Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Simon Gebremeskel is active.

Publication


Featured researches published by Simon Gebremeskel.


International Journal of Cancer | 2015

The reversible P2Y12 inhibitor ticagrelor inhibits metastasis and improves survival in mouse models of cancer

Simon Gebremeskel; Terry L. LeVatte; Robert Liwski; Brent Johnston; Michael Bezuhly

Tumor cells use activated platelets to promote their proliferation and metastatic potential. Because platelet activation is largely mediated through ADP engagement of purinergic P2Y12 receptors on platelets, we investigated the potential of the reversible P2Y12 inhibitor ticagrelor, a clinical agent used in the prevention of cardiovascular and cerebrovascular events, to inhibit tumor adhesion and metastasis. In B16‐F10 melanoma intravenous and intrasplenic metastasis models, mice treated with a clinical dose of ticagrelor (10 mg/kg) exhibited marked reductions in lung (84%) and liver (86%) metastases. Furthermore, ticagrelor treatment improved survival compared to saline‐treated animals. A similar effect was observed in a 4T1 breast cancer model, with reductions in lung (55%) and bone marrow (87%) metastases following ticagrelor treatment. In vitro, B16‐F10 cells exhibited decreased interaction with platelets from ticagrelor‐treated mice compared to saline‐treated mice, an effect similar to that observed with blockade of glycoprotein IIbIIIa. Similarly, B16‐F10 cells co‐incubated with platelets from ticagrelor‐treated mice exhibited reduced adhesion to endothelial monolayers compared to those co‐incubated with platelets from saline‐treated animals, an effect also observed in vivo. Interestingly, pretreatment of endothelial monolayers with ticagrelor did not result in reduced tumor cell adhesion. These findings support a role for P2Y12‐mediated platelet activation in promoting metastases, and provide proof‐of‐concept for the clinical use of ticagrelor in the prevention of tumor metastasis.


Oncotarget | 2015

Concepts and mechanisms underlying chemotherapy induced immunogenic cell death: impact on clinical studies and considerations for combined therapies.

Simon Gebremeskel; Brent Johnston

Chemotherapy has historically been thought to induce cancer cell death in an immunogenically silent manner. However, recent studies have demonstrated that therapeutic outcomes with specific chemotherapeutic agents (e.g. anthracyclines) correlate strongly with their ability to induce a process of immunogenic cell death (ICD) in cancer cells. This process generates a series of signals that stimulate the immune system to recognize and clear tumor cells. Extensive studies have revealed that chemotherapy-induced ICD occurs via the exposure/release of calreticulin (CALR), ATP, chemokine (C–X–C motif) ligand 10 (CXCL10) and high mobility group box 1 (HMGB1). This review provides an in-depth look into the concepts and mechanisms underlying CALR exposure, activation of the Toll-like receptor 3/IFN/CXCL10 axis, and the release of ATP and HMGB1 from dying cancer cells. Factors that influence the impact of ICD in clinical studies and the design of therapies combining chemotherapy with immunotherapy are also discussed.


OncoImmunology | 2015

Natural killer T cell activation overcomes immunosuppression to enhance clearance of postsurgical breast cancer metastasis in mice

Simon Gebremeskel; Daniel R. Clattenburg; Drew Slauenwhite; Lynnea Lobert; Brent Johnston

Metastatic lesions are responsible for over 90% of breast cancer associated deaths. Therefore, strategies that target metastasis are of particular interest. This study examined the efficacy of natural killer T (NKT) cell activation as a post-surgical immunotherapy in a mouse model of metastatic breast cancer. Following surgical resection of orthotopic 4T1 mammary carcinoma tumors, BALB/c mice were treated with NKT cell activating glycolipid antigens (α-GalCer, α-C-GalCer or OCH) or α-GalCer-loaded dendritic cells (DCs). Low doses of glycolipids transiently reduced metastasis but did not increase survival. A high dose of α-GalCer enhanced overall survival, but was associated with increased toxicity and mortality at early time points. Treatment with α-GalCer-loaded DCs limited tumor metastasis, prolonged survival, and provided curative outcomes in ∼45% of mice. However, survival was not increased further by additional DC treatments or co-transfer of expanded NKT cells. NKT cell activation via glycolipid-loaded DCs decreased the frequency and immunosuppressive activity of myeloid derived suppressor cells (MDSCs) in tumor-resected mice. In vitro, NKT cells were resistant to the immunosuppressive effects of MDSCs and were able to reverse the inhibitory effects of MDSCs on T cell proliferation. NKT cell activation enhanced antitumor immunity in tumor-resected mice, increasing 4T1-specific cytotoxic responses and IFNγ production from natural killer (NK) cells and CD8+ T cells. Consistent with increased tumor immunity, mice surviving to day 150 were resistant to a second tumor challenge. This work provides a clear rationale for manipulating NKT cells to target metastatic disease.


Plastic and Reconstructive Surgery | 2015

Reply: Alternatively Activated M2 Macrophages Improve Autologous Fat Graft Survival in a Mouse Model through Induction of Angiogenesis.

Kyle D. Phipps; Simon Gebremeskel; Joshua A. Gillis; Paul Hong; Brent Johnston; Michael Bezuhly

Background: Variability in graft retention with subsequent undercorrection remains a significant limitation of autologous fat grafting. The authors evaluated whether graft retention in a mouse model could be improved via graft supplementation with alternatively activated M2 macrophages, cells known to play a critical role in tissue repair. Methods: Grafts from C57BL/6 mouse inguinal fat pads were supplemented with M2 macrophages generated by intraperitoneal Brewer’s thioglycollate injection and in vitro culture. Grafts with saline or M2 macrophages were injected under recipient mouse scalps and assessed by serial micro–computed tomographic analysis. Explanted grafts underwent immunohistochemical and flow cytometric analyses. M2 culture supernatants were added to stromal vascular fraction adipose-derived stem cells to assess adipogenic gene expression induction. Results: One month after graft injection, no significant difference was noted between M2 macrophage–supplemented (105 ± 7.0 mm3) and control graft volumes (72 ± 22 mm3). By 3 months after injection, M2 macrophage–supplemented grafts remained stable, whereas controls experienced further volume loss (103 ± 8 mm3 versus 39.4 ± 15 mm3; p = 0.015). Presence of macrophages in supplemented grafts was confirmed by flow cytometry. M2 macrophage–supplemented grafts exhibited a 157 percent increase in vascular density compared with controls (p < 0.05). Induction of adipogenic C/EBP&agr; gene expression was observed with M2 supernatants addition to stromal vascular fraction adipose-derived stem cells. Conclusions: M2 macrophages improve autologous fat graft volume retention by stimulating angiogenesis. These findings provide proof-of-principle for development of fat grafting techniques that harness reparative properties of M2 macrophages.


OncoImmunology | 2016

CXCL16-positive dendritic cells enhance invariant natural killer T cell-dependent IFNγ production and tumor control

Linnea L. Veinotte; Simon Gebremeskel; Brent Johnston

ABSTRACT Crosstalk interactions between dendritic cells (DCs) and invariant natural killer T (iNKT) cells are important in regulating antitumor responses elicited by glycolipid antigens. iNKT cells constitutively express the chemokine receptor CXCR6, while cytokine-activated DCs upregulate the transmembrane chemokine ligand, CXCL16. This study examined the co-stimulatory role of CXCR6/CXCL16 interactions in glycolipid-dependent iNKT cell activation and tumor control. Spleen and liver DCs in wild-type mice, but not iNKT cell deficient (Jα18−/−) mice, transiently upregulated surface CXCL16 following in vivo administration of the glycolipid antigen α-galactosylceramide. Recombinant CXCL16 did not directly induce iNKT cell activation in vitro but enhanced interferon (IFN)-γ production when mouse or human iNKT cells were stimulated with plate-bound anti-CD3. Compared with glycolipid-loaded CXCL16neg DCs, CXCL16hi DCs induced higher levels of IFNγ production in iNKT cell cultures and following adoptive transfer in vivo. The number of IFNγ+ iNKT cells and expansion of T-bet+ iNKT cells were reduced in vivo when CXCL16−/− DCs were used to activate iNKT cells. Enhanced IFNγ production in vivo was not dependent on CXCR6 expression on natural killer (NK) cells. Adoptive transfer of glycolipid-loaded CXCL16hi DCs provided superior protection against tumor metastasis compared to CXCL16neg DC transfers. Similarly, wild-type DCs provided superior protection against metastasis compared with CXCL16−/− DCs. These experiments implicate an important role for CXCR6/CXCL16 interactions in regulating iNKT cell IFNγ production and tumor control. The selective use of CXCL16hi DCs in adoptive transfer immunotherapies may prove useful for enhancing T helper (Th) type 1 responses and clinical outcomes in cancer patients.


Arthritis & Rheumatism | 2014

Regulation of cytokine polarization and T cell recruitment to inflamed paws in mouse collagen-induced arthritis by the chemokine receptor CXCR6.

Drew Slauenwhite; Simon Gebremeskel; Carolyn D. Doucette; David W. Hoskin; Brent Johnston

The chemokine receptor CXCR6 is highly expressed on lymphocytes isolated from the synovium of patients with rheumatoid arthritis, psoriatic arthritis, or juvenile idiopathic arthritis, suggesting that CXCR6 regulates immune cell activation or infiltration into arthritic joints. This study was undertaken to examine the role of CXCR6 in T cell activation and arthritis development.


Neuropharmacology | 2017

Cannabinoid 2 receptor is a novel anti-inflammatory target in experimental proliferative vitreoretinopathy

Anna-Maria Szczesniak; Richard F. Porter; James Thomas Toguri; Joanna Borowska-Fielding; Simon Gebremeskel; Anuja Siwakoti; Brent Johnston; Christian Lehmann; Melanie E. M. Kelly

ABSTRACT Proliferative vitreoretinopathy (PVR) can develop after ocular trauma or inflammation and is a common complication of surgery to correct retinal detachment. Currently, there are no pharmacological treatments for PVR. Cannabinoids acting at cannabinoid 2 receptor (CB2R) can decrease inflammation and fibrosis. The objective of this study was to examine the anti‐inflammatory actions of CB2R as a candidate novel therapeutic target in experimental PVR. PVR was induced by intravitreal injection of dispase in wild‐type (WT) and CB2R genetic knockout (CB2R−/−) mice. Ocular pathology was studied at 24 h or one week after dispase injection. CB2R modulation was examined in WT mice, using the CB2R agonist, HU308, and the CB2R antagonist, AM630. Histopathological scoring and quantification of microglia was used to evaluate tissue pathology. Quantitative PCR and multiplex assays were used to assess changes in proinflammatory cytokines. Intravital microscopy (IVM) was used to visualize and quantify leukocyte‐endothelial adhesion to the iridial microcirculation. Activation of CB2R with HU308 in WT mice with PVR decreased mean histopathological scores, the number of microglia, and leukocyte adhesion compared to vehicle‐treated animals. Conversely, an increase in histopathological scores and activated microglia was observed in PVR animals after treatment with AM630. CB2R−/− mice with PVR exhibited exacerbated ocular histopathology, increased microglia numbers, and elevated protein levels of cytokines as compared to WT mice. In conclusion, our results indicate that intervention at early stage PVR with CB2R agonists reduces ocular inflammation and disease severity. CB2R may represent a therapeutic target to prevent PVR progression and vision loss. This article is part of the Special Issue entitled ‘Lipid Sensing G Protein‐Coupled Receptors in the CNS’. HIGHLIGHTSActivation of CB2R is anti‐inflammatory in a dispase model of PVR.CB2R activation reduces retinal microglia numbers and decreases PVR pathology.CB2R absence or blockade exacerbates ocular inflammation and PVR pathology.Absence of CB2R elevates inflammatory mediators in PVR.


Molecular Therapy - Oncolytics | 2017

Reovirus FAST Protein Enhances Vesicular Stomatitis Virus Oncolytic Virotherapy in Primary and Metastatic Tumor Models

Fabrice Le Boeuf; Simon Gebremeskel; Nichole M. McMullen; Han He; Anna L. Greenshields; David W. Hoskin; John C. Bell; Brent Johnston; Chungen Pan; Roy Duncan

The reovirus fusion-associated small transmembrane (FAST) proteins are the smallest known viral fusogens (∼100–150 amino acids) and efficiently induce cell-cell fusion and syncytium formation in multiple cell types. Syncytium formation enhances cell-cell virus transmission and may also induce immunogenic cell death, a form of apoptosis that stimulates immune recognition of tumor cells. These properties suggest that FAST proteins might serve to enhance oncolytic virotherapy. The oncolytic activity of recombinant VSVΔM51 (an interferon-sensitive vesicular stomatitis virus [VSV] mutant) encoding the p14 FAST protein (VSV-p14) was compared with a similar construct encoding GFP (VSV-GFP) in cell culture and syngeneic BALB/c tumor models. Compared with VSV-GFP, VSV-p14 exhibited increased oncolytic activity against MCF-7 and 4T1 breast cancer spheroids in culture and reduced primary 4T1 breast tumor growth in vivo. VSV-p14 prolonged survival in both primary and metastatic 4T1 breast cancer models, and in a CT26 metastatic colon cancer model. As with VSV-GFP, VSV-p14 preferentially replicated in vivo in tumors and was cleared rapidly from other sites. Furthermore, VSV-p14 increased the numbers of activated splenic CD4, CD8, natural killer (NK), and natural killer T (NKT) cells, and increased the number of activated CD4 and CD8 cells in tumors. FAST proteins may therefore provide a multi-pronged approach to improving oncolytic virotherapy via syncytium formation and enhanced immune stimulation.


Immunology and Cell Biology | 2016

Reconstitution models to evaluate natural killer T cell function in tumor control

Simon Gebremeskel; Drew Slauenwhite; Brent Johnston

Natural killer T (NKT) cells are glycolipid‐reactive T lymphocytes that function in immunosurveillance and immune regulation. However, reduced tumor control in NKT cell‐deficient Jα18−/− mice may be confounded by an overall reduction in T‐cell receptor (TCR) repertoire diversity in these animals. Mechanistic studies are also hindered by a lack of tools to target molecules specifically in NKT cells. To address these issues, we developed protocols to expand functional NKT cells and stably reconstitute them in Jα18−/− mice. In vivo delivery of α‐galactosylceramide (α‐GalCer)‐loaded dendritic cells expanded NKT cells in wild‐type mice without skewing CD4 or TCR Vβ expression profiles. Expanded NKT cells exhibited enhanced cytokine responses upon re‐stimulation with glycolipid or CD3 ligation. Adoptive transfer of recently expanded wild‐type or interferon (IFN)‐γ−/− NKT cells protected recipient Jα18−/− mice from B16 melanoma metastasis without the need for additional glycolipid stimulation. However, NKT cell reconstitution in recipient Jα18−/− mice was short lived. Long‐term reconstitution was only achieved when expanded NKT cells were transferred into sublethally irradiated recipients. Thirty days after transfer, NKT cell numbers, phenotype and α‐GalCer‐induced cytokine responses were equivalent to naive wild‐type mice. Jα18−/− recipients reconstituted with wild‐type or IFN‐γ−/− NKT cells were both protected from B16 melanoma metastasis following α‐GalCer treatment, and NK cell transactivation was intact in mice reconstituted with IFN‐γ−/− NKT cells. These studies validate the use of reconstitution protocols to investigate the mechanisms of NKT cell immune function, demonstrating that NKT cell‐derived IFN‐γ and the altered TCR repertoire in Jα18−/− mice do not impact NKT cell‐mediated antitumor responses.


Cancer immunology research | 2017

Natural Killer T-cell Immunotherapy in Combination with Chemotherapy-Induced Immunogenic Cell Death Targets Metastatic Breast Cancer

Simon Gebremeskel; Lynnea Lobert; Kaitlyn Tanner; Brynn Walker; Tora Oliphant; Livia E. Clarke; Graham Dellaire; Brent Johnston

Low-dose cyclophosphamide or gemcitabine treatments, in combination with natural killer T-cell activation therapy, modulated immune function and improved survival in a mouse model of metastatic breast cancer. Natural killer T (NKT) cells are glycolipid-reactive lymphocytes that promote cancer control. In previous studies, NKT-cell activation improved survival and antitumor immunity in a postsurgical mouse model of metastatic breast cancer. Herein, we investigated whether NKT-cell activation could be combined with chemotherapeutic agents to augment therapeutic outcomes. Gemcitabine and cyclophosphamide analogues enhanced the potential immunogenicity of 4T1 mammary carcinoma cells by increasing the expression of antigen-presenting molecules (MHC-I, MHC-II, and CD1d) and promoting exposure or release of immunogenic cell death markers (calreticulin, HMGB1, and ATP). In 4T1 primary tumor and postsurgical metastasis models, BALB/c mice were treated with cyclophosphamide or gemcitabine. NKT cells were then activated by transfer of dendritic cells loaded with the glycolipid antigen α-galactosylceramide (α-GalCer). Chemotherapeutic treatments did not impact NKT-cell activation but enhanced recruitment into primary tumors. Cyclophosphamide, gemcitabine, or α-GalCer–loaded dendritic cell monotherapies decreased tumor growth in the primary tumor model and reduced metastatic burden and prolonged survival in the metastasis model. Combining chemotherapeutics with NKT-cell activation therapy significantly enhanced survival, with surviving mice exhibiting attenuated tumor growth following a second tumor challenge. The frequency of myeloid-derived suppressor cells was reduced by gemcitabine, cyclophosphamide, or α-GalCer–loaded dendritic cell treatments; cyclophosphamide also reduced the frequency of regulatory T cells. Individual treatments increased immune cell activation, cytokine polarization, and cytotoxic responses, although these readouts were not enhanced further by combining therapies. These findings demonstrate that NKT-cell activation therapy can be combined with gemcitabine or cyclophosphamide to target tumor burden and enhance protection against tumor recurrence. Cancer Immunol Res; 5(12); 1086–97. ©2017 AACR.

Collaboration


Dive into the Simon Gebremeskel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge