Network


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

Hotspot


Dive into the research topics where Yana A. Wilson is active.

Publication


Featured researches published by Yana A. Wilson.


Journal of Immunology | 2010

Immune-mediated mechanisms of parasite tissue sequestration during experimental cerebral malaria

Fiona H. Amante; Ashraful Haque; Amanda C. Stanley; Fabian de Labastida Rivera; Louise M. Randall; Yana A. Wilson; Gladys Yeo; Christian Pieper; Brendan S. Crabb; Tania F. de Koning-Ward; Rachel J. Lundie; Michael F. Good; Alberto Pinzon-Charry; Mark S. Pearson; Mary Duke; Donald P McManus; Alex Loukas; Geoff R. Hill; Christian R. Engwerda

Cerebral malaria is a severe complication of malaria. Sequestration of parasitized RBCs in brain microvasculature is associated with disease pathogenesis, but our understanding of this process is incomplete. In this study, we examined parasite tissue sequestration in an experimental model of cerebral malaria (ECM). We show that a rapid increase in parasite biomass is strongly associated with the induction of ECM, mediated by IFN-γ and lymphotoxin α, whereas TNF and IL-10 limit this process. Crucially, we discovered that host CD4+ and CD8+ T cells promote parasite accumulation in vital organs, including the brain. Modulation of CD4+ T cell responses by helminth coinfection amplified CD4+ T cell-mediated parasite sequestration, whereas vaccination could generate CD4+ T cells that reduced parasite biomass and prevented ECM. These findings provide novel insights into immune-mediated mechanisms of ECM pathogenesis and highlight the potential of T cells to both prevent and promote infectious diseases.


Nature Medicine | 2012

Recipient nonhematopoietic antigen-presenting cells are sufficient to induce lethal acute graft-versus-host disease

Motoko Koyama; Rachel D. Kuns; Stuart D. Olver; Neil C. Raffelt; Yana A. Wilson; Alistair L. J. Don; Katie E. Lineburg; Melody Cheong; Renee J. Robb; Kate A. Markey; Antiopi Varelias; Bernard Malissen; Günter J. Hämmerling; Andrew D. Clouston; Christian R. Engwerda; Purnima Bhat; Kelli P. A. MacDonald; Geoffrey R. Hill

The presentation pathways by which allogeneic peptides induce graft-versus-host disease (GVHD) are unclear. We developed a bone marrow transplant (BMT) system in mice whereby presentation of a processed recipient peptide within major histocompatibility complex (MHC) class II molecules could be spatially and temporally quantified. Whereas donor antigen presenting cells (APCs) could induce lethal acute GVHD via MHC class II, recipient APCs were 100–1,000 times more potent in this regard. After myeloablative irradiation, T cell activation and memory differentiation occurred in lymphoid organs independently of alloantigen. Unexpectedly, professional hematopoietic-derived recipient APCs within lymphoid organs had only a limited capacity to induce GVHD, and dendritic cells were not required. In contrast, nonhematopoietic recipient APCs within target organs induced universal GVHD mortality and promoted marked alloreactive donor T cell expansion within the gastrointestinal tract and inflammatory cytokine generation. These data challenge current paradigms, suggesting that experimental lethal acute GVHD can be induced by nonhematopoietic recipient APCs.


Blood | 2010

Stem cell mobilization with G-CSF induces type 17 differentiation and promotes scleroderma

Geoffrey R. Hill; Stuart D. Olver; Rachel D. Kuns; Antiopi Varelias; Neil C. Raffelt; Alistair L. J. Don; Kate A. Markey; Yana A. Wilson; Mark J. Smyth; Yoichiro Iwakura; Joel Tocker; Andrew D. Clouston; Kelli P. A. MacDonald

The recent shift to the use of stem cells mobilized by granulocyte colony-stimulating factor (G-CSF) for hematopoietic transplantation has increased chronic graftversus-host disease (GVHD), although the mechanisms of this are unclear. We have found that G-CSF invokes potent type 17 rather than type 1 or type 2 differentiation. The amplification of interleukin-17 (IL-17) production by G-CSF occurs in both CD4 and CD8 conventional T cells and is dependent on, and downstream of, G-CSF-induced IL-21 signaling. Importantly, donor IL-17A controls the infiltration of macrophages into skin and cutaneous fibrosis, manifesting late after transplantation as scleroderma. Interestingly, donor CD8 T cells were the predominant source of IL-17A after transplantation and could mediate scleroderma independently of CD4 T cells. This study provides a logical explanation for the propensity of allogeneic stem cell transplantation to invoke sclerodermatous GVHD and suggests a therapeutic strategy for intervention.


Blood | 2012

Identification and expansion of highly suppressive CD8 +FoxP3 + regulatory T cells after experimental allogeneic bone marrow transplantation

Renee J. Robb; Katie E. Lineburg; Rachel D. Kuns; Yana A. Wilson; Neil C. Raffelt; Stuart D. Olver; Antiopi Varelias; Kylie A. Alexander; Bianca E. Teal; Tim Sparwasser; Günter J. Hämmerling; Kate A. Markey; Motoko Koyama; Andrew D. Clouston; Christian R. Engwerda; Geoffrey R. Hill; Kelli P. A. MacDonald

FoxP3(+) confers suppressive properties and is confined to regulatory T cells (T(reg)) that potently inhibit autoreactive immune responses. In the transplant setting, natural CD4(+) T(reg) are critical in controlling alloreactivity and the establishment of tolerance. We now identify an important CD8(+) population of FoxP3(+) T(reg) that convert from CD8(+) conventional donor T cells after allogeneic but not syngeneic bone marrow transplantation. These CD8(+) T(reg) undergo conversion in the mesenteric lymph nodes under the influence of recipient dendritic cells and TGF-β. Importantly, this population is as important for protection from GVHD as the well-studied natural CD4(+)FoxP3(+) population and is more potent in exerting class I-restricted and antigen-specific suppression in vitro and in vivo. Critically, CD8(+)FoxP3(+) T(reg) are exquisitely sensitive to inhibition by cyclosporine but can be massively and specifically expanded in vivo to prevent GVHD by coadministering rapamycin and IL-2 antibody complexes. CD8(+)FoxP3(+) T(reg) thus represent a new regulatory population with considerable potential to preferentially subvert MHC class I-restricted T-cell responses after bone marrow transplantation.


Blood | 2009

Conventional dendritic cells are the critical donor APC presenting alloantigen after experimental bone marrow transplantation

Kate A. Markey; Tatjana Banovic; Rachel D. Kuns; Stuart D. Olver; Alistair L. J. Don; Neil C. Raffelt; Yana A. Wilson; Liza J. Raggatt; Allison R. Pettit; Jonathan S. Bromberg; Geoffrey R. Hill; Kelli P. A. MacDonald

We have quantified the relative contribution of donor antigen-presenting cell populations to alloantigen presentation after bone marrow transplantation (BMT) by using transgenic T cells that can respond to host-derived alloantigen presented within the donor major histocompatibility complex. We also used additional transgenic/knockout donor mice and/or monoclonal antibodies that allowed conditional depletion of conventional dendritic cells (cDCs), plasmacytoid DC (pDCs), macrophages, or B cells. Using these systems, we demonstrate that donor cDCs are the critical population presenting alloantigen after BMT, whereas pDCs and macrophages do not make a significant contribution in isolation. In addition, alloantigen presentation was significantly enhanced in the absence of donor B cells, confirming a regulatory role for these cells early after transplantation. These data have major implications for the design of therapeutic strategies post-BMT, and suggest that cDC depletion and the promotion of B-cell reconstitution may be beneficial tools for the control of alloreactivity.


Nature Medicine | 2009

Induction of natural killer T cell–dependent alloreactivity by administration of granulocyte colony–stimulating factor after bone marrow transplantation

Edward S. Morris; Kelli P. A. MacDonald; Rachel D. Kuns; Helen M Morris; Tatjana Banovic; Alistair L. J. Don; Vanessa Rowe; Yana A. Wilson; Neil C. Raffelt; Christian R. Engwerda; Angela C. Burman; Kate A. Markey; Dale I. Godfrey; Mark J. Smyth; Geoffrey R. Hill

Granulocyte colony–stimulating factor (G-CSF) is often used to hasten neutrophil recovery after allogeneic bone marrow transplantation (BMT), but the clinical and immunological consequences evoked remain unclear. We examined the effect of G-CSF administration after transplantation in mouse models and found that exposure to either standard G-CSF or pegylated-G-CSF soon after BMT substantially increased graft-versus-host disease (GVHD). This effect was dependent on total body irradiation (TBI) rendering host dendritic cells (DCs) responsive to G-CSF by upregulating their expression of the G-CSF receptor. Stimulation of host DCs by G-CSF subsequently unleashed a cascade of events characterized by donor natural killer T cell (NKT cell) activation, interferon-γ secretion and CD40-dependent amplification of donor cytotoxic T lymphocyte function during the effector phase of GVHD. Crucially, the detrimental effects of G-CSF were only present when it was administered after TBI conditioning and at a time when residual host antigen presenting cells were still present, perhaps explaining the conflicting and somewhat controversial clinical studies from the large European and North American BMT registries. These data have major implications for the use of G-CSF in disease states where NKT cell activation may have effects on outcome.


The Journal of Infectious Diseases | 2011

Disseminated Varicella Infection Caused by Varicella Vaccine Strain in a Child With Low Invariant Natural Killer T Cells and Diminished CD1d Expression

Tatjana Banovic; Mayonelo Yanilla; Russell Simmons; Ian Robertson; Wayne A. Schroder; Neil C. Raffelt; Yana A. Wilson; Geoffrey R. Hill; Patrick Hogan; Clare Nourse

BACKGROUND Live attenuated varicella vaccine is considered a safe vaccine with serious adverse effects reported only in immunocompromised children. We describe a severe life-threatening infection with varicella vaccine virus causing rash and pneumonitis in a 6-year-old boy with no apparent immunodeficiency. METHODS AND RESULTS Polymerase chain reaction (PCR) analysis of vesicle swab samples demonstrated varicella zoster virus (VZV). Sequencing of the PCR product demonstrated 100% homology with human herpesvirus 3 strain VZV-Oka ORF62 gene. Routine immunologic investigations failed to demonstrate any abnormality. Total leukocyte, lymphocyte, and neutrophil counts and lymphocyte subsets were normal. Immunoglobulins, C3, C4, and CH50 were intact. Specific IgG to protein and polysaccharide antigens and to Epstein-Barr virus and cytomegalovirus were present. Normal lymphocyte proliferation to phytohemagglutinin and VZV antigens was detected. Neutrophil function and natural killer (NK) cell activity were normal. The analysis of invariant NK T (iNKT) cell numbers and function revealed diminished iNKT cells, reported once previously and unique to our patient, deficient expression of the cognate receptor, CD1d. CONCLUSIONS This report provides a further link between deficiency of the iNKT/CD1d pathway and increased susceptibility to varicella vaccine virus, suggesting an important role of this innate pathway in host defense against yet another member of the herpesvirus family.


Blood | 2011

Type I-IFNs control GVHD and GVL responses after transplantation

Renee J. Robb; Ellen Kreijveld; Rachel D. Kuns; Yana A. Wilson; Stuart D. Olver; Alistair L. J. Don; Neil C. Raffelt; Nicole Anne De Weerd; Katie E. Lineburg; Antiopi Varelias; Kate A. Markey; Motoko Koyama; Andrew D. Clouston; Paul J. Hertzog; Kelli P. A. MacDonald; Geoffrey R. Hill

Although the effects of type II-IFN (IFN-γ) on GVHD and leukemia relapse are well studied, the effects of type I-interferon (type I-IFN, IFN-α/β) remain unclear. We investigated this using type I-IFN receptor-deficient mice and exogenous IFN-α administration in established models of GVHD and GVL. Type I-IFN signaling in host tissue prevented severe colon-targeted GVHD in CD4-dependent models of GVHD directed toward either major histocompatibility antigens or multiple minor histocompatibility antigens. This protection was the result of suppression of donor CD4(+) T-cell proliferation and differentiation. Studies in chimeric recipients demonstrated this was due to type I-IFN signaling in hematopoietic tissue. Consistent with this finding, administration of IFN-α during conditioning inhibited donor CD4(+) proliferation and differentiation. In contrast, CD8-dependent GVHD and GVL effects were enhanced when type I-IFN signaling was intact in the host or donor, respectively. This finding reflected the ability of type I-IFN to both sensitize host target tissue/leukemia to cell-mediated cytotoxicity and augment donor CTL function. These data confirm that type I-IFN plays an important role in defining the balance of GVHD and GVL responses and suggests that administration of the cytokine after BM transplantation could be studied prospectively in patients at high risk of relapse.


Blood | 2010

Soluble lymphotoxin is an important effector molecule in GVHD and GVL

Kate A. Markey; Angela C. Burman; Tatjana Banovic; Rachel D. Kuns; Neil C. Raffelt; Rowe; Stuart D. Olver; Alj Don; Edward S. Morris; Allison R. Pettit; Yana A. Wilson; Renee J. Robb; Louise M. Randall; Heinrich Körner; Christian R. Engwerda; Andrew D. Clouston; Kpa MacDonald; Geoffrey R. Hill

Tumor necrosis factor (TNF) is a key cytokine in the effector phase of graft-versus-host disease (GVHD) after bone marrow transplantation, and TNF inhibitors have shown efficacy in clinical and experimental GVHD. TNF signals through the TNF receptors (TNFR), which also bind soluble lymphotoxin (LTalpha3), a TNF family member with a previously unexamined role in GVHD pathogenesis. We have used preclinical models to investigate the role of LT in GVHD. We confirm that grafts deficient in LTalpha have an attenuated capacity to induce GVHD equal to that seen when grafts lack TNF. This is not associated with other defects in cytokine production or T-cell function, suggesting that LTalpha3 exerts its pathogenic activity directly via TNFR signaling. We confirm that donor-derived LTalpha is required for graft-versus-leukemia (GVL) effects, with equal impairment in leukemic clearance seen in recipients of LTalpha- and TNF-deficient grafts. Further impairment in tumor clearance was seen using Tnf/Lta(-/-) donors, suggesting that these molecules play nonredundant roles in GVL. Importantly, donor TNF/LTalpha were only required for GVL where the recipient leukemia was susceptible to apoptosis via p55 TNFR signaling. These data suggest that antagonists neutralizing both TNF and LTalpha3 may be effective for treatment of GVHD, particularly if residual leukemia lacks the p55 TNFR.


Blood | 2010

SOCS3 regulates graft-versus-host disease

Geoffrey R. Hill; Rachel D. Kuns; Neil C. Raffelt; Alistair L. J. Don; Stuart D. Olver; Kate A. Markey; Yana A. Wilson; Joel Tocker; Warren S. Alexander; Andrew D. Clouston; Andrew W. Roberts; Kelli P. A. MacDonald

Suppressor of cytokine signaling-3 (SOCS3) is the main intracellular regulator of signaling by granulocyte colony-stimulating factor, an immune-modulatory cytokine used to mobilize stem cells for transplantation. We have therefore studied the contribution of SOCS3 to the spectrum of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Grafts from SOCS3(-/Deltavav) donor mice in which SOCS3 deficiency is restricted to the hematopoietic compartment had an augmented capacity to induce acute GVHD. With the use of SOCS3(-/DeltaLysM) and SOCS3(-/Deltalck) donors in which SOCS3 deficiency was restricted to the myeloid or T-cell lineage, respectively, we confirmed SOCS3 deficiency promoted acute GVHD mortality and histopathology within the gastrointestinal tract by effects solely within the donor T cell. SOCS3(-/Deltalck) donor T cells underwent enhanced alloantigen-dependent proliferation and generation of interleukin-10 (IL-10), IL-17, and interferon-gamma (IFNgamma) after SCT. The enhanced capacity of the SOCS3(-/Deltalck) donor T cell to induce acute GVHD was dependent on IFNgamma but independent of IL-10 or IL-17. Surprisingly, SOCS3(-/Deltalck) donor T cells also induced severe, transforming growth factor beta- and IFNgamma-dependent, sclerodermatous GVHD. Thus, the delivery of small molecule SOCS3 mimetics may prove to be useful for the inhibition of both acute and chronic GVHD.

Collaboration


Dive into the Yana A. Wilson's collaboration.

Top Co-Authors

Avatar

Neil C. Raffelt

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Rachel D. Kuns

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Kate A. Markey

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Stuart D. Olver

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Alistair L. J. Don

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Geoffrey R. Hill

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antiopi Varelias

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Christian R. Engwerda

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge