Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters










Publication year range
1.
Protein Sci ; 5(8): 1737-41, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8844860

ABSTRACT

Toxic shock syndrome toxin-1 (TSST-1) is one of a family of staphylococcal exotoxins recognized as microbial superantigens. The toxin plays a dominant role in the genesis of toxic shock in humans through a massive activation of the immune system. This potentially lethal illness occurs as a result of the interaction of TSST-1 with a significant proportion of the T-cell repertoire. TSST-1, like other superantigens, can bind directly to class II major histocompatibility (MHC class II) molecules prior to its interaction with entire families of V beta chains of the T-cell receptor (TCR). The three-dimensional structure of a mutant (His-135-Ala) TSST-1 was compared with the structure of the native (wild-type) TSST-1 at 2.5 A resolution. The replacement of His 135 of TSST-1 with an Ala residue results in the loss of T-cell mitogenicity and toxicity in experimental animals. This residue, postulated to be directly involved in the toxin-TCR interactions, is located on the major helix alpha 2, which forms the backbone of the molecule and is exposed to the solvent. In the molecular structure of the mutant toxin, the helix alpha 2 remains unaltered, but the His to Ala modification causes perturbations on the neighboring helix alpha 1 by disrupting helix-helix interactions. Thus, the effects on TCR binding of the His 135 residue could actually be mediated, wholly or in part, by the alpha 1 helix.


Subject(s)
Bacterial Toxins , Enterotoxins/chemistry , Protein Structure, Tertiary , Superantigens/chemistry , Crystallization , Crystallography, X-Ray , Enterotoxins/genetics , Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Staphylococcus aureus/chemistry , Staphylococcus aureus/genetics , Superantigens/genetics
3.
Infect Immun ; 63(6): 2141-6, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7768593

ABSTRACT

Toxic shock syndrome toxin 1 (TSST-1), a superantigen produced by Staphylococcus aureus, is a causative agent of toxic shock syndrome (TSS). This superantigen is a potent stimulator of T cells and macrophages/monocytes, resulting in the release of cytokines that are implicated in the pathogenesis of TSS. This study characterizes a mutant TSST-1, derived by site-directed mutagenesis, that has an alanine substitution at histidine 135 (mutant 135). This single-amino-acid change results in a mutant toxin that has lost mitogenic activity for T cells. In contrast to wild-type TSST-1, this mutant does not induce T cells to express interleukin-2, gamma interferon, or tumor necrosis factor beta (TNF-beta). The inability of mutant 135 to activate T cells is not due to a lack of binding to the class II major histocompatibility complex receptor. In addition, the mutant TSST-1 does not induce expression of TNF-alpha, which plays a role in the development of lethal shock. The lack of TNF-alpha induction by mutant 135 is likely due to its inability to activate T cells. These data suggest that the mutation at histidine 135 in TSST-1 affects toxin interactions with the T-cell receptor rather than the class II major histocompatibility complex receptor.


Subject(s)
Bacterial Toxins , Enterotoxins/physiology , Lymphocyte Activation , Staphylococcus aureus/pathogenicity , Superantigens/physiology , T-Lymphocytes/immunology , Animals , Base Sequence , Enterotoxins/chemistry , Histocompatibility Antigens Class II/metabolism , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/biosynthesis
4.
Infect Immun ; 63(4): 1229-34, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7890377

ABSTRACT

A recombinant of toxic shock syndrome toxin 1 (TSST-1) which contains a single histidine-to-alanine mutation at residue 135 (H135A) was analyzed for toxicity and vaccine potential in a lipopolysaccharide (LPS)-potentiated mouse lethality model. The 50% lethal dose (LD50) of TSST-1 in BALB/c mice was 47.2 micrograms/kg, but H135A was not lethal when tested at a dose equivalent to 10 LD50s of TSST-1. Levels of tumor necrosis factor (TNF) and gamma interferon (IFN-gamma) in serum were, respectively, 10- and 50-fold higher in LPS-potentiated mice injected with 15 LD50s of TSST-1 than in mice given H135A. Mice injected with only TSST-1 did not have elevated levels of TNF or IFN-gamma in serum, while H135A plus LPS or LPS alone elicited identical, yet very low, levels of TNF and IFN-gamma. An enzyme-linked immunosorbent assay of H135A and TSST-1 with anti-TSST-1 serum yielded very similar dose-response curves, which strongly suggests that H135A serologically and conformationally resembles the native toxin. Mice immunized with H135A developed antibodies that recognized TSST-1 in an enzyme-linked immunosorbent assay and afforded protection against a 15-LD50 challenge of TSST-1 plus LPS. The pooled sera of mice immunized with either TSST-1 or H135A also prevented lymphocyte proliferation due to TSST-1.


Subject(s)
Bacterial Toxins , Enterotoxins/toxicity , Lipopolysaccharides/pharmacology , Superantigens , Animals , Bacterial Vaccines/immunology , Enterotoxins/immunology , Immunization , Interferon-gamma/blood , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
5.
Infect Immun ; 63(3): 1095-101, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7532624

ABSTRACT

Staphylococcus aureus toxic shock syndrome toxin 1 (TSST-1) is involved in the pathogenesis of toxic shock syndrome and perhaps other staphylococcal diseases. Recently, the C-terminal part of the TSST-1 toxin has been shown to be responsible for mitogenic activity in animal models. We studied the role of the C-terminal structural unit of TSST-1 with regard to proliferation, cytokine release (tumor necrosis factor alpha [TNF-alpha], interleukin-6 [IL-6], and IL-8), mRNA expression for IL-6, IL-8, IL-10, TNF-alpha, and CD40 ligand (CD40L), synthesis of immunoglobulin E (IgE), IgA, IgG, and IgM, CD23 expression, and soluble CD23 (sCD23) release from human peripheral blood mononuclear cells (PBMC). For this purpose, we used the recombinant wild-type TSST-1 (p17) mutant toxin Y115A (tyrosine residue modified to alanine) and toxin H135A (histidine residue modified to alanine). Unmodified toxin p17 and mutant toxin Y115A, at a concentration below 5 ng, to a lesser degree, induced a strong proliferation. Toxin p17 followed by toxin Y115A was the most pronounced inducer for mRNA expression for IL-10 and CD40L and cytokine generation (mRNA and protein) for TNF-alpha, IL-6, and IL-8. Mutant protein H135A failed to activate human PBMC. Both toxins p17 and, to a lesser degree, Y115A significantly suppressed IL-4- and anti-CD40-induced synthesis of all four Igs as well as IL-4-induced CD23 expression and sCD23 release. Mutant toxin H135A failed to do so. Thus, our data show that a region in the C terminus of TSST-1 is responsible not only for mitogenic activity but also for additional immunomodulating biological activities of TSST-1. More specifically, histidine residue H135A of the 194-amino-acid toxin appears to be critical for the expression of biological activities in a human in vitro model.


Subject(s)
Bacterial Toxins , Enterotoxins/immunology , Leukocytes, Mononuclear/immunology , Staphylococcus aureus/immunology , Superantigens/immunology , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Base Sequence , CD40 Antigens , Cytokines/biosynthesis , Cytokines/metabolism , Humans , Immunoglobulin Isotypes/biosynthesis , Interleukin-10/genetics , Lymphocyte Activation , Molecular Sequence Data , RNA, Messenger/analysis , Receptors, IgE/biosynthesis , Recombinant Proteins/immunology , Structure-Activity Relationship
6.
Infect Immun ; 63(2): 509-15, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7822015

ABSTRACT

Structure-function studies have revealed that the region between amino acids 115 and 141 of toxic shock syndrome toxin 1 (TSST-1) constitutes a biologically active domain. A critical residue appears to be histidine 135, since a site-directed mutation that alters the histidine to alanine (H135A) results in a loss of mitogenic activity and an absence of toxicity as measured in a rabbit infection model of toxic shock syndrome. We have characterized the mutant toxin further and report here on its immunogenic activity in rabbits and on the protective ability of mutant-specific antibodies in two animal models of toxin-mediated shock. Antibodies raised in rabbits by immunization with the purified H135A are fully cross-reactive with staphylococcal TSST-1 and wild-type recombinant TSST-1 (rTSST-1) expressed in Escherichia coli. The H135A antibodies neutralized the mitogenic activity for murine splenic T cells equally well as did TSST-1-specific polyclonal and monoclonal antibodies. In addition, the H135A antibodies blocked the production of tumor necrosis factor by spleen cells stimulated with rTSST-1. The toxicities of rTSST-1 and H135A were compared in D-galactosamine (D-GalNH2)-sensitized MRL-lpr/lpr mice. The nontoxicity of H135A was confirmed in this murine model of superantigen-induced septic shock. No toxicity of H135A was demonstrable at doses of 60 micrograms, while doses of rTSST-1 as low as 2 micrograms caused significant mortality within 24 to 72 h after challenge. Furthermore, subsequent to challenge of mice with H135A, no elevation in the serum levels of interleukin-2 or tumor necrosis factor was measurable. Passive immunization with H135A antibodies also protected MRL-lpr/lpr mice against lethal challenge with rTSST-1. Finally, rabbits actively immunized with purified H135A did not succumb to infection with a transformed strain of Staphylococcus aureus expressing rTSST-1. Additional animal studies will be required to confirm the immunizing potential of H135A and the efficacy of H135A antibodies as a neutralizing antitoxin.


Subject(s)
Bacterial Toxins , Enterotoxins/immunology , Shock, Septic/prevention & control , Superantigens , Animals , Antibodies, Bacterial/immunology , Cross Reactions , Enterotoxins/chemistry , Enterotoxins/toxicity , Female , Histidine , Lymphocyte Activation/drug effects , Male , Mice , Mice, Mutant Strains , Mitogens , Mutagenesis, Site-Directed , Rabbits , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/biosynthesis
7.
Pathobiology ; 63(6): 293-304, 1995.
Article in English | MEDLINE | ID: mdl-8738468

ABSTRACT

Recombinant toxic shock syndrome toxin-1 (rTSST-1) administered to MRL-lpr/lpr TCR V beta 8.2 transgenic mice at doses of 0.1 microgram/mouse resulted in 100% mortality. This was an unexpected finding since TSST-1 does not activate V beta 8.2 T cells. In contrast, control mice heterozygous at the lpr locus and also for the transgene (MRL-lpr/+; V beta 8.2/0) survived doses of superantigen 100 times higher. The transgenic mice which succumbed to rTSST-1 challenge exhibited histopathology of the liver consistent with toxic shock (generalized inflammation and hepatocellular necrosis) as well as substantially elevated serum TNF-alpha, IL-2, and IL-6 cytokine levels. Splenic T cells derived from transgenic mice stimulated with rTSST-1 in vitro did not undergo detectable proliferation as measured in a standard mitogen assay. However, PCR amplification of cDNA prepared from the V beta 8.2 splenocytes revealed the presence of minor populations of TSST-1-reactive V beta elements (i.e. V beta 3 and V beta 15). Furthermore, an expansion of the V beta 3 and V beta 15 T-cell families was detected by PCR assay of spleen cell cultures stimulated with rTSST-1. These results suggested that the exquisite sensitivity of the MRL-lpr/lpr V beta 8.2 transgenic animals to rTSST-1 was not dependent exclusively on T-cell proliferation but was augmented by the influence of a defective fas antigen receptor expressed in homozygous lpr mice. To test this hypothesis more directly, we compared the sensitivity of MRL-lpr/lpr mice (not carrying the V beta 8.2 transgene) to MRL-+/+ mice. The MRL-lpr/lpr fas antigen-defective mice were substantially more susceptible to rTSST-1 challenge. Mice carrying the lpr mutation on another genetic background (C57BL/6.C3H-lpr/lpr) were also more sensitive to rTSST-1 challenge than were C57BL/6.C3H-+/+ mice. Although induction of toxic shock is clearly associated with T-cell proliferation, defects in fas antigen receptor or ligand may also contribute substantively to superantigen-mediated lethal shock by still undefined mechanisms.


Subject(s)
Adenine/analogs & derivatives , Bacterial Toxins , Enterotoxins/pharmacology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Shock, Septic/etiology , Superantigens , fas Receptor/genetics , Adenine/pharmacology , Animals , Base Sequence , Cells, Cultured , Cyclosporine/pharmacology , Female , Heterozygote , Interleukin-2/metabolism , Interleukin-6/metabolism , Isoquinolines/pharmacology , Lipopolysaccharides/pharmacology , Liver/pathology , Lymphocyte Activation , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Nitrogen Oxides/pharmacology , Recombinant Proteins/pharmacology , Shock, Septic/pathology , Spleen/cytology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
8.
Infect Immun ; 61(3): 793-9, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8432600

ABSTRACT

Menstrually associated toxic shock syndrome (TSS) is attributed primarily to the effects of staphylococcal exotoxin toxic shock syndrome toxin 1 (TSST-1). A region of the 194-amino-acid toxin spanning residues 115 through 144 constitutes a biologically active site. Several point mutations in the TSST-1 gene in that region result in gene products with reduced mitogenic activity for murine T cells. In this study we evaluated the toxicity of recombinant TSST-1 and several mutants of TSST-1 made by transformed Staphylococcus aureus during in vivo growth in a rabbit infection model of TSS. The toxicities of the transformed strains of S. aureus for rabbits correlated with the mitogenic activities of the recombinant toxins. An isolate originally obtained from a patient with a confirmed case of TSS (S. aureus 587) implanted in a subcutaneous chamber served as a positive control. TSST-1 produced in vivo led to lethal shock within 48 h, and a TSST-1-neutralizing antibody (monoclonal antibody 8-5-7) administered to rabbits challenged with S. aureus 587 prevented fatal illness. Rabbits infected with transformed S. aureus RN4220 expressing wild-type toxin (p17) or mutant toxins retaining mitogenic activity for T cells succumbed within a similar time frame. Blood chemistries of samples obtained from infected animals before death indicated abnormalities in renal and hepatic functions similar to those induced by parenteral injection of purified staphylococcal TSST-1. Mutant toxin 135 (histidine modified to alanine at residue 135) possessed only 5 to 10% of the mitogenic activity of wild-type toxin. Rabbits challenged with transformed S. aureus RN4220 expressing mutant toxin 135 exhibited only mild transient illness. Mutant toxin 135 retained reactivity with monoclonal antibody 8-5-7 and by several criteria was conformationally intact. Toxin from a double mutant, 141.144, with alanine substitutions at residues 141 (histidine) and 144 (tyrosine), also was devoid of mitogenic activity. In this case, antibody recognition was lost. Mutant toxins 115 and 141 were found to possess approximately half-maximal mitogenic activity. Rabbits challenged with S. aureus RN4220 expressing either 115 or 141 toxin succumbed to lethal shock. We conclude that the ability of TSST-1 to activate murine T cells in vitro and its expression of toxicity leading to lethal shock in rabbits are related phenomena.


Subject(s)
Bacterial Toxins , Enterotoxins/toxicity , Shock, Septic/physiopathology , Staphylococcus aureus/pathogenicity , Superantigens , Animals , Antibodies, Monoclonal , Blood Chemical Analysis , Enterotoxins/immunology , Mitogens , Rabbits , Recombinant Proteins/toxicity , Structure-Activity Relationship
9.
J Infect Dis ; 163(3): 524-9, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1995725

ABSTRACT

A 21-mer synthetic peptide (KGEKVDLNTKRTKKSQHTSEG), designated TSST-1(58-78), was constructed from the primary structure of the toxic shock syndrome toxin 1 (TSST-1). The peptide reacted with a panel of neutralizing monoclonal antibodies (MAbs) to whole TSST-1 in solid-phase immunoassays. TSST-1(58-78) promoted the in vitro proliferation of human peripheral blood mononuclear cells (PBMC) in a dose-dependent manner. Minimum dose required for stimulation (P less than or equal to .05 microM) was 0.75 microM peptide. This mitogenic effect was abrogated by incubation of the peptide with MAbs to whole TSST-1 before addition to PBMC. The ability of TSST-1(58-78) to stimulate the proliferation of highly purified resting human T cells was analyzed. Significant proliferation (P less than or equal to .01) was observed only in the presence of increasing populations of monocytes added to the cultures. Adherent human monocytes exposed to TSST-1(58-78) released tumor necrosis factor. Thus, some of the immunoregulatory properties attributed to TSST-1 are demonstrated by the region of the toxin represented by the peptide TSST-1(58-78).


Subject(s)
Bacterial Toxins , Enterotoxins/pharmacology , Peptide Fragments/pharmacology , Staphylococcus aureus , Superantigens , T-Lymphocytes/drug effects , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antigen-Presenting Cells/physiology , Cell Division/drug effects , Enterotoxins/immunology , Humans , In Vitro Techniques , Mitogens , Molecular Sequence Data , Peptide Fragments/immunology , T-Lymphocytes/physiology , Tumor Necrosis Factor-alpha/biosynthesis
10.
Infect Immun ; 58(9): 3151-3, 1990 Sep.
Article in English | MEDLINE | ID: mdl-2387638

ABSTRACT

Cyclosporin A prevents visceralization of Leishmania major infection of BALB/c mice (N. C. Behforouz, C. D. Wenger, and B. A. Mathison, J. Immunol. 136:3067-3075, 1986; W. Solbach, K. Forberg, E. Kammerer, C. Bogdan, and M. Rollinghoff, J. Immunol. 134:702-707, 1986). We report that cyclosporin A exacerbates disseminated leishmaniasis caused by L. donovani in C57BL/10 mice. Normal mice challenged with 5 x 10(6) amastigotes intravenously cleared the infection within several months by spontaneous acquisition of cell-mediated immunity. In contrast, cyclosporin A administered daily intraperitoneally at a dose of 1.25 mg per mouse prevented development of curative immunity and converted C57BL/10 (curing) mice to a noncuring phenotype. A rationale for the contrasting effects of cyclosporin A in the two murine models of leishmaniasis is provided.


Subject(s)
Cyclosporins/therapeutic use , Leishmaniasis, Visceral/drug therapy , Animals , Female , Immunity, Cellular , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
11.
Infect Immun ; 58(9): 3020-8, 1990 Sep.
Article in English | MEDLINE | ID: mdl-1696937

ABSTRACT

Toxic shock syndrome toxin 1 (TSST-1), a 22-kilodalton protein made by strains of Staphylococcus aureus harboring the chromosomal toxin gene, may elicit toxic shock syndrome in humans. In vitro, TSST-1 induces T cells to proliferate and macrophages to secrete interleukin-1. To conduct a structure-function analysis, point mutations on the TSST-1 gene were generated by site-directed mutagenesis to identify amino acids critical for activity of the toxin. Specific tyrosine and histidine residues were replaced by alanines. Wild-type and mutant TSST-1 gene constructs were expressed in Escherichia coli, and the products were tested for their mitogenic potential and reactivity with a TSST-1 neutralizing monoclonal antibody (MAb 8-5-7). Four of the mutants were similar to the wild type; i.e., the mutant toxins stimulated murine T cells and reacted with MAb 8-5-7 equally as well as the wild type. Two mutants exhibited a decrease in mitogenic activity, but one of these retained the capacity to bind with MAb 8-5-7 while the other was no longer recognized by the same antibody. One double mutant demonstrated minimal mitogenic activity and did not react in enzyme-linked immunosorbent and immunoblot assays with MAb 8-5-7. The data show that specific residues near the carboxy terminus of TSST-1 are essential for mitogenic activity and in forming the epitope recognized by neutralizing MAb 8-5-7.


Subject(s)
Antibodies, Monoclonal/immunology , Bacterial Toxins , Enterotoxins/genetics , Mitogens/immunology , Mutation , Superantigens , Animals , Base Sequence , Binding, Competitive , Blotting, Western , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Enterotoxins/immunology , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Escherichia coli/genetics , Mice , Mitogens/genetics , Molecular Sequence Data , Neutralization Tests , Spleen/cytology , Spleen/immunology
13.
Rev Infect Dis ; 11 Suppl 1: S289-93, 1989.
Article in English | MEDLINE | ID: mdl-2784586

ABSTRACT

The purpose of this study was to determine whether endotoxin could augment toxic shock syndrome toxin 1 (TSST-1)-induced production of interleukin 1 (IL-1) by murine macrophages. Macrophages from C3H/HeJ or C57Bl/6 mice were stimulated with purified TSST-1 alone or in combination with lipopolysaccharide (LPS). A dramatic synergistic thymocyte-proliferative response was induced by supernatants from C57Bl/6 macrophages stimulated with both TSST-1 and LPS. No enhanced response was induced by supernatants from C3H/HeJ macrophages. A portion of the enhanced response induced by C57Bl/6 macrophage supernatants was attributed to synergism between IL-1 and residual TSST-1 in the thymocyte assay. The addition of monoclonal antibody to TSST-1 to the supernatants eliminated the effects of residual TSST-1 in the thymocyte assay and demonstrated a synergistic induction of IL-1. These data (1) show that LPS can enhance macrophage responsiveness to TSST-1; (2) suggest that TSST-1 not only induces IL-1 secretion but also enhances target cell responsiveness to IL-1; and (3) further support the role of IL-1 in the pathogenesis of toxic shock syndrome.


Subject(s)
Bacterial Toxins , Endotoxins/pharmacology , Enterotoxins/pharmacology , Interleukin-1/biosynthesis , Macrophages/immunology , Superantigens , Animals , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Staphylococcus aureus , T-Lymphocytes/immunology
14.
Rev Infect Dis ; 11 Suppl 1: S214-7; discussion S217-8, 1989.
Article in English | MEDLINE | ID: mdl-2928638

ABSTRACT

The effectiveness of passive immunization was assessed in an infection model of toxic shock syndrome (TSS) in which monoclonal antibody to TSS toxin 1 (TSST-1) was administered intravenously to rabbits. Previously implanted infection chambers were inoculated with Staphylococcus aureus strains RN4710 and D4508. The former strain carries the TSST-1 gene on plasmid pRN6201; the latter is a TSST-1-negative clinical isolate obtained from a patient with nonmenstrual TSS. Purified monoclonal antibody, MAb 8-5-7 (IgG), was administered in two doses of approximately 1.25 mg each 24 hours before and 24 hours after infection. MAb 8-5-7 provided complete protection against both the TSS-like syndrome and the mortality that occurred in unprotected rabbits infected with strain RN4710 but did not provide complete protection in rabbits infected with strain D4508; three of the five rabbits either displayed signs of illness or died despite treatment. Western-blot analyses of the extracellular proteins produced by strains RN4710 and D4508 that used MAb 8-5-7 as a probe revealed that only TSST-1 produced by RN4710 reacted with the antibody. Thus, if MAb 8-5-7 partially protected animals against infections with strain D4508, the protection appears to have been nonspecific.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bacterial Toxins , Enterotoxins/immunology , Immunization, Passive , Shock, Septic/prevention & control , Staphylococcus aureus/immunology , Superantigens , Animals , Blotting, Western , Male , Plasmids , Rabbits , Staphylococcus aureus/genetics
16.
Rev Infect Dis ; 11 Suppl 1: S90-5, 1989.
Article in English | MEDLINE | ID: mdl-2928656

ABSTRACT

Toxic shock syndrome toxin 1 (TSST-1), plays a significant role in the pathogenesis of TSS. TSST-1 production is subject to physiologic and environmental constraints. Thus, DNA probes that detect the chromosomal gene encoding the toxin are of value diagnostically, epidemiologically, and for studies of gene expression. Several synthetic oligonucleotide probes complementary to two regions of the TSST-1 gene were used to ascertain the presence of this gene in the chromosomal DNA of 261 strains of S. aureus from various TSS-related and non-TSS-related sources. Isolates were from clinically confirmed menstrual and nonmenstrual cases of TSS and from healthy vaginal carriers of S. aureus. Other strains tested included clinical non-TSS isolates and food poisoning-associated staphylococcal isolates. Detection of the TSST-1 gene by the labeled gene probes correlated in all but two cases with production of TSST-1. Ten Centers for Disease Control (CDC) strains that were isolated from TSS patients and did not produce TSST-1 were also examined, as were several strains of Staphylococcus epidermidis isolated from patients with suspected TSS. Neither group of strains possessed the TSST-1 gene. Finally, a 7-kilobase DNA restriction fragment of S. aureus containing the entire TSST-1 gene was transformed into Escherichia coli strains HB101 and DH5 alpha via a plasmid vector.


Subject(s)
Bacterial Toxins , Enterotoxins/genetics , Gene Expression Regulation , Shock, Septic/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Superantigens , Base Sequence , Blotting, Southern , Carrier State/microbiology , DNA Probes , DNA, Bacterial/genetics , Enterotoxins/biosynthesis , Female , Humans , Immunoblotting , Molecular Sequence Data , Nucleic Acid Hybridization , Staphylococcal Food Poisoning/microbiology , Staphylococcus aureus/metabolism , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism , Vagina/microbiology
17.
Antimicrob Agents Chemother ; 32(7): 1012-8, 1988 Jul.
Article in English | MEDLINE | ID: mdl-3056244

ABSTRACT

The influence of pretreatment of Escherichia coli and Staphylococcus aureus with sub-MICs of the new beta-lactam antibiotic imipenem on phagocytosis and killing by murine peritoneal macrophages and the susceptibility of these organisms to serum bactericidal activity were studied. The effects of imipenem, a round form inducer in gram-negative rods, and piperacillin, a filamentous form inducer, were compared. Bacteria grown in the presence of sub-MICs of imipenem or piperacillin were incubated for 30 min with macrophage monolayers in the absence of antibiotic. Phagocytosis, killing, and survival within macrophages were evaluated by microbiological and fluorescence microscope assays. Bacteria grown in the presence of a sub-MIC of imipenem were phagocytized and killed in numbers significantly higher than untreated or piperacillin-treated bacteria were. Intracellular bacteria pretreated with a sub-MIC of imipenem were also readily killed by lymphokine-activated macrophages. Prior treatment with a sub-MIC of imipenem resulted in an increased susceptibility of E. coli but not S. aureus to the bactericidal activity of immune serum. Imipenem treatment and immune serum acted synergistically to enhance phagocytosis and killing. The data indicate that exposure of E. coli and S. aureus to a sub-MIC of imipenem enhances the susceptibility of these potential pathogens to cellular and humoral host defense mechanisms.


Subject(s)
Blood Bactericidal Activity/drug effects , Cell Survival/drug effects , Escherichia coli/drug effects , Imipenem/pharmacology , Macrophage Activation/drug effects , Phagocytosis/drug effects , Staphylococcus aureus/drug effects , Antibody Formation/drug effects , Immunity, Cellular/drug effects , Piperacillin/pharmacology
19.
Infect Immun ; 56(4): 998-9, 1988 Apr.
Article in English | MEDLINE | ID: mdl-3346079

ABSTRACT

An anti-TSST-1-specific monoclonal antibody (MAb 8-5-7) was tested for its protective capacity in a rabbit infection model to toxic shock syndrome (TSS). The challenge strain of Staphylococcus aureus (RN4710), which contained a plasmid encoding TSS toxin-1, was introduced into previously implanted chambers. Purified monoclonal antibody (1.25 mg of immunoglobulin G) administered parenterally 1 day before and 1 day after initiation of infection provided complete protection against the TSS-like syndrome and the mortality which occurred in unprotected rabbits.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bacterial Toxins , Enterotoxins/immunology , Shock, Septic/prevention & control , Superantigens , Animals , Antibodies, Bacterial/therapeutic use , Immunization, Passive , Male , Neutralization Tests , Rabbits
20.
Microbiologica ; 11(1): 13-20, 1988 Jan.
Article in English | MEDLINE | ID: mdl-3280948

ABSTRACT

In this study we evaluated the kinetics of phagocytosis and killing of E. coli by thioglycollate-elicited murine peritoneal macrophages and the role of specific antibodies and complement present in different serum preparations in modulating these processes. In our system phagocytosis of E. coli by macrophage monolayer was exponential for 180 min. The killing activity was high in the first 30-60 min and then virtually ceased. The least phagocytosis and killing occurred in presence of heat-inactivated fetal calf serum (HFCS). These activities were 2-fold increased in presence of normal mouse serum (NMS) or heat-inactivated newborn calf serum (HNCS) and were highly stimulated in presence of immune mouse serum (IMS). IMS without complement was less efficient in enhancing phagocytosis and killing by macrophages. However when IMS or HNCS were deprived of specific antibodies their activity was remarkably reduced. When macrophages containing phagocytized bacteria were reincubated with different sera, multiplication of intracellular E. coli occurred with HFCS, NMS or antibody-deprived IMS or HNCS. In contrast, a significant decrease in the survival of intracellular bacteria was seen in presence of IMS, HNCS or complement-deprived IMS. The results indicated that specific bacterial antibodies play a major role in the phagocytic process and in the activation of killing mechanisms. However optimal macrophage activity resulted from the presence of both specific antibodies and complement.


Subject(s)
Antibodies, Bacterial/immunology , Complement System Proteins/immunology , Escherichia coli/immunology , Macrophages/immunology , Phagocytosis , Animals , Female , Immune Sera/immunology , Kinetics , Mice , Opsonin Proteins/immunology , Peritoneal Cavity/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...