Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Front Immunol ; 15: 1277526, 2024.
Article En | MEDLINE | ID: mdl-38605961

This study evaluated a depot-formulated cytokine-based adjuvant to improve the efficacy of the recombinant F1V (rF1V) plague vaccine and examined the protective response following aerosol challenge in a murine model. The results of this study showed that co-formulation of the Alhydrogel-adsorbed rF1V plague fusion vaccine with the depot-formulated cytokines recombinant human interleukin 2 (rhuIL-2) and/or recombinant murine granulocyte macrophage colony-stimulating factor (rmGM-CSF) significantly enhances immunogenicity and significant protection at lower antigen doses against a lethal aerosol challenge. These results provide additional support for the co-application of the depot-formulated IL-2 and/or GM-CSF cytokines to enhance vaccine efficacy.


Plague Vaccine , Yersinia pestis , Humans , Animals , Mice , Cytokines , Antigens, Bacterial , Vaccines, Synthetic , Aerosols
2.
Antimicrob Agents Chemother ; 68(3): e0149723, 2024 Mar 06.
Article En | MEDLINE | ID: mdl-38358266

Bacillus anthracis is a Gram-positive Centers for Disease Control and Prevention category "A" biothreat pathogen. Without early treatment, inhalation of anthrax spores with progression to inhalational anthrax disease is associated with high fatality rates. Gepotidacin is a novel first-in-class triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action and is being evaluated for use against biothreat and conventional pathogens. Gepotidacin selectively inhibits bacterial DNA replication via a unique binding mode and has in vitro activity against a collection of B. anthracis isolates including antibacterial-resistant strains, with the MIC90 ranging from 0.5 to 1 µg/mL. In vivo activity of gepotidacin was also evaluated in the New Zealand White rabbit model of inhalational anthrax. The primary endpoint was survival, with survival duration and bacterial clearance as secondary endpoints. The trigger for treatment was the presence of anthrax protective antigen in serum. New Zealand White rabbits were dosed intravenously for 5 days with saline or gepotidacin at 114 mg/kg/d to simulate a dosing regimen of 1,000 mg intravenous (i.v.) three times a day (TID) in humans. Gepotidacin provided a survival benefit compared to saline control, with 91% survival (P-value: 0.0001). All control animals succumbed to anthrax and were found to be blood- and organ culture-positive for B. anthracis. The novel mode of action, in vitro microbiology, preclinical safety, and animal model efficacy data, which were generated in line with Food and Drug Administration Animal Rule, support gepotidacin as a potential treatment for anthrax in an emergency biothreat situation.


Acenaphthenes , Anthrax Vaccines , Anthrax , Bacillus anthracis , Heterocyclic Compounds, 3-Ring , Respiratory Tract Infections , Rabbits , Humans , Animals , Anthrax/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Anthrax Vaccines/therapeutic use
3.
Hum Vaccin Immunother ; 19(3): 2290345, 2023 Dec 15.
Article En | MEDLINE | ID: mdl-38115181

Pulmonary anthrax caused by exposure to inhaled Bacillus anthracis, the most lethal form of anthrax disease, is a continued military and public health concern for the United States. The vaccine AV7909, consisting of the licensed anthrax drug substance AVA adjuvanted with CpG7909, induces high levels of toxin neutralizing antibodies in healthy adults using fewer doses than AVA. This study compares the ability of one- or two-dose regimens of AV7909 to induce a protective immune response in guinea pigs challenged with a lethal dose of aerosolized B. anthracis spores 6 weeks after the last vaccine dose. The results indicated that AV7909 was less effective when delivered as a single dose compared to the two-dose regimen that resulted in dose-dependent protection against death. The toxin neutralizing assay (TNA) titer and anti-PA IgG responses were proportional to the protective efficacy, with a 50% TNA neutralizing factor (NF50) greater than 0.1 associated with survival in animals receiving two doses of vaccine. The strong protection at relatively low TNA NF50 titers in this guinea pig model supports the exploration of lower doses in clinical trials to determine if these protective levels of neutralizing antibodies can be achieved in humans; however, protection with a single dose may not be feasible.


Anthrax Vaccines , Anthrax , Bacillus anthracis , Adult , Humans , Animals , Guinea Pigs , Anthrax/prevention & control , Antibodies, Bacterial , Antibodies, Neutralizing , Antigens, Bacterial
4.
Antibiotics (Basel) ; 12(8)2023 Aug 19.
Article En | MEDLINE | ID: mdl-37627757

Francisella tularensis subspecies tularensis is a category-A biothreat agent that can cause lethal tularemia. Ceftobiprole medocaril is being explored as a medical countermeasure for the treatment of pneumonic tularemia. The efficacy of ceftobiprole medocaril against inhalational tularemia was evaluated in the Fischer 344 rat model of infection. The dose was expected to be effective against F. tularensis isolates with ceftobiprole minimum inhibitory concentrations ≤0.5 µg/mL. Animals treated with ceftobiprole medocaril exhibited a 92% survival rate 31 days post-challenge, identical to the survival of levofloxacin-treated rats. By comparison, rats receiving placebo experienced 100% mortality. Terminally collected blood, liver, lung, and spleen samples confirmed disseminated F. tularensis infections in most animals that died prior to completing treatments (placebo animals and a rat treated with ceftobiprole medocaril), although levels of bacteria detected in the placebo samples were significantly elevated compared to the ceftobiprole-medocaril-treated group geometric mean. Furthermore, no evidence of infection was detected in any rat that completed ceftobiprole medocaril or levofloxacin treatment and survived to the end of the post-treatment observation period. Overall, survival rates, body weights, and bacterial burdens consistently demonstrated that treatment with ceftobiprole medocaril is efficacious against otherwise fatal cases of pneumonic tularemia in the rat model.

5.
Antimicrob Agents Chemother ; 67(5): e0138122, 2023 05 17.
Article En | MEDLINE | ID: mdl-37097147

Francisella tularensis (F. tularensis) is a Centers for Disease Control (CDC) category "A" Gram-negative biothreat pathogen. Inhalation of F. tularensis can cause pneumonia and respiratory failure and is associated with high mortality rates without early treatment. Gepotidacin is a novel, first-in-class triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action. Gepotidacin selectively inhibits bacterial DNA replication via a unique binding mode, has activity against multidrug-resistant target pathogens, and has demonstrated in vitro activity against diverse collections of F. tularensis isolates (MIC90 of 0.5 to 1 µg/mL). Gepotidacin was evaluated in the cynomolgus macaque model of inhalational tularemia, using the SCHU S4 strain, with treatment initiated after exposure and sustained fever. Macaques were dosed via intravenous (i.v.) infusion with saline or gepotidacin at 72 mg/kg/day to support a human i.v. infusion dosing regimen of 1,000 mg three times daily. The primary study endpoint was survival, with survival duration and bacterial clearance as secondary endpoints. Gepotidacin treatment resulted in 100% survival compared to 12.5% in the saline-treated control group (P < 0.0001) at Day 43 postinhalational challenge. All gepotidacin-treated animals were blood and organ culture negative for F. tularensis at the end of the study. In contrast, none of the saline control animals were blood and organ culture negative. Gepotoidacin's novel mechanism of action and the efficacy data reported here (aligned with the Food and Drug Administration Animal Rule) support gepotidacin as a potential treatment for pneumonic tularemia in an emergency biothreat situation.


Francisella tularensis , Tularemia , Animals , Humans , Tularemia/microbiology , Disease Models, Animal , Macaca fascicularis , Bacterial Vaccines
6.
Sci Rep ; 13(1): 4047, 2023 03 10.
Article En | MEDLINE | ID: mdl-36899021

Melioidosis is an endemic disease in numerous tropical regions. Additionally, the bacterium that causes melioidosis, Burkholderia pseudomallei, has potential to be used as a biological weapon. Therefore, development of effective and affordable medical countermeasures to serve regions affected by the disease and to have medical countermeasures available in the event of a bioterrorism attack remains critical. The current study evaluated the efficacy of eight distinct acute phase ceftazidime treatment regimens administered therapeutically in the murine model. At the conclusion of the treatment period, survival rates were significantly greater in several of the treated groups when compared to the control group. Pharmacokinetics of a single dose of ceftazidime were examined at 150 mg/kg, 300 mg/kg, and 600 mg/kg and were compared to an intravenous clinical dose administered at 2000 mg every eight hours. The clinical dose has an estimated 100% fT > 4*MIC which exceeded the highest murine dose of 300 mg/kg every six hours at 87.2% fT > 4*MIC. Based upon survival at the end of the treatment regimen and supplemented by pharmacokinetic modeling, a daily dose of 1200 mg/kg of ceftazidime, administered every 6 h at 300 mg/kg, provides protection in the acute phase of inhalation melioidosis in the murine model.


Burkholderia pseudomallei , Melioidosis , Animals , Mice , Ceftazidime/pharmacology , Melioidosis/microbiology , Disease Models, Animal , Aerosols/pharmacology , Anti-Bacterial Agents/pharmacology
7.
Vaccines (Basel) ; 10(2)2022 Jan 19.
Article En | MEDLINE | ID: mdl-35214604

Background: The need for an updated plague vaccine is highlighted by outbreaks in endemic regions together with the pandemic potential of this disease. There is no easily available, approved vaccine. Methods: Here we have used a murine model of pneumonic plague to examine the factors that maximise immunogenicity and contribute to survival following vaccination. We varied vaccine type, as either a genetic fusion of the F1 and V protein antigens or a mixture of these two recombinant antigens, as well as antigen dose-level and formulation in order to correlate immune response to survival. Results: Whilst there was interaction between each of the variables of vaccine type, dose level and formulation and these all contributed to survival, vaccine formulation in protein-coated microcrystals (PCMCs) was the key contributor in inducing antibody titres. From these data, we propose a cut-off in total serum antibody titre to the F1 and V proteins of 100 µg/mL and 200 µg/mL, respectively. At these thresholds, survival is predicted in this murine pneumonic model to be >90%. Within the total titre of antibody to the V antigen, the neutralising antibody component correlated with dose level and was enhanced when the V antigen in free form was formulated in PCMCs. Antibody titre to F1 was limited by fusion to V, but this was compensated for by PCMC formulation. Conclusions: These data will enable clinical assessment of this and other candidate plague vaccines that utilise the same vaccine antigens by identifying a target antibody titre from murine models, which will guide the evaluation of clinical titres as serological surrogate markers of efficacy.

8.
Vaccine ; 39(1): 1-5, 2021 01 03.
Article En | MEDLINE | ID: mdl-33199078

The anthrax vaccine candidate AV7909 is being developed as a next-generation vaccine for post-exposure prophylaxis (PEP) against inhalational anthrax. In clinical studies, two vaccinations with AV7909 administered either two or four weeks apart induced an enhanced immune response compared to BioThrax® (Anthrax Vaccine Adsorbed) (AVA). Anthrax toxin-neutralizing antibody (TNA) levels on Day 70 following initial vaccination that were associated with protection of animals exposed to inhalational anthrax were previously reported for the 0, 4-week AV7909 vaccination regimen. The current study shows that a 0, 2-week AV7909 vaccination regimen protected guinea pigs (GPs) and nonhuman primates (NHPs) against a lethal inhalational anthrax challenge on Days 28 and 70 after the first immunization. An earlier induction of protective TNA levels using a 0, 2-week AV7909 vaccination regimen may provide benefit over the currently approved AVA PEP 0, 2, and 4-week vaccination regimen.


Anthrax Vaccines , Anthrax , Bacillus anthracis , Animals , Anthrax/prevention & control , Antibodies, Bacterial , Antibodies, Neutralizing , Antigens, Bacterial , Guinea Pigs , Post-Exposure Prophylaxis , Primates
9.
Front Immunol ; 11: 598847, 2020.
Article En | MEDLINE | ID: mdl-33542715

Venezuelan, eastern and western equine encephalitis viruses (EEV) can cause severe disease of the central nervous system in humans, potentially leading to permanent damage or death. Yet, no licensed vaccine for human use is available to protect against these mosquito-borne pathogens, which can be aerosolized and therefore pose a bioterror threat in addition to the risk of natural outbreaks. Using the mouse aerosol challenge model, we evaluated the immunogenicity and efficacy of EEV vaccines that are based on the modified vaccinia Ankara-Bavarian Nordic (MVA-BN®) vaccine platform: three monovalent vaccines expressing the envelope polyproteins E3-E2-6K-E1 of the respective EEV virus, a mixture of these three monovalent EEV vaccines (Triple-Mix) as a first approach to generate a multivalent vaccine, and a true multivalent alphavirus vaccine (MVA-WEV, Trivalent) encoding the polyproteins of all three EEVs in a single non-replicating MVA viral vector. BALB/c mice were vaccinated twice in a four-week interval and samples were assessed for humoral and cellular immunogenicity. Two weeks after the second immunization, animals were exposed to aerosolized EEV. The majority of vaccinated animals exhibited VEEV, WEEV, and EEEV neutralizing antibodies two weeks post-second administration, whereby the average VEEV neutralizing antibodies induced by the monovalent and Trivalent vaccine were significantly higher compared to the Triple-Mix vaccine. The same statistical difference was observed for VEEV E1 specific T cell responses. However, all vaccinated mice developed comparable interferon gamma T cell responses to the VEEV E2 peptide pools. Complete protective efficacy as evaluated by the prevention of mortality and morbidity, lack of clinical signs and viremia, was demonstrated for the respective monovalent MVA-EEV vaccines, the Triple-Mix and the Trivalent single vector vaccine not only in the homologous VEEV Trinidad Donkey challenge model, but also against heterologous VEEV INH-9813, WEEV Fleming, and EEEV V105-00210 inhalational exposures. These EEV vaccines, based on the safe MVA vector platform, therefore represent promising human vaccine candidates. The trivalent MVA-WEV construct, which encodes antigens of all three EEVs in a single vector and can potentially protect against all three encephalitic viruses, is currently being evaluated in a human Phase 1 trial.


Encephalitis Virus, Eastern Equine/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Western Equine/immunology , Encephalomyelitis, Equine/prevention & control , Viral Vaccines/immunology , Aerosols , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cross Protection/immunology , Disease Models, Animal , Encephalomyelitis, Equine/immunology , Encephalomyelitis, Equine/mortality , Female , Immunization , Mice , Mortality , Neutralization Tests , Vaccines, DNA , Viral Vaccines/administration & dosage
10.
J Pediatr Orthop ; 39(8): 387-393, 2019 Sep.
Article En | MEDLINE | ID: mdl-31393289

BACKGROUND: Throwing requires proper stability and orientation of the pelvis and scapula for efficient energy transfer during pitching. Fatigue of the pelvis and scapular musculature throughout the course of a game can impair pitching performance, and place excessive demands on the throwing arm leading to injury. The purpose of this study was to examine differences in pelvis, torso, and upper extremity pitching mechanics and muscle activations between the fastball, change-up, and curveball pitches in youth baseball pitchers following a simulated game. METHODS: Fourteen youth baseball pitchers with no history of injury participated. Pitching mechanics were collected using an electromagnetic tracking system. Surface electromyography data were collected on the bilateral gluteus medius and maximus; and throwing arm side latissimus dorsi, lower trapezius, upper trapezius, and serratus anterior. Participants were instructed to throw maximum effort pitches during a simulated game that provided random game situations similar to those that occur in competition. Participants were limited to 85 pitches based on age-restricted pitch counts. Data from 3 fastballs, curveballs, and change-ups thrown in the first and last innings were selected for analysis. RESULTS: Repeated measures multivariate analyses of variance revealed that neither pitch type nor the effect of a simulated game resulted in statistically significant changes in pitching mechanics (F(10,600)=0.55, P=0.85), or muscle activations (pelvic: F(4,195)=0.07, P=0.85; scapular: F(4,118)=0.09, P=0.52). CONCLUSIONS: The principle findings of this study revealed that pitching to the age-restricted pitch count limit did not result in altered pitching mechanics or muscle activations, and no differences occurred between the 3 pitches. These results support previous research that indicate the curveball pitch is no more dangerous for youth than the other pitches commonly thrown. This is supported by the pitcher's ability to maintain a proper arm slot during all 3 pitches and indicates that they are obtaining the spin on the ball from their grip and not by altering upper extremity mechanics. LEVEL OF EVIDENCE: Level 5.


Baseball/physiology , Electromyography/methods , Muscle, Skeletal/physiology , Scapula/physiology , Shoulder Joint/physiology , Upper Extremity/physiology , Adolescent , Biomechanical Phenomena , Humans , Male
11.
Biophys J ; 116(3): 406-418, 2019 02 05.
Article En | MEDLINE | ID: mdl-30558886

Based on our recent finding that FBP21 regulates human Brr2 helicase activity involved in the activation of the spliceosomal B-complex, we investigated the structural and dynamic contribution of FBP21 to the interaction. By using NMR spectroscopy, we could show that the 50 C-terminal residues of FBP21 (FBP21326-376), which are sufficient to fully form the interaction with the C-terminal Sec63 unit of Brr2 (Brr2C-Sec63), adopt a random-coil conformation in their unbound state. Upon interaction with Brr2C-Sec63, 42 residues of FBP21326-376 cover the large binding site on Brr2C-Sec63 in an extended conformation. Short charged motifs are steering complex formation, still allowing the bound state to retain dynamics. Based on fragment docking in combination with experimental restraints, we present models of the complex structure. The FBP21326-376/Brr2C-Sec63 interaction thus presents an example of an intrinsically disordered protein/ordered-protein interaction in which a large binding site provides high specificity and, in combination with conformational disorder, displays a relatively high affinity.


Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Humans , Molecular Docking Simulation , Protein Domains , Thermodynamics
12.
Biophys J ; 114(4): 788-799, 2018 02 27.
Article En | MEDLINE | ID: mdl-29490241

Precursor messenger RNA splicing is mediated by the spliceosome, a large and dynamic molecular machine composed of five small nuclear RNAs and numerous proteins. Many spliceosomal proteins are predicted to be intrinsically disordered or contain large disordered regions, but experimental validation of these predictions is scarce, and the precise functions of these proteins are often unclear. Here, we show via circular dichroism spectroscopy, dynamic light scattering, and NMR spectroscopy that the yeast spliceosomal disassembly factor Ntr2 is largely intrinsically disordered. Peptide SPOT analyses, analytical size-exclusion chromatography, and surface plasmon resonance measurements revealed that Ntr2 uses an N-terminal region to bind the C-terminal helicase unit of the Brr2 RNA helicase, an enzyme involved in spliceosome activation and implicated in splicing catalysis and spliceosome disassembly. NMR analyses suggested that Ntr2 does not adopt a tertiary structure and likely remains disordered upon complex formation. RNA binding and unwinding studies showed that Ntr2 downregulates Brr2 helicase activity in vitro by modulating the fraction of helicase molecules productively bound to the RNA substrate. Our data clarify the nature of a physical link between Brr2 and Ntr2, and point to the possibility of a functional Ntr2-Brr2 interplay during splicing.


Intrinsically Disordered Proteins/metabolism , RNA Helicases/metabolism , RNA, Fungal/metabolism , RNA, Small Nuclear/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism , Catalysis , Intrinsically Disordered Proteins/chemistry , RNA Helicases/chemistry , Saccharomyces cerevisiae Proteins/chemistry
13.
Article En | MEDLINE | ID: mdl-29133571

The recommended management of inhalational anthrax, a high-priority bioterrorist threat, includes antibiotics and antitoxins. Obiltoxaximab, a chimeric monoclonal antibody against anthrax protective antigen (PA), is licensed under the U.S. Food and Drug Administration's (FDA's) Animal Rule for the treatment of inhalational anthrax. Because of spore latency, disease reemergence after treatment cessation is a concern, and there is a need to understand the development of endogenous protective immune responses following antitoxin-containing anthrax treatment regimens. Here, acquired protective immunity was examined in New Zealand White (NZW) rabbits challenged with a targeted lethal dose of Bacillus anthracis spores and treated with antibiotics, obiltoxaximab, or a combination of both. Survivors of the primary challenge were rechallenged 9 months later and monitored for survival. Survival rates after primary and rechallenge for controls and animals treated with obiltoxaximab, levofloxacin, or a combination of both were 0, 65, 100, and 95%, and 0, 100, 95, and 89%, respectively. All surviving immune animals had circulating antibodies to PA and serum toxin-neutralizing titers prior to rechallenge. Following rechallenge, systemic bacteremia and toxemia were not detected in most animals, and the levels of circulating anti-PA IgG titers increased starting at 5 days postrechallenge. We conclude that treatment with obiltoxaximab, alone or combined with antibiotics, significantly improves the survival of rabbits that received a lethal inhalation B. anthracis spore challenge dose and does not interfere with the development of immunity. Survivors of primary challenge are protected against reexposure, have rare incidents of systemic bacteremia and toxemia, and have evidence of an anamnestic response.


Anthrax , Anti-Bacterial Agents , Antibodies, Monoclonal , Antitoxins , Bacillus anthracis , Levofloxacin , Respiratory Tract Infections , Spores, Bacterial , Animals , Female , Male , Rabbits , Anthrax/immunology , Anthrax/microbiology , Anthrax/mortality , Anthrax/prevention & control , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/biosynthesis , Antibodies, Monoclonal/pharmacology , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Antitoxins/pharmacology , Bacillus anthracis/drug effects , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/blood , Bacterial Toxins/immunology , Drug Therapy, Combination , Immunization, Passive/methods , Immunoglobulin G/biosynthesis , Immunologic Memory/drug effects , Levofloxacin/pharmacology , Random Allocation , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/mortality , Respiratory Tract Infections/prevention & control , Spores, Bacterial/drug effects , Spores, Bacterial/immunology , Spores, Bacterial/pathogenicity , Survival Analysis
14.
Article En | MEDLINE | ID: mdl-28784679

The fluorocycline TP-271 was evaluated in mouse and nonhuman primate (NHP) models of inhalational anthrax. BALB/c mice were exposed by nose-only aerosol to Bacillus anthracis Ames spores at a level of 18 to 88 lethal doses sufficient to kill 50% of exposed individuals (LD50). When 21 days of once-daily dosing was initiated at 24 h postchallenge (the postexposure prophylaxis [PEP] study), the rates of survival for the groups treated with TP-271 at 3, 6, 12, and 18 mg/kg of body weight were 90%, 95%, 95%, and 84%, respectively. When 21 days of dosing was initiated at 48 h postchallenge (the treatment [Tx] study), the rates of survival for the groups treated with TP-271 at 6, 12, and 18 mg/kg TP-271 were 100%, 91%, and 81%, respectively. No deaths of TP-271-treated mice occurred during the 39-day posttreatment observation period. In the NHP model, cynomolgus macaques received an average dose of 197 LD50 of B. anthracis Ames spore equivalents using a head-only inhalation exposure chamber, and once-daily treatment of 1 mg/kg TP-271 lasting for 14 or 21 days was initiated within 3 h of detection of protective antigen (PA) in the blood. No (0/8) animals in the vehicle control-treated group survived, whereas all 8 infected macaques treated for 21 days and 4 of 6 macaques in the 14-day treatment group survived to the end of the study (56 days postchallenge). All survivors developed toxin-neutralizing and anti-PA IgG antibodies, indicating an immunologic response. On the basis of the results obtained with the mouse and NHP models, TP-271 shows promise as a countermeasure for the treatment of inhalational anthrax.


Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Bacillus anthracis/drug effects , Respiratory Tract Infections/drug therapy , Tetracyclines/therapeutic use , Animals , Anthrax/microbiology , Anthrax/mortality , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Disease Models, Animal , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Macaca fascicularis , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Post-Exposure Prophylaxis/methods , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/mortality , Spores, Bacterial , Survival Rate , Tetracyclines/pharmacokinetics
15.
Nucleic Acids Res ; 45(13): 7922-7937, 2017 Jul 27.
Article En | MEDLINE | ID: mdl-28838205

Splicing of eukaryotic pre-mRNA is carried out by the spliceosome, which assembles stepwise on each splicing substrate. This requires the concerted action of snRNPs and non-snRNP accessory proteins, the functions of which are often not well understood. Of special interest are B complex factors that enter the spliceosome prior to catalytic activation and may alter splicing kinetics and splice site selection. One of these proteins is FBP21, for which we identified several spliceosomal binding partners in a yeast-two-hybrid screen, among them the RNA helicase Brr2. Biochemical and biophysical analyses revealed that an intrinsically disordered region of FBP21 binds to an extended surface of the C-terminal Sec63 unit of Brr2. Additional contacts in the C-terminal helicase cassette are required for allosteric inhibition of Brr2 helicase activity. Furthermore, the direct interaction between FBP21 and the U4/U6 di-snRNA was found to reduce the pool of unwound U4/U6 di-snRNA. Our results suggest FBP21 as a novel key player in the regulation of Brr2.


Carrier Proteins/metabolism , Nuclear Proteins/metabolism , RNA Helicases/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Allosteric Regulation , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Humans , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Interaction Domains and Motifs , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Precursors/metabolism , RNA Splicing , RNA, Small Nuclear/metabolism , RNA-Binding Proteins , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism , Two-Hybrid System Techniques
16.
Article En | MEDLINE | ID: mdl-28559261

TP-271 is a novel, fully synthetic fluorocycline in development for complicated bacterial respiratory infections. TP-271 was active in vitro against a panel of 29 Francisella tularensis isolates, showing MICs against 50% and 90% of isolates of 0.25 and 0.5 µg/ml, respectively. In a mouse model of inhalational tularemia, animals were exposed by aerosol to 91 to 283 50% lethal doses (LD50)/mouse of F. tularensis SCHU S4. Following 21 days of once-daily intraperitoneal dosing with TP-271 at 3, 6, 12, and 18 mg/kg of body weight/day, initiating at 24 h postchallenge, survival was 80%, 100%, 100%, and 100%, respectively. When treatment was initiated at 72 h postchallenge, survival was 89%, 100%, 100%, and 100% in the 3-, 6-, 12-, and 18-mg/kg/day TP-271 groups, respectively. No mice treated with the vehicle control survived. Surviving mice treated with TP-271 showed little to no relapse during 14 days posttreatment. In a nonhuman primate model of inhalational tularemia, cynomolgus macaques received an average aerosol exposure of 1,144 CFU of F. tularensis SCHU S4. Once-daily intravenous infusion with 1 or 3 mg/kg TP-271, or vehicle control, for 21 days was initiated within 6 h of confirmed fever. All animals treated with TP-271 survived to the end of the study, with no relapse during 14 days after the last treatment, whereas no vehicle control-treated animals survived. The protection and low relapse afforded by TP-271 treatment in these studies support continued investigation of TP-271 for use in the event of aerosolized exposure to F. tularensis.


Anti-Bacterial Agents/therapeutic use , Francisella tularensis/drug effects , Respiratory Tract Infections/drug therapy , Tetracyclines/therapeutic use , Tularemia/drug therapy , Animals , Disease Models, Animal , Female , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Respiratory Tract Infections/microbiology , Tularemia/microbiology
17.
Antimicrob Agents Chemother ; 60(10): 5796-805, 2016 10.
Article En | MEDLINE | ID: mdl-27431219

The Centers for Disease Control and Prevention recommend adjunctive antitoxins when systemic anthrax is suspected. Obiltoxaximab, a monoclonal antibody against protective antigen (PA), is approved for treatment of inhalational anthrax in combination with antibiotics and for prophylaxis when alternative therapies are not available. The impact of toxin neutralization with obiltoxaximab during pre- and postexposure prophylaxis was explored, and efficacy results that supported the prophylaxis indication are presented here. New Zealand White rabbits and cynomolgus macaques received obiltoxaximab as a single intramuscular or intravenous dose of 2 to 16 mg/kg of body weight at various times relative to Bacillus anthracis aerosol spore challenge. The primary endpoint was survival, and effect of treatment timing was explored. In rabbits, obiltoxaximab administration 9 h postchallenge singly or combined with a 5-day levofloxacin regimen protected 89% to 100% of animals compared to 33% with levofloxacin monotherapy. In cynomolgus macaques, a single intramuscular dose of 16 mg/kg obiltoxaximab led to 100% survival when given 1 to 3 days preexposure and 83% to 100% survival when given 18 to 24 h postexposure and prior to systemic bacteremia onset. Obiltoxaximab administration after bacteremia onset resulted in lower (25% to 50%) survival rates reflective of treatment setting. Prophylactic administration of obiltoxaximab before spore challenge or to spore-challenged animals before systemic bacterial dissemination is efficacious in promoting survival, ameliorating toxemia, and inhibiting bacterial spread to the periphery.


Anthrax/mortality , Anthrax/prevention & control , Antibodies, Monoclonal/pharmacology , Antitoxins/pharmacology , Bacillus anthracis/pathogenicity , Respiratory Tract Infections/mortality , Respiratory Tract Infections/prevention & control , Animals , Anthrax/drug therapy , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antitoxins/administration & dosage , Bacillus anthracis/drug effects , Bacteremia/drug therapy , Bacteremia/microbiology , Disease Models, Animal , Female , Injections, Intramuscular , Injections, Intravenous , Macaca fascicularis , Male , Post-Exposure Prophylaxis , Pre-Exposure Prophylaxis , Rabbits , Respiratory Tract Infections/drug therapy , Survival Rate
18.
Antimicrob Agents Chemother ; 60(10): 5787-95, 2016 10.
Article En | MEDLINE | ID: mdl-27431222

Inhalational anthrax has high mortality even with antibiotic treatment, and antitoxins are now recommended as an adjunct to standard antimicrobial regimens. The efficacy of obiltoxaximab, a monoclonal antibody against anthrax protective antigen (PA), was examined in multiple studies conducted in two animal models of inhalational anthrax. A single intravenous bolus of 1 to 32 mg/kg of body weight obiltoxaximab or placebo was administered to New Zealand White rabbits (two studies) and cynomolgus macaques (4 studies) at disease onset (significant body temperature increase or detection of serum PA) following lethal challenge with aerosolized Bacillus anthracis spores. The primary endpoint was survival. The relationship between efficacy and disease severity, defined by pretreatment bacteremia and toxemia levels, was explored. In rabbits, single doses of 1 to 16 mg/kg obiltoxaximab led to 17 to 93% survival. In two studies, survival following 16 mg/kg obiltoxaximab was 93% and 62% compared to 0% and 0% for placebo (P = 0.0010 and P = 0.0013, respectively). Across four macaque studies, survival was 6.3% to 78.6% following 4 to 32 mg/kg obiltoxaximab. In two macaque studies, 16 mg/kg obiltoxaximab reduced toxemia and led to survival rates of 31%, 35%, and 47% versus 0%, 0%, and 6.3% with placebo (P = 0.0085, P = 0.0053, P = 0.0068). Pretreatment bacteremia and toxemia levels inversely correlated with survival. Overall, obiltoxaximab monotherapy neutralized PA and increased survival across the range of disease severity, indicating clinical benefit of toxin neutralization with obiltoxaximab in both early and late stages of inhalational anthrax.


Anthrax/drug therapy , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antitoxins/pharmacology , Respiratory Tract Infections/drug therapy , Animals , Anthrax/etiology , Anthrax/mortality , Anti-Bacterial Agents/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Female , Macaca fascicularis , Male , Rabbits , Respiratory Tract Infections/etiology , Respiratory Tract Infections/mortality , Survival Rate , Treatment Outcome
19.
Int J Sports Phys Ther ; 11(1): 108-14, 2016 Feb.
Article En | MEDLINE | ID: mdl-26900505

BACKGROUND: With shoulder pain and injury on the rise in overhead athletes, clinicians are often examining preventative exercises to address functional abnormalities. Because shoulder impingement is prevalent in overhead athletes, much focus is on scapular stability and the function of the stabilizing force couple of the upper and lower trapezius and serratus anterior. HYPOTHESIS/PURPOSE: The purpose of this study was to examine scapular muscle activation during a series of throws and holds (throwing without releasing) with two different ball weights (7oz and 12oz). It was hypothesized that the holds exercises would elicit greater activation of the scapular musculature than the throw, irrespective of ball weight. STUDY DESIGN: Case control laboratory study. METHODS: Twenty-two NCAA Division I, right hand dominant, softball players (19.91 + 1.04 years; 169.24 + 7.36 cm; 72.09 + 10.61 kg) volunteered to participate. Surface EMG was utilized to measure muscle activity in the upper, middle and lower trapezius and serratus anterior muscles during three different throwing activities. RESULTS: MANOVA results revealed no significant differences in muscle activity between throwing conditions, F(16,82) = 1.02, p = 0.446, Wilks' Λ = 0.696, Cohen's d = 0.44 (7oz holds), 0.24 (12oz holds), power = 0.625. CONCLUSION/CLINICAL RELEVANCE: The results may provide some clinical insight in advocating the use of holds with different ball weights. The holds throw may be an effective step in shoulder strengthening that can more closely mimic the functional movement of throwing without the element of ball release. LEVELS OF EVIDENCE: Level 3.

20.
J Strength Cond Res ; 30(2): 415-20, 2016 Feb.
Article En | MEDLINE | ID: mdl-26815175

It is generally accepted that playing with fatigue is a primary predictor of injury in youth baseball because muscular fatigue is believed to alter mechanics during the arm cocking and acceleration phases. Therefore, the purpose of this study was to quantitatively describe gluteal and upper extremity muscle activations in youth baseball pitchers during a simulated game. Twenty-three youth baseball players (11.2 ± 0.8 years; 151.4 ± 8.7 cm; 47.5 ± 10.8 kg) participated. Data were collected through a Delsys Bagnoli-8-channel electromyography system. Single differential electrodes (interelectrode distance: 10 mm) were attached to the bilateral gluteus maximus and medius and throwing side latissimus dorsi, lower trapezius, and serratus anterior and upper trapezius. After warm-up, participants were instructed to throw randomly provided game situations over a regulation distance (46 feet; 14.02 meters) to a catcher. Three, 4-seam fastballs for strikes, thrown in the first and last innings of the simulated game were selected for analysis. A multivariate analysis of variance revealed no statistically significant differences in muscle activity at the 3 phases of the throw, between first and last innings of the simulated game with an observed power of 0.274 (phase 1: foot contact to maximum shoulder external rotation), 0.297 (phase 2: maximum shoulder external rotation to ball release), and 0.226 (phase 3: ball release to maximum shoulder internal rotation). Examining muscle activations as a pitcher approaches fatigue provides information on how long a pitcher can perform before mechanical alterations occur. Although this study did not reveal significant changes, it did reiterate the fact that pitch counts may be working in possibly preventing a youth pitcher throwing to fatigue.


Baseball/physiology , Electromyography , Muscle, Skeletal/physiology , Adolescent , Child , Humans , Male , Muscle Fatigue/physiology
...