cellular receptors and mediates the translocation of the two enzymatic moieties of the toxin to the cytosol. Two PA receptors are known, with capillary morphogenesis protein 2 (CMG2) being the more important for pathogenesis and tumor endothelial marker 8 (TEM8) playing a minor role. The C-terminal PA domain 4 (PAD4) has extensive interactions with the receptors and is required for binding. Our previous study identified PAD4 variants having enhanced TEM8 binding specificity. To obtain PA variants that selectively bind to CMG2, here we performed phage display selections using magnetic beads having bound CMG2. We found that PA residue isoleucine 656 plays a critical role in PA binding to TEM8 but has a much lesser effect on PA binding to CMG2. We further characterized the role of residue 656 in distinguishing PA binding to CMG2 versus TEM8 by substituting it with the other 19 amino acids. Of the resulting variants, PA I656Q and PA I656V had significantly reduced activity on TEM8-expressing CHO cells but maintained their activity on CMG2-expressing CHO cells. The preference of these PA mutants for CMG2 over TEM8 was further demonstrated using mouse embryonic fibroblast cells and mice deficient in the CMG2 and/or the TEM8 receptors. The structural basis of the alterations in the receptor binding activities of these mutants is also discussed.


Subject(s)
Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Biomarkers, Tumor/metabolism , Mutation, Missense , Receptors, Peptide/metabolism , Amino Acid Substitution , Animals , Antigens, Bacterial/genetics , Bacillus anthracis/genetics , Bacterial Toxins/genetics , Biomarkers, Tumor/genetics , CHO Cells , Cricetinae , Cricetulus , Embryo, Mammalian , Fibroblasts , Mice , Microfilament Proteins , Protein Binding , Protein Domains , Receptors, Cell Surface , Receptors, Peptide/genetics , Structure-Activity Relationship
3.
J Biol Chem ; 290(10): 6584-95, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25564615

ABSTRACT

Anthrax disease is caused by a toxin consisting of protective antigen (PA), lethal factor, and edema factor. Antibodies against PA have been shown to be protective against the disease. Variable domains of camelid heavy chain-only antibodies (VHHs) with affinity for PA were obtained from immunized alpacas and screened for anthrax neutralizing activity in macrophage toxicity assays. Two classes of neutralizing VHHs were identified recognizing distinct, non-overlapping epitopes. One class recognizes domain 4 of PA at a well characterized neutralizing site through which PA binds to its cellular receptor. A second neutralizing VHH (JKH-C7) recognizes a novel epitope. This antibody inhibits conversion of the PA oligomer from "pre-pore" to its SDS and heat-resistant "pore" conformation while not preventing cleavage of full-length 83-kDa PA (PA83) by cell surface proteases to its oligomer-competent 63-kDa form (PA63). The antibody prevents endocytosis of the cell surface-generated PA63 subunit but not preformed PA63 oligomers formed in solution. JKH-C7 and the receptor-blocking VHH class (JIK-B8) were expressed as a heterodimeric VHH-based neutralizing agent (VNA2-PA). This VNA displayed improved neutralizing potency in cell assays and protected mice from anthrax toxin challenge with much better efficacy than the separate component VHHs. The VNA protected virtually all mice when separately administered at a 1:1 ratio to toxin and protected mice against Bacillus anthracis spore infection. Thus, our studies show the potential of VNAs as anthrax therapeutics. Due to their simple and stable nature, VNAs should be amenable to genetic delivery or administration via respiratory routes.


Subject(s)
Anthrax/immunology , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Immunoglobulin Heavy Chains/immunology , Animals , Anthrax/microbiology , Anthrax/pathology , Anthrax/therapy , Antibodies, Bacterial/administration & dosage , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/antagonists & inhibitors , Camelids, New World/immunology , Epitopes/immunology , Humans , Mice , Spores/immunology , Spores/pathogenicity
4.
Sci Rep ; 4: 4754, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24755540

ABSTRACT

We characterized an anti-cancer fusion protein consisting of anthrax lethal factor (LF) and the catalytic domain of Pseudomonas exotoxin A by (i) mutating the N-terminal amino acids and by (ii) reductive methylation to dimethylate all lysines. Dimethylation of lysines was achieved quantitatively and specifically without affecting binding of the fusion protein to PA or decreasing the enzymatic activity of the catalytic moiety. Ubiquitination in vitro was drastically decreased for both the N-terminally mutated and dimethylated variants, and both appeared to be slightly more stable in the cytosol of treated cells. The dimethylated variant showed greatly reduced neutralization by antibodies to LF. The two described modifications offer unique advantages such as increased cytotoxic activity and diminished antibody recognition, and thus may be applicable to other therapeutic proteins that act in the cytosol of cells.


Subject(s)
ADP Ribose Transferases/genetics , Antigens, Bacterial/genetics , Bacterial Toxins/genetics , Exotoxins/genetics , Mutation , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/toxicity , Virulence Factors/genetics , ADP Ribose Transferases/chemistry , Animals , Antigens, Bacterial/chemistry , Antineoplastic Agents , Bacterial Toxins/chemistry , Cell Line , Cricetinae , Cytosol/metabolism , Epitopes/genetics , Epitopes/immunology , Epitopes/metabolism , Exotoxins/chemistry , Humans , Kinetics , Mass Spectrometry , Methylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Ubiquitination , Virulence Factors/chemistry , Pseudomonas aeruginosa Exotoxin A
5.
Infect Immun ; 81(12): 4592-603, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24082082

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) is a major cause of severe food-borne disease worldwide, and two Shiga toxins, Stx1 and Stx2, are primarily responsible for the serious disease consequence, hemolytic-uremic syndrome (HUS). Here we report identification of a panel of heavy-chain-only antibody (Ab) V(H) (VHH) domains that neutralize Stx1 and/or Stx2 in cell-based assays. VHH heterodimer toxin-neutralizing agents containing two linked Stx1-neutralizing VHHs or two Stx2-neutralizing VHHs were generally much more potent at Stx neutralization than a pool of the two-component monomers tested in cell-based assays and in vivo mouse models. We recently reported that clearance of toxins can be promoted by coadministering a VHH-based toxin-neutralizing agent with an antitag monoclonal antibody (MAb), called the "effector Ab," that indirectly decorates each toxin molecule with four Ab molecules. Decoration occurs because the Ab binds to a common epitopic tag present at two sites on each of the two VHH heterodimer molecules that bind to each toxin molecule. Here we show that coadministration of effector Ab substantially improved the efficacy of Stx toxin-neutralizing agents to prevent death or kidney damage in mice following challenge with Stx1 or Stx2. A single toxin-neutralizing agent consisting of a double-tagged VHH heterotrimer--one Stx1-specific VHH, one Stx2-specific VHH, and one Stx1/Stx2 cross-specific VHH--was effective in preventing all symptoms of intoxication from Stx1 and Stx2 when coadministered with effector Ab. Overall, the availability of simple, defined, recombinant proteins that provide cost-effective protection against HUS opens up new therapeutic approaches to managing disease.


Subject(s)
Escherichia coli Infections/immunology , Hemolytic-Uremic Syndrome/immunology , Shiga Toxin 1/immunology , Shiga Toxin 2/immunology , Single-Domain Antibodies/immunology , Animals , Antibodies, Monoclonal/immunology , Enteropathogenic Escherichia coli/immunology , Enteropathogenic Escherichia coli/metabolism , Female , Mice , Molecular Sequence Data , Shiga Toxin 1/metabolism , Shiga Toxin 2/metabolism , Shiga-Toxigenic Escherichia coli/immunology , Shiga-Toxigenic Escherichia coli/metabolism
6.
PLoS One ; 8(8): e74474, 2013.
Article in English | MEDLINE | ID: mdl-24015319

ABSTRACT

Anthrax edema factor (EF) is a calmodulin-dependent adenylate cyclase that converts adenosine triphosphate (ATP) into 3'-5'-cyclic adenosine monophosphate (cAMP), contributing to the establishment of Bacillus anthracis infections and the resulting pathophysiology. We show that EF adenylate cyclase toxin activity is strongly mediated by the N-end rule, and thus is dependent on the identity of the N-terminal amino acid. EF variants having different N-terminal residues varied by more than 100-fold in potency in cultured cells and mice. EF variants having unfavorable, destabilizing N-terminal residues showed much greater activity in cells when the E1 ubiquitin ligase was inactivated or when proteasome inhibitors were present. Taken together, these results show that EF is uniquely affected by ubiquitination and/or proteasomal degradation.


Subject(s)
Adenylyl Cyclases/metabolism , Antigens, Bacterial/metabolism , Bacillus anthracis/enzymology , Bacterial Toxins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Adenylyl Cyclases/genetics , Animals , Antigens, Bacterial/genetics , Bacillus anthracis/genetics , Bacterial Toxins/genetics , Cell Line , Mice
7.
J Mol Recognit ; 26(8): 376-81, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23784994

ABSTRACT

The monoclonal antibody S9.6 binds DNA-RNA hybrids with high affinity, making it useful in research and diagnostic applications, such as in microarrays and in the detection of R-loops. A single-chain variable fragment (scFv) of S9.6 was produced, and its affinities for various synthetic nucleic acid hybrids were measured by surface plasmon resonance (SPR). S9.6 exhibits dissociation constants of approximately 0.6 nM for DNA-RNA and, surprisingly, 2.7 nM for RNA-RNA hybrids that are AU-rich. The affinity of the S9.6 scFv did not appear to be strongly influenced by various buffer conditions or by ionic strength below 500 mM NaCl. The smallest epitope that was strongly bound by the S9.6 scFv contained six base pairs of DNA-RNA hybrid. Published 2013. This article is a U.S. Government work and is in the public domain in the USA.


Subject(s)
Antibodies, Monoclonal/metabolism , DNA/metabolism , Nucleic Acid Heteroduplexes/metabolism , RNA/metabolism , Single-Chain Antibodies/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/isolation & purification , Antibody Affinity , Antibody Specificity , Buffers , Cations, Divalent/chemistry , DNA/chemistry , Epitopes/chemistry , Epitopes/metabolism , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Nucleic Acid Conformation , Nucleic Acid Heteroduplexes/chemistry , Osmolar Concentration , RNA/chemistry , Single-Chain Antibodies/isolation & purification , Surface Plasmon Resonance
8.
Antimicrob Agents Chemother ; 57(9): 4139-45, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23774434

ABSTRACT

Bacillus anthracis, the causative agent of anthrax, manifests its pathogenesis through the action of two secreted toxins. The bipartite lethal and edema toxins, a combination of lethal factor or edema factor with the protein protective antigen, are important virulence factors for this bacterium. We previously developed small-molecule inhibitors of lethal factor proteolytic activity (LFIs) and demonstrated their in vivo efficacy in a rat lethal toxin challenge model. In this work, we show that these LFIs protect against lethality caused by anthrax infection in mice when combined with subprotective doses of either antibiotics or neutralizing monoclonal antibodies that target edema factor. Significantly, these inhibitors provided protection against lethal infection when administered as a monotherapy. As little as two doses (10 mg/kg) administered at 2 h and 8 h after spore infection was sufficient to provide a significant survival benefit in infected mice. Administration of LFIs early in the infection was found to inhibit dissemination of vegetative bacteria to the organs in the first 32 h following infection. In addition, neutralizing antibodies against edema factor also inhibited bacterial dissemination with similar efficacy. Together, our findings confirm the important roles that both anthrax toxins play in establishing anthrax infection and demonstrate the potential for small-molecule therapeutics targeting these proteins.


Subject(s)
Anthrax/drug therapy , Anti-Bacterial Agents/pharmacology , Antibodies, Neutralizing/pharmacology , Bacillus anthracis/drug effects , Bacterial Toxins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Spores, Bacterial/drug effects , Animals , Anthrax/microbiology , Anthrax/mortality , Anti-Bacterial Agents/pharmacokinetics , Antigens, Bacterial , Bacillus anthracis/growth & development , Drug Administration Schedule , Female , Mice , Mice, Inbred C57BL , Molecular Weight , Protease Inhibitors/pharmacokinetics , Spores, Bacterial/growth & development , Survival Analysis , Time Factors
9.
J Biol Chem ; 288(13): 9058-65, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23393143

ABSTRACT

Anthrax toxin protective antigen (PA) delivers its effector proteins into the host cell cytosol through formation of an oligomeric pore, which can assume heptameric or octameric states. By screening a highly directed library of PA mutants, we identified variants that complement each other to exclusively form octamers. These PA variants were individually nontoxic and demonstrated toxicity only when combined with their complementary partner. We then engineered requirements for activation by matrix metalloproteases and urokinase plasminogen activator into two of these variants. The resulting therapeutic toxin specifically targeted cells expressing both tumor associated proteases and completely stopped tumor growth in mice when used at a dose far below that which caused toxicity. This scheme for obtaining intercomplementing subunits can be employed with other oligomeric proteins and potentially has wide application.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Neoplasms/drug therapy , Animals , Bacillus anthracis/metabolism , Cell Line, Tumor , Female , Gene Library , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Molecular Conformation , Mutation , Neoplasms/metabolism , Plasmids/metabolism , Protein Conformation , Protein Engineering/methods , Protein Interaction Mapping/methods , Protein Structure, Tertiary , Proteins/chemistry , Ultracentrifugation
10.
PLoS One ; 7(1): e29941, 2012.
Article in English | MEDLINE | ID: mdl-22238680

ABSTRACT

Antitoxins are needed that can be produced economically with improved safety and shelf life compared to conventional antisera-based therapeutics. Here we report a practical strategy for development of simple antitoxin therapeutics with substantial advantages over currently available treatments. The therapeutic strategy employs a single recombinant 'targeting agent' that binds a toxin at two unique sites and a 'clearing Ab' that binds two epitopes present on each targeting agent. Co-administration of the targeting agent and the clearing Ab results in decoration of the toxin with up to four Abs to promote accelerated clearance. The therapeutic strategy was applied to two Botulinum neurotoxin (BoNT) serotypes and protected mice from lethality in two different intoxication models with an efficacy equivalent to conventional antitoxin serum. Targeting agents were a single recombinant protein consisting of a heterodimer of two camelid anti-BoNT heavy-chain-only Ab V(H) (VHH) binding domains and two E-tag epitopes. The clearing mAb was an anti-E-tag mAb. By comparing the in vivo efficacy of treatments that employed neutralizing vs. non-neutralizing agents or the presence vs. absence of clearing Ab permitted unprecedented insight into the roles of toxin neutralization and clearance in antitoxin efficacy. Surprisingly, when a post-intoxication treatment model was used, a toxin-neutralizing heterodimer agent fully protected mice from intoxication even in the absence of clearing Ab. Thus a single, easy-to-produce recombinant protein was as efficacious as polyclonal antiserum in a clinically-relevant mouse model of botulism. This strategy should have widespread application in antitoxin development and other therapies in which neutralization and/or accelerated clearance of a serum biomolecule can offer therapeutic benefit.


Subject(s)
Antitoxins/biosynthesis , Antitoxins/therapeutic use , Botulism/therapy , Immunotherapy/trends , Animals , Antibody Affinity , Antitoxins/metabolism , Botulinum Antitoxin/biosynthesis , Botulinum Antitoxin/metabolism , Botulinum Antitoxin/therapeutic use , Botulism/immunology , Botulism/mortality , Botulism/pathology , Disease Models, Animal , Drug Discovery/methods , Drugs, Investigational/metabolism , Drugs, Investigational/therapeutic use , Female , Immunotherapy/methods , Mice , Models, Biological , Protein Multimerization/physiology , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Survival Analysis , Treatment Outcome
11.
Infect Immun ; 79(11): 4609-16, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21911463

ABSTRACT

Bacillus anthracis is the causative agent of anthrax, and the tripartite anthrax toxin is an essential element of its pathogenesis. Edema factor (EF), a potent adenylyl cyclase, is one of the toxin components. In this work, anti-EF monoclonal antibodies (MAb) were produced following immunization of mice, and four of the antibodies were fully characterized. MAb 3F2 has an affinity of 388 pM, was most effective for EF detection, and appears to be the first antibody reported to neutralize EF by binding to the catalytic C(B) domain. MAb 7F10 shows potent neutralization of edema toxin activity in vitro and in vivo; it targets the N-terminal protective antigen binding domain. The four MAb react with three different domains of edema factor, and all were able to detect purified edema factor in Western blot analysis. None of the four MAb cross-reacted with the lethal factor toxin component. Three of the four MAb protected mice in both a systemic edema toxin challenge model and a subcutaneous spore-induced foreleg edema model. A combination of three of the MAb also significantly delayed the time to death in a third subcutaneous spore challenge model. This appears to be the first direct evidence that monoclonal antibody-mediated neutralization of EF alone is sufficient to delay anthrax disease progression.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Animals , Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Cell Line , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/prevention & control , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Hybridomas , Immunization , Immunoglobulin G , Macrophages/immunology , Mice , Mice, Inbred BALB C
12.
Proc Natl Acad Sci U S A ; 107(32): 14070-4, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660775

ABSTRACT

The tripartite protein exotoxin secreted by Bacillus anthracis, a major contributor to its virulence and anthrax pathogenesis, consists of binary complexes of the protective antigen (PA) heptamer (PA63h), produced by proteolytic cleavage of PA, together with either lethal factor or edema factor. The mouse monoclonal anti-PA antibody 1G3 was previously shown to be a potent antidote that shares F(C) domain dependency with the human monoclonal antibody MDX-1303 currently under clinical development. Here we demonstrate that 1G3 instigates severe perturbation of the PA63h structure and creates a PA supercomplex as visualized by electron microscopy. This phenotype, produced by the unconventional mode of antibody action, highlights the feasibility for optimization of vaccines based on analogous structural modification of PA63h as an additional strategy for future remedies against anthrax.


Subject(s)
Antibodies, Neutralizing , Antigen-Antibody Complex/chemistry , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Animals , Antibodies, Monoclonal , Humans , Mice , Protein Conformation , Protein Multimerization
13.
J Mol Biol ; 387(3): 680-93, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19361425

ABSTRACT

The virulence of Bacillus anthracis is critically dependent on the cytotoxic components of the anthrax toxin, lethal factor (LF) and edema factor (EF). LF and EF gain entry into host cells through interactions with the protective antigen (PA), which binds to host cellular receptors such as CMG2. Antibodies that neutralize PA have been shown to confer protection in animal models and are undergoing intense clinical development. A murine monoclonal antibody, 14B7, has been reported to interact with domain 4 of PA (PAD4) and block its binding to CMG2. More recently, the 14B7 antibody was used as the platform for the selection of very high affinity, single-chain antibodies that have tremendous potential as a combination anthrax prophylactic and treatment. Here, we report the high-resolution X-ray structures of three high-affinity, single-chain antibodies in the 14B7 family; 14B7 and two high-affinity variants 1H and M18. In addition, we present the first neutralizing antibody-PA structure, M18 in complex with PAD4 at 3.8 A resolution. These structures provide insights into the mechanism of neutralization, and the effect of various mutations on antibody affinity, and enable a comparison between the binding of the M18 antibody and CMG2 with PAD4.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Peptide Fragments/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody Affinity , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Crystallography, X-Ray , Immunoglobulin Variable Region , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/genetics , Peptide Fragments/metabolism
14.
Protein Sci ; 18(2): 259-67, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19177559

ABSTRACT

Disulfide bonds play a critical role in the stabilization of the immunoglobulin beta-sandwich sandwich. Under reducing conditions, such as those that prevail in the cytoplasm, disulfide bonds do not normally form and as a result most antibodies expressed in that compartment (intrabodies) accumulate in a misfolded and inactive state. We have developed a simple method for the quantitative isolation of antibody fragments that retain full activity under reducing conditions from large mutant libraries. In E. coli, inactivation of the cysteine oxidoreductase DsbA abolishes protein oxidation in the periplasm, which leads to the accumulation of scFvs and other disulfide-containing proteins in a reduced form. Libraries of mutant scFvs were tethered onto the inner membrane of dsbA cells and mutants that could bind fluorescently labeled antigen in the reducing periplasm were screened by Anchored Periplasmic Expression (APEx; Harvey et al., Proc Natl Acad Sci USA 2004;101:9193-9198.). Using this approach, we isolated scFv antibody variants that are fully active when expressed in the cytoplasm or when the four Cys residues that normally form disulfides are substituted by Ser residues.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Disulfides/metabolism , Immunoglobulin Variable Region/metabolism , Protein Folding , Amino Acid Sequence , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Cysteine/metabolism , Directed Molecular Evolution , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Flow Cytometry , Gene Library , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Molecular Sequence Data , Mutation , Oxidation-Reduction , Protein Conformation , Protein Disulfide-Isomerases/genetics , Protein Engineering , Sequence Alignment , Serine/metabolism
15.
Mol Cancer Ther ; 7(1): 48-58, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18202009

ABSTRACT

Chronic myelogenous leukemia (CML) is driven by constitutively activated Bcr-Abl tyrosine kinase, which causes the defective adhesion of CML cells to bone marrow stroma. The overexpression of p210Bcr-Abl was reported to down-regulate CXCR4 expression, and this is associated with the cell migration defects in CML. We proposed that tyrosine kinase inhibitors, imatinib or INNO-406, may restore CXCR4 expression and cause the migration of CML cells to bone marrow microenvironment niches, which in turn results in acquisition of stroma-mediated chemoresistance of CML progenitor cells. In KBM5 and K562 cells, imatinib, INNO-406, or IFN-alpha increased CXCR4 expression and migration. This increase in CXCR4 levels on CML progenitor cells was likewise found in samples from CML patients treated with imatinib or IFN-alpha. Imatinib induced G0-G1 cell cycle block in CML cells, which was further enhanced in a mesenchymal stem cell (MSC) coculture system. MSC coculture protected KBM-5 cells from imatinib-induced cell death. These antiapoptotic effects were abrogated by the CXCR4 antagonist AMD3465 or by inhibitor of integrin-linked kinase QLT0267. Altogether, these findings suggest that the up-regulation of CXCR4 by imatinib promotes migration of CML cells to bone marrow stroma, causing the G0-G1 cell cycle arrest and hence ensuring the survival of quiescent CML progenitor cells.


Subject(s)
Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Movement/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptors, CXCR4/metabolism , Antigens, CD34/metabolism , Benzamides , Cell Line, Tumor , Cell Survival/drug effects , Humans , Imatinib Mesylate , Interferon-alpha/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Up-Regulation/drug effects
16.
Stem Cells ; 22(2): 188-201, 2004.
Article in English | MEDLINE | ID: mdl-14990858

ABSTRACT

The aim of this study was to investigate factors influencing the engraftment potential of acute myeloid leukemia (AML) CD34+ cells in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. We examined the relationship between engraftment, CXCR4 expression on CD34+ and CD34+CD38- cells, and patient (Pt) clinical/laboratory characteristics in 44 samples from 11 Pts. Engraftment, evaluated by Southern blot and CD45 flow cytometric analyses, was observed in murine bone marrow of 6 of 11 Pt samples, ranging from 0.1% to 73.9% by Southern blot and from 0.1%-36.8% by flow cytometry. Poor Pt prognosis was inversely correlated with engraftment; the median overall survival was 95.9 weeks for Pts whose cells did not engraft and 26.1 weeks for those whose cells did engraft (p = 0.012, log-rank test). No other clinical/laboratory variable predicted engraftment. No correlation between the level of CXCR4 expression on AML cells and engraftment was observed. Cells with virtually absent CXCR4 expression were able to engraft, and cells from two Pts with high expression levels of CXCR4 did not engraft. Furthermore, anti-CXCR4 antibody failed to block the engraftment of AML cells into NOD/SCID mice. In conclusion, we demonstrated that CXCR4 is not critical for the engraftment of AML CD34+ cells in NOD/SCID mice. The model may, however, reflect the clinical course of the disease.


Subject(s)
Bone Marrow/metabolism , Graft Survival/physiology , Leukemia, Myeloid, Acute , Receptors, CXCR4/metabolism , Animals , Antigens, CD34/immunology , Flow Cytometry , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation/physiology
17.
Blood ; 99(9): 3461-4, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11964319

ABSTRACT

Recent studies suggest that the Bcl-2 and mitogen-activated protein kinase (MAPK) pathways together confer an aggressive, apoptosis-resistant phenotype on acute myelogenous leukemia (AML) cells. In this study, we analyzed the effects of simultaneous inhibition of these 2 pathways. In AML cell lines with constitutively activated MAPK, MAPK kinase (MEK) blockade by PD184352 strikingly potentiated the apoptosis induced by the small-molecule Bcl-2 inhibitor HA14-1 or by Bcl-2 antisense oligonucleotides. Isobologram analysis confirmed the synergistic nature of this interaction. Moreover, MEK blockade overcame Bcl-2 overexpression-mediated resistance to the proapoptotic effects of HA14-1. Most importantly, simultaneous exposure to PD184352 significantly (P =.01) potentiated HA14-1-mediated inhibition of clonogenic growth in all primary AML samples tested. These findings show that the Bcl-2 and MAPK pathways are relevant molecular targets in AML and that their concurrent inhibition could be developed into a new therapeutic strategy for this disease.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Leukemia, Myeloid, Acute/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Benzamides/pharmacology , Benzopyrans/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Humans , Nitriles/pharmacology , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Cells, Cultured/drug effects
18.
Blood ; 99(1): 326-35, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11756188

ABSTRACT

It has been shown that the novel synthetic triterpenoid CDDO inhibits proliferation and induces differentiation and apoptosis in myeloid leukemia cells. In the current study the effects of the C-28 methyl ester of CDDO, CDDO-Me, were analyzed on cell growth and apoptosis of leukemic cell lines and primary acute myelogenous leukemia (AML). CDDO-Me decreased the viability of leukemic cell lines, including multidrug resistant (MDR)-1-overexpressing, p53(null) HL-60-Dox and of primary AML cells, and it was 3- to 5-fold more active than CDDO. CDDO-Me induced a loss of mitochondrial membrane potential, induction of caspase-3 cleavage, increase in annexin V binding and DNA fragmentation, suggesting the induction of apoptosis. CDDO-Me induced pro-apoptotic Bax protein that preceded caspase activation. Furthermore, CDDO-Me inhibited the activation of ERK1/2, as determined by the inhibition of mitochondrial ERK1/2 phosphorylation, and it blocked Bcl-2 phosphorylation, rendering Bcl-2 less anti-apoptotic. CDDO-Me induced granulo-monocytic differentiation in HL-60 cells and monocytic differentiation in primary cells. Of significance, colony formation of AML progenitors was significantly inhibited in a dose-dependent fashion, whereas normal CD34(+) progenitor cells were less affected. Combinations with ATRA or the RXR-specific ligand LG100268 enhanced the effects of CDDO-Me on cell viability and terminal differentiation of myeloid leukemic cell lines. In conclusion, CDDO-Me is an MDR-1- and a p53-independent compound that exerts strong antiproliferative, apoptotic, and differentiating effects in myeloid leukemic cell lines and in primary AML samples when given in submicromolar concentrations. Differential effects of CDDO-Me on leukemic and normal progenitor cells suggest that CDDO-Me has potential as a novel compound in the treatment of hematologic malignancies.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Leukemia, Myeloid, Acute/pathology , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Annexin A5/metabolism , Blast Crisis/pathology , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cytarabine/pharmacology , DNA Fragmentation/drug effects , Drug Interactions , Flow Cytometry , HL-60 Cells/drug effects , Humans , Membrane Potentials/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinoids/pharmacology , Tretinoin/pharmacology , Tumor Cells, Cultured , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...