Network


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

Hotspot


Dive into the research topics where Rachel D. Kuns is active.

Publication


Featured researches published by Rachel D. Kuns.


Blood | 2010

An antibody against the colony-stimulating factor 1 receptor depletes the resident subset of monocytes and tissue- and tumor-associated macrophages but does not inhibit inflammation

Kelli P. A. MacDonald; James S. Palmer; Stephen L. Cronau; Elke Seppanen; Stuart D. Olver; Neil C. Raffelt; Rachel D. Kuns; Allison R. Pettit; Andrew D. Clouston; Brandon J. Wainwright; Dan Branstetter; Jeffrey Smith; Raymond J. Paxton; Douglas Pat Cerretti; Lynn Bonham; Geoffrey R. Hill; David A. Hume

The development of the mononuclear phagocyte system requires macrophage colony-stimulating factor (CSF-1) signaling through the CSF-1 receptor (CSF1R, CD115). We examined the effect of an antibody against CSF1R on macrophage homeostasis and function using the MacGreen transgenic mouse (csf1r-enhanced green fluorescent protein) as a reporter. The administration of a novel CSF1R blocking antibody selectively reduced the CD115(+)Gr-1(neg) monocyte precursor of resident tissue macrophages. CD115(+)Gr-1(+) inflammatory monocytes were correspondingly increased, supporting the view that monocytes are a developmental series. Within tissue, the antibody almost completely depleted resident macrophage populations in the peritoneum, gastrointestinal tract, liver, kidney, and skin, but not in the lung or female reproductive organs. CSF1R blockade reduced the numbers of tumor-associated macrophages in syngeneic tumor models, suggesting that these cells are resident type macrophages. Conversely, it had no effect on inflammatory monocyte recruitment in models, including lipopolysaccharide-induced lung inflammation, wound healing, peritonitis, and severe acute graft-versus-host disease. Depletion of resident tissue macrophages from bone marrow transplantation recipients actually resulted in accelerated pathology and exaggerated donor T-cell activation. The data indicate that CSF1R signaling is required only for the maturation and replacement of resident-type monocytes and tissue macrophages, and is not required for monocyte production or inflammatory function.


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.


Journal of Immunology | 2005

Cytokine Expanded Myeloid Precursors Function as Regulatory Antigen-Presenting Cells and Promote Tolerance through IL-10-Producing Regulatory T Cells

Kelli P. A. MacDonald; Vanessa Rowe; Andrew D. Clouston; Joseph K. Welply; Rachel D. Kuns; James L.M. Ferrara; Ranjeny Thomas; Geoffrey R. Hill

The initiation of graft-vs-host disease (GVHD) after stem cell transplantation is dependent on direct Ag presentation by host APCs, whereas the effect of donor APC populations is unclear. We studied the role of indirect Ag presentation in allogenic T cell responses by adding populations of cytokine-expanded donor APC to hemopoietic grafts that would otherwise induce lethal GVHD. Progenipoietin-1 (a synthetic G-CSF/Flt-3 ligand molecule) and G-CSF expanded myeloid dendritic cells (DC), plasmacytoid DC, and a novel granulocyte-monocyte precursor population (GM) that differentiate into class II+,CD80/CD86+,CD40− APC during GVHD. Whereas addition of plasmacytoid and myeloid donor DC augmented GVHD, GM cells promoted transplant tolerance by MHC class II-restricted generation of IL-10-secreting, Ag-specific regulatory T cells. Importantly, although GM cells abrogated GVHD, graft-vs-leukemia effects were preserved. Thus, a population of cytokine-expanded GM precursors function as regulatory APCs, suggesting that G-CSF derivatives may have application in disorders characterized by a loss of self-tolerance.


Journal of Clinical Investigation | 2005

NKT cell–dependent leukemia eradication following stem cell mobilization with potent G-CSF analogs

Edward S. Morris; Kelli P. A. MacDonald; Vanessa Rowe; Tatjana Banovic; Rachel D. Kuns; Alistair L. J. Don; Helen M. Bofinger; Angela C. Burman; Stuart D. Olver; Norbert Kienzle; Steven A. Porcelli; Daniel G. Pellicci; Dale I. Godfrey; Mark J. Smyth; Geoffrey R. Hill

NKT cells have pivotal roles in immune regulation and tumor immunosurveillance. We report that the G-CSF and FMS-like tyrosine kinase 3 ligand (Flt-3L) chimeric cytokine, progenipoietin-1, markedly expands the splenic and hepatic NKT cell population and enhances functional responses to alpha-galactosylceramide. In a murine model of allogeneic stem cell transplantation, donor NKT cells promoted host DC activation and enhanced perforin-restricted CD8+ T cell cytotoxicity against host-type antigens. Following leukemic challenge, donor treatment with progenipoietin-1 significantly improved overall survival when compared with G-CSF or control, attributable to reduced graft-versus-host disease mortality and paradoxical augmentation of graft-versus-leukemia (GVL) effects. Enhanced cellular cytotoxicity was dependent on donor NKT cells, and leukemia clearance was profoundly impaired in recipients of NKT cell-deficient grafts. Enhanced cytotoxicity and GVL effects were not associated with Flt-3L signaling or effects on DCs but were reproduced by prolonged G-CSF receptor engagement with pegylated G-CSF. Thus, modified G-CSF signaling during stem cell mobilization augments NKT cell-dependent CD8+ cytotoxicity, effectively separating graft-versus-host disease and GVL and greatly expanding the potential applicability of allogeneic stem cell transplantation for the therapy of malignant disease.


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.


Journal of Immunology | 2007

Cutting Edge: Conventional Dendritic Cells Are the Critical APC Required for the Induction of Experimental Cerebral Malaria

Saskia deWalick; Fiona H. Amante; Karli A. McSweeney; Louise M. Randall; Amanda C. Stanley; Ashraful Haque; Rachel D. Kuns; Kelli P. A. MacDonald; Geoff R. Hill; Christian R. Engwerda

Cerebral malaria (CM) is a serious complication of Plasmodium falciparum infection, causing significant morbidity and mortality among young children and nonimmune adults in the developing world. Although previous work on experimental CM has identified T cells as key mediators of pathology, the APCs and subsets therein required to initiate immunopathology remain unknown. In this study, we show that conventional dendritic cells but not plasmacytoid dendritic cells are required for the induction of malaria parasite-specific CD4+ T cell responses and subsequent experimental CM. These data have important implications for the development of malaria vaccines and the therapeutic management of CM.


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.


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.

Collaboration


Dive into the Rachel D. Kuns's collaboration.

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

Kate A. Markey

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
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

Katie E. Lineburg

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

Alistair L. J. Don

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge