Network


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

Hotspot


Dive into the research topics where Erika A. Eksioglu is active.

Publication


Featured researches published by Erika A. Eksioglu.


Journal of Clinical Investigation | 2013

Induction of myelodysplasia by myeloid-derived suppressor cells

Xianghong Chen; Erika A. Eksioglu; Junmin Zhou; Ling Zhang; Julie Y. Djeu; Nicole Fortenbery; Pearlie K. Epling-Burnette; Sandra van Bijnen; Harry Dolstra; John P. Cannon; Je-In Youn; Sarah S. Donatelli; Dahui Qin; Theo de Witte; Jianguo Tao; Huaquan Wang; Pingyan Cheng; Dmitry I. Gabrilovich; Alan F. List; Sheng Wei

Myelodysplastic syndromes (MDS) are age-dependent stem cell malignancies that share biological features of activated adaptive immune response and ineffective hematopoiesis. Here we report that myeloid-derived suppressor cells (MDSC), which are classically linked to immunosuppression, inflammation, and cancer, were markedly expanded in the bone marrow of MDS patients and played a pathogenetic role in the development of ineffective hematopoiesis. These clonally distinct MDSC overproduce hematopoietic suppressive cytokines and function as potent apoptotic effectors targeting autologous hematopoietic progenitors. Using multiple transfected cell models, we found that MDSC expansion is driven by the interaction of the proinflammatory molecule S100A9 with CD33. These 2 proteins formed a functional ligand/receptor pair that recruited components to CD33’s immunoreceptor tyrosine-based inhibition motif (ITIM), inducing secretion of the suppressive cytokines IL-10 and TGF-β by immature myeloid cells. S100A9 transgenic mice displayed bone marrow accumulation of MDSC accompanied by development of progressive multilineage cytopenias and cytological dysplasia. Importantly, early forced maturation of MDSC by either all-trans-retinoic acid treatment or active immunoreceptor tyrosine-based activation motif–bearing (ITAM-bearing) adapter protein (DAP12) interruption of CD33 signaling rescued the hematologic phenotype. These findings indicate that primary bone marrow expansion of MDSC driven by the S100A9/CD33 pathway perturbs hematopoiesis and contributes to the development of MDS.


Proceedings of the National Academy of Sciences of the United States of America | 2014

TGF-β–inducible microRNA-183 silences tumor-associated natural killer cells

Sarah S. Donatelli; Junmin Zhou; Danielle L. Gilvary; Erika A. Eksioglu; Xianghong Chen; W. Douglas Cress; Eric B. Haura; Matthew B. Schabath; Domenico Coppola; Sheng Wei; Julie Y. Djeu

Significance Natural killer (NK) cells are potent tumor-cell killers, but exposure to transforming growth factor beta-1 (TGF-β) abrogates their effectiveness. Here, we show that this suppression is a result of TGF-β induction of microRNA (miR)-183, which binds and represses DNAX activating protein 12 kDa (DAP12), a signal adaptor for lytic function in NK cells. Because introduction of miR-183 alone or its functional blockade in the presence of TGF-β reduced or restored DAP12 levels in NK cells, we define miR-183 as a key factor in TGF-β–mediated immunosuppression. Since DAP12 is required for signaling through multiple NK cytotoxicity receptors and TGF-β is overexpressed by diverse solid malignancies, our data may have significant importance in the development of NK-based cancer immunotherapies. Transforming growth factor β1 (TGF-β), enriched in the tumor microenvironment and broadly immunosuppressive, inhibits natural killer (NK) cell function by yet-unknown mechanisms. Here we show that TGF-β–treated human NK cells exhibit reduced tumor cytolysis and abrogated perforin polarization to the immune synapse. This result was accompanied by loss of surface expression of activating killer Ig-like receptor 2DS4 and NKp44, despite intact cytoplasmic stores of these receptors. Instead, TGF-β depleted DNAX activating protein 12 kDa (DAP12), which is critical for surface NK receptor stabilization and downstream signal transduction. Mechanistic analysis revealed that TGF-β induced microRNA (miR)-183 to repress DAP12 transcription/translation. This pathway was confirmed with luciferase reporter constructs bearing the DAP12 3′ untranslated region as well as in human NK cells by use of sense and antisense miR-183. Moreover, we documented reduced DAP12 expression in tumor-associated NK cells in lung cancer patients, illustrating this pathway to be consistently perturbed in the human tumor microenvironment.


Journal of Medicinal Chemistry | 2009

Grassystatins A–C from Marine Cyanobacteria, Potent Cathepsin E Inhibitors that Reduce Antigen Presentation

Jason C. Kwan; Erika A. Eksioglu; Chen Liu; Valerie J. Paul; Hendrik Luesch

In our efforts to explore marine cyanobacteria as a source of novel bioactive compounds, we discovered a statine unit-containing linear decadepsipeptide, grassystatin A (1), which we screened against a diverse set of 59 proteases. We describe the structure determination of 1 and two natural analogues, grassystatins B (2) and C (3), using NMR, MS, and chiral HPLC techniques. Compound 1 selectively inhibited cathepsins D and E with IC(50)s of 26.5 nM and 886 pM, respectively. Compound 2 showed similar potency and selectivity against cathepsins D and E (IC(50)s of 7.27 nM and 354 pM, respectively), whereas the truncated peptide analogue grassystatin C (3), which consists of two fewer residues than 1 and 2, was less potent against both but still selective for cathepsin E. The selectivity of compounds 1-3 for cathepsin E over D (20-38-fold) suggests that these natural products may be useful tools to probe cathepsin E function. We investigated the structural basis of this selectivity using molecular docking. We also show that 1 can reduce antigen presentation by dendritic cells, a process thought to rely on cathepsin E.


Blood | 2016

The NLRP3 Inflammasome functions as a driver of the myelodysplastic syndrome phenotype.

Ashley A. Basiorka; Kathy L. McGraw; Erika A. Eksioglu; Xianghong Chen; Joseph O. Johnson; Ling Zhang; Qing Zhang; Brittany Irvine; Thomas Cluzeau; David Sallman; Eric Padron; Rami S. Komrokji; Lubomir Sokol; Rebecca C. Coll; Avril A. B. Robertson; Matthew A. Cooper; John L. Cleveland; Luke A. J. O'Neill; Sheng Wei; Alan F. List

Despite genetic heterogeneity, myelodysplastic syndromes (MDSs) share features of cytological dysplasia and ineffective hematopoiesis. We report that a hallmark of MDSs is activation of the NLRP3 inflammasome, which drives clonal expansion and pyroptotic cell death. Independent of genotype, MDS hematopoietic stem and progenitor cells (HSPCs) overexpress inflammasome proteins and manifest activated NLRP3 complexes that direct activation of caspase-1, generation of interleukin-1β (IL-1β) and IL-18, and pyroptotic cell death. Mechanistically, pyroptosis is triggered by the alarmin S100A9 that is found in excess in MDS HSPCs and bone marrow plasma. Further, like somatic gene mutations, S100A9-induced signaling activates NADPH oxidase (NOX), increasing levels of reactive oxygen species (ROS) that initiate cation influx, cell swelling, and β-catenin activation. Notably, knockdown of NLRP3 or caspase-1, neutralization of S100A9, and pharmacologic inhibition of NLRP3 or NOX suppress pyroptosis, ROS generation, and nuclear β-catenin in MDSs and are sufficient to restore effective hematopoiesis. Thus, alarmins and founder gene mutations in MDSs license a common redox-sensitive inflammasome circuit, which suggests new avenues for therapeutic intervention.


PLOS ONE | 2011

Characterization of HCV interactions with Toll-like receptors and RIG-I in liver cells.

Erika A. Eksioglu; Haizhen Zhu; Lilly Bayouth; Jennifer Bess; Hongyan Liu; David R. Nelson; Chen Liu

Background and Aim The aim of this study was to examine the mechanisms of IFN induction and viral escape. In order to accomplish the goal we compared our new hepatoma cell line LH86, which has intact TLR3 and RIG-I expression and responds to HCV by inducing IFN, with Huh7.5 cells which lack those features. Methods The initial interaction of LH86 cells, Huh7.5 cells or their transfected counter parts (LH86 siRIG-I, siTLR3 or siTLR7 and Huh7.5 RIG-I, TLR3 or TLR7) after infection with HCV (strain JFH-1) was studied by measuring the expression levels of IFNβ, TRAIL, DR4, DR5 and their correlation to viral replication. Results HCV replicating RNA induces IFN in LH86 cells. The IFN induction system is functional in LH86, and the expression of the RIG-I and TLR3 in LH86 is comparable to the primary hepatocytes. Both proteins appear to play important roles in suppression of viral replication. We found that innate immunity against HCV is associated with the induction of apoptosis by RIG-I through the TRAIL pathway and the establishment of an antiviral state by TLR3. HCV envelope proteins interfere with the expression of TLR3 and RIG-I. Conclusion These findings correlate with the lower expression level of PRRs in HCV chronic patients and highlight the importance of the PRRs in the initial interaction of the virus and its host cells. This work represents a novel mechanism of viral pathogenesis for HCV and demonstrates the role of PRRs in viral infection.


Cellular and Molecular Life Sciences | 2015

The inflammatory microenvironment in MDS

Lili Yang; Yaqin Qian; Erika A. Eksioglu; Pearlie K. Epling-Burnette; Sheng Wei

Myelodysplastic syndromes (MDS) are a collection of pre-malignancies characterized by impaired proliferation and differentiation of hematopoietic stem cells and a tendency to evolve into leukemia. Among MDS’s pathogenic mechanisms are genetic, epigenetic, apoptotic, differentiation, and cytokine milieu abnormalities. Inflammatory changes are a prominent morphologic feature in some cases, with increased populations of plasma cells, mast cells, and lymphocytes in bone marrow aspirates. Accumulating evidence suggests that the bone marrow microenvironment contributes to MDS disease pathology, with microenvironment alterations and abnormality preceding, and facilitating clonal evolution in MDS patients. In this review, we focus on the inflammatory changes involved in the pathology of MDS, with an emphasis on immune dysfunction, stromal microenvironment, and cytokine imbalance in the microenvironment as well as activation of innate immune signaling in MDS patients. A better understanding of the mechanism of MDS pathophysiology will be beneficial to the development of molecular-targeted therapies in the future.


Scandinavian Journal of Immunology | 2010

Influence of serum and soluble CD25 (sCD25) on regulatory and effector T-cell function in hepatocellular carcinoma.

Roniel Cabrera; Miguel Ararat; Erika A. Eksioglu; Mengde Cao; Yiling Xu; Clive Wasserfall; Mark A. Atkinson; Chen Liu; David R. Nelson

Our previous studies showed that high levels of soluble CD25 (sCD25) in the serum of patients with hepatocellular carcinoma (HCC) correlated with blunted effector T‐cells (Teff) responses, tumour burden and poor survival. Understanding the interactions between Teff, CD4+CD25+ regulatory T cells (Treg) and soluble factors can identify novel therapeutic targets. In this study, we characterize the mechanisms by which HCC serum and sCD25 mediate suppression of Teff and evaluate the effect of sCD25 on the suppression assays with normal healthy control cells (NHC) at a 1:1 Treg to Teff cell ratio to determine whether sCD25 has any impact on Treg suppression. HCC serum and sCD25 suppressed Teff proliferation and downregulated CD25 expression on HCC Teff in a dose‐dependent fashion with sCD25 doses above 3000 pg/ml. Treg from HCC and cirrhosis patients suppressed proliferation of target CD4+CD25− Teff in serum‐free medium (SFM). HCC Treg showed a higher degree of suppression than cirrhosis‐derived Treg. In contrast, Treg from NHC did not suppress target Teff in SFM. However, isolated Treg from all three study subjects (HCC, cirrhosis and NHC) suppressed CD4+CD25− Teff in serum conditions or in the presence of sCD25 in the range 6000–12,000 pg/ml. In conclusion, downregulation of CD25 cell surface expression on Teff is part of the overall suppressive mechanism of sCD25 and HCC serum on Teff responses. The observed sCD25 and HCC serum‐mediated suppression is further influenced via novel immune‐inhibitory interaction between CD4+CD25+ Treg and sCD25.


Journal of Viral Hepatitis | 2010

Hepatitis C virus modulates human monocyte-derived dendritic cells.

Erika A. Eksioglu; Jennifer Bess; Haizhen Zhu; Yiling Xu; Hui-Jia Dong; John Elyar; David R. Nelson; Chen Liu

Summary.  This study is to examine the monocyte‐derived dendritic cell (DC) response to hepatitis C virus (HCV) in a cell culture system. Adherence‐derived DCs were incubated with various titres of JFH‐1 (HCV genotype 2a), generated from transfected Huh 7.5 cells or co‐incubated with Newcastle disease virus (NDV). Infection and the type 1 interferon (IFN) response were assessed by real‐time reverse transcriptase‐polymerase chain reaction, morphology by light microscopy and immunophenotype by flow cytometry. Our data demonstrated no viral replication or particle release from DC after HCV infection. Morphologically, monocytes showed a tendency to shift to immature DCs when cultured with HCV, when compared with control monocytes. This shift was confirmed by flow cytometry and appeared to be related to viral titres. There was also an increase in immature DC numbers. HCV infection induced IFNβ expression in DCs, and the amount seemed to be inversely correlated with viral titres indicating that HCV has the capacity to negatively regulate such cells. However, IFNα does not appear to be affected by direct contact with the virus. A strong IFNβ signal induced by NDV in DC was substantially diminished by HCV. HCV negatively affects the maturation of DCs and suppresses the type 1 IFN response of DC. Our results suggest a mechanism of viral evasion of host immunity.


Frontiers in Bioscience | 2010

Dendritic cells as therapeutic agents against cancer.

Erika A. Eksioglu; Eisen S; Reddy

Dendritic cells (DC) are antigen-presenting cells whose immunobiology has been proven to be central to the function of the immune system. Further understanding of these cells is leading the way to the manipulation of the immune system as a tool to cure and prevent a vast array of diseases including cancers. These cells have been used in trials as vaccine adjuvants in therapies that aim to break the bodys tolerance to the tumor. From the first 1000 DC vaccinees in 2003 there has been a breadth of information on safety that is paving the way to the study of the efficiency of these therapies. This review aims to explore recent updates to the current literature on DC vaccine therapies in clinical trials and analyze their future. At this crossroads is where intricacies of the technique are being revised to explore the most efficient and effective parameters for the enhancement of DC adjuvant therapies.


Scientific Reports | 2015

HMGB1 induction of clusterin creates a chemoresistant niche in human prostate tumor cells

Junmin Zhou; Xianghong Chen; Danielle L. Gilvary; Melba Marie Tejera; Erika A. Eksioglu; Sheng Wei; Julie Y. Djeu

Development of chemoresistance, especially to docetaxel (DTX), is the primary barrier to the cure of castration-resistant prostate cancer but its mechanism is obscure. Here, we report a seminal crosstalk between dying and residual live tumor cells during treatment with DTX that can result in outgrowth of a chemoresistant population. Survival was due to the induction of secretory/cytoplasmic clusterin (sCLU), which is a potent anti-apoptotic protein known to bind and sequester Bax from mitochondria, to prevent caspase 3 activation. sCLU induction in live cells depended on HMGB1 release from dying cells. Supernatants from DTX-treated DU145 tumor cells, which were shown to contain HMGB1, effectively induced sCLU from newly-plated DU145 tumor cells and protected them from DTX toxicity. Addition of anti-HMBG1 to the supernatant or pretreatment of newly-plated DU145 tumor cells with anti-TLR4 or anti-RAGE markedly abrogated sCLU induction and protective effect of the supernatant. Mechanistically, HMGB1 activated NFκB to promote sCLU gene expression and prevented the translocation of activated Bax to mitochondria to block cell death. Importantly, multiple currently-used chemotherapeutic drugs could release HMGB1 from tumor cells. These results suggest that acquisition of chemoresistance may involve the HMGB1/TLR4-RAGE/sCLU pathway triggered by dying cells to provide survival advantage to remnant live tumor cells.

Collaboration


Dive into the Erika A. Eksioglu's collaboration.

Top Co-Authors

Avatar

Sheng Wei

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Xianghong Chen

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Junmin Zhou

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Julie Y. Djeu

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Nicole Fortenbery

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Alan F. List

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kathy L. McGraw

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge