Network


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

Hotspot


Dive into the research topics where Hiu Kiu is active.

Publication


Featured researches published by Hiu Kiu.


Seminars in Cell & Developmental Biology | 2008

SOCS regulation of the JAK/STAT signalling pathway

Ben A. Croker; Hiu Kiu; Sandra E. Nicholson

The suppressor of cytokine signalling (SOCS) proteins were, as their name suggests, first described as inhibitors of cytokine signalling. While their actions clearly now extend to other intracellular pathways, they remain key negative regulators of cytokine and growth factor signalling. In this review we focus on the mechanics of SOCS action and the complexities of the mouse models that have underpinned our current understanding of SOCS biology.


Journal of Clinical Investigation | 2006

SOCS-3 negatively regulates innate and adaptive immune mechanisms in acute IL-1–dependent inflammatory arthritis

Peter K. K. Wong; Paul J. Egan; Ben A. Croker; Kristy O’Donnell; Natalie A. Sims; Sarah F. Drake; Hiu Kiu; Edward J. McManus; Warren S. Alexander; Andrew W. Roberts; Ian P. Wicks

RA is an autoimmune disease characterized by sustained imbalance between pro- and antiinflammatory immune mechanisms. The SOCS proteins are negative regulators of cytokine signaling, but to date there has been little information on their function in disease. The generation of Socs3(-/Delta vav) mice, which lack SOCS-3 in the hematopoietic and endothelial cell compartment, allowed us to explore the role of endogenous SOCS-3 during acute inflammatory arthritis. Joint inflammation in Socs3(-/Delta vav) mice was particularly severe and was characterized by increased numbers of neutrophils in the inflamed synovium, bone marrow, peripheral blood, and spleen. These features were most likely due to increased production of and enhanced responsiveness to G-CSF and IL-6 during arthritis in these mice. Local osteoclast generation and bone destruction were also dramatically increased in the absence of SOCS-3, as was macrophage activation. Finally, SOCS-3 was found to negatively regulate CD4+ T lymphocyte activation, including production of the pleiotropic cytokine IL-17. The absence of SOCS-3 therefore had dramatic effects in this disease model, with a broader impact on cellular responses than SOCS-1 deficiency. These findings provide direct in vivo evidence that endogenous SOCS-3 is a critical negative regulator of multiple cell types orchestrating inflammatory joint disease.


Growth Factors Journal | 2012

Biology and significance of the JAK/STAT signalling pathways

Hiu Kiu; Sandra E. Nicholson

Since its discovery two decades ago, the activation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway by numerous cytokines and growth factors has resulted in it becoming one of the most well-studied intracellular signalling networks. The field has progressed from the identification of the individual components to high-resolution crystal structures of both JAK and STAT, and an understanding of the complexities of the molecular activation and deactivation cycle which results in a diverse, yet highly specific and regulated pattern of transcriptional responses. While there is still more to learn, we now appreciate how disruption and deregulation of this pathway can result in clinical disease and look forward to adoption of the next generation of JAK inhibitors in routine clinical treatment.


Immunology and Cell Biology | 2012

IL-6 promotes acute and chronic inflammatory disease in the absence of SOCS3

Ben A. Croker; Hiu Kiu; Marc Pellegrini; Jesse G. Toe; Simon Preston; Donald Metcalf; Joanne A. O'Donnell; Louise H. Cengia; Kate McArthur; Nicos A. Nicola; Warren S. Alexander; Andrew W. Roberts

The lack of expression of the suppressor of cytokine signalling‐3 (SOCS3) or inactivation of the negative regulatory capacity of SOCS3 has been well documented in rheumatoid arthritis, viral hepatitis and cancer. The specific qualitative and quantitative consequences of SOCS3 deficiency on interleukin‐6 (IL‐6)‐mediated pro‐ and anti‐inflammatory responses remain controversial in vitro and unknown in vivo. Mice with a conditional deletion of SOCS3 in hematopoietic cells develop lethal inflammatory disease during adult life and develop gross histopathological changes during experimental arthritis, typified by elevated IL‐6 levels. To clarify the nature of the IL‐6 responses in vivo, we generated mice deficient in SOCS3 (SOCS3−/Δvav) or both SOCS3 and IL‐6 (IL‐6−/−/SOCS3−/Δvav), and examined responses in models of acute and chronic inflammation. Acute responses to IL‐1β were lethal to SOCS3−/Δvav mice but not IL‐6−/−/SOCS3−/Δvav mice, indicating that IL‐6 was required for the lethal inflammation induced by IL‐1β. Administration of IL‐1β to SOCS3−/Δvav mice induced systemic apoptosis of lymphocytes in the thymus, spleen and lymph nodes that was dependent on the presence of IL‐6. IL‐6 deficiency prolonged survival of SOCS3−/Δvav mice and ameliorated spontaneous inflammatory disease developing during adult life. Infection of SOCS3−/Δvav mice with LCMV induced a lethal inflammatory response that was dependent on IL‐6, despite SOCS3−/Δvav mice controlling viral replication. We conclude that SOCS3 is required for survival during inflammatory responses and is a critical regulator of IL‐6 in vivo.


Immunology and Cell Biology | 2009

SOCS1 negatively regulates the production of Foxp3 + CD4 + T cells in the thymus

Yifan Zhan; Gayle M. Davey; Kate L. Graham; Hiu Kiu; Nadine L. Dudek; Thomas W. H. Kay; Andrew M. Lew

SOCS1 profoundly influences the development and peripheral homeostasis of CD8+ T cells but has less impact on CD4+ T cells. Despite the moderate influence of SOCS1 in the development of the total CD4 T‐cell lineage, we show here that SOCS1 deficiency resulted in a 10‐fold increase in Foxp3+ CD4+ T cells in the thymus. Increased numbers of Foxp3+ thymocytes occurred in mice with T‐cell‐specific ablation of SOCS1, suggesting that the effect is T‐cell intrinsic. This increase in Foxp3+ CD4+cells in SOCS1‐deficient mice also occurred in the absence of IFN‐γ or/and IL‐7 signaling. Increase in CD25+CD4+ T cells in the absence of SOCS1 could be partly due to enhanced survival by CD25+CD4+cells, to a lesser degree CD25−CD4+ T cells, from SOCS1‐deficient mice with or without T‐cell growth factors.


Experimental Hematology | 2008

Socs3 maintains the specificity of biological responses to cytokine signals during granulocyte and macrophage differentiation

Ben A. Croker; Lisa A. Mielke; Samuel Wormald; Donald Metcalf; Hiu Kiu; Warren S. Alexander; Douglas J. Hilton; Andrew W. Roberts

Granulocyte colony-stimulating factor (G-CSF) and interleukin-6 (IL-6) play key roles in regulating emergency granulopoiesis and inflammation, and are both negatively regulated by the inducible intracellular protein suppressor of cytokine signaling-3 (Socs3). Mice with Socs3 deleted specifically in hematopoietic cells succumb to severe neutrophil and macrophage-driven inflammation by 1 year of age, and responses to G-CSF are grossly exacerbated. In order to determine which elements of cellular responses to cytokines require Socs3, we have examined the differentiative and proliferative capacity of hematopoietic progenitor cells stimulated by G-CSF and IL-6. The differentiation of Socs3-deficient progenitor cells is skewed toward macrophage production in response to G-CSF or IL-6, whereas wild-type progenitor cells produce mainly neutrophils. The proliferative capacity of Socs3-deficient progenitor cells is greatly enhanced in response to G-CSF at all concentrations, but only at low concentrations for IL-6. Strikingly, synergistic responses to costimulation with stem cell factor and IL-6 (but not G-CSF) are lost at higher concentrations in Socs3-deficient progenitor cells. Cytokine-induced expression of transcriptional regulators including cebpb, Ets2, Bcl3, c-Myc, Jun, and Fosl2 are differentially regulated in Socs3-deficient cells. The tight regulation by Socs3 of signal transducer and activator of transcription 3 phosphorylation and gene transcription after cytokine receptor ligation significantly influences the fate of myeloid progenitor cells.


PLOS ONE | 2012

Induction of Protective CD4+ T Cell-Mediated Immunity by a Leishmania Peptide Delivered in Recombinant Influenza Viruses

Katherine Kedzierska; Joan M. Curtis; Sophie A. Valkenburg; Lauren A. Hatton; Hiu Kiu; Peter C. Doherty; Lukasz Kedzierski

The available evidence suggests that protective immunity to Leishmania is achieved by priming the CD4+ Th1 response. Therefore, we utilised a reverse genetics strategy to generate influenza A viruses to deliver an immunogenic Leishmania peptide. The single, immunodominant Leishmania-specific LACK158–173 CD4+ peptide was engineered into the neuraminidase stalk of H1N1 and H3N2 influenza A viruses. These recombinant viruses were used to vaccinate susceptible BALB/c mice to determine whether the resultant LACK158–173-specific CD4+ T cell responses protected against live L. major infection. We show that vaccination with influenza-LACK158–173 triggers LACK158–173-specific Th1-biased CD4+ T cell responses within an appropriate cytokine milieu (IFN-γ, IL-12), essential for the magnitude and quality of the Th1 response. A single intraperitoneal exposure (non-replicative route of immunisation) to recombinant influenza delivers immunogenic peptides, leading to a marked reduction (2–4 log) in parasite burden, albeit without reduction in lesion size. This correlated with increased numbers of IFN-γ-producing CD4+ T cells in vaccinated mice compared to controls. Importantly, the subsequent prime-boost approach with a serologically distinct strain of influenza (H1N1->H3N2) expressing LACK158–173 led to a marked reduction in both lesion size and parasite burdens in vaccination trials. This protection correlated with high levels of IFN-γ producing cells in the spleen, which were maintained for 6 weeks post-challenge indicating the longevity of this protective effector response. Thus, these experiments show that Leishmania-derived peptides delivered in the context of recombinant influenza viruses are immunogenic in vivo, and warrant investigation of similar vaccine strategies to generate parasite-specific immunity.


PLOS Genetics | 2015

Early Lineage Priming by Trisomy of Erg Leads to Myeloproliferation in a Down Syndrome Model

Ashley P. Ng; Yifang Hu; Donald Metcalf; Craig D. Hyland; Helen Ierino; Belinda Phipson; Di Wu; Tracey M. Baldwin; Maria Kauppi; Hiu Kiu; Ladina Di Rago; Douglas J. Hilton; Gordon K. Smyth; Warren S. Alexander

Down syndrome (DS), with trisomy of chromosome 21 (HSA21), is the commonest human aneuploidy. Pre-leukemic myeloproliferative changes in DS foetal livers precede the acquisition of GATA1 mutations, transient myeloproliferative disorder (DS-TMD) and acute megakaryocytic leukemia (DS-AMKL). Trisomy of the Erg gene is required for myeloproliferation in the Ts(1716)65Dn DS mouse model. We demonstrate here that genetic changes specifically attributable to trisomy of Erg lead to lineage priming of primitive and early multipotential progenitor cells in Ts(1716)65Dn mice, excess megakaryocyte-erythroid progenitors, and malignant myeloproliferation. Gene expression changes dependent on trisomy of Erg in Ts(1716)65Dn multilineage progenitor cells were correlated with those associated with trisomy of HSA21 in human DS hematopoietic stem and primitive progenitor cells. These data suggest a role for ERG as a regulator of hematopoietic lineage potential, and that trisomy of ERG in the context of DS foetal liver hemopoiesis drives the pre-leukemic changes that predispose to subsequent DS-TMD and DS-AMKL.


Growth Factors Journal | 2009

Regulation of multiple cytokine signalling pathways by SOCS3 is independent of SOCS2

Hiu Kiu; Christopher J. Greenhalgh; Anne L. Thaus; Douglas J. Hilton; Nicos A. Nicola; Warren S. Alexander; Andrew W. Roberts

Suppressor of cytokine signalling (SOCS) 3 is an essential regulator of cytokine signalling, and in turn its expression is tightly regulated. Data from overexpression studies in cell lines suggest that SOCS2 regulates SOCS3 protein degradation, by forming a molecular bridge to an E3 ubiquitin-ligase complex. Whether this regulation is relevant in primary cells is unknown. In this study, we utilized Socs2 − / − mice to examine the role of SOCS2 in modulating SOCS3 expression and degradation, and its impact on interleukin-2 (IL-2) and IL-6 signalling in primary haemopoietic cells. Both biochemical and biological analyses demonstrated unperturbed SOCS3 expression and cytokine signalling in the absence of SOCS2. Our results suggest that SOCS2 is not a physiological regulator of SOCS3 expression and action in primary haemopoietic cells.


Growth Factors Journal | 2007

Mechanism of crosstalk inhibition of IL-6 signaling in response to LPS and TNFα

Hiu Kiu; Douglas J. Hilton; Nicos A. Nicola; Matthias Ernst; Rodolfo Marquez; Warren S. Alexander; Andrew W. Roberts; Edward J. McManus

Negative regulation of cytokine signaling is critical for the generation of the appropriate cellular outcome in response to signals, and can be modulated by other concomitant extracellular stimuli (“crosstalk”). Using both genetic and pharmacological manipulations we have investigated the mechanisms by which the pro-inflammatory stimuli, lipopolysaccharide (LPS) and Tumor necrosis factor α (TNFα), negatively regulate interleukin-6 (IL-6) signaling in primary mouse macrophages. Analysis of suppressor of cytokine signalling 3 (SOCS3)-deficient macrophages reveal that SOCS3 is necessary but surprisingly, not sufficient for the complete crosstalk inhibition of IL-6 signaling induced by LPS and TNFα. Analysis of macrophages from gp130 (Y757F) mutant mice suggest that SH2 domain-containing tyrosine phosphatase (SHP2) activity does not explain the residual inhibitory effect of these pro-inflammatory stimuli. In addition, p38 mitogen-activated protein kinase (p38) activation also negatively regulates IL-6 signaling independent of its parallel and necessary action to induce SOCS3 expression. Finally, we have identified an additional, novel mechanism of crosstalk inhibition: a reduction in total cellular levels of gp130 following stimulation with LPS and TNFα.

Collaboration


Dive into the Hiu Kiu's collaboration.

Top Co-Authors

Avatar

Warren S. Alexander

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donald Metcalf

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Ben A. Croker

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nicos A. Nicola

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Edward J. McManus

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jeff Szer

Royal Melbourne Hospital

View shared research outputs
Top Co-Authors

Avatar

Lisa A. Mielke

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Louise H. Cengia

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge