@article{conley_till_berglund_jones_sheats_2023, title={A myristoylated alanine-rich C-kinase substrate (MARCKS) inhibitor peptide attenuates neutrophil outside-in & beta;(2)-integrin activation and signaling}, volume={17}, ISSN={["1933-6926"]}, url={https://doi.org/10.1080/19336918.2023.2233204}, DOI={10.1080/19336918.2023.2233204}, abstractNote={ABSTRACT MARCKS is an actin and PIP2-binding protein that plays an essential role in neutrophil migration and adhesion; however, the molecular details regarding MARCKS function in these processes remains unclear. Neutrophil adhesion and migration also require the cell surface receptors β2-integrins. We hypothesized that MARCKS inhibition would alter neutrophil β2-integrin activation and signaling. We utilized a MARCKS-targeting peptide to inhibit MARCKS in inside-out and outside-in β2-integrin activation in neutrophils. MANS-mediated MARCKS inhibition had no significant effect on inside-out β2-integrin activation. MANS treatment significantly attenuated ICAM-1/Mn2+-stimulated static adhesion, cell spreading and β2-integrin clustering, suggesting a role for MARCKS function in outside-in β2-integrin activation. Additional work is needed to better understand the molecular mechanisms of MARCKS role in outside-in β2-integrin activation and signaling.}, number={1}, journal={CELL ADHESION & MIGRATION}, author={Conley, Haleigh and Till, Rebecca L. and Berglund, Alix K. and Jones, Samuel L. and Sheats, M. Katie}, year={2023}, month={Dec}, pages={1–16} } @article{smith_berglund_robertson_schnabel_mcmullen_gilger_oh_2023, title={Effect of gentamicin on CD3+ T‐lymphocyte proliferation for treatment of equine recurrent uveitis: An in vitro study}, volume={26}, ISSN={1463-5216 1463-5224}, url={http://dx.doi.org/10.1111/vop.13098}, DOI={10.1111/vop.13098}, abstractNote={Abstract}, number={4}, journal={Veterinary Ophthalmology}, publisher={Wiley}, author={Smith, Hannah L. and Berglund, Alix K. and Robertson, James B. and Schnabel, Lauren V. and McMullen, Richard J., Jr and Gilger, Brian C. and Oh, Annie}, year={2023}, month={Apr}, pages={347–354} } @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{koch_schnabel_ellis_bates_berglund_2022, title={TGF-beta 2 enhances expression of equine bone marrow-derived mesenchymal stem cell paracrine factors with known associations to tendon healing}, volume={13}, ISSN={["1757-6512"]}, DOI={10.1186/s13287-022-03172-9}, abstractNote={Abstract}, number={1}, journal={STEM CELL RESEARCH & THERAPY}, author={Koch, Drew W. and Schnabel, Lauren V and Ellis, Ilene M. and Bates, Rowan E. and Berglund, Alix K.}, year={2022}, month={Sep} } @article{cameron_even_linardi_berglund_schnabel_engiles_ortved_2021, title={Adeno-Associated Virus-Mediated Overexpression of Interleukin-10 Affects the Immunomodulatory Properties of Equine Bone Marrow-Derived Mesenchymal Stem Cells}, volume={32}, ISSN={["1557-7422"]}, DOI={10.1089/hum.2020.319}, abstractNote={Joint injury can cause posttraumatic inflammation, which if severe enough can lead to posttraumatic osteoarthritis (PTOA), a progressive and debilitating condition. Posttraumatic inflammation is characterized by an influx of T lymphocytes and upregulation of inflammatory cytokines and degradative enzymes by activated chondrocytes and synoviocytes. Intra-articular bone marrow-derived mesenchymal stem cell (BM-MSC) injection for the treatment of OA has been of interest due to the immunomodulatory properties of these cells. Interleukin-10, a potent immunomodulatory cytokine, has also been investigated as an OA therapeutic. Therefore, the objective of this study was to evaluate the combinatorial effects of BM-MSCs and IL-10 in OA using a gene therapy approach. We hypothesized that BM-MSCs overexpressing IL-10 would have superior immunomodulatory effects leading to increased suppression of T-cell proliferation and decreased production of pro-inflammatory cytokines providing protection of the extra-cellular matrix (ECM) in a stimulated, co-culture OA model. Treatment groups included: untransduced BM-MSC, AAV-IL10 transduced BM-MSC, and AAV-null transduced BM-MSC, which were unstimulated or stimulated with IL-1β/TNF-α. T-cell proliferation was significantly decreased by the presence of BM-MSCs especially when these BM-MSCs were AAV transduced. There was no significant difference in T-cell suppression when cells were cultured with AAV-IL10 transduced or AAV-null transduced BM-MSCs. AAV transduction itself was associated with decreased synthesis of IL-1β, IL-6 and TNF-α. Expression of IL-1β and MMP13 was downregulated in AAV-transduced BM-MSCs and MMP13 expression was downregulated in cartilage explants co-cultured with AAV-transduced BM-MSCs. Despite mitigation of some pro-inflammatory cascades, rescue of ECM loss, as determined by glycosaminoglycan quantification and histological evaluation, did not occur in either AAV-IL10 transduced or AAV-null transduced co-cultures. Although IL-10 overexpression may enhance BM-MSC mediated T-cell suppression, we did not observe significant modulation of inflammation-driven cartilage degradation in cultures containing AAV-IL10 transduced BM-MSCs. AAV transduction itself does appear to affect paracrine signaling by BM-MSCs which warrants further investigation.}, number={17-18}, journal={HUMAN GENE THERAPY}, author={Cameron, Ashley D. and Even, Kayla M. and Linardi, Renata L. and Berglund, Alix K. and Schnabel, Lauren V. and Engiles, Julie B. and Ortved, Kyla F.}, year={2021}, month={Sep}, pages={907–918} } @article{rowland_miller_berglund_schnabel_levine_antczak_watts_2021, title={Cross-matching of allogeneic mesenchymal stromal cells eliminates recipient immune targeting}, volume={10}, ISSN={["2157-6580"]}, DOI={10.1002/sctm.20-0435}, abstractNote={Abstract}, number={5}, journal={STEM CELLS TRANSLATIONAL MEDICINE}, author={Rowland, Aileen L. and Miller, Donald and Berglund, Alix and Schnabel, Lauren V and Levine, Gwendolyn J. and Antczak, Douglas F. and Watts, Ashlee E.}, year={2021}, month={May}, pages={694–710} } @article{connard_linardi_even_berglund_schnabel_ortved_2021, title={Effects of continuous passage on the immunomodulator properties of equine bone marrow-derived mesenchymal stem cells in vitro (vol 234, 110203, 2021)}, volume={242}, ISSN={["1873-2534"]}, DOI={10.1016/j.vetimm.2021.110340}, journal={VETERINARY IMMUNOLOGY AND IMMUNOPATHOLOGY}, author={Connard, Shannon S. and Linardi, Renata L. and Even, Kayla M. and Berglund, Alix K. and Schnabel, Lauren V. and Ortved, Kyla F.}, year={2021}, month={Dec} } @article{connard_linardi_even_berglund_schnabel_ortved_2021, title={Effects of continuous passage on the immunomodulatory properties of equine bone marrow-derived mesenchymal stem cells in vitro}, volume={234}, ISSN={["1873-2534"]}, DOI={10.1016/j.vetimm.2021.110203}, abstractNote={The immunomodulatory properties of mesenchymal stem cells (MSCs) have been studied extensively due to their increasing clinical application for tissue regeneration and repair following culture expansion. We have studied the effect of continuous passage on the immunomodulatory capacity of equine bone marrow-derived MSCs (BM-MSCs). Equine BM-MSCs were isolated and culture expanded to passage three, six, and nine (P3, P6, P9). Immunomodulatory properties of each passage were assessed using a T cell proliferation assay and cytokine synthesis following stimulation with interferon gamma (IFN-γ). Equine BM-MSCs maintained their primary cell morphology and immunophenotype throughout all passages. T cell proliferation was suppressed by all passages of BM-MSCs, compared to peripheral blood mononuclear cells (PBMCs) alone. There was no significant difference in suppression of T cell proliferation between P3, P6, and P9 BM-MSCs. All passages of BM-MSCs significantly increased cytokine synthesis in response to stimulation with IFN-γ. There were no significant differences in production of interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1) or regulate on activation, normal T cell expressed and secreted (RANTES) following stimulation with IFN-γ between P3, P6, and P9 BM-MSCs. P9 BM-MSCs had significantly increased production of tumor necrosis factor alpha (TNF-α), (IL-1β), and (IL-10) compared to P3 BM-MSCs. Additionally, there was a significant increase in production of (IL-8) in P6 and P9 BM-MSCs in comparison to P3 BM-MSCs. Our findings demonstrate that culture expansion affects some of the immunomodulatory properties of BM-MSCs in vitro, which may suggest that MSCs isolated from a single collection of bone marrow may be culture expanded, but only those from lower passage numbers would be ideal for clinical application.}, journal={VETERINARY IMMUNOLOGY AND IMMUNOPATHOLOGY}, author={Connard, Shannon S. and Linardi, Renata L. and Even, Kayla M. and Berglund, Alix K. and Schnabel, Lauren V and Ortved, Kyla F.}, year={2021}, month={Apr} } @article{berglund_long_robertson_schnabel_2021, title={TGF-beta 2 Reduces the Cell-Mediated Immunogenicity of Equine MHC-Mismatched Bone Marrow-Derived Mesenchymal Stem Cells Without Altering Immunomodulatory Properties}, volume={9}, ISSN={["2296-634X"]}, DOI={10.3389/fcell.2021.628382}, abstractNote={Allogeneic mesenchymal stem cells (MSCs) are a promising cell therapy for treating numerous diseases, but major histocompatibility complex (MHC)-mismatched MSCs can be rejected by the recipient’s immune system. Pre-treating MSCs with transforming growth factor-β2 (TGF-β2) to downregulate surface expression of MHC molecules may enhance the ability of allogeneic MSCs to evade immune responses. We used lymphocyte proliferation assays and ELISAs to analyze the immunomodulatory potential of TGF-β2-treated equine bone marrow-derived MSCs. T cell activation and cytotoxicity assays were then used to measure thein vitrocell-mediated immunogenicity. Similar to untreated MSCs, TGF-β2-treated MSCs inhibited T cell proliferation and did not stimulate MHC-mismatched T cells to proliferate. Additionally, similar quantities of prostaglandin E2 and TGF-β1 were detected in assays with untreated and TGF-β2-treated MSCs supporting that TGF-β2-treated MSCs retain their strong immunomodulatory propertiesin vitro. Compared to untreated MSCs, TGF-β2-treated MSCs induced less T cell activation and had reduced cell-mediated cytotoxicityin vitro. These results indicate that treating MSCs with TGF-β2 is a promising strategy to reduce the cell-mediated immunogenicity of MHC-mismatched MSCs and facilitate allogeneic MSC therapy.}, journal={FRONTIERS IN CELL AND DEVELOPMENTAL BIOLOGY}, author={Berglund, Alix K. and Long, Julie M. and Robertson, James B. and Schnabel, Lauren V}, year={2021}, month={Feb} } @article{gilbertie_long_schubert_berglund_schaer_schnabel_2018, title={Pooled Platelet-Rich Plasma Lysate Therapy Increases Synoviocyte Proliferation and Hyaluronic Acid Production While Protecting Chondrocytes From Synoviocyte-Derived Inflammatory Mediators}, volume={5}, ISSN={["2297-1769"]}, DOI={10.3389/fvets.2018.00150}, abstractNote={Platelet-rich plasma (PRP) preparations are being used with moderate success to treat osteoarthritis (OA) in humans and in veterinary species. Such preparations are hindered, however, by being autologous in nature and subject to tremendous patient and processing variability. For this reason, there has been increasing interest in the use of platelet lysate preparations instead of traditional PRP. Platelet lysate preparations are acellular, thereby reducing concerns over immunogenicity, and contain high concentrations of growth factors and cytokines. In addition, platelet lysate preparations can be stored frozen for readily available use. The purpose of this study was to evaluate the effects of a pooled allogeneic platelet-rich plasma lysate (PRP-L) preparation on equine synoviocytes and chondrocytes challenged with inflammatory mediators in-vitro to mimic the OA joint environment. Our hypothesis was that PRP-L treatment of inflamed synoviocytes would protect chondrocytes challenged with synoviocyte conditioned media by reducing synoviocyte pro-inflammatory cytokine production while increasing synoviocyte anti-inflammatory cytokine production. Synoviocytes were stimulated with either interleukin-1β (IL-1β) or lipopolysaccharide (LPS) for 24 h followed by no treatment or treatment with platelet-poor plasma lysate (PPP-L) or PRP-L for 48 h. Synoviocyte growth was evaluated at the end of the treatment period and synoviocyte conditioned media was assessed for concentrations of hyaluronic acid (HA), IL-1β, tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6). Chondrocytes were then challenged for 48 h with synoviocyte conditioned media from each stimulation and treatment group and examined for gene expression of collagen types I (COL1A1), II (COL2A1), and III (COL3A1), aggrecan (ACAN), lubricin (PRG4), and matrix metallopeptidase 3 (MMP-3) and 13 (MMP-13). Treatment of inflamed synoviocytes with PRP-L resulted in increased synoviocyte growth and increased synoviocyte HA and IL-6 production. Challenge of chondrocytes with conditioned media from PRP-L treated synoviocytes resulted in increased collagen type II and aggrecan gene expression as well as decreased MMP-13 gene expression. The results of this study support continued investigation into the use of pooled PRP-L for the treatment of osteoarthritis and warrant further in-vitro studies to discern the mechanisms of action of PRP-L.}, journal={FRONTIERS IN VETERINARY SCIENCE}, author={Gilbertie, Jessica M. and Long, Julie M. and Schubert, Alicia G. and Berglund, Alix K. and Schaer, Thomas P. and Schnabel, Lauren V.}, year={2018}, month={Jul} } @article{berglund_schnabel_2017, title={Allogeneic major histocompatibility complex-mismatched equine bone marrow-derived mesenchymal stem cells are targeted for death by cytotoxic anti-major histocompatibility complex antibodies}, volume={49}, ISSN={["2042-3306"]}, DOI={10.1111/evj.12647}, abstractNote={Summary}, number={4}, journal={EQUINE VETERINARY JOURNAL}, author={Berglund, A. K. and Schnabel, L. V.}, year={2017}, month={Jul}, pages={539–544} } @article{sherman_gilger_berglund_schnabel_2017, title={Effect of bone marrow-derived mesenchymal stem cells and stem cell supernatant on equine corneal wound healing in vitro}, volume={8}, ISSN={1757-6512}, url={http://dx.doi.org/10.1186/s13287-017-0577-3}, DOI={10.1186/s13287-017-0577-3}, abstractNote={We aimed to determine and compare the in vitro effects of autologous bone marrow-derived mesenchymal stem cells (BM-MSCs) and mesenchymal stem cell supernatant (MSC-Sp) on the wound healing capacity of equine corneal fibroblasts using a scratch assay. Bone marrow aspirates and eyes were collected from normal, euthanized horses with subsequent isolation and culture of BM-MSCs and corneal stromal cells. Corneal stromal cells were culture-expanded in the culture well of transwell plates and then treated with an autologous BM-MSC suspension (dose: 2.5 × 105/100 μL media with the BM-MSCs contained within the insert well), MSC-Sp solution, or naive culture media (control) for 72 h. A linear defect in confluent cell cultures was created (i.e., corneal scratch assay) to assess the cellular closure (“healing”) over time. Three representative areas of the scratch in each culture were photographed at each time point and the scratch area was quantitated using image analysis software (ImageJ). Media from the scratches were analyzed for various growth factors using human enzyme-linked immunosorbent assay (ELISA) kits that crossreact with the horse. There was a significant percentage decrease in the scratch area remaining in the BM-MSC and MSC-Sp groups compared to the control group. There was also a significant percentage decrease in the scratch area remaining in the BM-MSC group compared to the MSC-Sp group at 36 h post-scratch and all time points thereafter. The concentration of transforming growth factor (TGF)-β1 in the media was significantly higher in the BM-MSC group compared to the control group. The significant decrease in scratch area in equine corneal fibroblast cultures treated with autologous BM-MSCs compared to MSC-Sp or control treatments suggests that BM-MSCs may substantially improve corneal wound healing in horses. MSC-Sp may also improve corneal wound healing given the significant decrease in scratch area compared to control treatments, and would be an immediately available and cost-effective treatment option.}, number={1}, journal={Stem Cell Research & Therapy}, publisher={Springer Science and Business Media LLC}, author={Sherman, Amanda B. and Gilger, Brian C. and Berglund, Alix K. and Schnabel, Lauren V.}, year={2017}, month={May} }