Network


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

Hotspot


Dive into the research topics where Kylie R. James is active.

Publication


Featured researches published by Kylie R. James.


Journal of Clinical Investigation | 2014

Type I IFN signaling in CD8– DCs impairs Th1-dependent malaria immunity

Ashraful Haque; Shannon E. Best; Marcela Montes de Oca; Kylie R. James; Anne Ammerdorffer; Chelsea L. Edwards; Fabian de Labastida Rivera; Fiona H. Amante; Patrick T. Bunn; Meru Sheel; Ismail Sebina; Motoko Koyama; Antiopi Varelias; Paul J. Hertzog; Ulrich Kalinke; Sin Yee Gun; Laurent Rénia; Christiane Ruedl; Kelli P. A. MacDonald; Geoffrey R. Hill; Christian R. Engwerda

Many pathogens, including viruses, bacteria, and protozoan parasites, suppress cellular immune responses through activation of type I IFN signaling. Recent evidence suggests that immune suppression and susceptibility to the malaria parasite, Plasmodium, is mediated by type I IFN; however, it is unclear how type I IFN suppresses immunity to blood-stage Plasmodium parasites. During experimental severe malaria, CD4+ Th cell responses are suppressed, and conventional DC (cDC) function is curtailed through unknown mechanisms. Here, we tested the hypothesis that type I IFN signaling directly impairs cDC function during Plasmodium infection in mice. Using cDC-specific IFNAR1-deficient mice, and mixed BM chimeras, we found that type I IFN signaling directly affects cDC function, limiting the ability of cDCs to prime IFN-γ-producing Th1 cells. Although type I IFN signaling modulated all subsets of splenic cDCs, CD8- cDCs were especially susceptible, exhibiting reduced phagocytic and Th1-promoting properties in response to type I IFNs. Additionally, rapid and systemic IFN-α production in response to Plasmodium infection required type I IFN signaling in cDCs themselves, revealing their contribution to a feed-forward cytokine-signaling loop. Together, these data suggest abrogation of type I IFN signaling in CD8- splenic cDCs as an approach for enhancing Th1 responses against Plasmodium and other type I IFN-inducing pathogens.


PLOS ONE | 2013

Schistosoma japonicum Eggs Induce a Proinflammatory, Anti-Fibrogenic Phenotype in Hepatic Stellate Cells

Barrie J. Anthony; Kylie R. James; Geoffrey N. Gobert; Grant A. Ramm; Donald P. McManus

Hepatic fibrosis induced by egg deposition is the most serious pathology associated with chronic schistosomiasis, in which the hepatic stellate cell (HSC) plays a central role. While the effect of Schistosoma mansoni eggs on the fibrogenic phenotype of HSCs has been investigated, studies determining the effect of eggs of S . japonicum on HSCs are lacking. Disease caused by S . japonicum is much more severe than that resulting from S. mansoni infection so it is important to compare the pathologies caused by these two parasites, to determine whether this phenotype is due to the species interacting differently with the mammalian host. Accordingly, we investigated the effect of S . japonicum eggs on the human HSC cell line, LX-2, with and without TGF-β (Transforming Growth Factor beta) co-treatment, so as to determine the impact on genes associated with fibrogenesis, inflammation and matrix re-organisation. Activation status of HSCs was assessed by αSMA (Alpha Smooth Muscle Actin) immunofluorescence, accumulation of Oil Red O-stained lipid droplets and the relative expression of selected genes associated with activation. The fibrogenic phenotype of HSCs was inhibited by the presence of eggs both with or without TGF-β treatment, as evidenced by a lack of αSMA staining and reduced gene expression of αSMA and Col1A1 (Collagen 1A1). Unlike S. mansoni-treated cells, however, expression of the quiescent HSC marker PPAR-γ (Peroxisome Proliferator-Activated Receptor gamma) was not increased, nor was there accumulation of lipid droplets. In contrast, S . japonicum eggs induced the mRNA expression of MMP-9 (Matrix Metalloproteinase 9), CCL2 (Chemokine (C-C motif) Ligand 2) and IL-6 (Interleukin 6) in HSCs indicating that rather than inducing complete HSC quiescence, the eggs induced a proinflammatory phenotype. These results suggest HSCs in close proximity to S . japonicum eggs in the liver may play a role in the proinflammatory regulation of hepatic granuloma formation.


Parasite Immunology | 2017

IL-6 promotes CD4+ T-cell and B-cell activation during Plasmodium infection

Ismail Sebina; Lily G. Fogg; Kylie R. James; Megan S. F. Soon; Jasmin Akter; Bryce S. Thomas; Geoffrey R. Hill; Christian R. Engwerda; Ashraful Haque

Humoral immunity develops in the spleen during blood‐stage Plasmodium infection. This elicits parasite‐specific IgM and IgG, which control parasites and protect against malaria. Studies in mice have elucidated cells and molecules driving humoral immunity to Plasmodium, including CD4+ T cells, B cells, interleukin (IL)‐21 and ICOS. IL‐6, a cytokine readily detected in Plasmodium‐infected mice and humans, is recognized in other systems as a driver of humoral immunity. Here, we examined the effect of infection‐induced IL‐6 on humoral immunity to Plasmodium. Using P. chabaudi chabaudi AS (PcAS) infection of wild‐type and IL‐6−/− mice, we found that IL‐6 helped to control parasites during primary infection. IL‐6 promoted early production of parasite‐specific IgM but not IgG. Notably, splenic CD138+ plasmablast development was more dependent on IL‐6 than germinal centre (GC) B‐cell differentiation. IL‐6 also promoted ICOS expression by CD4+ T cells, as well as their localization close to splenic B cells, but was not required for early Tfh‐cell development. Finally, IL‐6 promoted parasite control, IgM and IgG production, GC B‐cell development and ICOS expression by Tfh cells in a second model, Py17XNL infection. IL‐6 promotes CD4+ T‐cell activation and B‐cell responses during blood‐stage Plasmodium infection, which encourages parasite‐specific antibody production.


PLOS Pathogens | 2016

IFNAR1-Signalling Obstructs ICOS-mediated Humoral Immunity during Non-lethal Blood-Stage Plasmodium Infection

Ismail Sebina; Kylie R. James; Megan S. F. Soon; Lily G. Fogg; Shannon E. Best; Fabian de Labastida Rivera; Marcela Montes de Oca; Fiona H. Amante; Bryce S. Thomas; Lynette Beattie; Fernando Souza-Fonseca-Guimaraes; Mark J. Smyth; Paul J. Hertzog; Geoffrey R. Hill; Andreas Hutloff; Christian R. Engwerda; Ashraful Haque

Parasite-specific antibodies protect against blood-stage Plasmodium infection. However, in malaria-endemic regions, it takes many months for naturally-exposed individuals to develop robust humoral immunity. Explanations for this have focused on antigenic variation by Plasmodium, but have considered less whether host production of parasite-specific antibody is sub-optimal. In particular, it is unclear whether host immune factors might limit antibody responses. Here, we explored the effect of Type I Interferon signalling via IFNAR1 on CD4+ T-cell and B-cell responses in two non-lethal murine models of malaria, P. chabaudi chabaudi AS (PcAS) and P. yoelii 17XNL (Py17XNL) infection. Firstly, we demonstrated that CD4+ T-cells and ICOS-signalling were crucial for generating germinal centre (GC) B-cells, plasmablasts and parasite-specific antibodies, and likewise that T follicular helper (Tfh) cell responses relied on B cells. Next, we found that IFNAR1-signalling impeded the resolution of non-lethal blood-stage infection, which was associated with impaired production of parasite-specific IgM and several IgG sub-classes. Consistent with this, GC B-cell formation, Ig-class switching, plasmablast and Tfh differentiation were all impaired by IFNAR1-signalling. IFNAR1-signalling proceeded via conventional dendritic cells, and acted early by limiting activation, proliferation and ICOS expression by CD4+ T-cells, by restricting the localization of activated CD4+ T-cells adjacent to and within B-cell areas of the spleen, and by simultaneously suppressing Th1 and Tfh responses. Finally, IFNAR1-deficiency accelerated humoral immune responses and parasite control by boosting ICOS-signalling. Thus, we provide evidence of a host innate cytokine response that impedes the onset of humoral immunity during experimental malaria.


Infection and Immunity | 2014

Effect of Mature Blood-Stage Plasmodium Parasite Sequestration on Pathogen Biomass in Mathematical and In Vivo Models of Malaria

David S. Khoury; Deborah Cromer; Shannon E. Best; Kylie R. James; P. Kim; Christian R. Engwerda; Ashraful Haque; Miles P. Davenport

ABSTRACT Parasite biomass and microvasculature obstruction are strongly associated with disease severity and death in Plasmodium falciparum-infected humans. This is related to sequestration of mature, blood-stage parasites (schizonts) in peripheral tissue. The prevailing view is that schizont sequestration leads to an increase in pathogen biomass, yet direct experimental data to support this are lacking. Here, we first studied parasite population dynamics in inbred wild-type (WT) mice infected with the rodent species of malaria, Plasmodium berghei ANKA. As is commonly reported, these mice became moribund due to large numbers of parasites in multiple tissues. We then studied infection dynamics in a genetically targeted line of mice, which displayed minimal tissue accumulation of parasites. We constructed a mathematical model of parasite biomass dynamics, incorporating schizont-specific host clearance, both with and without schizont sequestration. Combined use of mathematical and in vivo modeling indicated, first, that the slowing of parasite growth in the genetically targeted mice can be attributed to specific clearance of schizonts from the circulation and, second, that persistent parasite growth in WT mice can be explained solely as a result of schizont sequestration. Our work provides evidence that schizont sequestration could be a major biological process driving rapid, early increases in parasite biomass during blood-stage Plasmodium infection.


Journal of Immunology | 2014

Tissue Requirements for Establishing Long-Term CD4+ T Cell–Mediated Immunity following Leishmania donovani Infection

Patrick T. Bunn; Amanda C. Stanley; Fabian de Labastida Rivera; Alexander Mulherin; Meru Sheel; Clare E. Alexander; Rebecca J. Faleiro; Fiona H. Amante; Marcela Montes de Oca; Shannon E. Best; Kylie R. James; Paul M. Kaye; Ashraful Haque; Christian R. Engwerda

Organ-specific immunity is a feature of many infectious diseases, including visceral leishmaniasis caused by Leishmania donovani. Experimental visceral leishmaniasis in genetically susceptible mice is characterized by an acute, resolving infection in the liver and chronic infection in the spleen. CD4+ T cell responses are critical for the establishment and maintenance of hepatic immunity in this disease model, but their role in chronically infected spleens remains unclear. In this study, we show that dendritic cells are critical for CD4+ T cell activation and expansion in all tissue sites examined. We found that FTY720-mediated blockade of T cell trafficking early in infection prevented Ag-specific CD4+ T cells from appearing in lymph nodes, but not the spleen and liver, suggesting that early CD4+ T cell priming does not occur in liver-draining lymph nodes. Extended treatment with FTY720 over the first month of infection increased parasite burdens, although this associated with blockade of lymphocyte egress from secondary lymphoid tissue, as well as with more generalized splenic lymphopenia. Importantly, we demonstrate that CD4+ T cells are required for the establishment and maintenance of antiparasitic immunity in the liver, as well as for immune surveillance and suppression of parasite outgrowth in chronically infected spleens. Finally, although early CD4+ T cell priming appeared to occur most effectively in the spleen, we unexpectedly revealed that protective CD4+ T cell–mediated hepatic immunity could be generated in the complete absence of all secondary lymphoid tissues.


European Journal of Immunology | 2015

Spatiotemporal requirements for IRF7 in mediating type I IFN-dependent susceptibility to blood-stage Plasmodium infection

Chelsea L. Edwards; Shannon E. Best; Sin Yee Gun; Carla Claser; Kylie R. James; Marcela Montes de Oca; Ismail Sebina; Fabian de Labastida Rivera; Fiona H. Amante; Paul J. Hertzog; Christian R. Engwerda; Laurent Rénia; Ashraful Haque

Type I IFN signaling suppresses splenic T helper 1 (Th1) responses during blood‐stage Plasmodium berghei ANKA (PbA) infection in mice, and is crucial for mediating tissue accumulation of parasites and fatal cerebral symptoms via mechanisms that remain to be fully characterized. Interferon regulatory factor 7 (IRF7) is considered to be a master regulator of type I IFN responses. Here, we assessed IRF7 for its roles during lethal PbA infection and nonlethal Plasmodium chabaudi chabaudi AS (PcAS) infection as two distinct models of blood‐stage malaria. We found that IRF7 was not essential for tissue accumulation of parasites, cerebral symptoms, or brain pathology. Using timed administration of anti‐IFNAR1 mAb, we show that late IFNAR1 signaling promotes fatal disease via IRF7‐independent mechanisms. Despite this, IRF7 significantly impaired early splenic Th1 responses and limited control of parasitemia during PbA infection. Finally, IRF7 also suppressed antiparasitic immunity and Th1 responses during nonlethal PcAS infection. Together, our data support a model in which IRF7 suppresses antiparasitic immunity in the spleen, while IFNAR1‐mediated, but IRF7‐independent, signaling contributes to pathology in the brain during experimental blood‐stage malaria.


Scientific Reports | 2015

Reduced erythrocyte susceptibility and increased host clearance of young parasites slows Plasmodium growth in a murine model of severe malaria

David S. Khoury; Deborah Cromer; Shannon E. Best; Kylie R. James; Ismail Sebina; Ashraful Haque; Miles P. Davenport

The best correlate of malaria severity in human Plasmodium falciparum (Pf) infection is the total parasite load. Pf-infected humans could control parasite loads by two mechanisms, either decreasing parasite multiplication, or increasing parasite clearance. However, few studies have directly measured these two mechanisms in vivo. Here, we have directly quantified host clearance of parasites during Plasmodium infection in mice. We transferred labelled red blood cells (RBCs) from Plasmodium infected donors into uninfected and infected recipients, and tracked the fate of donor parasites by frequent blood sampling. We then applied age-based mathematical models to characterise parasite clearance in the recipient mice. Our analyses revealed an increased clearance of parasites in infected animals, particularly parasites of a younger developmental stage. However, the major decrease in parasite multiplication in infected mice was not mediated by increased clearance alone, but was accompanied by a significant reduction in the susceptibility of RBCs to parasitisation.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Host-mediated impairment of parasite maturation during blood-stage Plasmodium infection

David S. Khoury; Deborah Cromer; Jasmin Akter; Ismail Sebina; Trish Elliott; Bryce S. Thomas; Megan S. F. Soon; Kylie R. James; Shannon E. Best; Ashraful Haque; Miles P. Davenport

Significance Adaptive immunity to Plasmodium falciparum takes years to develop in endemic regions, leaving young children vulnerable to high parasite burdens and severe malaria. Host innate immune responses clearly occur during infection and may control parasite numbers in nonimmune individuals, for example by accelerating parasite removal from circulation. However, evidence of whether and how this occurs in vivo remains sparse. We set out to measure host removal of parasites during acute blood-stage Plasmodium infection in mice. However, rather than being removed more rapidly, parasites unexpectedly persisted in circulation. Persistence resulted from host-dependent slowing of parasite maturation. Thus Plasmodium maturation within red blood cells does not occur at a constant rate in vivo and can be influenced by the host itself. Severe malaria and associated high parasite burdens occur more frequently in humans lacking robust adaptive immunity to Plasmodium falciparum. Nevertheless, the host may partly control blood-stage parasite numbers while adaptive immunity is gradually established. Parasite control has typically been attributed to enhanced removal of parasites by the host, although in vivo quantification of this phenomenon remains challenging. We used a unique in vivo approach to determine the fate of a single cohort of semisynchronous, Plasmodium berghei ANKA- or Plasmodium yoelii 17XNL-parasitized red blood cells (pRBCs) after transfusion into naive or acutely infected mice. As previously shown, acutely infected mice, with ongoing splenic and systemic inflammatory responses, controlled parasite population growth more effectively than naive controls. Surprisingly, however, this was not associated with accelerated removal of pRBCs from circulation. Instead, transfused pRBCs remained in circulation longer in acutely infected mice. Flow cytometric assessment and mathematical modeling of intraerythrocytic parasite development revealed an unexpected and substantial slowing of parasite maturation in acutely infected mice, extending the life cycle from 24 h to 40 h. Importantly, impaired parasite maturation was the major contributor to control of parasite growth in acutely infected mice. Moreover, by performing the same experiments in rag1−/− mice, which lack T and B cells and mount weak inflammatory responses, we revealed that impaired parasite maturation is largely dependent upon the host response to infection. Thus, impairment of parasite maturation represents a host-mediated, immune system-dependent mechanism for limiting parasite population growth during the early stages of an acute blood-stage Plasmodium infection.


Journal of Immunology | 2017

Development of a Novel CD4+ TCR Transgenic Line That Reveals a Dominant Role for CD8+ Dendritic Cells and CD40 Signaling in the Generation of Helper and CTL Responses to Blood-Stage Malaria

Daniel Fernandez-Ruiz; Lei Shong Lau; Nazanin Ghazanfari; Claerwen M. Jones; Wei Yi Ng; Gayle M. Davey; Dorothee Berthold; Lauren E. Holz; Yu Kato; Matthias H. Enders; Ganchimeg Bayarsaikhan; Sanne H. Hendriks; Lianne I. M. Lansink; Jessica A. Engel; Megan S. F. Soon; Kylie R. James; Anton J. Cozijnsen; Vanessa Mollard; Alessandro D. Uboldi; Christopher J. Tonkin; Tania F. de Koning-Ward; Paul R. Gilson; Tsuneyasu Kaisho; Ashraful Haque; Brendan S. Crabb; Francis R. Carbone; Geoffrey I. McFadden; William R. Heath

We describe an MHC class II (I-Ab)–restricted TCR transgenic mouse line that produces CD4+ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4+ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, Plasmodium chabaudi AS, and Plasmodium yoelii 17XNL) and human (Plasmodium falciparum) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of Ab to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8+ T cell–mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8+ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8+ DC (a subset of XCR1+ DC) were the major APC responsible for activation of both T cell subsets, although other DC also contributed to CD4+ T cell responses. Depletion of CD8+ DC at the beginning of infection prevented ECM development and impaired both Th1 and follicular Th cell responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4+ T cell immunity during malaria and provides evidence that CD4+ T cell help, acting via CD40L signaling, can promote immunity or pathology to blood-stage malaria largely through Ag presentation by CD8+ DC.

Collaboration


Dive into the Kylie R. James's collaboration.

Top Co-Authors

Avatar

Ashraful Haque

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Shannon E. Best

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
Top Co-Authors

Avatar

Ismail Sebina

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Megan S. F. Soon

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Fabian de Labastida Rivera

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Fiona H. Amante

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Bryce S. Thomas

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Chelsea L. Edwards

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge