Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
eNeuro ; 10(6)2023 Jun.
Article in English | MEDLINE | ID: mdl-37321845

ABSTRACT

Peripheral administration of tissue inhibitor of metalloproteinases 2 (TIMP2), a protein inhibitor of matrix metalloproteinases (MMPs), has previously been shown to have beneficial effects on cognition and neurons in aged mice. Here, to better understand the potential of recombinant TIMP2 proteins, an IgG4Fc fusion protein (TIMP2-hIgG4) was developed to extend the plasma half-life of TIMP2. Following one month of administration of TIMP2 or TIMP2-hIgG4 via intraperitoneal injections, 23-month-old male C57BL/6J mice showed improved hippocampal-dependent memory in a Y-maze, increased hippocampal cfos gene expression, and increased excitatory synapse density in the CA1 and dentate gyrus (DG) of the hippocampus. Thus, fusion to hIgG4 extended the half-life of TIMP2 while retaining the beneficial cognitive and neuronal effects. Moreover, it retained its ability to cross the blood-brain barrier. To deepen the mechanistic understanding of the beneficial function of TIMP2 on neuronal activity and cognition, a TIMP2 construct lacking MMP inhibitory activity, Ala-TIMP2, was generated, which provides steric hindrance that prevents inhibition of MMPs by the TIMP2 protein while still allowing MMP binding. A comprehensive assessment of the MMP inhibitory and binding capacity of these engineered proteins is outlined. Surprisingly, MMP inhibition by TIMP2 was not essential for its beneficial effects on cognition and neuronal function. These findings both confirm previously published research, expand on the potential mechanism for the beneficial effects of TIMP2, and provide important details for a therapeutic path forward for TIMP2 recombinant proteins in aging-related cognitive decline.


Subject(s)
Cognition , Matrix Metalloproteinases , Animals , Male , Mice , Aging , Matrix Metalloproteinases/metabolism , Mice, Inbred C57BL
3.
Sci Rep ; 9(1): 8420, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182754

ABSTRACT

Human CLDN18.2 is highly expressed in a significant proportion of gastric and pancreatic adenocarcinomas, while normal tissue expression is limited to the epithelium of the stomach. The restricted expression makes it a potential drug target for the treatment of gastric and pancreatic adenocarcinoma, as evidenced by efforts to target CLDN18.2 via naked antibody and CAR-T modalities. Herein we describe CLDN18.2-targeting via a CD3-bispecific and an antibody drug conjugate and the characterization of these potential therapeutic molecules in efficacy and preliminary toxicity studies. Anti-hCLDN18.2 ADC, CD3-bispecific and diabody, targeting a protein sequence conserved in rat, mouse and monkey, exhibited in vitro cytotoxicity in BxPC3/hCLDN18.2 (IC50 = 1.52, 2.03, and 0.86 nM) and KATO-III/hCLDN18.2 (IC50 = 1.60, 0.71, and 0.07 nM) respectively and inhibited tumor growth of pancreatic and gastric patient-derived xenograft tumors. In a rat exploratory toxicity study, the ADC was tolerated up to 10 mg/kg. In a preliminary assessment of tolerability, the anti-CLDN18.2 diabody (0.34 mg/kg) did not produce obvious signs of toxicity in the stomach of NSG mice 4 weeks after dosing. Taken together, our data indicate that targeting CLDN18.2 with an ADC or bispecific modality could be a valid therapeutic approach for the treatment of gastric and pancreatic cancer.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , Claudins/immunology , Immunoconjugates/therapeutic use , Pancreatic Neoplasms/therapy , Stomach Neoplasms/therapy , Adenocarcinoma/therapy , Animals , Carcinoma, Pancreatic Ductal/therapy , Cell Line, Tumor , Humans , Immunoconjugates/blood , Mice , Pancreatic Neoplasms/blood , Rats , Stomach Neoplasms/blood
4.
Nat Commun ; 7: 13376, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27857134

ABSTRACT

Staphylococcus aureus is both an important pathogen and a human commensal. To explore this ambivalent relationship between host and microbe, we analysed the memory humoral response against IsdB, a protein involved in iron acquisition, in four healthy donors. Here we show that in all donors a heavily biased use of two immunoglobulin heavy chain germlines generated high affinity (pM) antibodies that neutralize the two IsdB NEAT domains, IGHV4-39 for NEAT1 and IGHV1-69 for NEAT2. In contrast to the typical antibody/antigen interactions, the binding is primarily driven by the germline-encoded hydrophobic CDRH-2 motifs of IGHV1-69 and IGHV4-39, with a binding mechanism nearly identical for each antibody derived from different donors. Our results suggest that IGHV1-69 and IGHV4-39, while part of the adaptive immune system, may have evolved under selection pressure to encode a binding motif innately capable of recognizing and neutralizing a structurally conserved protein domain involved in pathogen iron acquisition.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Proteins/immunology , Staphylococcal Infections/microbiology , Virulence Factors/immunology , Antibodies, Neutralizing , B-Lymphocytes , Humans , Immunologic Memory , Models, Molecular , Protein Conformation , Protein Domains , RNA, Long Noncoding , Staphylococcal Infections/immunology , Staphylococcus aureus
5.
J Biol Chem ; 291(23): 12254-70, 2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27129281

ABSTRACT

Purinergic homomeric P2X3 and heteromeric P2X2/3 receptors are ligand-gated cation channels activated by ATP. Both receptors are predominantly expressed in nociceptive sensory neurons, and an increase in extracellular ATP concentration under pathological conditions, such as tissue damage or visceral distension, induces channel opening, membrane depolarization, and initiation of pain signaling. Hence, these receptors are considered important therapeutic targets for pain management, and development of selective antagonists is currently progressing. To advance the search for novel analgesics, we have generated a panel of monoclonal antibodies directed against human P2X3 (hP2X3). We have found that these antibodies produce distinct functional effects, depending on the homomeric or heteromeric composition of the target, its kinetic state, and the duration of antibody exposure. The most potent antibody, 12D4, showed an estimated IC50 of 16 nm on hP2X3 after short term exposure (up to 18 min), binding to the inactivated state of the channel to inhibit activity. By contrast, with the same short term application, 12D4 potentiated the slow inactivating current mediated by the heteromeric hP2X2/3 channel. Extending the duration of exposure to ∼20 h resulted in a profound inhibition of both homomeric hP2X3 and heteromeric hP2X2/3 receptors, an effect mediated by efficient antibody-induced internalization of the channel from the plasma membrane. The therapeutic potential of mAb12D4 was assessed in the formalin, complete Freund's adjuvant, and visceral pain models. The efficacy of 12D4 in the visceral hypersensitivity model indicates that antibodies against P2X3 may have therapeutic potential in visceral pain indications.


Subject(s)
Antibodies, Monoclonal/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X2/immunology , Receptors, Purinergic P2X3/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Cell Line, Tumor , Cells, Cultured , Female , Freund's Adjuvant , HEK293 Cells , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/prevention & control , Ion Channels/chemistry , Ion Channels/metabolism , Ion Channels/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred BALB C , Microscopy, Confocal , Pain/chemically induced , Pain/metabolism , Pain/prevention & control , Protein Multimerization/immunology , Rats , Receptors, Purinergic P2X2/chemistry , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X3/chemistry , Receptors, Purinergic P2X3/metabolism , Trinitrobenzenesulfonic Acid , Visceral Pain/chemically induced , Visceral Pain/metabolism , Visceral Pain/prevention & control
6.
Protein Sci ; 25(2): 442-55, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26481561

ABSTRACT

Applications of microbial transglutaminase (mTGase) produced from Streptomyces mobarensis (S. mobarensis) were recently extended from food to pharmaceutical industry. To use mTGase for clinical applications, like generation of site specific antibody drug conjugates, it would be beneficial to manufacture mTGase in Escherichia coli (E. coli). To date, attempts to express recombinant soluble and active S. mobarensis mTGase have been largely unsuccessful. mTGase from S. mobarensis is naturally expressed as proenzyme and stepwise proteolytically processed into its active mature form outside of the bacterial cell. The pro-domain is essential for correct folding of mTGase as well as for inhibiting activity of mTGase inside the cell. Here, we report a genetically modified mTGase that has full activity and can be expressed at high yields in the cytoplasm of E. coli. To achieve this we performed an alanine-scan of the mTGase pro-domain and identified mutants that maintain its chaperone function but destabilize the cleaved pro-domain/mTGase interaction in a temperature dependent fashion. This allows proper folding of mTGase and keeps the enzyme inactive during expression at 20°C, but results in full activity when shifted to 37°C due to loosen domain interactions. The insertion of the 3C protease cleavage site together with pro-domain alanine mutants Tyr14, Ile24, or Asn25 facilitate high yields (30-75 mg/L), and produced an enzyme with activity identical to wild type mTGase from S. mobarensis. Site-specific antibody drug conjugates made with the E .coli produced mTGase demonstrated identical potency in an in vitro cell assay to those made with mTGase from S. mobarensis.


Subject(s)
Escherichia coli/genetics , Immunoconjugates/metabolism , Protein Engineering , Streptomyces/enzymology , Transglutaminases/genetics , Transglutaminases/metabolism , Amino Acid Sequence , Base Sequence , Enzyme Precursors/chemistry , Enzyme Precursors/genetics , Enzyme Precursors/isolation & purification , Enzyme Precursors/metabolism , Gene Expression , Industrial Microbiology , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutation , Plasmids/genetics , Protein Structure, Tertiary , Solubility , Streptomyces/chemistry , Streptomyces/genetics , Transglutaminases/chemistry , Transglutaminases/isolation & purification
7.
PLoS One ; 10(7): e0132282, 2015.
Article in English | MEDLINE | ID: mdl-26161543

ABSTRACT

The efficacy of an antibody-drug conjugate (ADC) is dependent on the properties of its linker-payload which must remain stable while in systemic circulation but undergo efficient processing upon internalization into target cells. Here, we examine the stability of a non-cleavable Amino-PEG6-based linker bearing the monomethyl auristatin D (MMAD) payload site-specifically conjugated at multiple positions on an antibody. Enzymatic conjugation with transglutaminase allows us to create a stable amide linkage that remains intact across all tested conjugation sites on the antibody, and provides us with an opportunity to examine the stability of the auristatin payload itself. We report a position-dependent degradation of the C terminus of MMAD in rodent plasma that has a detrimental effect on its potency. The MMAD cleavage can be eliminated by either modifying the C terminus of the toxin, or by selection of conjugation site. Both approaches result in improved stability and potency in vitro and in vivo. Furthermore, we show that the MMAD metabolism in mouse plasma is likely mediated by a serine-based hydrolase, appears much less pronounced in rat, and was not detected in cynomolgus monkey or human plasma. Clarifying these species differences and controlling toxin degradation to optimize ADC stability in rodents is essential to make the best ADC selection from preclinical models. The data presented here demonstrate that site selection and toxin susceptibility to mouse plasma degradation are important considerations in the design of non-cleavable ADCs, and further highlight the benefits of site-specific conjugation methods.


Subject(s)
Aminobenzoates/pharmacokinetics , Drug Carriers/pharmacokinetics , Oligopeptides/pharmacokinetics , Aminobenzoates/administration & dosage , Aminobenzoates/chemistry , Animals , Antibodies/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Stability , Female , HEK293 Cells , Humans , Macaca fascicularis , Mice, SCID , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Rats
8.
Bioconjug Chem ; 26(4): 650-9, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25643134

ABSTRACT

The systemic stability of the antibody-drug linker is crucial for delivery of an intact antibody-drug conjugate (ADC) to target-expressing tumors. Linkers stable in circulation but readily processed in the target cell are necessary for both safety and potency of the delivered conjugate. Here, we report a range of stabilities for an auristatin-based payload site-specifically attached through a cleavable valine-citrulline-p-aminobenzylcarbamate (VC-PABC) linker across various sites on an antibody. We demonstrate that the conjugation site plays an important role in determining VC-PABC linker stability in mouse plasma, and that the stability of the linker positively correlates with ADC cytotoxic potency both in vitro and in vivo. Furthermore, we show that the VC-PABC cleavage in mouse plasma is not mediated by Cathepsin B, the protease thought to be primarily responsible for linker processing in the lysosomal degradation pathway. Although the VC-PABC cleavage is not detected in primate plasma in vitro, linker stabilization in the mouse is an essential prerequisite for designing successful efficacy and safety studies in rodents during preclinical stages of ADC programs. The divergence of linker metabolism in mouse plasma and its intracellular cleavage offers an opportunity for linker optimization in the circulation without compromising its efficient payload release in the target cell.


Subject(s)
Aminobenzoates/chemistry , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Immunoconjugates/chemistry , Oligopeptides/chemistry , Pancreatic Neoplasms/drug therapy , Aminobenzoates/blood , Aminobenzoates/pharmacokinetics , Aminobenzoates/pharmacology , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Carbamates/chemistry , Cathepsin B/chemistry , Cathepsin B/metabolism , Cell Line, Tumor , Dipeptides/chemistry , Drug Delivery Systems/methods , Drug Stability , Female , Humans , Immunoconjugates/blood , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Mice , Mice, Nude , Models, Molecular , Oligopeptides/blood , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
J Mol Biol ; 427(6 Pt B): 1513-1534, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25284753

ABSTRACT

The ability of antibodies to bind an antigen with a high degree of affinity and specificity has led them to become the largest and fastest growing class of therapeutic proteins. Clearly identifying the epitope at which they bind their cognate antigen provides insight into their mechanism of action and helps differentiate antibodies that bind the same antigen. Here, we describe a method to precisely and efficiently map the epitopes of a panel of antibodies in parallel over the course of several weeks. This method relies on the combination of rational library design, quantitative yeast surface display and next-generation DNA sequencing and was demonstrated by mapping the epitopes of several antibodies that neutralize alpha toxin from Staphylococcus aureus. The accuracy of this method was confirmed by comparing the results to the co-crystal structure of one antibody and alpha toxin and was further refined by the inclusion of a lower-affinity variant of the antibody. In addition, this method produced quantitative insight into the epitope residues most critical for the antibody-antigen interaction and enabled the relative affinities of each antibody toward alpha toxin variants to be estimated. This affinity estimate serves as a predictor of neutralizing antibody potency and was used to anticipate the ability of each antibody to effectively bind and neutralize naturally occurring alpha toxin variants secreted by strains of S. aureus, including clinically relevant strains. Ultimately this type information can be used to help select the best clinical candidate among a set of antibodies against a given antigen.


Subject(s)
Antibodies, Monoclonal/immunology , Bacterial Toxins/immunology , Epitopes/analysis , Hemolysin Proteins/immunology , High-Throughput Nucleotide Sequencing , Peptide Library , Saccharomyces cerevisiae/immunology , Staphylococcal Infections/prevention & control , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Bacterial Toxins/genetics , Epitope Mapping/methods , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Flow Cytometry , Hemolysin Proteins/genetics , Humans , Molecular Sequence Data , Protein Conformation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology
10.
PLoS One ; 8(1): e54886, 2013.
Article in English | MEDLINE | ID: mdl-23365682

ABSTRACT

Assays for bacterial ribosomal RNA precursors (pre-rRNA) have been shown to distinguish viable from inactivated bacterial cells in drinking water samples. Because the synthesis of pre-rRNA is rapidly induced by nutritional stimulation, viable bacteria can be distinguished from inactivated cells and free nucleic acids by measuring the production of species-specific pre-rRNA in samples that have been briefly stimulated with nutrients. Here, pre-rRNA analysis was applied to bacteria from serum, a human sample matrix. In contrast to drinking water, serum is rich in nutrients that might be expected to mask the effects of nutritional stimulation. Reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) assays were used to detect pre-rRNA of four bacterial species: Acinetobacter baumannii, Pseudomonas aeruginosa, Staphylococcus aureus, and the Mycobacterium tuberculosis complex. These species were chosen for their clinical significance and phylogenetic diversity (Proteobacteria, Firmicutes, and Actinobacteria). To maximize resolving power, pre-rRNA was normalized to genomic DNA of each pathogen. When viable cells were shifted from serum to bacteriological culture medium, rapid replenishment of pre-rRNA was always observed. Cells of P. aeruginosa that were inactivated in the presence of serum exhibited no pre-rRNA response to nutritional stimulation, despite strong genomic DNA signals in these samples. When semi-automated methods were used, pre-rRNA analysis detected viable A. baumannii cells in serum at densities of ≤100 CFU/mL in <5.5 hours. Originally developed for rapid microbiological analysis of drinking water, ratiometric pre-rRNA analysis can also assess the viability of bacterial cells derived from human specimens, without requiring bacteriological culture.


Subject(s)
DNA, Bacterial/blood , Microbial Viability , RNA Precursors/blood , RNA, Bacterial/blood , Serum/microbiology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/isolation & purification , Colony Count, Microbial , Culture Media , Humans , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/isolation & purification , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification
11.
Infect Immun ; 80(6): 2221-30, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22451517

ABSTRACT

Listeriolysin O (LLO) is a pore-forming toxin of the cholesterol-dependent cytolysin (CDC) family and a primary virulence factor of the intracellular pathogen Listeria monocytogenes. LLO mediates rupture of phagosomal membranes, thereby releasing bacteria into the growth-permissive host cell cytosol. Several unique features of LLO allow its activity to be precisely regulated in order to facilitate phagosomal escape, intracellular growth, and cell-to-cell spread. To improve our understanding of the multifaceted contribution of LLO to the pathogenesis of L. monocytogenes, we developed a screen that combined saturation mutagenesis and signature tags, termed in vivo analysis by saturation mutagenesis and signature tags (IVASS). We generated a library of LLO mutant strains, each harboring a single amino acid substitution and a signature tag, by using the previously described pPL2 integration vector. The signature tags acted as molecular barcodes, enabling high-throughput, parallel analysis of 40 mutants in a single animal and identification of attenuated mutants by negative selection. Using the IVASS technique we were able to screen over 90% of the 505 amino acids present in LLO and identified 60 attenuated mutants. Of these, 39 LLO residues were previously uncharacterized and potentially revealed novel functions of the toxin during infection. The mutants that were subsequently analyzed in vivo each conferred a 2- to 4-orders of magnitude loss in virulence compared to wild type, thereby validating the screening methods. Phenotypic analysis of the LLO mutant library using common in vitro techniques suggested that the functional contributions of some residues could only have been revealed through in vivo analysis.


Subject(s)
Bacterial Toxins/metabolism , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Listeria monocytogenes/pathogenicity , Alanine , Amino Acid Sequence , Animals , Bacterial Toxins/genetics , DNA, Bacterial , Female , Gene Expression Regulation, Bacterial/physiology , Heat-Shock Proteins/genetics , Hemolysin Proteins/genetics , Listeria monocytogenes/genetics , Mice , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Phenotype , Polymerase Chain Reaction , Protein Conformation , Protein Structure, Tertiary
12.
Infect Immun ; 80(2): 720-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22083714

ABSTRACT

Listeria monocytogenes causes a serious food-borne disease due to its ability to spread from the intestine to other organs, a process that is poorly understood. In this study we used 20 signature-tagged wild-type clones of L. monocytogenes in guinea pigs in combination with extensive quantitative data analysis to gain insight into extraintestinal dissemination. We show that L. monocytogenes colonized the liver in all asymptomatic animals. Spread to the liver occurred as early as 4 h after ingestion via a direct pathway from the intestine to the liver. This direct pathway contributed significantly to the bacterial load in the liver and was followed by a second wave of dissemination via the mesenteric lymph nodes (indirect pathway). Furthermore, bacteria were eliminated in the liver, whereas small intestinal villi provided a niche for bacterial replication, indicating organ-specific differences in net bacterial growth. Bacteria were shed back from intestinal villi into the small intestinal lumen and reinfected the Peyer's patches. Together, these results support a novel dissemination model where L. monocytogenes replicates in intestinal villi, is shed into the lumen, and reinfects intestinal immune cells that traffic to liver and mesenteric lymph nodes, a process that occurs even during asymptomatic colonization.


Subject(s)
Intestines/microbiology , Listeria monocytogenes , Listeriosis/microbiology , Liver/microbiology , Lymph Nodes/microbiology , Animals , Bacterial Load , Female , Guinea Pigs , Listeriosis/pathology , Spleen/microbiology , Time Factors
13.
Infect Immun ; 77(3): 943-51, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19139192

ABSTRACT

Clostridium septicum alpha-toxin is a beta-barrel pore-forming cytolysin that is functionally similar to aerolysin. Residues important in receptor binding, oligomerization, and pore formation have been identified; however, little is known about the activity of the toxin in an infection, although it is essential for disease. We have now shown that deletion of a small portion of the transmembrane domain, so that the toxin is no longer able to form pores, completely abrogates its ability to contribute to disease, as does replacement of the sole cysteine residue with leucine. However, although previous biochemical and cytotoxicity assays clearly indicated that mutations in residues important in oligomerization, binding, and prepore conversion greatly reduced activity or rendered the toxin inactive, once the mutated toxins were overexpressed by the natural host in the context of an infection it was found they were able to cause disease in a mouse model of myonecrosis. These results highlight the importance of testing the activity of virulence determinants in the normal host background and in an infectious disease context and provide unequivocal evidence that it is the ability of alpha-toxin to form a pore that confers its toxicity in vivo.


Subject(s)
Bacterial Toxins/metabolism , Clostridium Infections/pathology , Clostridium septicum/pathogenicity , Muscle, Skeletal/microbiology , Muscle, Skeletal/pathology , Pore Forming Cytotoxic Proteins/metabolism , Animals , Bacterial Toxins/genetics , Blotting, Western , Clostridium Infections/genetics , Clostridium Infections/metabolism , Mice , Necrosis , Pore Forming Cytotoxic Proteins/genetics
14.
Infect Immun ; 76(8): 3439-50, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18474644

ABSTRACT

While recombinant Listeria monocytogenes strains can be explored as vaccine candidates, it is important to develop attenuated but highly immunogenic L. monocytogenes vaccine vectors. Here, prfA* mutations selected on the basis of upregulated expression of L. monocytogenes PrfA-dependent genes and proteins were assessed to determine their abilities to augment expression of foreign immunogens in recombinant L. monocytogenes vectors and therefore enhance vaccine-elicited immune responses (a prfA* mutation is a mutation that results in constitutive overexpression of PrfA and PrfA-dependent virulence genes; the asterisk distinguishes the mutation from inactivation or stop mutations). A total of 63 recombinant L. monocytogenes vaccine vectors expressing seven individual viral or bacterial immunogens each in nine different L. monocytogenes strains carrying wild-type prfA or having prfA* mutations were constructed and investigated. Mutations selected on the basis of increased PrfA activation in recombinant L. monocytogenes prfA* vaccine vectors augmented expression of seven individual protein immunogens remarkably. Consistently, prime and boost vaccination studies with mice indicated that the prfA(G155S) mutation in recombinant L. monocytogenes DeltaactA prfA* strains enhanced vaccine-elicited cellular immune responses. Surprisingly, the prfA(G155S) mutation was found to enhance vaccine-elicited humoral immune responses as well. The highly immunogenic recombinant L. monocytogenes DeltaactA prfA* vaccine strains were as attenuated as the recombinant parent L. monocytogenes DeltaactA vaccine vector. Thus, recombinant attenuated L. monocytogenes DeltaactA prfA* vaccine vectors potentially are better antimicrobial and anticancer vaccines.


Subject(s)
Bacterial Vaccines/immunology , Listeria monocytogenes/genetics , Mutation , Peptide Termination Factors/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Viral/blood , Bacterial Vaccines/genetics , Enzyme-Linked Immunosorbent Assay , Female , Immunization, Secondary , Interferon-gamma/biosynthesis , Listeria monocytogenes/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
15.
Biochemistry ; 45(48): 14347-54, 2006 Dec 05.
Article in English | MEDLINE | ID: mdl-17128973

ABSTRACT

Alpha toxin (AT) is the major virulence factor of Clostridium septicum that is a proteolytically activated pore-forming toxin that belongs to the aerolysin-like family of toxins. AT is predicted to be a three-domain molecule on the basis of its functional and sequence similarity with aerolysin, for which the crystal structure has been determined. In this study, we have substituted the entire primary structure of AT with alanine or cysteine to identify those amino acids that comprise functional domains involved in receptor binding, oligomerization, and pore formation. These studies revealed that receptor binding is restricted to domain 1 of the AT structure, whereas domains 1 and 3 are involved in oligomerization. These studies also revealed the presence of a putative functional region of AT proximal to the receptor-binding domain but distal from the pore-forming domain that is proposed to regulate the insertion of the transmembrane beta-hairpin of the prepore oligomer.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Clostridium septicum/chemistry , Clostridium septicum/metabolism , Amino Acid Sequence , Bacterial Toxins/genetics , Binding Sites , Clostridium septicum/genetics , Conserved Sequence , Crystallography, X-Ray , Cysteine/genetics , Cysteine/metabolism , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Protein Structure, Tertiary , Sequence Alignment , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...