Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biomech Eng ; 146(10)2024 10 01.
Article in English | MEDLINE | ID: mdl-38652582

ABSTRACT

Penn State University is developing a pediatric total artificial heart (TAH) as a bridge-to-transplant device that supports infants and small children with single ventricle anomalies or biventricular heart failure to address high waitlist mortality rates for pediatric patients with severe congenital heart disease (CHD). Two issues with mechanical circulatory support devices are thrombus formation and thromboembolic events. This in vitro study characterizes flow within Penn State's pediatric total artificial heart under physiological operating conditions. Particle image velocimetry (PIV) is used to quantify flow within the pump and to calculate wall shear rates (WSRs) along the internal pump surface to identify potential thrombogenic regions. Results show that the diastolic inflow jets produce sufficient wall shear rates to reduce thrombus deposition potential along the inlet side of the left and right pumps. The inlet jet transitions to rotational flow, which promotes wall washing along the apex of the pumps, prevents flow stasis, and aligns flow with the outlet valve prior to systolic ejection. However, inconsistent high wall shear rates near the pump apex cause increased thrombogenic potential. Strong systolic outflow jets produce high wall shear rates near the outlet valve to reduce thrombus deposition risk. The right pump, which has a modified outlet port angle to improve anatomical fit, produces lower wall shear rates and higher thrombus susceptibility potential (TSP) compared to the left pump. In summary, this study provides a fluid dynamic understanding of a new pediatric total artificial heart and indicates thrombus susceptibility is primarily confined to the apex, consistent with similar pulsatile heart pumps.


Subject(s)
Heart, Artificial , Hydrodynamics , Humans , Rheology , Child , Thrombosis , Models, Cardiovascular
2.
J Biomech Eng ; 144(7)2022 07 01.
Article in English | MEDLINE | ID: mdl-34897373

ABSTRACT

For children born with a single functional ventricle, the Fontan operation bypasses the right ventricle by forming a four-way total cavopulmonary connection and adapts the existing ventricle for the systemic circulation. However, upon reaching adulthood, many Fontan patients exhibit low cardiac output and elevated venous pressure, eventually requiring a heart transplantation. Despite efforts in developing a new device or using an existing device for failing Fontan support, there is still no Food and Drug Administration-approved device for subpulmonary support. Penn State University is developing a hydrodynamically levitated Fontan circulatory assist device (FCAD) for bridge-to-transplant or destination therapy. The hemodynamics within the FCAD, at both steady and patient averaged pulsatile conditions for three physiological pump operating conditions, were quantified using particle image velocimetry (PIV) to determine the velocity magnitudes and Reynolds normal and shear stresses within the device. Data were acquired at three planes (0 mm and ±25% of the radius) for the inferior and superior vena cavae inlets and the pulmonary artery outlet. The inlets had a blunt velocity profile that became skewed toward the collecting volute as fluid approached the rotor. At the outlet, regardless of the flow condition, a high-velocity jet exited the volute and moved downstream in a helical pattern. Turbulent stresses observed at the volute exit were influenced by the rotor's rotation. Regardless of inlet conditions, the pump demonstrated advantageous behavior for clinical use with a predictable flow field and a low risk of platelet adhesion and hemolysis based on calculated wall shear rates and turbulent stresses, respectively.


Subject(s)
Fontan Procedure , Heart-Assist Devices , Adult , Child , Fontan Procedure/methods , Heart Ventricles , Hemodynamics , Humans , Models, Cardiovascular
3.
Antimicrob Agents Chemother ; 65(7): e0272420, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33972247

ABSTRACT

Development and validation of large animal models of Pseudomonas aeruginosa ventilator-associated pneumonia are needed for testing new drug candidates in a manner that mimics how they will be used clinically. We developed a new model in which rabbits were ventilated with low tidal volume and challenged with P. aeruginosa to recapitulate hallmark clinical features of acute respiratory distress syndrome (ARDS): acute lung injury and inflammation, progressive decrease in arterial oxygen partial pressure to fractional inspired oxygen PaO2:FiO2, leukopenia, neutropenia, thrombocytopenia, hyperlactatemia, severe hypotension, bacterial dissemination from lung to other organs, multiorgan dysfunction, and ultimately death. We evaluated the predictive power of this rabbit model for antibiotic efficacy testing by determining whether a humanized dosing regimen of meropenem, a potent antipseudomonal ß-lactam antibiotic, when administered with or without intensive care unit (ICU)-supportive care (fluid challenge and norepinephrine), could halt or reverse natural disease progression. Our humanized meropenem dosing regimen produced a plasma concentration-time profile in the rabbit model similar to those reported in patients with ventilator-associated bacterial pneumonia. In this rabbit model, treatment with humanized meropenem and ICU-supportive care achieved the highest level of survival, halted the worsening of ARDS biomarkers, and reversed lethal hypotension, although treatment with humanized meropenem alone also conferred some protection compared to treatment with placebo (saline) alone or placebo plus ICU-supportive care. In conclusion, this rabbit model could help predict whether an antibiotic will be efficacious for the treatment of human ventilator-associated pneumonia.


Subject(s)
Pneumonia, Ventilator-Associated , Pseudomonas aeruginosa , Animals , Anti-Bacterial Agents/therapeutic use , Drug Development , Humans , Meropenem , Pneumonia, Ventilator-Associated/drug therapy , Rabbits
4.
Antimicrob Agents Chemother ; 65(8): e0055221, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34001510

ABSTRACT

There is an urgent need for oral agents to combat resistant Gram-negative pathogens. Here, we describe the characterization of VNRX-5236, a broad-spectrum boronic acid ß-lactamase inhibitor (BLI), and its orally bioavailable etzadroxil prodrug, VNRX-7145. VNRX-7145 is being developed in combination with ceftibuten, an oral cephalosporin, to combat strains of Enterobacterales expressing extended-spectrum ß-lactamases (ESBLs) and serine carbapenemases. VNRX-5236 is a reversible covalent inhibitor of serine ß-lactamases, with inactivation efficiencies on the order of 104 M-1 · sec-1, and prolonged active site residence times (t1/2, 5 to 46 min). The spectrum of inhibition includes Ambler class A ESBLs, class C cephalosporinases, and class A and D carbapenemases (KPC and OXA-48, respectively). Rescue of ceftibuten by VNRX-5236 (fixed at 4 µg/ml) in isogenic strains of Escherichia coli expressing class A, C, or D ß-lactamases demonstrated an expanded spectrum of activity relative to oral comparators, including investigational penems, sulopenem, and tebipenem. VNRX-5236 rescued ceftibuten activity in clinical isolates of Enterobacterales expressing ESBLs (MIC90, 0.25 µg/ml), KPCs (MIC90, 1 µg/ml), class C cephalosporinases (MIC90, 1 µg/ml), and OXA-48-type carbapenemases (MIC90, 1 µg/ml). Frequency of resistance studies demonstrated a low propensity for recovery of resistant variants at 4× the MIC of the ceftibuten/VNRX-5236 combination. In vivo, whereas ceftibuten alone was ineffective (50% effective dose [ED50], >128 mg/kg), ceftibuten/VNRX-7145 administered orally protected mice from lethal septicemia caused by Klebsiella pneumoniae producing KPC carbapenemase (ED50, 12.9 mg/kg). The data demonstrate potent, broad-spectrum rescue of ceftibuten activity by VNRX-5236 in clinical isolates of cephalosporin-resistant and carbapenem-resistant Enterobacterales.


Subject(s)
Cephalosporins , beta-Lactamase Inhibitors , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins , Carbapenems/pharmacology , Ceftibuten , Cephalosporins/pharmacology , Mice , Microbial Sensitivity Tests , Serine , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/genetics
5.
J Org Chem ; 85(3): 1365-1375, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31670958

ABSTRACT

C1-CBP-vancomycin (3) was examined alongside CBP-vancomycin for susceptibility to acquired resistance upon serial exposure against two vancomycin-resistant enterococci strains where its activity proved more durable and remarkably better than many current therapies. Combined with earlier studies, this observation confirmed an added mechanism of action was introduced by incorporation of the trimethylammonium cation and that C1-CBP-vancomycin exhibits activity against vancomycin-resistant organisms through two synergistic mechanisms of action, both independent of d-Ala-d-Ala/d-Lac binding. New insights into this added mechanism of action, induced cell membrane permeabilization, can be inferred from studies that show added exogenous lipoteichoic acid reduces antimicrobial activity, rescues bacteria cell growth inhibition, and blocks induced cell permeabilization properties of C1-CBP-vancomycin, suggesting a direct binding interaction with embedded teichoic acid is responsible for the added mechanism of action and enhanced antimicrobial activity. Further studies indicate that the trimethylammonium cation does not introduce new liabilities in common pharmacological properties of the analogue and established that 3 is well tolerated in mice, displays substantial PK improvements over both vancomycin and CBP-vancomycin, and exhibits in vivo efficacy against a challenging multidrug-resistant and vancomycin-resistant S. aureus strain that is representative of the resistant pathogens all fear will emerge in the general population.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Vancomycin , Animals , Anti-Bacterial Agents/pharmacology , Cations , Humans , Mice , Microbial Sensitivity Tests , Vancomycin/pharmacology , Vancomycin Resistance
6.
Article in English | MEDLINE | ID: mdl-30670434

ABSTRACT

Lantibiotics present an attractive scaffold for the development of novel antibiotics. We report here a novel lantibiotic for the treatment of Clostridium difficile infection. The lead compounds were selected from a library of over 700 single- and multiple-substitution variants of the lantibiotic mutacin 1140 (MU1140). The best performers in vitro and in vivo were further used to challenge Golden Syrian hamsters orally in a Golden Syrian hamster model of Clostridium difficile-associated disease (CDAD) in a dose-response format, resulting in the selection of OG716 as the lead compound. This lantibiotic was characterized by a 50% effective dose of 23.85 mg/kg of body weight/day (10.97 µmol/kg/day) in this model. Upon oral administration of the maximum feasible dose (≥1,918 mg/kg/day), no observable toxicities or side effects were noted, and no effect on intestinal motility was observed. Compartmentalization to the gastrointestinal tract was confirmed. MU1140-derived variants offer a large pipeline for the development of novel antibiotics for the treatment of several indications and are particularly attractive considering their novel mechanism of action. Based on the currently available data, OG716 has an acceptable profile for further development for the treatment of CDAD.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriocins/pharmacology , Clostridium Infections/drug therapy , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemistry , Bacteriocins/administration & dosage , Bacteriocins/adverse effects , Bacteriocins/chemistry , Biological Availability , Cecum/microbiology , Clostridium Infections/mortality , Colony Count, Microbial , Dose-Response Relationship, Drug , Female , Gastric Emptying/drug effects , Male , Maximum Tolerated Dose , Mesocricetus , Rats, Wistar
7.
Article in English | MEDLINE | ID: mdl-29038270

ABSTRACT

The recently approved combination of meropenem and vaborbactam (Vabomere) is highly active against Gram-negative pathogens, especially Klebsiella pneumoniae carbapenemase (KPC)-producing, carbapenem-resistant Enterobacteriaceae We evaluated the efficacy of meropenem-vaborbactam against three clinically relevant isolates in a murine pyelonephritis model. The data indicate that the combination of meropenem and vaborbactam significantly increased bacterial killing compared to that with the untreated controls. These data suggest that this combination may have utility in the treatment of complicated urinary tract infections due to KPC-producing, carbapenem-resistant Enterobacteriaceae.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Boronic Acids/therapeutic use , Carbapenem-Resistant Enterobacteriaceae/drug effects , Escherichia coli/drug effects , Klebsiella pneumoniae/drug effects , Meropenem/therapeutic use , Pyelonephritis/drug therapy , Urinary Tract Infections/drug therapy , beta-Lactamase Inhibitors/therapeutic use , Animals , Bacterial Proteins/metabolism , Carbapenem-Resistant Enterobacteriaceae/isolation & purification , Disease Models, Animal , Drug Combinations , Humans , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/metabolism , Mice , Microbial Sensitivity Tests , Pyelonephritis/microbiology , Urinary Tract Infections/microbiology , beta-Lactamases/metabolism
8.
Artif Organs ; 42(12): 1119-1124, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30370640

ABSTRACT

The existence of acquired von Willebrand syndrome (AVWS) in patients with continuous flow left ventricular assist devices (LVADs) is well documented and has been verified by numerous investigators. AVWS has not been observed to occur in pulsatile devices such as the SynCardia total artificial heart (TAH), the HeartMate XVE, and the Thoratec pulsatile ventricular assist device (PVAD) used as a single pump. AVWS can also occur in patients with aortic stenosis, ventricular septal defect, mitral stenosis, and patent ductus arteriosus. It has been experimentally verified that supraphysiologic shear stress that occurs under these conditions can cleave the von Willebrand molecule, but the critical magnitude of stress and duration is unclear. Limited experimental results demonstrate that shear stresses as low as 5 Pa (50 dyne/cm2 ) can cause cleavage. Stresses in current centrifugal pumps can be as high as two orders of magnitude greater than this value. Pulsatile LVADs have stresses almost two orders of magnitude less than continuous flow LVADs. In order to improve continuous flow LVADs, the challenge for designers is to first determine the magnitude and duration of stress that is causing AVWS and then, if possible, design a pump below these stresses.


Subject(s)
Heart-Assist Devices/adverse effects , von Willebrand Diseases/etiology , Humans , Pulsatile Flow
9.
Article in English | MEDLINE | ID: mdl-28167541

ABSTRACT

Based upon knowledge of the hydrolytic profile of major ß-lactamases found in Gram-negative bacteria, we tested the efficacy of the combination of ceftazidime-avibactam (CAZ-AVI) with aztreonam (ATM) against carbapenem-resistant enteric bacteria possessing metallo-ß-lactamases (MBLs). Disk diffusion and agar-based antimicrobial susceptibility testing were initially performed to determine the in vitro efficacy of a unique combination of CAZ-AVI and ATM against 21 representative Enterobacteriaceae isolates with a complex molecular background that included blaIMP, blaNDM, blaOXA-48, blaCTX-M, blaAmpC, and combinations thereof. Time-kill assays were conducted, and the in vivo efficacy of this combination was assessed in a murine neutropenic thigh infection model. By disk diffusion assay, all 21 isolates were resistant to CAZ-AVI alone, and 19/21 were resistant to ATM. The in vitro activity of CAZ-AVI in combination with ATM against diverse Enterobacteriaceae possessing MBLs was demonstrated in 17/21 isolates, where the zone of inhibition was ≥21 mm. All isolates demonstrated a reduction in CAZ-AVI agar dilution MICs with the addition of ATM. At 2 h, time-kill assays demonstrated a ≥4-log10-CFU decrease for all groups that had CAZ-AVI with ATM (8 µg/ml) added, compared to the group treated with CAZ-AVI alone. In the murine neutropenic thigh infection model, an almost 4-log10-CFU reduction was noted at 24 h for CAZ-AVI (32 mg/kg every 8 h [q8h]) plus ATM (32 mg/kg q8h) versus CAZ-AVI (32 mg/kg q8h) alone. The data presented herein require us to carefully consider this new therapeutic combination to treat infections caused by MBL-producing Enterobacteriaceae.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azabicyclo Compounds/pharmacology , Aztreonam/pharmacology , Ceftazidime/pharmacology , Enterobacteriaceae Infections/drug therapy , Enterobacteriaceae/drug effects , Animals , Colony Count, Microbial , Cyclophosphamide , Drug Administration Schedule , Drug Combinations , Drug Therapy, Combination , Enterobacteriaceae/enzymology , Enterobacteriaceae/genetics , Enterobacteriaceae/growth & development , Enterobacteriaceae Infections/microbiology , Female , Gene Expression , Humans , Klebsiella Infections/complications , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/growth & development , Mice , Microbial Sensitivity Tests , Neutropenia/chemically induced , Neutropenia/complications , Neutropenia/drug therapy , Neutropenia/microbiology , Plasmids/chemistry , Plasmids/metabolism , Soft Tissue Infections/complications , Soft Tissue Infections/drug therapy , Soft Tissue Infections/microbiology , Thigh , beta-Lactam Resistance/drug effects , beta-Lactam Resistance/genetics , beta-Lactamases/genetics , beta-Lactamases/metabolism
10.
Antimicrob Agents Chemother ; 59(1): 299-309, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348518

ABSTRACT

Alpha-toxin (AT) is a major virulence determinant in Staphylococcus aureus skin and soft tissue infection models. We previously demonstrated that prophylactic administration of 2A3, an AT-neutralizing monoclonal antibody (MAb), prevents S. aureus disease in a mouse dermonecrosis model by neutralizing AT-mediated tissue necrosis and immune evasion. In the present study, MEDI4893*, an affinity-optimized version of 2A3, was characterized for therapeutic activity in the dermonecrosis model as a single agent and in combination with two frontline antibiotics, vancomycin and linezolid. MEDI4893* postinfection therapy was found to exhibit a therapeutic treatment window similar to that for linezolid but longer than that for vancomycin. Additionally, when combined with either vancomycin or linezolid, MEDI4893* resulted in reduced tissue damage, increased neutrophil and macrophage infiltration and abscess formation, and accelerated healing relative to those with the antibiotic monotherapies. These data suggest that AT neutralization with a potent MAb holds promise for both prophylaxis and adjunctive therapy with antibiotics and may be a valuable addition to currently available options for the treatment of S. aureus skin and soft tissue infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Staphylococcal Skin Infections/drug therapy , Animals , Anti-Bacterial Agents/pharmacokinetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Broadly Neutralizing Antibodies , Disease Models, Animal , Drug Therapy, Combination , Female , Linezolid/pharmacokinetics , Linezolid/pharmacology , Mice, Inbred BALB C , Necrosis/drug therapy , Necrosis/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Vancomycin/pharmacokinetics , Vancomycin/pharmacology
11.
Antimicrob Agents Chemother ; 57(5): 2182-90, 2013 May.
Article in English | MEDLINE | ID: mdl-23459481

ABSTRACT

This study examines the alteration in Staphylococcus aureus gene expression following treatment with the type 2 fatty acid synthesis inhibitor AFN-1252. An Affymetrix array study showed that AFN-1252 rapidly increased the expression of fatty acid synthetic genes and repressed the expression of virulence genes controlled by the SaeRS 2-component regulator in exponentially growing cells. AFN-1252 did not alter virulence mRNA levels in a saeR deletion strain or in strain Newman expressing a constitutively active SaeS kinase. AFN-1252 caused a more pronounced increase in fabH mRNA levels in cells entering stationary phase, whereas the depression of virulence factor transcription was attenuated. The effect of AFN-1252 on gene expression in vivo was determined using a mouse subcutaneous granuloma infection model. AFN-1252 was therapeutically effective, and the exposure (area under the concentration-time curve from 0 to 48 h [AUC(0-48)]) of AFN-1252 in the pouch fluid was comparable to the plasma levels in orally dosed animals. The inhibition of fatty acid biosynthesis by AFN-1252 in the infected pouches was signified by the substantial and sustained increase in fabH mRNA levels in pouch-associated bacteria, whereas depression of virulence factor mRNA levels in the AFN-1252-treated pouch bacteria was not as evident as it was in exponentially growing cells in vitro. The trends in fabH and virulence factor gene expression in the animal were similar to those in slower-growing bacteria in vitro. These data indicate that the effects of AFN-1252 on virulence factor gene expression depend on the physiological state of the bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Benzofurans/pharmacology , Enoyl-(Acyl-Carrier Protein) Reductase (NADPH, B-Specific)/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Pyrones/pharmacology , Staphylococcus aureus/drug effects , Acetyltransferases/genetics , Acetyltransferases/metabolism , Animals , Anti-Bacterial Agents/pharmacokinetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzofurans/pharmacokinetics , Enoyl-(Acyl-Carrier Protein) Reductase (NADPH, B-Specific)/genetics , Enoyl-(Acyl-Carrier Protein) Reductase (NADPH, B-Specific)/metabolism , Enzyme Inhibitors/pharmacokinetics , Fatty Acids/metabolism , Gene Expression Profiling , Granuloma/drug therapy , Granuloma/microbiology , Lipid Metabolism/drug effects , Mice , Protein Kinases/genetics , Protein Kinases/metabolism , Pyrones/pharmacokinetics , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
12.
J Antimicrob Chemother ; 68(12): 2852-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23873647

ABSTRACT

OBJECTIVES: The purpose of this study was to conduct a pharmacokinetic and pharmacodynamic evaluation of high (320/1600 mg) and standard (160/800 mg) doses of trimethoprim/sulfamethoxazole and linezolid in outpatients with mild diabetic foot infections (DFIs). METHODS: Both viable skin/soft tissue from the infection site and serum were obtained at various times after antibiotic administration from 18 patients (6 per study group) being treated with linezolid, standard doses of trimethoprim/sulfamethoxazole or high doses of trimethoprim/sulfamethoxazole during a follow-up clinic visit. These samples were assayed for drug concentrations by liquid chromatography in tandem with mass spectrometry. Patient sera were also utilized in time-kill assays against two strains of Staphylococcus aureus and three strains of ß-haemolytic streptococci. RESULTS: The mean tissue/serum ratio for linezolid was 0.46 (range, 0.18-0.71). The mean tissue/serum ratio for trimethoprim was 1.2 (range, 0.3-4.5) for both standard and high doses, and 0.23 (range, 0.1-0.46) and 0.36 (range, 0.14-1.28) for standard and high doses of sulfamethoxazole, respectively. Linezolid exhibited inhibitory activity in time-kill assays against strains of S. aureus (0.45 ± 0.5 log10 cfu/mL) and ß-haemolytic streptococci (2.2 ± 0.6 log10 cfu/mL), while trimethoprim/sulfamethoxazole exhibited bactericidal (>3 log kill) activity against all of these isolates. These findings were consistent for each sampling time and for high as well as standard doses of trimethoprim/sulfamethoxazole. CONCLUSIONS: This pharmacokinetic/pharmacodynamic study found that trimethoprim/sulfamethoxazole exhibits good skin/soft tissue penetration in patients with DFIs as well as bactericidal activity in serum against strains of S. aureus and ß-haemolytic streptococci.


Subject(s)
Acetamides/pharmacokinetics , Anti-Bacterial Agents/pharmacokinetics , Bacterial Infections/drug therapy , Diabetic Foot/complications , Oxazolidinones/pharmacokinetics , Trimethoprim, Sulfamethoxazole Drug Combination/pharmacokinetics , Acetamides/administration & dosage , Acetamides/pharmacology , Aged , Aged, 80 and over , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Chromatography, Liquid , Female , Humans , Linezolid , Male , Middle Aged , Outpatients , Oxazolidinones/administration & dosage , Oxazolidinones/pharmacology , Serum/chemistry , Skin/chemistry , Staphylococcus aureus/drug effects , Streptococcus/drug effects , Tandem Mass Spectrometry , Time Factors , Trimethoprim, Sulfamethoxazole Drug Combination/administration & dosage , Trimethoprim, Sulfamethoxazole Drug Combination/pharmacology
13.
PLoS Pathog ; 7(2): e1001287, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347352

ABSTRACT

Methicillin-resistant Staphylococcus aureus is estimated to cause more U.S. deaths annually than HIV/AIDS. The emergence of hypervirulent and multidrug-resistant strains has further amplified public health concern and accentuated the need for new classes of antibiotics. RNA degradation is a required cellular process that could be exploited for novel antimicrobial drug development. However, such discovery efforts have been hindered because components of the Gram-positive RNA turnover machinery are incompletely defined. In the current study we found that the essential S. aureus protein, RnpA, catalyzes rRNA and mRNA digestion in vitro. Exploiting this activity, high through-put and secondary screening assays identified a small molecule inhibitor of RnpA-mediated in vitro RNA degradation. This agent was shown to limit cellular mRNA degradation and exhibited antimicrobial activity against predominant methicillin-resistant S. aureus (MRSA) lineages circulating throughout the U.S., vancomycin intermediate susceptible S. aureus (VISA), vancomycin resistant S. aureus (VRSA) and other Gram-positive bacterial pathogens with high RnpA amino acid conservation. We also found that this RnpA-inhibitor ameliorates disease in a systemic mouse infection model and has antimicrobial activity against biofilm-associated S. aureus. Taken together, these findings indicate that RnpA, either alone, as a component of the RNase P holoenzyme, and/or as a member of a more elaborate complex, may play a role in S. aureus RNA degradation and provide proof of principle for RNA catabolism-based antimicrobial therapy.


Subject(s)
Anti-Infective Agents/pharmacology , RNA Processing, Post-Transcriptional/drug effects , RNA, Messenger/metabolism , Ribonuclease P/antagonists & inhibitors , Staphylococcal Infections/prevention & control , Staphylococcus aureus , Animals , Anti-Infective Agents/therapeutic use , Female , Hep G2 Cells , Humans , Mice , Models, Biological , Ribonuclease P/physiology , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Staphylococcal Infections/genetics , Staphylococcal Infections/pathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Vancomycin/pharmacology , Vancomycin/therapeutic use , Virulence/drug effects , Virulence/genetics
14.
Toxicol Pathol ; 41(3): 519-27, 2013.
Article in English | MEDLINE | ID: mdl-22700648

ABSTRACT

Repeated bolus intravenous (IV) administration of large doses of beta-lactams and aminoglycosides has previously been associated with the development of eosinophilic and occlusive arterial lesions limited to the lungs in calves. Reviewing 13 years worth of records from left ventricular assist device implantation studies, morphologically identical segmental arterial lesions were present in 32 of the 56 calves receiving IV antibiotics, affecting lungs (6/50), kidneys (12/56), or lungs and kidneys (14/50). In 16 of these calves, renal arterial lesions spatially colocalized with renal cortical infarctions. Lesions were noted in additional abdominal organs in 4 of the 50 calves and were exclusively present in the liver in a single calf. Similar arterial lesions were also noted in the lungs (3/4), kidneys (1/4), liver (1/4), and spleen (1/4) of unimplanted calves receiving similar IV antibiotic regimens for bacterial infections. Lesions were observed with therapeutic IV doses of cephalosporins with or without aminoglycosides over shorter intervals than previously implicated. Lesions were significantly associated with increased peripheral eosinophil counts and mildly elevated, not reduced, arterial pulse pressures. This report documents the features of an idiosyncratic drug reaction with features strongly suggestive of an acute type-I hypersensitivity in this species.


Subject(s)
Anti-Bacterial Agents/adverse effects , Arteritis/chemically induced , Eosinophilia/chemically induced , Heart-Assist Devices , Animals , Arteritis/etiology , Arteritis/physiopathology , Blood Pressure , Cattle , Clinical Trials as Topic , Eosinophilia/etiology , Eosinophilia/physiopathology , Eosinophils/cytology , Eosinophils/drug effects , Infarction/pathology , Kidney/blood supply , Kidney/pathology , Kidney Cortex/blood supply , Kidney Cortex/pathology , Leukocyte Count , Lung/blood supply , Lung/pathology , Male , Pulmonary Artery/pathology , beta-Lactams/adverse effects
15.
J Biomater Appl ; 38(2): 302-310, 2023 08.
Article in English | MEDLINE | ID: mdl-37470381

ABSTRACT

Segmented polyurethane (PU) block copolymers are widely used in implantable cardiovascular medical devices due to their good biocompatibility and excellent mechanical properties. More specifically, PU Biospan MS/0.4 was used in ventricular assist devices over the past decades. However, this product is being discontinued and it has become necessary to find an alternative PU biomaterial for application in cardiovascular devices. One important criterion for assessing cardiac biomaterials is blood compatibility. In this study, we characterized the surface properties of four medical-grade PU biomaterials: Biospan MS/0.4, BioSpan S, BioSpan 2F, and CarboSil 20 80A, including surface chemistry, topography, microphase separation structure and wettability, and then measured the blood plasma coagulation responses using bovine and human blood plasma. Results showed that BioSpan 2F contains high amounts of fluorine and has the lowest surface free energy while the other materials have surfaces with silicone present. An in vitro coagulation assay shows that these materials demonstrated improved blood coagulation responses compared to the polystyrene control and there were no significant differences in coagulation time among all PU biomaterials. The chromogenic assay showed all PU materials led to low FXII contact activation, and there were no significant differences in FXII contact activation, consistent with plasma coagulation responses.


Subject(s)
Polymers , Polyurethanes , Animals , Cattle , Humans , Polymers/chemistry , Polyurethanes/chemistry , Blood Coagulation , Biocompatible Materials/chemistry , Plasma/chemistry , Surface Properties
16.
ASAIO J ; 69(12): 1065-1073, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37549654

ABSTRACT

Congenital heart disease affects approximately 40,000 infants annually in the United States with 25% requiring invasive treatment. Due to limited number of donor hearts and treatment options available for children, pediatric ventricular assist devices (PVADs) are used as a bridge to transplant. The 12cc pneumatic Penn State PVAD is optimized to prevent platelet adhesion and thrombus formation at patient nominal conditions; however, children demonstrate variable blood hematocrit and elevated heart rates. Therefore, with pediatric patients exhibiting greater variability, particle image velocimetry is used to evaluate the PVAD with three non-Newtonian hematocrit blood analogs (20%, 40%, and 60%) and at two beat rates (75 and 120 bpm) to understand the device's performance. The flow fields demonstrate a strong inlet jet that transitions to a solid body rotation during diastole. During systole, the rotation dissipates and reorganizes into an outlet jet. This flow field is consistent across all hematocrits and beat rates but at a higher velocity magnitude during 120 bpm. There are also minor differences in flow field timing and surface washing due to hematocrit. Therefore, despite patient differences in hematocrit or required pumping output, thorough surface washing can be achieved in the PVAD by altering operating conditions, thus reducing platelet adhesion potential.


Subject(s)
Heart Transplantation , Heart-Assist Devices , Infant , Child , Humans , Hematocrit , Pulsatile Flow , Tissue Donors , Blood Flow Velocity
17.
ASAIO J ; 69(5): 467-474, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36399789

ABSTRACT

The loss of high molecular weight multimers (HMWM) of von Willebrand factor (vWF) in aortic stenosis (AS) and continuous-flow left ventricular assist devices (cf-LVADs) is believed to be associated with high turbulent blood shear. The objective of this study is to understand the degradation mechanism of HMWM in terms of exposure time (kinetic) and flow regime (dynamics) within clinically relevant pathophysiologic conditions. A custom high-shear rotary device capable of creating fully controlled exposure times and flows was used. The system was set so that human platelet-poor plasma flowed through at 1.75 ml/sec, 0.76 ml/sec, or 0.38 ml/sec resulting in the exposure time ( texp ) of 22, 50, or 100 ms, respectively. The flow was characterized by the Reynolds number (Re). The device was run under laminar (Re = 1,500), transitional (Re = 3,000; Re = 3,500), and turbulent (Re = 4,500) conditions at a given texp followed by multimer analysis. No degradation was observed at laminar flow at all given texp . Degradation of HMWM at a given texp increases with the Re. Re ( p < 0.0001) and texp ( p = 0.0034) are significant factors in the degradation of HMWM. Interaction between Re and texp , however, is not always significant ( p = 0.73).


Subject(s)
Heart-Assist Devices , von Willebrand Diseases , Humans , von Willebrand Factor/metabolism , Kinetics , Molecular Weight
18.
Cell Rep Med ; 4(11): 101253, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37918405

ABSTRACT

Colonization of the gut and airways by pathogenic bacteria can lead to local tissue destruction and life-threatening systemic infections, especially in immunologically compromised individuals. Here, we describe an mRNA-based platform enabling delivery of pathogen-specific immunoglobulin A (IgA) monoclonal antibodies into mucosal secretions. The platform consists of synthetic mRNA encoding IgA heavy, light, and joining (J) chains, packaged in lipid nanoparticles (LNPs) that express glycosylated, dimeric IgA with functional activity in vitro and in vivo. Importantly, mRNA-derived IgA had a significantly greater serum half-life and a more native glycosylation profile in mice than did a recombinantly produced IgA. Expression of an mRNA encoded Salmonella-specific IgA in mice resulted in intestinal localization and limited Peyer's patch invasion. The same mRNA-LNP technology was used to express a Pseudomonas-specific IgA that protected from a lung challenge. Leveraging the mRNA antibody technology as a means to intercept bacterial pathogens at mucosal surfaces opens up avenues for prophylactic and therapeutic interventions.


Subject(s)
Mucous Membrane , Peyer's Patches , Mice , Animals , Immunoglobulin A , Antibodies, Monoclonal
19.
Front Cell Infect Microbiol ; 13: 1297281, 2023.
Article in English | MEDLINE | ID: mdl-38149013

ABSTRACT

Background: New drugs targeting antimicrobial resistant pathogens, including Pseudomonas aeruginosa, have been challenging to evaluate in clinical trials, particularly for the non-ventilated hospital-acquired pneumonia and ventilator-associated pneumonia indications. Development of new antibacterial drugs is facilitated by preclinical animal models that could predict clinical efficacy in patients with these infections. Methods: We report here an FDA-funded study to develop a rabbit model of non-ventilated pneumonia with Pseudomonas aeruginosa by determining the extent to which the natural history of animal disease reproduced human pathophysiology and conducting validation studies to evaluate whether humanized dosing regimens of two antibiotics, meropenem and tobramycin, can halt or reverse disease progression. Results: In a rabbit model of non-ventilated pneumonia, endobronchial challenge with live P. aeruginosa strain 6206, but not with UV-killed Pa6206, caused acute respiratory distress syndrome, as evidenced by acute lung inflammation, pulmonary edema, hemorrhage, severe hypoxemia, hyperlactatemia, neutropenia, thrombocytopenia, and hypoglycemia, which preceded respiratory failure and death. Pa6206 increased >100-fold in the lungs and then disseminated from there to infect distal organs, including spleen and kidneys. At 5 h post-infection, 67% of Pa6206-challenged rabbits had PaO2 <60 mmHg, corresponding to a clinical cut-off when oxygen therapy would be required. When administered at 5 h post-infection, humanized dosing regimens of tobramycin and meropenem reduced mortality to 17-33%, compared to 100% for saline-treated rabbits (P<0.001 by log-rank tests). For meropenem which exhibits time-dependent bactericidal activity, rabbits treated with a humanized meropenem dosing regimen of 80 mg/kg q2h for 24 h achieved 100% T>MIC, resulting in 75% microbiological clearance rate of Pa6206 from the lungs. For tobramycin which exhibits concentration-dependent killing, rabbits treated with a humanized tobramycin dosing regimen of 8 mg/kg q8h for 24 h achieved Cmax/MIC of 9.8 ± 1.4 at 60 min post-dose, resulting in 50% lung microbiological clearance rate. In contrast, rabbits treated with a single tobramycin dose of 2.5 mg/kg had Cmax/MIC of 7.8 ± 0.8 and 8% (1/12) microbiological clearance rate, indicating that this rabbit model can detect dose-response effects. Conclusion: The rabbit model may be used to help predict clinical efficacy of new antibacterial drugs for the treatment of non-ventilated P. aeruginosa pneumonia.


Subject(s)
Pneumonia , Pseudomonas Infections , Humans , Animals , Rabbits , Meropenem/therapeutic use , Pseudomonas aeruginosa , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Tobramycin/pharmacology , Tobramycin/therapeutic use , Pneumonia/drug therapy , Drug Development
20.
Artif Organs ; 36(9): 820-4, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22428807

ABSTRACT

Evaluation of thrombogenicity is a critical component in the preclinical testing and development of blood pumps. Left ventricular assist devices (LVADs), because of their device routing, can produce thromboembolic showers to the kidney resulting in renal cortical ischemia or infarctions. Although postmortem evaluation of renal pathology can confirm ischemic events and infarctions, there are no validated and highly sensitive real-time measures of renal ischemia in the preclinical models. In this article, we report the evaluation of urinary biomarkers of ischemic tubular damage in a lamb preclinical LVAD model. We found that urinary excretion of glutathione-S-transferase-π, heat shock protein 1B, and hepatitis A virus cellular receptor 1 homologue precursor (HAVCR1/kidney injury molecule 1) were upregulated in toxic ischemic renal injury as well as in the immediate postoperative period in an LVAD-implanted lamb. These markers were consistent with both gross and histologic pathology, and proved far more sensitive for renal injury than serum blood urea nitrogen or creatinine concentrations.


Subject(s)
Heart-Assist Devices/adverse effects , Ischemia/urine , Kidney Diseases/etiology , Kidney Diseases/urine , Kidney/blood supply , Kidney/pathology , Animals , Glutathione S-Transferase pi/urine , HSP70 Heat-Shock Proteins/urine , Ischemia/etiology , Ischemia/pathology , Kidney Diseases/pathology , Receptors, Virus/metabolism , Sheep, Domestic
SELECTION OF CITATIONS
SEARCH DETAIL