Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Radiat Res ; 186(2): 162-74, 2016 08.
Article in English | MEDLINE | ID: mdl-27388041

ABSTRACT

There is increasing evidence that radiation-induced damage to endothelial cells and loss of endothelial function may contribute to both acute radiation syndromes and long-term effects of whole-body nuclear irradiation. Therefore, several drugs are being developed to mitigate the effects of nuclear radiation, most of these drugs will target and protect or regenerate leukocytes and platelets. Our laboratory has demonstrated that TP508, a 23-amino acid thrombin peptide, activates endothelial cells and stem cells to revascularize and regenerate tissues. We now show that TP508 can mitigate radiation-induced damage to endothelial cells in vitro and in vivo. Our in vitro results demonstrate that human endothelial cells irradiation attenuates nitric oxide (NO) signaling, disrupts tube formation and induces DNA double-strand breaks (DSB). TP508 treatment reverses radiation effects on NO signaling, restores tube formation and accelerates the repair of radiation-induced DSB. The radiation-mitigating effects of TP508 on endothelial cells were also seen in CD-1 mice where systemic injection of TP508 stimulated endothelial cell sprouting from aortic explants after 8 Gy irradiation. Systemic doses of TP508 that mitigated radiation-induced endothelial cell damage, also significantly increased survival of CD-1 mice when injected 24 h after 8.5 Gy exposure. These data suggest that increased survival observed with TP508 treatment may be due to its effects on vascular and microvascular endothelial cells. Our study supports the usage of a regenerative drug such as TP508 to activate endothelial cells as a countermeasure for mitigating the effects of nuclear radiation.


Subject(s)
DNA Repair/drug effects , DNA Repair/radiation effects , Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Peptide Fragments/pharmacology , Thrombin/pharmacology , Amino Acid Sequence , Animals , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , Dose-Response Relationship, Radiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Male , Mice , Nitric Oxide/biosynthesis , Survival Analysis
2.
Toxins (Basel) ; 4(11): 1288-300, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23202316

ABSTRACT

Antibiotic treatment may fail to protect individuals, if not started early enough, after infection with Bacillus anthracis, due to the continuing activity of toxins that the bacterium produces. Stable and easily stored inhibitors of the edema factor toxin (EF), an adenylyl cyclase, could save lives in the event of an outbreak, due to natural causes or a bioweapon attack. The toxin's basic activity is to convert ATP to cAMP, and it is thus in principle a simple phosphatase, which means that many mammalian enzymes, including intracellular adenylcyclases, may have a similar activity. While nucleotide based inhibitors, similar to its natural substrate, ATP, were identified early, these compounds had low activity and specificity for EF. We used a combined structural and computational approach to choose small organic molecules in large, web-based compound libraries that would, based on docking scores, bind to residues within the substrate binding pocket of EF. A family of fluorenone-based inhibitors was identified that inhibited the release of cAMP from cells treated with EF. The lead inhibitor was also shown to inhibit the diarrhea caused by enterotoxigenic E. coli (ETEC) in a murine model, perhaps by serving as a quorum sensor. These inhibitors are now being tested for their ability to inhibit Anthrax infection in animal models and may have use against other pathogens that produce toxins similar to EF, such as Bordetella pertussis or Vibrio cholera.


Subject(s)
Anthrax/drug therapy , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Drug Design , Small Molecule Libraries/pharmacology , Adenosine Triphosphate/metabolism , Adenylyl Cyclase Inhibitors , Animals , Anthrax/microbiology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Antigens, Bacterial/chemistry , Bacillus anthracis/drug effects , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Toxins/chemistry , Binding Sites , Cyclic AMP/metabolism , Disease Models, Animal , Ligands , Models, Molecular , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use
3.
Infect Immun ; 77(4): 1649-63, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19168734

ABSTRACT

Bacillus anthracis is the causative agent of anthrax. We have developed a novel whole-bacterial-cell anthrax vaccine utilizing B. anthracis that is killed but metabolically active (KBMA). Vaccine strains that are asporogenic and nucleotide excision repair deficient were engineered by deleting the spoIIE and uvrAB genes, rendering B. anthracis extremely sensitive to photochemical inactivation with S-59 psoralen and UV light. We also introduced point mutations into the lef and cya genes, which allowed inactive but immunogenic toxins to be produced. Photochemically inactivated vaccine strains maintained a high degree of metabolic activity and secreted protective antigen (PA), lethal factor, and edema factor. KBMA B. anthracis vaccines were avirulent in mice and induced less injection site inflammation than recombinant PA adsorbed to aluminum hydroxide gel. KBMA B. anthracis-vaccinated animals produced antibodies against numerous anthrax antigens, including high levels of anti-PA and toxin-neutralizing antibodies. Vaccination with KBMA B. anthracis fully protected mice against challenge with lethal doses of toxinogenic unencapsulated Sterne 7702 spores and rabbits against challenge with lethal pneumonic doses of fully virulent Ames strain spores. Guinea pigs vaccinated with KBMA B. anthracis were partially protected against lethal Ames spore challenge, which was comparable to vaccination with the licensed vaccine anthrax vaccine adsorbed. These data demonstrate that KBMA anthrax vaccines are well tolerated and elicit potent protective immune responses. The use of KBMA vaccines may be broadly applicable to bacterial pathogens, especially those for which the correlates of protective immunity are unknown.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/immunology , Antibodies, Bacterial/blood , Bacillus anthracis , Vaccines, Inactivated/immunology , Animals , Anthrax/microbiology , Anthrax/prevention & control , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/genetics , Antigens, Bacterial/immunology , Bacillus anthracis/genetics , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacillus anthracis/radiation effects , Female , Furocoumarins , Guinea Pigs , Immunity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mutation , Rabbits , Spores, Bacterial/genetics , Ultraviolet Rays , Vaccination , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/genetics , Virulence
4.
J Immune Based Ther Vaccines ; 5: 11, 2007 Oct 22.
Article in English | MEDLINE | ID: mdl-17953756

ABSTRACT

BACKGROUND: There is a clear need for vaccines and therapeutics for potential biological weapons of mass destruction and emerging diseases. Anthrax, caused by the bacterium Bacillus anthracis, has been used as both a biological warfare agent and bioterrorist weapon previously. Although antibiotic therapy is effective in the early stages of anthrax infection, it does not have any effect once exposed individuals become symptomatic due to B. anthracis exotoxin accumulation. The bipartite exotoxins are the major contributing factors to the morbidity and mortality observed in acute anthrax infections. METHODS: Using recombinant B. anthracis protective antigen (PA83), covalently coupled to a novel non-toxic muramyl dipeptide (NT-MDP) derivative we hyper-immunized goats three times over the course of 14 weeks. Goats were plasmapheresed and the IgG fraction (not affinity purified) and F(ab')2 derivatives were characterized in vitro and in vivo for protection against lethal toxin mediated intoxication. RESULTS: Anti-PA83 IgG conferred 100% protection at 7.5 mug in a cell toxin neutralization assay. Mice exposed to 5 LD50 of Bacillus anthracis Ames spores by intranares inoculation demonstrated 60% survival 14 d post-infection when administered a single bolus dose (32 mg/kg body weight) of anti-PA83 IgG at 24 h post spore challenge. Anti-PA83 F(ab')2 fragments retained similar neutralization and protection levels both in vitro and in vivo. CONCLUSION: The protection afforded by these GMP-grade caprine immunotherapeutics post-exposure in the pilot murine model suggests they could be used effectively to treat post-exposure, symptomatic human anthrax patients following a bioterrorism event. These results also indicate that recombinant PA83 coupled to NT-MDP is a potent inducer of neutralizing antibodies and suggest it would be a promising vaccine candidate for anthrax. The ease of production, ease of covalent attachment, and immunostimulatory activity of the NT-MDP indicate it would be a superior adjuvant to alum or other traditional adjuvants in vaccine formulations.

5.
Infect Immun ; 75(8): 4020-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17502384

ABSTRACT

The currently available commercial human anthrax vaccine requires multiple injections for efficacy and has side effects due to its alum adjuvant. These factors limit its utility when immunizing exposed populations in emergent situations. We evaluated a novel mucosal adjuvant that consists of a nontoxic, water-in-oil nanoemulsion (NE). This material does not contain a proinflammatory component but penetrates mucosal surfaces to load antigens into dendritic cells. Mice and guinea pigs were intranasally immunized with recombinant Bacillus anthracis protective antigen (rPA) mixed in NE as an adjuvant. rPA-NE immunization was effective in inducing both serum anti-PA immunoglobulin G (IgG) and bronchial anti-PA IgA and IgG antibodies after either one or two mucosal administrations. Serum anti-PA IgG2a and IgG2b antibodies and PA-specific cytokine induction after immunization indicate a Th1-polarized immune response. rPA-NE immunization also produced high titers of lethal-toxin-neutralizing serum antibodies in both mice and guinea pigs. Guinea pigs nasally immunized with rPA-NE vaccine were protected against an intradermal challenge with approximately 1,000 times the 50% lethal dose ( approximately 1,000x LD(50)) of B. anthracis Ames strain spores (1.38 x 10(3) spores), which killed control animals within 96 h. Nasal immunization also resulted in 70% and 40% survival rates against intranasal challenge with 10x LD(50) and 100x LD(50) (1.2 x 10(6) and 1.2 x 10(7)) Ames strain spores. Our results indicate that NE can effectively adjuvant rPA for intranasal immunization. This potentially could lead to a needle-free anthrax vaccine requiring fewer doses and having fewer side effects than the currently available human vaccine.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Immunity, Mucosal , Nanoparticles , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antitoxins/blood , Bacillus anthracis/immunology , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Emulsions , Female , Guinea Pigs , Humans , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Survival Analysis , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology
6.
Transplantation ; 75(10): 1631-5, 2003 May 27.
Article in English | MEDLINE | ID: mdl-12777847

ABSTRACT

BACKGROUND: Certain transplantation immunosuppressive strategies are primarily based on the interruption of interleukin (IL)-2 signaling by calcineurin inhibition or anti-IL-2 receptor-antibody blockade. However, recent evidence suggests that IL-2 is necessary for peripheral deletion of allograft-specific lymphocytes. STUDY DESIGN: In this study, we examined the apoptotic effects of the calcineurin inhibitor, cyclosporine A, the chimeric anti-interleukin-2 receptor monoclonal antibody, basiliximab, and the rabbit antihuman thymocyte preparation Thymoglobulin (rATG) on phytohemagglutinin-activated human lymphocyte models designed to simulate initial exposure to the graft or ongoing rejection of the graft. RESULTS: We found that rATG increases Fas expression, decreases Bcl-2 expression, and induces early apoptosis in naïve lymphocytes. However, rATG has more of a necrotic effect on activated lymphocytes. Basiliximab and cyclosporine had little effect on apoptosis, but did alter Bcl-2 and Fas expression. CONCLUSIONS: Compared with IL-2 pathway inhibitors, rATG increases lymphocyte apoptosis, probably via Bcl-2 pathway inhibition. Because apoptosis is required for the development of graft tolerance, induction strategies that use IL-2-independent pathways may be advantageous.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antilymphocyte Serum/pharmacology , Apoptosis/drug effects , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-2/antagonists & inhibitors , Lymphocyte Activation/physiology , Lymphocytes/physiology , Recombinant Fusion Proteins , Animals , Basiliximab , Calcineurin Inhibitors , Cells, Cultured , Humans , Lymphocytes/pathology , Necrosis , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Rabbits , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...