Network


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

Hotspot


Dive into the research topics where Engin Ozkaynak is active.

Publication


Featured researches published by Engin Ozkaynak.


Nature Medicine | 2007

Deacetylase inhibition promotes the generation and function of regulatory T cells

Ran Tao; Edwin F. de Zoeten; Engin Ozkaynak; Chunxia Chen; Liqing Wang; Paige M. Porrett; Bin Li; Laurence A. Turka; Eric N. Olson; Mark I. Greene; Andrew D. Wells; Wayne W. Hancock

Histone/protein deacetylases (HDACs) regulate chromatin remodeling and gene expression as well as the functions of more than 50 transcription factors and nonhistone proteins. We found that administration of an HDAC inhibitor (HDACi) in vivo increased Foxp3 gene expression, as well as the production and suppressive function of regulatory T cells (Treg cells). Although Treg cells express multiple HDACs, HDAC9 proved particularly important in regulating Foxp3-dependent suppression. Optimal Treg function required acetylation of several lysines in the forkhead domain of Foxp3, and Foxp3 acetylation enhanced binding of Foxp3 to the Il2 promoter and suppressed endogenous IL-2 production. HDACi therapy in vivo enhanced Treg-mediated suppression of homeostatic proliferation, decreased inflammatory bowel disease through Treg-dependent effects, and, in conjunction with a short course of low-dose rapamycin, induced permanent, Treg-dependent cardiac and islet allograft survival and donor-specific allograft tolerance. Our data show that use of HDACi allows the beneficial pharmacologic enhancement of both the numbers and suppressive function of Foxp3+ Treg cells.


Journal of Experimental Medicine | 2005

Recruitment of Foxp3+ T regulatory cells mediating allograft tolerance depends on the CCR4 chemokine receptor

Iris Lee; Liqing Wang; Andrew D. Wells; Martin E. Dorf; Engin Ozkaynak; Wayne W. Hancock

Although certain chemokines and their receptors guide homeostatic recirculation of T cells and others promote recruitment of activated T cells to inflammatory sites, little is known of the mechanisms underlying a third function, migration of Foxp3+ regulatory T (T reg) cells to sites where they maintain unresponsiveness. We studied how T reg cells are recruited to cardiac allografts in recipients tolerized with CD154 monoclonal antibody (mAb) plus donor-specific transfusion (DST). Real-time polymerase chain reaction showed that intragraft Foxp3 levels in tolerized recipients were ∼100-fold higher than rejecting allografts or allografts associated with other therapies inducing prolonged survival but not tolerance. Foxp3+ cells were essential for tolerance because pretransplant thymectomy or peritransplant depletion of CD25+ cells prevented long-term survival, as did CD25 mAb therapy in well-functioning allografts after CD154/DST therapy. Analysis of multiple chemokine pathways showed that tolerance was accompanied by intragraft up-regulation of CCR4 and one of its ligands, macrophage-derived chemokine (CCL22), and that tolerance induction could not be achieved in CCR4−/− recipients. We conclude that Foxp3 expression is specifically up-regulated within allografts of mice displaying donor-specific tolerance, that recruitment of Foxp3-expressing T reg cells to an allograft tissue is dependent on the chemokine receptor, CCR4, and that, in the absence of such recruitment, tolerizing strategies such as CD154 mAb therapy are ineffectual.


Nature Immunology | 2001

Importance of ICOS–B7RP-1 costimulation in acute and chronic allograft rejection

Engin Ozkaynak; Wei Gao; Nida Shemmeri; Chichung Wang; Jose-Carlos Gutierrez-Ramos; Jillian Amaral; Shixin Qin; James B. Rottman; Anthony J. Coyle; Wayne W. Hancock

Primary T cell activation requires B7-CD28 and CD40-CD154 costimulation, but effector T cell functions are considered to be largely independent of these costimulatory pathways. Although blockade of costimulation with cytolytic T lymphocyte–associated antigen 4–immunoglobulin (CTLA-4–Ig) or monoclonal antibody (mAb) to CD154 prolongs allograft survival, chronic rejection follows, which suggests that additional key costimulatory pathways are active in vivo. We found that both antibody to inducible costimulator (anti-ICOS) and an ICOS-Ig fusion protein suppressed intragraft T cell activation and cytokine expression and prolonged allograft survival in a manner similar to that in ICOS−/− allograft recipients. The combination of anti-ICOS therapy and cyclosporin A led to permanent engraftment. In addition, ICOS–B7RP-1 costimulation was required for the development of chronic rejection after CD40-CD154 blockade. These data demonstrate a key role for the ICOS–B7RP-1 pathway in acute and chronic rejection and highlight the benefits of targeting this pathway in combination with the use of conventional immunosuppressive agent.


Nature Immunology | 2001

The costimulatory molecule ICOS plays an important role in the immunopathogenesis of EAE

James B. Rottman; Tammy Smith; James R. Tonra; Kenneth Ganley; Troy Bloom; Robert Silva; Barbara Pierce; Jose-Carlos Gutierrez-Ramos; Engin Ozkaynak; Anthony J. Coyle

The inducible costimulatory molecule (ICOS) is expressed on activated T cells and participates in a variety of important immunoregulatory functions. After the induction of experimental allergic encephalomyelitis in SJL mice with proteolipid protein (PLP), brain ICOS mRNA and protein were up-regulated on infiltrating CD3+ T cells before disease onset. ICOS blockade during the efferent immune response (9–20 days after immunization) abrogated disease, but blockade during antigen priming (1–10 days after immunization) exacerbated disease. Upon culture with PLP and compared with immunized controls, splenocytes produced either decreased interferon-γ (IFN-γ, in efferent blockade) or excessive IFN-γ (in priming blockade). PLP-specific immunoglobulin G1 was decreased in animals treated with anti-ICOS during antigen priming, but not in other groups.


Journal of Immunology | 2002

Programmed Death-1 Targeting Can Promote Allograft Survival

Engin Ozkaynak; Liqing Wang; Andrew D. J. Goodearl; Kevin Mcdonald; Shixin Qin; Theresa O'keefe; Thao Duong; Tammy Smith; Jose-Carlos Gutierrez-Ramos; James B. Rottman; Anthony J. Coyle; Wayne W. Hancock

The recently identified CD28 homolog and costimulatory molecule programmed death-1 (PD-1) and its ligands, PD-L1 and PD-L2, which are homologs of B7, constitute an inhibitory regulatory pathway of potential therapeutic use in immune-mediated diseases. We examined the expression and functions of PD-1 and its ligands in experimental cardiac allograft rejection. In initial studies, we found that most normal tissues and cardiac isografts had minimal expression of PD-1, PD-L1, or PD-L2, but intragraft induction of all three molecules occurred during development of cardiac allograft rejection. Intragraft expression of all three genes was maintained despite therapy with cyclosporin A or rapamycin, but was prevented in the early posttransplant period by costimulation blockade using CD154 or anti-inducible costimulator mAb. We prepared PD-L1.Ig and PD-L2.Ig fusion proteins and showed that each bound to activated PD-1+ T cells and inhibited T cell functions in vitro, thereby allowing us to test the effects of PD-1 targeting on allograft survival in vivo. Neither agent alone modulated allograft rejection in wild-type recipients. However, use of PD-L1.Ig administration in CD28−/− recipients, or in conjunction with immunosuppression in fully MHC-disparate combinations, markedly prolonged cardiac allograft survival, in some cases causing permanent engraftment, and was accompanied by reduced intragraft expression of IFN-γ and IFN-γ-induced chemokines. PD-L1.Ig use also prevented development of transplant arteriosclerosis post-CD154 mAb therapy. These data show that when combined with limited immunosuppression, or in the context of submaximal TCR or costimulatory signals, targeting of PD-1 can block allograft rejection and modulate T and B cell-dependent pathologic immune responses in vivo.


Biochemical and Biophysical Research Communications | 1991

Murine osteogenic protein (OP-1): High levels of mRNA in kidney

Engin Ozkaynak; Patrick N.J. Schnegelsberg; Hermann Oppermann

The murine OP-1 gene (EMBL accession No. X56906) encoding the homolog of human osteogenic protein-1 was isolated from cDNA and genomic libraries using human OP-1 cDNA as probe. The deduced murine OP-1 amino acid sequence revealed 11 amino acids changes, three of them in the mature protein. Murine OP-1 probes were used for analysis of OP-1 mRNA in mouse embryo and organ tissues. Northern blot hybridization revealed multiple mRNA species: the major species of 2.2 kb, minor species of 1.8 and 2.4 kb and a large 4 kb species, which may represent alternative splices. Tissue specific expression was studied in brain, lung, heart, liver, spleen, kidney, adrenal and bladder tissue. Maximal levels of OP-1 mRNA were found in kidney which may explain the phenomenon of epithelial osteogenesis, first described by Huggins in 1931 using epithelium from the urinary tract. Moreover, our data suggest that kidneys may be the main site of OP-1 synthesis, even though it is distant from its physiological site of action, skeletal bone.


Transplantation | 2005

Systemic transforming growth factor-β1 gene therapy induces Foxp3+ regulatory cells, restores self-tolerance, and facilitates regeneration of beta cell function in overtly diabetic nonobese diabetic mice

Xunrong Luo; Hua Yang; Il Soo Kim; Fludd Saint-Hilaire; Dolca Thomas; Bishnu P. De; Engin Ozkaynak; Thangamani Muthukumar; Wayne W. Hancock; Ronald G. Crystal; Manikkam Suthanthiran

Background. Type 1 diabetes results from auto-aggressive T–cell-mediated destruction of beta cells of the pancreas. Recent data suggest that restoration of self-tolerance may facilitate islet-cell regeneration/recovery. In view of the immunoregulatory activity of transforming growth factor (TGF)-&bgr;1, we investigated whether systemic TGF-&bgr;1 gene therapy blocks islet destructive autoimmunity and facilitates regeneration of beta-cell function in overtly diabetic nonobese diabetic (NOD) mice. Methods. We used site-directed mutagenesis to create cysteine to serine mutation at sites 224 and 226 and constructed a replication deficient adenovirus (Ad) vector encoding active form of human TGF-&bgr;1 (Ad-hTGF-&bgr;1). Overtly diabetic NOD mice received intravenous injection of Ad-hTGF-&bgr;1. Seven to 14 days after the injection, the mice received transplants with 500 syngeneic islets under the kidney capsule. Islet-graft survival and regeneration of endogenous beta-cell function were examined. Results. Syngeneic islet grafts failed by day 17 in all untreated mice, whereas Ad-hTGF-&bgr;1 therapy prolonged survival of islet grafts. Islet grafts from treated mice showed well-preserved islets with a peri-islet infiltrate primarily of CD4+ T cells and expression of CD25 and Foxp3. Systemic TGF-&bgr;1 gene therapy was associated with islet regeneration in the native pancreas. Native pancreas of treated mice revealed islets staining strongly for insulin. Similar to what was found in the syngeneic islet graft, there were well-demarcated peri-islet infiltrates that were positive for CD4, TGF-&bgr;1, and Foxp3. Conclusions. Our data demonstrate that systemic TGF-&bgr;1 gene therapy blocks islet destructive autoimmunity, facilitates islet regeneration, and cures diabetes in diabetic NOD mice.


Infection and Immunity | 2003

Disruption of the ICOS-B7RP-1 costimulatory pathway leads to enhanced hepatic immunopathology and increased gamma interferon production by CD4 T cells in murine schistosomiasis.

Laura I. Rutitzky; Engin Ozkaynak; James B. Rottman

ABSTRACT Morbidity and mortality in schistosomiasis are largely due to an immune response mediated by CD4 T lymphocytes. Since lymphocyte activation is shaped by costimulatory signals, the specific functions of different costimulatory pathways are of increasing interest. We now examined the role of the inducible costimulatory molecule (ICOS) and its ligand B7-related protein 1 (B7RP-1) in the experimental murine schistosome infection by blocking this costimulatory pathway with monoclonal antibody against ICOS, administered daily by intraperitoneal injection during the patent phase of the disease. The treated mice exhibited enhanced hepatic immunopathology characterized by enlarged egg granulomas and pronounced parenchymal inflammation with hepatocellular necrosis, resulting in elevated liver enzyme levels in serum. Most strikingly, there was a sharp increase in gamma interferon (IFN-γ) production by schistosome egg antigen-stimulated granuloma cells, bulk mesenteric lymph node (MLN) cells, and purified MLN CD4 T cells, which contrasted with a more discreet change in the Th2-type cytokines interleukin 4 (IL-4) and IL-10. These findings suggest that the ICOS-B7RP-1 costimulatory pathway serves primarily to control IFN-γ production, thereby promoting a cytokine environment conducive to limited hepatic damage.


Transplantation | 2006

Alloimmunity in Primate Heart Recipients with Cd154 Blockade: Evidence for Alternative Costimulation Mechanisms

Agnes M. Azimzadeh; Steffen Pfeiffer; Guosheng Wu; Carsten Schröder; George L. Zorn; Shahrooz S. Kelishadi; Engin Ozkaynak; Marilyn R. Kehry; James B. Atkinson; Geraldine G. Miller; Richard N. Pierson

Background. CD154 mediates key facets of humoral and cellular immunity to alloantigens, and is tolerogenic to influenza antigens in primates. Barriers to CD154-based tolerance induction for primate cardiac allografts have not previously been defined. Methods. Heterotopic cardiac allograft outcomes in cynomolgus monkeys treated with a CD154 inhibitor, IDEC-131 (n=27), were compared to no treatment (n=4) or cyclosporine A (n=6). Results. CD154 blockade significantly prolonged median allograft survival, from 6.2 (range 6, 7, n=4) days in untreated controls, to 39 (8,112, n=16) days with intensive monotherapy and 93 (>25, 386; n=3) days with added antithymocyte globulin (ATG), but did not yield tolerance. Alloantibody production was delayed but not prevented by IDEC-131 alone or with ATG, and was exacerbated by infusion of donor bone marrow (n=8). Expression of ICOS was prominent in graft infiltrating lymphocytes, and preceded elaboration of antidonor antibody and vasculopathy. Conclusion. CD154 monotherapy modulates primate cardiac alloimmunity, but does not readily induce tolerance. Targeting alternative costimulation pathways, including ICOS, may facilitate tolerance induction based on CD154 blockade.


PLOS ONE | 2009

Three distinct domains contribute to nuclear transport of murine Foxp3.

Wayne W. Hancock; Engin Ozkaynak

Foxp3, a 47-kDa transcription factor, is necessary for the function of CD4+CD25+ regulatory T cells (Tregs), with an essential role in the control of self-reactive T cells and in preventing autoimmunity. Activation of Tregs by TCR engagement results in upregulation of Foxp3 expression, followed by its rapid nuclear transport and binding to chromatin. Here, we identify three distinct Foxp3 domains that contribute to nuclear transport. The first domain (Domain 1) comprises the C-terminal 12 amino acids. The second domain (Domain 2) is located immediately N-terminal to the forkhead domain (FHD), recently reported to be a binding site for the runt-related transcription factor 1/acute myeloid leukemia 1 (Runx1/AML1). The third domain (Domain 3) is located within the N-terminal first 51 amino acids. Unlike the known nuclear localization signals (NLSs), none of these three regions are rich in basic residues and do not bear any similarity to known monopartite or bipartite NLSs that have one or more clusters of basic amino acids. The basic arginine-lysine-lysine-arginine (RKKR) sequence, located 12-aa from the C-terminal end of Foxp3 was previously reported to be a nuclear localization signal (NLS) for several proteins, including for a GFP-Foxp3 hybrid. Evidence is provided here that in the full-length native Foxp3 RKKR does not function as an NLS. The data reported in this study indicates that Foxp3 achieves nuclear transport by binding to other nuclear factors and co-transporting with them to the nucleus.

Collaboration


Dive into the Engin Ozkaynak's collaboration.

Top Co-Authors

Avatar

Wayne W. Hancock

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Liqing Wang

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Andrew D. Wells

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Edwin F. de Zoeten

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chunxia Chen

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Iris Lee

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge