Network


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

Hotspot


Dive into the research topics where Karin A Sedelies is active.

Publication


Featured researches published by Karin A Sedelies.


Oncogene | 2003

HScrib is a functional homologue of the Drosophila tumour suppressor Scribble

Lukas E. Dow; Anthony M. Brumby; Rosa Muratore; Michelle Coombe; Karin A Sedelies; Joseph A. Trapani; Sarah M. Russell; Helena E. Richardson; Patrick O. Humbert

Scribble (scrib), discs large (dlg) and lethal giant larvae (lgl) encode proteins that regulate cell polarity and have been identified as neoplastic tumour suppressor genes in Drosophila melanogaster. Here, we have used the Drosophila model system to provide the first functional evidence that human Scribble (hScrib) can act as a tumour suppressor. We show that hScrib protein displays highly polarized localization in mammalian epithelial cells and colocalizes with mammalian Dlg, similar to D. melanogaster Scribble (DmScrib) distribution in Drosophila epithelium. Furthermore, hScrib can rescue the polarity and tumorous overgrowth defects of scrib mutant Drosophila. hScrib therefore can act as an effective tumour suppressor in vivo, regulating both apical–basal polarity and cellular proliferation in a manner similar to that of DmScrib in Drosophila. These data demonstrate that hScrib is a functional homologue of DmScrib and therefore predict an important role for hScrib in the suppression of mammalian tumorigenesis.


Journal of Biological Chemistry | 2004

Discordant Regulation of Granzyme H and Granzyme B Expression in Human Lymphocytes

Karin A Sedelies; Thomas J. Sayers; Kirsten M. Edwards; Weisan Chen; Daniel G. Pellicci; Dale I. Godfrey; Joseph A. Trapani

We analyzed the expression of granzyme H in human blood leukocytes, using a novel monoclonal antibody raised against recombinant granzyme H. 33-kDa granzyme H was easily detected in unfractionated peripheral blood mononuclear cells, due to its high constitutive expression in CD3-CD56+ natural killer (NK) cells, whereas granzyme B was less abundant. The NK lymphoma cell lines, YT and Lopez, also expressed high granzyme H levels. Unstimulated CD4+ and particularly CD8+ T cells expressed far lower levels of granzyme H than NK cells, and various agents that classically induce T cell activation, proliferation, and enhanced granzyme B expression failed to induce granzyme H expression in T cells. Also, granzyme H was not detected in NK T cells, monocytes, or neutrophils. There was a good correlation between mRNA and protein expression in cells that synthesize both granzymes B and H, suggesting that gzmH gene transcription is regulated similarly to gzmB. Overall, our data indicate that although the gzmB and gzmH genes are tightly linked, expression of the proteins is quite discordant in T and NK cells. The finding that granzyme H is frequently more abundant than granzyme B in NK cells is consistent with a role for granzyme H in complementing the pro-apoptotic function of granzyme B in human NK cells.


Immunology and Cell Biology | 2006

Role of Bid-induced mitochondrial outer membrane permeabilization in granzyme B-induced apoptosis

Nigel J. Waterhouse; Karin A Sedelies; Joseph A. Trapani

Cytotoxic lymphocytes (CL) induce death of their targets by granule exocytosis. During this process, enzymes contained within cytotoxic granules (granzymes) are delivered to the target cell where the enzymes trigger the cell death by cleaving specific substrates. Granzyme B is the only granzyme that has been shown to induce cell death by apoptosis, but the exact pathway by which this is achieved has been the subject of hot debate. Furthermore, several other death‐inducing granzymes have been identified; therefore, the exact contribution of granzyme B to CL‐induced death is unclear. In this study, we discuss our recent findings on granzyme B‐induced cell death and discuss the potential relevance of this pathway to CL‐induced death of viral‐infected and transformed cells.


Journal of Cell Biology | 2007

Residual active granzyme B in cathepsin C-null lymphocytes is sufficient for perforin-dependent target cell apoptosis.

Vivien R. Sutton; Nigel J. Waterhouse; Kylie A. Browne; Karin A Sedelies; Annette Ciccone; Desiree A. Anthony; Aulikki Koskinen; Arno Müllbacher; Joseph A. Trapani

Cathepsin C activates serine proteases expressed in hematopoietic cells by cleaving an N-terminal dipeptide from the proenzyme upon granule packaging. The lymphocytes of cathepsin C–null mice are therefore proposed to totally lack granzyme B activity and perforin-dependent cytotoxicity. Surprisingly, we show, using live cell microscopy and other methodologies, that cells targeted by allogenic CD8+ cytotoxic T lymphocyte (CTL) raised in cathepsin C–null mice die through perforin-dependent apoptosis indistinguishable from that induced by wild-type CTL. The cathepsin C–null CTL expressed reduced but still appreciable granzyme B activity, but minimal granzyme A activity. Also, in contrast to mice with inactivation of both their granzyme A/B genes, cathepsin C deficiency did not confer susceptibility to ectromelia virus infection in vivo. Overall, our results indicate that although cathepsin C clearly generates the majority of granzyme B activity, some is still generated in its absence, pointing to alternative mechanisms for granzyme B processing and activation. Cathepsin C deficiency also results in considerably milder immune deficiency than perforin or granzyme A/B deficiency.


Cell Death & Differentiation | 2006

Functional dissociation of ΔΨm and cytochrome c release defines the contribution of mitochondria upstream of caspase activation during granzyme B-induced apoptosis

Nigel J. Waterhouse; Karin A Sedelies; Vivien R. Sutton; M Pinkoski; Kevin Thia; Ricky W. Johnstone; Phillip I. Bird; D Green; Joseph A. Trapani

Loss of Bid confers clonogenic survival to granzyme B-treated cells, however the exact role of Bid-induced mitochondrial damage – upstream or downstream of caspases – remains controversial. Here we show that direct cleavage of Bid by granzyme B, but not caspases, was required for granzyme B-induced apoptosis. Release of cytochrome c and SMAC, but not AIF or endonuclease G, occurred in the absence of caspase activity and correlated with the onset of apoptosis and loss of clonogenic potential. Loss of mitochondrial trans-membrane potential (ΔΨm) was also caspase independent, however if caspase activity was blocked the mitochondria regenerated their ΔΨm. Loss of ΔΨm was not required for rapid granzyme B-induced apoptosis and regeneration of ΔΨm following cytochrome c release did not confer clonogenic survival. This functional dissociation of cytochrome c and SMAC release from loss of ΔΨm demonstrates the essential contribution of Bid upstream of caspase activation during granzyme B-induced apoptosis.


Journal of Experimental Medicine | 2007

Residual active granzyme B in cathepsin C–null lymphocytes is sufficient for perforin-dependent target cell apoptosis

Vivien R. Sutton; Nigel J. Waterhouse; Kylie A. Browne; Karin A Sedelies; Annette Ciccone; Desiree A. Anthony; Aulikki Koskinen; Arno Müllbacher; Joseph A. Trapani

Sutton et al. 2007. J. Cell Biol. doi:10.1083/jcb.200609077nn[OpenUrl][1][Abstract/FREE Full Text][2]nn [1]: {openurl}?query=rft_id%253Dinfo%253Adoi%252F10.1083%252Fjcb.200609077%26rft_id%253Dinfo%253Apmid%252F17283185%26rft.genre%253Darticle%26rft_val_fmt%253Dinfo%253Aofi%252Ffmt%253Akev%253Amtx%


Cell Death & Differentiation | 2008

Blocking granule-mediated death by primary human NK cells requires both protection of mitochondria and inhibition of caspase activity

Karin A Sedelies; Annette Ciccone; Christopher J. Clarke; Jane Oliaro; Vivien R. Sutton; F. L. Scott; John Silke; Olivia Susanto; Douglas R. Green; Ricky W. Johnstone; Phillip I. Bird; Joseph A. Trapani; Nigel J. Waterhouse

Human GraB (hGraB) preferentially induces apoptosis via Bcl-2-regulated mitochondrial damage but can also directly cleave caspases and caspase substrates in cell-free systems. How hGraB kills cells when it is delivered by cytotoxic lymphocytes (CL) and the contribution of hGraB to CL-induced death is still not clear. We show that primary human natural killer (hNK) cells, which specifically used hGraB to induce target cell death, were able to induce apoptosis of cells whose mitochondria were protected by Bcl-2. Purified hGraB also induced apoptosis of Bcl-2-overexpressing targets but only when delivered at 5- to 10-fold the concentration required to kill cells expressing endogenous Bcl-2. Caspases were critical in this process as inhibition of caspase activity permitted clonogenic survival of Bcl-2-overexpressing cells treated with hGraB or hNK cells but did not protect cells that only expressed endogenous Bcl-2. Our data therefore show that hGraB triggers caspase activation via mitochondria-dependent and mitochondria-independent mechanisms that are activated in a hierarchical manner, and that the combined effects of Bcl-2 and direct caspase inhibition can block cell death induced by hGraB and primary hNK cells.


Cell Death & Differentiation | 2013

Mouse granzyme A induces a novel death with writhing morphology that is mechanistically distinct from granzyme B-induced apoptosis.

Olivia Susanto; Sarah Elizabeth Stewart; Ilia Voskoboinik; Daniella Brasacchio; Magdalena Hagn; Sarah Ellis; S Asquith; Karin A Sedelies; Phillip I. Bird; Nigel J. Waterhouse; Joseph A. Trapani

Human and mouse granzyme (Gzm)B both induce target cell apoptosis in concert with pore-forming perforin (Pfp); however the mechanisms by which other Gzms induce non-apoptotic death remain controversial and poorly characterised. We used timelapse microscopy to document, quantitatively and in real time, the death of target cells exposed to primary natural killer (NK) cells from mice deficient in key Gzms. We found that in the vast majority of cases, NK cells from wild-type mice induced classic apoptosis. However, NK cells from syngeneic Gzm B-deficient mice induced a novel form of cell death characterised by slower kinetics and a pronounced, writhing, ‘worm-like’ morphology. Dying cells initially contracted but did not undergo membrane blebbing, and annexin-V staining was delayed until the onset of secondary necrosis. As it is different from any cell death process previously reported, we tentatively termed this cell death ‘athetosis’. Two independent lines of evidence showed this alternate form of death was due to Gzm A: first, cell death was revealed in the absence of Gzm B, but was completely lost when the NK cells were deficient in both Gzm A and B; second, the athetotic morphology was precisely reproduced when recombinant mouse Gzm A was delivered by an otherwise innocuous dose of recombinant Pfp. Gzm A-mediated athetosis did not require caspase activation, early mitochondrial disruption or generation of reactive oxygen species, but did require an intact actin cytoskeleton and was abolished by latrunculin B and mycalolide B. This work defines an authentic role for mouse Gzm A in granule-induced cell death by cytotoxic lymphocytes.


Journal of Immunology | 2011

A Critical Role for Granzymes in Antigen Cross-Presentation through Regulating Phagocytosis of Killed Tumor Cells

Sabine Hoves; Vivien R. Sutton; Nicole M. Haynes; Edwin D. Hawkins; Daniel Fernandez Ruiz; Nikola Baschuk; Karin A Sedelies; Maximilian Schnurr; John Stagg; Daniel M. Andrews; Jose A. Villadangos; Joseph A. Trapani

Granzymes A and B (GrAB) are known principally for their role in mediating perforin-dependent death of virus-infected or malignant cells targeted by CTL. In this study, we show that granzymes also play a critical role as inducers of Ag cross-presentation by dendritic cells (DC). This was demonstrated by the markedly reduced priming of naive CD8+ T cells specific for the model Ag OVA both in vitro and in vivo in response to tumor cells killed in the absence of granzymes. Reduced cross-priming was due to impairment of phagocytosis of tumor cell corpses by CD8α+ DC but not CD8α− DC, demonstrating the importance of granzymes in inducing the exposure of prophagocytic “eat-me” signals on the dying target cell. Our data reveal a critical and previously unsuspected role for granzymes A and B in dictating immunogenicity by influencing the mode of tumor cell death and indicate that granzymes contribute to the efficient generation of immune effector pathways in addition to their well-known role in apoptosis induction.


Journal of Translational Medicine | 2004

Granzyme B; the chalk-mark of a cytotoxic lymphocyte

Nigel J. Waterhouse; Karin A Sedelies; Christopher J. Clarke

During cytotoxic lymphocyte (CL) mediated killing of target cells, granzyme B is released from the CL into the immune synapse. Recent studies have found that ELISPOT-detection of granzyme B correlated well with conventional assays for CL mediated killing. In this way, the released granzyme B can be used to mark the spot where a target cell was murdered. We discuss the benefits and potential limitations of using this assay to measure CL mediated killing of target cells.

Collaboration


Dive into the Karin A Sedelies's collaboration.

Top Co-Authors

Avatar

Joseph A. Trapani

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Nigel J. Waterhouse

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Vivien R. Sutton

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Ricky W. Johnstone

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annette Ciccone

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Kylie A. Browne

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Melinda E. Christensen

Translational Research Institute

View shared research outputs
Top Co-Authors

Avatar

Olivia Susanto

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge