Network


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

Hotspot


Dive into the research topics where Andrew J. Spence is active.

Publication


Featured researches published by Andrew J. Spence.


International Journal of Biomaterials | 2012

A Preliminary Study on the Potential of Manuka Honey and Platelet-Rich Plasma in Wound Healing

Scott A. Sell; Patricia S. Wolfe; Andrew J. Spence; Isaac A. Rodriguez; Jennifer M. McCool; Rebecca L. Petrella; Koyal Garg; Jeffery J. Ericksen; Gary L. Bowlin

Aim. The purpose of this study was to determine the in vitro response of cells critical to the wound healing process in culture media supplemented with a lyophilized preparation rich in growth factors (PRGF) and Manuka honey. Materials and Methods. This study utilized cell culture media supplemented with PRGF, as well as whole Manuka honey and the medical-grade Medihoney (MH), a Manuka honey product. The response of human fibroblasts (hDF), macrophages, and endothelial cells (hPMEC) was evaluated, with respect to cell proliferation, chemotaxis, collagen matrix production, and angiogenic potential, when subjected to culture with media containing PRGF, MH, Manuka honey, and a combination of PRGF and MH. Results. All three cell types demonstrated increases in cellular activity in the presence of PRGF, with further increases in activity seen in the presence of PRGF+MH. hDFs proved to be the most positively responsive cells, as they experienced enhanced proliferation, collagen matrix production, and migration into an in vitro wound healing model with the PRGF+MH-supplemented media. Conclusion. This preliminary in vitro study is the first to evaluate the combination of PRGF and Manuka honey, two products with the potential to increase regeneration individually, as a combined product to enhance dermal regeneration.


Cells | 2013

A Preliminary Evaluation of Lyophilized Gelatin Sponges, Enhanced with Platelet-Rich Plasma, Hydroxyapatite and Chitin Whiskers for Bone Regeneration

Isaac A. Rodriguez; Scott A. Sell; Jennifer M. McCool; Gunjan Saxena; Andrew J. Spence; Gary L. Bowlin

The purpose of this study was to perform a number of preliminary in vitro evaluations on an array of modified gelatin gel sponge scaffolds for use in a bone graft application. The gelatin gels were modified through the addition of a number of components which each possess unique properties conducive to the creation and regeneration of bone: a preparation rich in growth factors (PRGF, a bioactive, lyophilized form of platelet-rich plasma), hydroxyapatite, and chitin whiskers. Platelet-rich plasma therapy is an emerging practice that has proven effective in a number of clinical applications, including enhancing bone repair through improved deposition of new bony matrix and angiogenesis. As such, the inclusion of PRGF in our gelatin scaffolds was intended to significantly enhance scaffold bioactivity, while the addition of hydroxyapatite and chitin whiskers were anticipated to increase scaffold strength. Additionally, the gelatin sponges, which readily dissolve in aqueous solutions, were subjected to 1-Ethyl-3-[3-dimethylaminopropyl]carbodiimide hydrochloride (EDC) cross-linking, either during or post-gelation, to control their rate of degradation. Scaffolds were evaluated in vitro with respect to compressive strength, mass loss/degradation, protein release, and cellular interaction, with results demonstrating the potential of the gelatin gel sponge scaffold for use in the regeneration of bone.


Journal of Immunology | 2016

IL-10–Induced miR-155 Targets SOCS1 To Enhance IgE-Mediated Mast Cell Function

Amina Abdul Qayum; Anuya Paranjape; Daniel Abebayehu; Elizabeth Motunrayo Kolawole; Tamara T. Haque; Jamie Josephine Avila McLeod; Andrew J. Spence; Heather L. Caslin; Marcela Taruselli; Alena P. Chumanevich; Bianca Baker; Carole A. Oskeritzian; John J. Ryan

IL-10 is an important regulatory cytokine that modulates a wide range of immune cells. Whereas it is best known for its ability to suppress immune responses, IL-10 has been found to be pathogenic in several human and animal studies of immune-mediated diseases. There is a considerable gap in our understanding of the molecular mechanisms behind the stimulatory effects of IL-10 during allergic inflammation. IL-10 treatment has been shown to suppress mast cell TNF production. In this study, we report that whereas TNF secretion was reduced, IL-10 surprisingly enhanced IgE-mediated protease and cytokine production both in vitro and in vivo. This stimulatory effect was consistent in mouse and human skin mast cells. IL-10 enhanced activation of the key FcεRI signaling proteins Stat5, JNK, and ERK. We demonstrate that IL-10 effects are dependent on Stat3 activation, eliciting miR-155 expression, with a resulting loss of suppressor of cytokine signaling-1. The importance of miR-155 was demonstrated by the inability of IL-10 to enhance anaphylaxis in miR-155–deficient mice. Taken together, our results reveal an IL-10–induced, Stat3–miR-155 signaling pathway that can promote mast cell responses.


Journal of Immunology | 2017

TGF-β1 Suppresses IL-33–Induced Mast Cell Function

Victor Ndaw; Daniel Abebayehu; Andrew J. Spence; Patrick A Paez; E. Motunrayo Kolawole; Marcela Taruselli; Heather L. Caslin; Alena P. Chumanevich; Anuya Paranjape; Bianca Baker; Brian Barnstein; Tamara T. Haque; Kasalina N. Kiwanuka; Carole A. Oskeritzian; John J. Ryan

TGF-β1 is involved in many pathological conditions, including autoimmune disorders, cancer, and cardiovascular and allergic diseases. We have previously found that TGF-β1 can suppress IgE-mediated mast cell activation of human and mouse mast cells. IL-33 is a member of the IL-1 family capable of inducing mast cell responses and enhancing IgE-mediated activation. In this study, we investigated the effects of TGF-β on IL-33–mediated mast cell activation. Bone marrow–derived mast cells cultured in TGF-β1, β2, or β3 showed reduced IL-33–mediated production of TNF, IL-6, IL-13, and MCP-1 in a concentration-dependent manner. TGF-β1 inhibited IL-33–mediated Akt and ERK phosphorylation as well as NF-κB– and AP-1–mediated transcription. These effects were functionally important, as TGF-β1 injection suppressed IL-33–induced systemic cytokines in vivo and inhibited IL-33–mediated cytokine release from human mast cells. TGF-β1 also suppressed the combined effects of IL-33 and IgE-mediated activation on mouse and human mast cells. The role of IL-33 in the pathogenesis of allergic diseases is incompletely understood. These findings, consistent with our previously reported effects of TGF-β1 on IgE-mediated activation, demonstrate that TGF-β1 can provide broad inhibitory signals to activated mast cells.


Journal of Immunology | 2016

Fluvastatin Suppresses Mast Cell and Basophil IgE Responses: Genotype-Dependent Effects

Elizabeth Motunrayo Kolawole; Jamie Josephine Avila McLeod; Victor Ndaw; Daniel Abebayehu; Brian Barnstein; Travis Faber; Andrew J. Spence; Marcela Taruselli; Anuya Paranjape; Tamara T. Haque; Amina Abdul Qayum; Qasim A. Kazmi; Dayanjan S. Wijesinghe; Jamie Sturgill; Charles E. Chalfant; David B. Straus; Carole A. Oskeritzian; John J. Ryan

Mast cell (MC)– and basophil-associated inflammatory diseases are a considerable burden to society. A significant portion of patients have symptoms despite standard-of-care therapy. Statins, used to lower serum cholesterol, have immune-modulating activities. We tested the in vitro and in vivo effects of statins on IgE-mediated MC and basophil activation. Fluvastatin showed the most significant inhibitory effects of the six statins tested, suppressing IgE-induced cytokine secretion among mouse MCs and basophils. The effects of fluvastatin were reversed by mevalonic acid or geranylgeranyl pyrophosphatase, and mimicked by geranylgeranyl transferase inhibition. Fluvastatin selectively suppressed key FcεRI signaling pathways, including Akt and ERK. Although MCs and basophils from the C57BL/6J mouse strain were responsive to fluvastatin, those from 129/SvImJ mice were completely resistant. Resistance correlated with fluvastatin-induced upregulation of the statin target HMG-CoA reductase. Human MC cultures from eight donors showed a wide range of fluvastatin responsiveness. These data demonstrate that fluvastatin is a potent suppressor of IgE-mediated MC activation, acting at least partly via blockade of geranyl lipid production downstream of HMG-CoA reductase. Importantly, consideration of statin use for treating MC–associated disease needs to incorporate genetic background effects, which can yield drug resistance.


Journal of Leukocyte Biology | 2016

Dexamethasone rapidly suppresses IL‐33‐stimulated mast cell function by blocking transcription factor activity

Anuya Paranjape; Oksana Chernushevich; Amina Abdul Qayum; Andrew J. Spence; Marcela Taruselli; Daniel Abebayehu; Brian Barnstein; Jamie Josephine Avila McLeod; Bianca Baker; Gurjas S. Bajaj; Alena P. Chumanevich; Carole A. Oskeritzian; John J. Ryan

Mast cells are critical effectors of allergic disease and can be activated by IL‐33, a proinflammatory member of the IL‐1 cytokine family. IL‐33 worsens the pathology of mast cell–mediated diseases, but therapies to antagonize IL‐33 are still forthcoming. Because steroids are the mainstay of allergic disease treatment and are well known to suppress mast cell activation by other stimuli, we examined the effects of the steroid dexamethasone on IL‐33‐mediated mast cell function. We found that dexamethasone potently and rapidly suppressed cytokine production elicited by IL‐33 from murine bone marrow–derived and peritoneal mast cells. IL‐33 enhances IgE‐mediated mast cell cytokine production, an activity that was also antagonized by dexamethasone. These effects were consistent in human mast cells. We additionally observed that IL‐33 augmented migration of IgE‐sensitized mast cells toward antigen. This enhancing effect was similarly reversed by dexamethasone. Simultaneous addition of dexamethasone with IL‐33 had no effect on the phosphorylation of MAP kinases or NFκB p65 subunit; however, dexamethasone antagonized AP‐1‐ and NFκB‐mediated transcriptional activity. Intraperitoneal administration of dexamethasone completely abrogated IL‐33‐mediated peritoneal neutrophil recruitment and prevented plasma IL‐6 elevation. These data demonstrate that steroid therapy may be an effective means of antagonizing the effects of IL‐33 on mast cells in vitro and in vivo, acting partly by suppressing IL‐33‐induced NFκB and AP‐1 activity.


Journal of Immunology | 2016

Lactic Acid Suppresses IL-33–Mediated Mast Cell Inflammatory Responses via Hypoxia-Inducible Factor-1α–Dependent miR-155 Suppression

Daniel Abebayehu; Andrew J. Spence; Amina Abdul Qayum; Marcela Taruselli; Jamie Josephine Avila McLeod; Heather L. Caslin; Alena P. Chumanevich; Elizabeth Motunrayo Kolawole; Anuya Paranjape; Bianca Baker; Victor Ndaw; Brian Barnstein; Carole A. Oskeritzian; Scott A. Sell; John J. Ryan

Lactic acid (LA) is present in tumors, asthma, and wound healing, environments with elevated IL-33 and mast cell infiltration. Although IL-33 is a potent mast cell activator, how LA affects IL-33–mediated mast cell function is unknown. To investigate this, mouse bone marrow–derived mast cells were cultured with or without LA and activated with IL-33. LA reduced IL-33–mediated cytokine and chemokine production. Using inhibitors for monocarboxylate transporters (MCT) or replacing LA with sodium lactate revealed that LA effects are MCT-1– and pH-dependent. LA selectively altered IL-33 signaling, suppressing TGF-β–activated kinase-1, JNK, ERK, and NF-κB phosphorylation, but not p38 phosphorylation. LA effects in other contexts have been linked to hypoxia-inducible factor (HIF)-1α, which was enhanced in bone marrow–derived mast cells treated with LA. Because HIF-1α has been shown to regulate the microRNA miR-155 in other systems, LA effects on miR-155-5p and miR-155-3p species were measured. In fact, LA selectively suppressed miR-155-5p in an HIF-1α–dependent manner. Moreover, overexpressing miR-155-5p, but not miR-155-3p, abolished LA effects on IL-33–induced cytokine production. These in vitro effects of reducing cytokines were consistent in vivo, because LA injected i.p. into C57BL/6 mice suppressed IL-33–induced plasma cytokine levels. Lastly, IL-33 effects on primary human mast cells were suppressed by LA in an MCT-dependent manner. Our data demonstrate that LA, present in inflammatory and malignant microenvironments, can alter mast cell behavior to suppress inflammation.


Cellular Immunology | 2017

Didox (3,4-dihydroxybenzohydroxamic acid) suppresses IL-33-induced cytokine production in primary mouse mast cells

Heather L. Caslin; Jamie Josephine Avila McLeod; Andrew J. Spence; Amina Abdul Qayum; Elizabeth Motunrayo Kolawole; Marcela Taruselli; Anuya Paranjape; Howard L. Elford; John J. Ryan

While IgE is considered the primary mediator of mast cell activation, IL-33 contributes substantially in asthma, allergic rhinitis, and atopic dermatitis. To develop effective treatments for allergic disease, it is important to understand the role of therapeutic agents on IL-33 activation. We examined the effect of Didox (3,4-dihydroxybenzohydroxamic acid), an antioxidant and ribonucleotide reductase (RNR) inhibitor, on IL-33-mediated mast cell activation. Didox suppressed IL-6, IL-13, TNF, and MIP-1α (CCL3) production in bone marrow derived mast cells following IL-33 activation. This suppression was observed in different genetic backgrounds and extended to peritoneal mast cells. The antioxidant N-acetylcysteine mimicked the suppression of Didox, albeit at a much higher dose, while the RNR inhibitor hydroxyurea had no effect. Didox substantially suppressed IL-33-mediated NFκB and AP-1 transcriptional activities. These results suggest that Didox attenuates IL-33-induced mast cell activation and should be further studied as a potential therapeutic agent for inflammatory diseases involving IL-33.


Cellular Immunology | 2017

Didox (3,4-dihydroxybenzohydroxamic acid) suppresses IgE-mediated mast cell activation through attenuation of NFκB and AP-1 transcription

Jamie Josephine Avila McLeod; Heather L. Caslin; Andrew J. Spence; Elizabeth Motunrayo Kolawole; Amina Abdul Qayum; Anuya Paranjape; Marcela Taruselli; Tamara T. Haque; Kasalina N. Kiwanuka; Howard L. Elford; John J. Ryan

Mast cell activation via the high-affinity IgE receptor (FcεRI) elicits production of inflammatory mediators central to allergic disease. As a synthetic antioxidant and a potent ribonucleotide reductase (RNR) inhibitor, Didox (3,4-dihyroxybenzohydroxamic acid) has been tested in clinical trials for cancer and is an attractive therapeutic for inflammatory disease. We found that Didox treatment of mouse bone marrow-derived mast cells (BMMC) reduced IgE-stimulated degranulation and cytokine production, including IL-6, IL-13, TNF and MIP-1a (CCL3). These effects were consistent using BMMC of different genetic backgrounds and peritoneal mast cells. While the RNR inhibitor hydroxyurea had little or no effect on IgE-mediated function, high concentrations of the antioxidant N-acetylcysteine mimicked Didox-mediated suppression. Furthermore, Didox increased expression of the antioxidant genes superoxide dismutase and catalase, and suppressed DCFH-DA fluorescence, indicating reduced reactive oxygen species production. Didox effects were not due to changes in FcεRI expression or cell viability, suggesting it inhibits signaling required for inflammatory cytokine production. In support of this, we found that Didox reduced FcεRI-mediated AP-1 and NFκB transcriptional activity. Finally, Didox suppressed mast cell-dependent, IgE-mediated passive systemic anaphylaxis in vivo. These data demonstrate the potential use for Didox asa means of antagonizing mast cell responses in allergic disease.


Journal of Biomedical Materials Research Part A | 2017

Galectin-1 promotes an M2 macrophage response to polydioxanone scaffolds

Daniel Abebayehu; Andrew J. Spence; Barbara D. Boyan; Zvi Schwartz; John J. Ryan; Michael J. McClure

Regulating soft tissue repair to prevent fibrosis and promote regeneration is central to creating a microenvironment conducive to soft tissue development. Macrophages play an important role in this process. The macrophage response can be modulated using biomaterials, altering cytokine and growth factor secretion to promote regeneration. Electrospun polydioxanone (PDO) fiber scaffolds promoted an M2 phenotype when macrophages were cultured on large diameter, highly porous scaffolds, but an M1 phenotype on smaller diameter fibers. In this study, we investigated whether incorporation of galectin-1, an immunosuppressive protein that enhances muscle regeneration, could promote the M2 response. Galectin-1 was incorporated into large and small fiber PDO scaffolds during electrospinning. Galectin-1 incorporation increased arginase-1 and reduced iNOS and IL-6 production in mouse bone-marrow derived macrophages compared with PDO alone for both scaffold types. Inhibition of ERK mitogen-activated protein kinase did not alter galectin-1 effects on arginase-1 and iNOS expression, but reversed IL-6 suppression, indicating that IL-6 is mediated by a different mechanism. Our results suggest that galectin-1 can be used to modulate macrophage commitment to a pro-regenerative M2 phenotype, which may positively impact tissue regeneration when using small diameter PDO scaffolds.

Collaboration


Dive into the Andrew J. Spence's collaboration.

Top Co-Authors

Avatar

John J. Ryan

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Daniel Abebayehu

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Marcela Taruselli

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amina Abdul Qayum

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Anuya Paranjape

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carole A. Oskeritzian

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Heather L. Caslin

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Bianca Baker

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge