Network


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

Hotspot


Dive into the research topics where Kate A. Markey is active.

Publication


Featured researches published by Kate A. Markey.


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 | 2014

The biology of graft-versus-host disease: experimental systems instructing clinical practice

Kate A. Markey; Kelli P. A. MacDonald; Geoffrey R. Hill

The last 6 decades have seen major advances in the understanding of immunologic diseases, driven by preclinical animal models. Indeed, bone marrow transplantation (BMT) has its genesis in rodent models dating back to the 1950s. Allogeneic BMT and its major complication, graft-versus-host disease (GVHD), represent a paradigm for the translation of preclinical concepts into clinical practice. The appreciation that GVHD can be thought of as a stepwise escalation in immune activation characterized by eventual massive target tissue apoptosis has allowed the design of rational approaches to better manage patients. Here, we describe the pathophysiology of GVHD as defined in preclinical models, focusing on the successes and failures of this research to instruct and translate clinical practice. We also provide a commentary on the limitations of these models so that they may be better appreciated and addressed in future studies. Notable preclinical successes include the definition of modern immune suppression, reductions in conditioning intensity, posttransplant cyclophosphamide, and the promotion of regulatory T-cell reconstitution. New strategies including naïve T-cell depletion, focused cytokine and chemokine inhibition, and the blockade of costimulation now also appear highly promising and very likely to translate into patients in the near future.


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.


Journal of Clinical Investigation | 2014

CSF-1-dependant donor-derived macrophages mediate chronic graft-versus-host disease.

Kylie A. Alexander; Ryan Flynn; Katie E. Lineburg; Rachel D. Kuns; Bianca E. Teal; Stuart D. Olver; Mary Lor; Neil C. Raffelt; Motoko Koyama; Lucie Leveque; Laetitia Le Texier; Michelle Melino; Kate A. Markey; Antiopi Varelias; Christian R. Engwerda; Jonathan S. Serody; Baptiste Janela; Florent Ginhoux; Andrew D. Clouston; Bruce R. Blazar; Geoffrey R. Hill; Kelli P. A. MacDonald

Chronic GVHD (cGVHD) is the major cause of late, nonrelapse death following stem cell transplantation and characteristically develops in organs such as skin and lung. Here, we used multiple murine models of cGVHD to investigate the contribution of macrophage populations in the development of cGVHD. Using an established IL-17-dependent sclerodermatous cGVHD model, we confirmed that macrophages infiltrating the skin are derived from donor bone marrow (F4/80+CSF-1R+CD206+iNOS-). Cutaneous cGVHD developed in a CSF-1/CSF-1R-dependent manner, as treatment of recipients after transplantation with CSF-1 exacerbated macrophage infiltration and cutaneous pathology. Additionally, recipients of grafts from Csf1r-/- mice had substantially less macrophage infiltration and cutaneous pathology as compared with those receiving wild-type grafts. Neither CCL2/CCR2 nor GM-CSF/GM-CSFR signaling pathways were required for macrophage infiltration or development of cGVHD. In a different cGVHD model, in which bronchiolitis obliterans is a prominent manifestation, F4/80+ macrophage infiltration was similarly noted in the lungs of recipients after transplantation, and lung cGVHD was also IL-17 and CSF-1/CSF-1R dependent. Importantly, depletion of macrophages using an anti-CSF-1R mAb markedly reduced cutaneous and pulmonary cGVHD. Taken together, these data indicate that donor macrophages mediate the development of cGVHD and suggest that targeting CSF-1 signaling after transplantation may prevent and treat cGVHD.


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.


Blood | 2013

CD8α+ DCs can be induced in the absence of transcription factors Id2, Nfil3, and Batf3

Cyril Seillet; Jacob T. Jackson; Kate A. Markey; Hugh J. M. Brady; Geoffrey R. Hill; Kelli P. A. MacDonald; Stephen L. Nutt; Gabrielle T. Belz

Antiviral immunity and cross-presentation is mediated constitutively through CD8α+ and CD103+ DCs. Development of these DC subsets is thought to require the transcription factors Irf8, Id2, Nfil3, and Batf3, although how this network is regulated is poorly defined. We addressed the nature of the differentiation blocks observed in the absence of these factors and found that although all 4 factors are required for CD103+ DC development, only Irf8 is essential for CD8α+ DCs. CD8α+ DCs emerged in the absence of Id2, Nfil3 and Batf3 in short-term bone marrow reconstitution. These “induced” CD8α+ DCs exhibit several hallmarks of classic CD8α+ DCs including the expression of CD24, Tlr3, Xcr1, Clec9A, and the capacity to cross-present soluble, cell-associated antigens and viral antigens even in the absence of Batf3. Collectively, these results uncover a previously undescribed pathway by which CD8α+ DCs emerge independent of Id2, Nfil3, and Batf3, but dependent on Irf8.


Journal of Immunology | 2013

Induced regulatory T cells promote tolerance when stabilized by rapamycin and IL-2 in vivo

Ping Zhang; Siok-Keen Tey; Motoko Koyama; Rachel D. Kuns; Stuart D. Olver; Katie E. Lineburg; Mary Lor; Bianca E. Teal; Neil C. Raffelt; Jyothy Raju; Lucie Leveque; Kate A. Markey; Antiopi Varelias; Andrew D. Clouston; Steven W. Lane; Kelli P. A. MacDonald; Geoffrey R. Hill

Natural regulatory T cells (nTregs) play an important role in tolerance; however, the small numbers of cells obtainable potentially limit the feasibility of clinical adoptive transfer. Therefore, we studied the feasibility and efficacy of using murine-induced regulatory T cells (iTregs) for the induction of tolerance after bone marrow transplantation. iTregs could be induced in large numbers from conventional donor CD4 and CD8 T cells within 1 wk and were highly suppressive. During graft-versus-host disease (GVHD), CD4 and CD8 iTregs suppressed the proliferation of effector T cells and the production of proinflammatory cytokines. However, unlike nTregs, both iTreg populations lost Foxp3 expression within 3 wk in vivo, reverted to effector T cells, and exacerbated GVHD. The loss of Foxp3 in iTregs followed homeostatic and/or alloantigen-driven proliferation and was unrelated to GVHD. However, the concurrent administration of rapamycin, with or without IL-2/anti–IL-2 Ab complexes, to the transplant recipients significantly improved Foxp3 stability in CD4 iTregs (and, to a lesser extent, CD8 iTregs), such that they remained detectable 12 wk after transfer. Strikingly, CD4, but not CD8, iTregs could then suppress Teff proliferation and proinflammatory cytokine production and prevent GVHD in an equivalent fashion to nTregs. However, at high numbers and when used as GVHD prophylaxis, Tregs potently suppress graft-versus-leukemia effects and so may be most appropriate as a therapeutic modality to treat GVHD. These data demonstrate that CD4 iTregs can be produced rapidly in large, clinically relevant numbers and, when transferred in the presence of systemic rapamycin and IL-2, induce tolerance in transplant recipients.


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.


Journal of Immunology | 2009

Graft-versus-Host Disease Prevents the Maturation of Plasmacytoid Dendritic Cells

Tatjana Banovic; Kate A. Markey; Rachel D. Kuns; Stuart D. Olver; Neil C. Raffelt; Alistair L. J. Don; Mariapia A. Degli-Esposti; Christian R. Engwerda; Kelli P. A. MacDonald; Geoffrey R. Hill

The role of Ag presenting cell subsets in graft-versus-host disease (GVHD) remains unclear. We have thus examined the ability of plasmacytoid dendritic cells (pDC) to modulate transplant outcome. Surprisingly, host pDC were exquisitely sensitive to total body irradiation and were depleted before transplantation, thus allowing us to focus on donor pDC. The depletion of all pDC from bone marrow grafts resulted in an acceleration of GVHD mortality while the depletion of mature pDC from G-CSF mobilized splenic grafts had no effect. Thus, donor bone marrow pDC, but not mature pDC contained within stem cell grafts attenuate acute GVHD. In the presence of GVHD, donor pDC completely failed to reconstitute although a CD11clow120G8+ precursor DC reconstituted in an exaggerated and transient manner. These cells expressed Flt-3, the macrophage colony stimulating factor receptor and, consistent with a common dendritic cell (DC) precursor, were capable of differentiation into pDC and conventional DC in vivo in the absence of GVHD. These precursors were MHC class II+ and CD80/86+ but lacked CD40, were actively presenting host Ag and inhibited GVHD and T cell proliferation in a contact-dependent fashion. These data demonstrate that GVHD prevents the maturation of pDC and instead promotes the generation of a suppressive precursor DC, further contributing to the state of immune paralysis after transplantation.

Collaboration


Dive into the Kate A. Markey's collaboration.

Top Co-Authors

Avatar

Rachel D. Kuns

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Kelli P. A. MacDonald

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Geoffrey R. Hill

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Neil C. Raffelt

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

Antiopi Varelias

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Motoko Koyama

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Katie E. Lineburg

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alistair L. J. Don

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge