@article{nie_valdes-pena_frohock_smits_daiker_gilbertie_v. schnabel_pierce_2024, title={Expanded library of novel 2,3-pyrrolidinedione analogues exhibit anti-biofilm activity}, volume={99}, ISSN={["1464-3405"]}, url={https://doi.org/10.1016/j.bmcl.2024.129609}, DOI={10.1016/j.bmcl.2024.129609}, abstractNote={Herein we report a new library of 2,3-pyrrolidinedione analogues that expands on our previous report on the antimicrobial studies of this heterocyclic scaffold. The novel 2,3-pyrrolidinediones reported herein have been evaluated against S. aureus and methicillin-resistant S. aureus (MRSA) biofilms, and this work constitutes our first report on the antibiofilm properties of this class of compounds. The antibiofilm activity of these 2,3-pyrrolidinediones has been assessed through minimum biofilm eradication concentration (MBEC) and minimum biofilm inhibition concentration (MBIC) assays. The compounds displayed antibiofilm properties and represent intriguing scaffolds for further optimization and development.}, journal={BIOORGANIC & MEDICINAL CHEMISTRY LETTERS}, author={Nie, Minhua and Valdes-Pena, M. Alejandro and Frohock, Bram H. and Smits, Emma and Daiker, Jennifer C. and Gilbertie, Jessica M. and V. Schnabel, Lauren and Pierce, Joshua G.}, year={2024}, month={Feb} } @article{pezzanite_chow_dow_goodrich_gilbertie_schnabel_2023, title={Antimicrobial Properties of Equine Stromal Cells and Platelets and Future Directions}, volume={39}, ISSN={["1558-4224"]}, DOI={10.1016/j.cveq.2023.06.005}, abstractNote={Increasing antimicrobial resistance in veterinary practice has driven the investigation of novel therapeutic strategies including regenerative and biologic therapies to treat bacterial infection. Integration of biological approaches such as platelet lysate and mesenchymal stromal cell (MSC) therapy may represent adjunctive treatment strategies for bacterial infections that minimize systemic side effects and local tissue toxicity associated with traditional antibiotics and that are not subject to antibiotic resistance. In this review, we will discuss mechanisms by which biological therapies exert antimicrobial effects, as well as potential applications and challenges in clinical implementation in equine practice.}, number={3}, journal={VETERINARY CLINICS OF NORTH AMERICA-EQUINE PRACTICE}, author={Pezzanite, Lynn M. and Chow, Lyndah and Dow, Steven W. and Goodrich, Laurie R. and Gilbertie, Jessica M. and Schnabel, Lauren V}, year={2023}, month={Dec}, pages={565–578} } @article{gilbertie_schaer_engiles_seiler_deddens_schubert_jacob_stefanovski_ruthel_hickok_et al._2022, title={A Platelet-Rich Plasma-Derived Biologic Clears Staphylococcus aureus Biofilms While Mitigating Cartilage Degeneration and Joint Inflammation in a Clinically Relevant Large Animal Infectious Arthritis Model}, volume={12}, ISSN={2235-2988}, url={http://dx.doi.org/10.3389/fcimb.2022.895022}, DOI={10.3389/fcimb.2022.895022}, abstractNote={The leading cause of treatment failure in Staphylococcus aureus infections is the development of biofilms. Biofilms are highly tolerant to conventional antibiotics which were developed against planktonic cells. Consequently, there is a lack of antibiofilm agents in the antibiotic development pipeline. To address this problem, we developed a platelet-rich plasma (PRP)-derived biologic, termed BIO-PLY (for the BIOactive fraction of Platelet-rich plasma LYsate) which has potent in vitro bactericidal activity against S. aureus synovial fluid free-floating biofilm aggregates. Additional in vitro studies using equine synoviocytes and chondrocytes showed that BIO-PLY protected these cells of the joint from inflammation. The goal of this study was to test BIO-PLY for in vivo efficacy using an equine model of infectious arthritis. We found that horses experimentally infected with S. aureus and subsequently treated with BIO-PLY combined with the antibiotic amikacin (AMK) had decreased bacterial concentrations within both synovial fluid and synovial tissue and exhibited lower systemic and local inflammatory scores compared to horses treated with AMK alone. Most importantly, AMK+BIO-PLY treatment reduced the loss of infection-associated cartilage proteoglycan content in articular cartilage and decreased synovial tissue fibrosis and inflammation. Our results demonstrate the in vivo efficacy of AMK+BIO-PLY and represents a new approach to restore and potentiate antimicrobial activity against synovial fluid biofilms.}, journal={Frontiers in Cellular and Infection Microbiology}, publisher={Frontiers Media SA}, author={Gilbertie, Jessica M. and Schaer, Thomas P. and Engiles, Julie B. and Seiler, Gabriela S. and Deddens, Bennett L. and Schubert, Alicia G. and Jacob, Megan E. and Stefanovski, Darko and Ruthel, Gordon and Hickok, Noreen J. and et al.}, year={2022}, month={May} } @article{frias-de-diego_gilbertie_scholle_dejarnette_crisci_2022, title={Effect of BIO-PLYTM, a Platelet-Rich Plasma Derived Biologic on PRRSV-2-Infected Macrophages}, volume={14}, ISSN={1999-4915}, url={http://dx.doi.org/10.3390/v14122666}, DOI={10.3390/v14122666}, abstractNote={Porcine Reproductive and Respiratory Syndrome (PRRS) is the one of the most devastating diseases impacting the swine industry worldwide. Control and prevention methods rely on biosafety measures and vaccination. As an RNA virus with a high rate of mutation, vaccines are only partially effective against circulating and newly emerging strains. To reduce the burden of this disease, research on alternative control methods is needed. Here, we assess the in vitro antiviral effect of a novel platelet-rich plasma-derived biologic termed BIO-PLYTM (for the BIOactive fraction of Platelet-rich plasma LYsate) from both swine and equine origin. Our results show that BIO-PLYTM significantly reduces the amount of PRRSV viral load determined by RT-qPCR and the number of infectious viral particles measured by TCID50 in infected porcine alveolar and parenchymal macrophages. This study also showed limited toxicity of BIO-PLYTM in vitro and aspects of its immunomodulatory capacity evaluating the regulation of reactive oxygen species and cytokines production in infected cells. Finally, this study presents promising data on the effect of BIO-PLYTM on other RNA viruses such as human A influenza viruses and coronavirus.}, number={12}, journal={Viruses}, publisher={MDPI AG}, author={Frias-De-Diego, Alba and Gilbertie, Jessica M. and Scholle, Frank and Dejarnette, Sarah and Crisci, Elisa}, year={2022}, month={Nov}, pages={2666} } @article{koch_berglund_messenger_gilbertie_ellis_schnabel_2022, title={Interleukin-1 beta in tendon injury enhances reparative gene and protein expression in mesenchymal stem cells}, volume={9}, ISSN={["2297-1769"]}, DOI={10.3389/fvets.2022.963759}, abstractNote={Tendon injury in the horse carries a high morbidity and monetary burden. Despite appropriate therapy, reinjury is estimated to occur in 50–65% of cases. Although intralesional mesenchymal stem cell (MSC) therapy has improved tissue architecture and reinjury rates, the mechanisms by which they promote repair are still being investigated. Additionally, reevaluating our application of MSCs in tendon injury is necessary given recent evidence that suggests MSCs exposed to inflammation (deemed MSC licensing) have an enhanced reparative effect. However, applying MSC therapy in this context is limited by the inadequate quantification of the temporal cytokine profile in tendon injury, which hinders our ability to administer MSCs into an environment that could potentiate their effect. Therefore, the objectives of this study were to define the temporal cytokine microenvironment in a surgically induced model of equine tendon injury using ultrafiltration probes and subsequently evaluate changes in MSC gene and protein expression following in vitro inflammatory licensing with cytokines of similar concentration as identified in vivo. In our in vivo surgically induced tendon injury model, IL-1β and IL-6 were the predominant pro-inflammatory cytokines present in tendon ultrafiltrate where a discrete peak in cytokine concentration occurred within 48 h following injury. Thereafter, MSCs were licensed in vitro with IL-1β and IL-6 at a concentration identified from the in vivo study; however, only IL-1β induced upregulation of multiple genes beneficial to tendon healing as identified by RNA-sequencing. Specifically, vascular development, ECM synthesis and remodeling, chemokine and growth factor function alteration, and immunomodulation and tissue reparative genes were significantly upregulated. A significant increase in the protein expression of IL-6, VEGF, and PGE2 was confirmed in IL-1β-licensed MSCs compared to naïve MSCs. This study improves our knowledge of the temporal tendon cytokine microenvironment following injury, which could be beneficial for the development and determining optimal timing of administration of regenerative therapies. Furthermore, these data support the need to further study the benefit of MSCs administered within the inflamed tendon microenvironment or exogenously licensed with IL-1β in vitro prior to treatment as licensed MSCs could enhance their therapeutic benefit in the healing tendon.}, journal={FRONTIERS IN VETERINARY SCIENCE}, author={Koch, Drew W. W. and Berglund, Alix K. K. and Messenger, Kristen M. M. and Gilbertie, Jessica M. M. and Ellis, Ilene M. M. and Schnabel, Lauren V. V.}, year={2022}, month={Aug} } @article{gilbertie_ulloa_daiker_nguyen_smelter_rose_geriak_schnabel_nizet_sakoulas_2022, title={Potent Activity of Ertapenem Plus Cefazolin Within Staphylococcal Biofilms: A Contributing Factor in the Treatment of Methicillin-Susceptible Staphylococcus aureus Endocarditis}, volume={9}, ISSN={["2328-8957"]}, DOI={10.1093/ofid/ofac159}, abstractNote={Abstract}, number={5}, journal={OPEN FORUM INFECTIOUS DISEASES}, author={Gilbertie, Jessica and Ulloa, Erlinda R. and Daiker, Jennifer C. and Nguyen, Khanh and Smelter, Dan and Rose, Warren and Geriak, Matthew and Schnabel, Lauren V and Nizet, Victor and Sakoulas, George}, year={2022}, month={May} } @article{prasad_negron_du_mullins_palumbo_gilbertie_hook_grover_pawlinski_mackman_et al._2021, title={Host fibrinogen drives antimicrobial function in Staphylococcus aureus peritonitis through bacterial-mediated prothrombin activation}, volume={118}, ISSN={["1091-6490"]}, DOI={10.1073/pnas.2009837118}, abstractNote={Significance}, number={1}, journal={PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA}, author={Prasad, Joni M. and Negron, Oscar and Du, Xinli and Mullins, Eric S. and Palumbo, Joseph S. and Gilbertie, Jessica M. and Hook, Magnus and Grover, Steven P. and Pawlinski, Rafal and Mackman, Nigel and et al.}, year={2021}, month={Jan} } @article{copp_flanders_gagliardi_gilbertie_sessions_chubinskaya_loeser_schnabel_diekman_2021, title={The combination of mitogenic stimulation and DNA damage induces chondrocyte senescence}, volume={29}, ISSN={["1522-9653"]}, DOI={10.1016/j.joca.2020.11.004}, abstractNote={Cellular senescence is a phenotypic state characterized by stable cell-cycle arrest, enhanced lysosomal activity, and the secretion of inflammatory molecules and matrix degrading enzymes. Senescence has been implicated in osteoarthritis (OA) pathophysiology; however, the mechanisms that drive senescence induction in cartilage and other joint tissues are unknown. While numerous physiological signals are capable of initiating senescence, one emerging theme is that damaged cells convert to senescence in response to sustained mitogenic stimulation. The goal of this study was to develop an in vitro articular cartilage explant model to investigate the mechanisms of senescence induction.This study utilized healthy cartilage derived from cadaveric equine stifles and human ankles. Explants were irradiated to initiate DNA damage, and mitogenic stimulation was provided through serum-containing medium and treatment with transforming growth factor β1 and basic fibroblastic growth factor. Readouts of senescence were a quantitative flow cytometry assay to detect senescence-associated β galactosidase activity (SA-β-gal), immunofluorescence for p16 and γH2AX, and qPCR for the expression of inflammatory genes.Human cartilage explants required both irradiation and mitogenic stimulation to induce senescence as compared to baseline control conditions (7.16% vs 2.34% SA-β-gal high, p = 0.0007). These conditions also resulted in chondrocyte clusters within explants, a persistent DNA damage response, increased p16, and gene expression changes.Treatment of cartilage explants with mitogenic stimuli in the context of cellular damage reliably induces high levels of SA-β-gal activity and other senescence markers, which provides a physiologically relevant model system to investigate the mechanisms of senescence induction.}, number={3}, journal={OSTEOARTHRITIS AND CARTILAGE}, author={Copp, M. E. and Flanders, M. C. and Gagliardi, R. and Gilbertie, J. M. and Sessions, G. A. and Chubinskaya, S. and Loeser, R. F. and Schnabel, L. and Diekman, B. O.}, year={2021}, month={Mar}, pages={402–412} } @article{gilbertie_levent_norman_vinasco_scott_jacob_2020, title={Comprehensive phenotypic and genotypic characterization and comparison of virulence, biofilm, and antimicrobial resistance in urinary Escherichia coli isolated from canines}, volume={249}, ISSN={["1873-2542"]}, DOI={10.1016/j.vetmic.2020.108822}, abstractNote={Urinary tract infections (UTIs) affect nearly half of women and an estimated 14 % of the canine companion animal population at least once in their lifetime. As with humans, Escherichia coli is the most commonly isolated bacteria from canine UTIs and infections are dominated by specific phylogenetic groups with notable virulence attributes. In this study, we evaluated uropathogenic E. coli (UPEC) (n = 69) isolated from canine UTIs phenotypically and genotypically for virulence factors, biofilm formation and antimicrobial resistance profiles. Biofilm formation in UPEC strains was positively associated with common virulence factors including papG (p = 0.006), fimH (p < 0.0001), sfaS (p = 0.004), focA (p = 0.004), cnf-1 (p = 0.009) and hlyA (p = 0.006). There was a negative association between biofilm formation and phenotypic antimicrobial resistance for ampicillin (p < 0.0004), ciprofloxacin (p < 0.0001), and trimethoprim-sulfamethoxazole (p < 0.02), as well as multidrug resistance (isolates resistant to ≥ 3 classes of antimicrobials) (p < 0.0002), and the presence of extended spectrum beta-lactamase (ESBL)-producing genes (p < 0.05). In conclusion, UPECs isolated from clinical cases of canine UTIs show a broad negative association between antimicrobial resistance and biofilm formation, and this observation is supported both by phenotypic and genotypic endpoints. As the biofilm formation may result in antimicrobial tolerance, this could be a secondary evasive tactic of UPEC lacking traditional antimicrobial resistance traits. This observation is important for veterinary practitioners to consider when treating puzzling chronic intractable and/or recurrent cases of UTI that appear to be susceptible to antimicrobial therapy via traditional antimicrobial susceptibility testing (AST) methods.}, journal={VETERINARY MICROBIOLOGY}, author={Gilbertie, Jessica M. and Levent, Gizem and Norman, Keri N. and Vinasco, Javier and Scott, H. Morgan and Jacob, Megan E.}, year={2020}, month={Oct} } @article{gilbertie_schaer_schubert_jacob_menegatti_lavoie_schnabel_2020, title={Platelet-rich plasma lysate displays antibiofilm properties and restores antimicrobial activity against synovial fluid biofilms in vitro}, volume={38}, ISSN={["1554-527X"]}, DOI={10.1002/jor.24584}, abstractNote={Abstract}, number={6}, journal={JOURNAL OF ORTHOPAEDIC RESEARCH}, author={Gilbertie, Jessica M. and Schaer, Thomas P. and Schubert, Alicia G. and Jacob, Megan E. and Menegatti, Stefano and Lavoie, R. Ashton and Schnabel, Lauren V}, year={2020}, month={Jun}, pages={1365–1374} } @article{frohock_gilbertie_daiker_schnabel_pierce_2019, title={5-Benzylidene-4-Oxazolidinones Are Synergistic with Antibiotics for the Treatment of Staphylococcus aureus Biofilms}, volume={12}, ISBN={1439-7633}, url={https://doi.org/10.1002/cbic.201900633}, DOI={10.1002/cbic.201900633}, abstractNote={Abstract}, journal={CHEMBIOCHEM}, publisher={Wiley}, author={Frohock, Bram H. and Gilbertie, Jessica M. and Daiker, Jennifer C. and Schnabel, Lauren V and Pierce, Joshua G.}, year={2019} }