Network


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

Hotspot


Dive into the research topics where Ashleigh R. Poh is active.

Publication


Featured researches published by Ashleigh R. Poh.


Cell | 2014

RIPK1 Regulates RIPK3-MLKL-Driven Systemic Inflammation and Emergency Hematopoiesis

James A Rickard; Joanne A. O’Donnell; Joseph M Evans; Najoua Lalaoui; Ashleigh R. Poh; TeWhiti Rogers; James E. Vince; Kate E. Lawlor; Robert L. Ninnis; Holly Anderton; Cathrine Hall; Sukhdeep Kaur Spall; Toby J. Phesse; Helen E. Abud; Louise H. Cengia; Jason Corbin; Sandra Mifsud; Ladina Di Rago; Donald Metcalf; Matthias Ernst; Grant Dewson; Andrew W. Roberts; Warren S. Alexander; James M. Murphy; Paul G. Ekert; Seth L. Masters; David L. Vaux; Ben A. Croker; Motti Gerlic; John Silke

Upon ligand binding, RIPK1 is recruited to tumor necrosis factor receptor superfamily (TNFRSF) and Toll-like receptor (TLR) complexes promoting prosurvival and inflammatory signaling. RIPK1 also directly regulates caspase-8-mediated apoptosis or, if caspase-8 activity is blocked, RIPK3-MLKL-dependent necroptosis. We show that C57BL/6 Ripk1(-/-) mice die at birth of systemic inflammation that was not transferable by the hematopoietic compartment. However, Ripk1(-/-) progenitors failed to engraft lethally irradiated hosts properly. Blocking TNF reversed this defect in emergency hematopoiesis but, surprisingly, Tnfr1 deficiency did not prevent inflammation in Ripk1(-/-) neonates. Deletion of Ripk3 or Mlkl, but not Casp8, prevented extracellular release of the necroptotic DAMP, IL-33, and reduced Myd88-dependent inflammation. Reduced inflammation in the Ripk1(-/-)Ripk3(-/-), Ripk1(-/-)Mlkl(-/-), and Ripk1(-/-)Myd88(-/-) mice prevented neonatal lethality, but only Ripk1(-/-)Ripk3(-/-)Casp8(-/-) mice survived past weaning. These results reveal a key function for RIPK1 in inhibiting necroptosis and, thereby, a role in limiting, not only promoting, inflammation.


Science Signaling | 2014

Partial inhibition of gp130-Jak-Stat3 signaling prevents Wnt-β-catenin-mediated intestinal tumor growth and regeneration.

Toby J. Phesse; Michael Buchert; Emma Stuart; Dustin J. Flanagan; Maree C. Faux; Shoukat Afshar-Sterle; Francesca Walker; Hui-Hua Zhang; Cameron J. Nowell; Robert N. Jorissen; Chin Wee Tan; Yumiko Hirokawa; Moritz F. Eissmann; Ashleigh R. Poh; Jordane Malaterre; Helen B. Pearson; David G. Kirsch; Paolo Provero; Valeria Poli; Robert G. Ramsay; Oliver M. Sieber; Antony W. Burgess; Dennis Huszar; Elizabeth Vincan; Matthias Ernst

Partial suppression of the inflammatory gp130-Jak-Stat pathway inhibits intestinal tumor growth. A Novel Strategy for Treating Colon Cancer In most patients, colon cancer arises from a mutation in the gene encoding APC, which results in constitutive activation of the β-catenin pathway. Inhibition of this pathway interferes with the continuous renewal of the epithelial cells that line the intestinal tract and therefore may confer only limited therapeutic benefit. Phesse et al. discovered that the signaling pathway involving the receptor gp130, the associated Jak kinases, and the transcription factor Stat3 enhanced the growth of intestinal tumors in mice. Conversely, genetic or pharmacological inhibition of this pathway reduced tumor growth by increasing the expression of genes encoding the p21 and p16 proteins that halt cell division, through a cell-intrinsic mechanism. Thus, drugs targeting the Jak-Stat3 pathway, which are currently in clinical trials for the treatment of hematological malignancies, may also be useful for treating colon cancer. Most colon cancers arise from somatic mutations in the tumor suppressor gene APC (adenomatous polyposis coli), and these mutations cause constitutive activation of the Wnt–to–β-catenin pathway in the intestinal epithelium. Because Wnt–β-catenin signaling is required for homeostasis and regeneration of the adult intestinal epithelium, therapeutic targeting of this pathway is challenging. We found that genetic activation of the cytokine-stimulated pathway mediated by the receptor gp130, the associated Jak (Janus kinase) kinases, and the transcription factor Stat3 (signal transducer and activator of transcription 3) was required for intestinal regeneration in response to irradiation-induced damage in wild-type mice and for tumorigenesis in Apc-mutant mice. Systemic pharmacological or partial genetic inhibition of gp130-Jak-Stat3 signaling suppressed intestinal regeneration, the growth of tumors in Apc-mutant mice, and the growth of colon cancer xenografts. The growth of Apc-mutant tumors depended on gp130-Jak-Stat3 signaling for induction of the polycomb repressor Bmi-1, and the associated repression of genes encoding the cell cycle inhibitors p16 and p21. However, suppression of gp130-Jak-Stat3 signaling did not affect Wnt–β-catenin signaling or homeostasis in the intestine. Thus, these data not only suggest a molecular mechanism for how the gp130-Jak-Stat3 pathway can promote cancer but also provide a rationale for therapeutic inhibition of Jak in colon cancer.


Cancer Research | 2016

Stomach-Specific Activation of Oncogenic KRAS and STAT3-Dependent Inflammation Cooperatively Promote Gastric Tumorigenesis in a Preclinical Model

Stefan Thiem; Moritz F. Eissmann; Joachim Elzer; Anna Jonas; Tracy Putoczki; Ashleigh R. Poh; Paul M. Nguyen; Adele Preaudet; Dustin J. Flanagan; Elizabeth Vincan; Paul Waring; Michael Buchert; Andrew G. Jarnicki; Matthias Ernst

About 5% to 10% of human gastric tumors harbor oncogenic mutations in the KRAS pathway, but their presence alone is often insufficient for inducing gastric tumorigenesis, suggesting a requirement for additional mutagenic events or microenvironmental stimuli, including inflammation. Assessing the contribution of such events in preclinical mouse models requires Cre recombinase-mediated conditional gene expression in stem or progenitor cells of normal and transformed gastric epithelium. We therefore constructed a bacterial artificial chromosome containing transgene (Tg), comprising the regulatory elements of the trefoil factor 1 (Tff1) gene and the tamoxifen-inducible Cre recombinase (CreERT2)-coding sequence. The resulting Tg(Tff1-CreERT2) mice were crossed with mice harboring conditional oncogenic mutations in Kras or Braf The administration of tamoxifen to the resulting adult Tg(Tff1-CreERT2);Kras(LSL-G12D/+) and Tg(Tff1-CreERT2);Braf(LSL-V600E/+) mice resulted in gastric metaplasia, inflammation, and adenoma development, characterized by excessive STAT3 activity. To assess the contribution of STAT3 to the spontaneously developing gastric adenomas in gp130(F/F) mice, which carry a knockin mutation in the Il6 signal transducer (Il6st), we generated Tg(Tff1-CreERT2);Stat3(fl/fl);gp130(F/F) mice that also harbor a conditional Stat3 knockout allele and found that tamoxifen administration conferred a significant reduction in their tumor burden. Conversely, excessive Kras activity in Tg(Tff1-CreERT2);Kras(LSL-G12D/+);gp130(F/F) mice promoted more extensive gastric inflammation, metaplastic transformation, and tumorigenesis than observed in Tg(Tff1-CreERT2);Kras(LSL-G12D/+) mice. Collectively, our findings demonstrate that advanced gastric tumorigenesis requires oncogenic KRAS or BRAF in concert with aberrant STAT3 activation in epithelial precursor cells of the glandular stomach, providing a new conditional model of gastric cancer in which to investigate candidate therapeutic targets and treatment strategies. Cancer Res; 76(8); 2277-87. ©2016 AACR.


Frontiers in Oncology | 2018

Targeting Macrophages in Cancer: From Bench to Bedside

Ashleigh R. Poh; Matthias Ernst

Macrophages are a major component of the tumor microenvironment and orchestrate various aspects of immunity. Within tumors, macrophages can reversibly alter their endotype in response to environmental cues, including hypoxia and stimuli derived from other immune cells, as well as the extracellular matrix. Depending on their activation status, macrophages can exert dual influences on tumorigenesis by either antagonizing the cytotoxic activity immune cells or by enhancing antitumor responses. In most solid cancers, increased infiltration with tumor-associated macrophages (TAMs) has long been associated with poor patient prognosis, highlighting their value as potential diagnostic and prognostic biomarkers in cancer. A number of macrophage-centered approaches to anticancer therapy have been investigated, and include strategies to block their tumor-promoting activities or exploit their antitumor effector functions. Integrating therapeutic strategies to target TAMs to complement conventional therapies has yielded promising results in preclinical trials and warrants further investigation to determine its translational benefit in human cancer patients. In this review, we discuss the molecular mechanisms underlying the pro-tumorigenic programming of macrophages and provide a comprehensive update of macrophage-targeted therapies for the treatment of solid cancers.


Cancer Cell | 2017

Inhibition of Hematopoietic Cell Kinase Activity Suppresses Myeloid Cell-Mediated Colon Cancer Progression

Ashleigh R. Poh; Christopher G. Love; Frederick Masson; Adele Preaudet; Cary Tsui; Lachlan Whitehead; Simon Monard; Yelena Khakham; Lotta Burstroem; Guillaume Lessene; Oliver M. Sieber; Clifford A. Lowell; Tracy Putoczki; Robert J.J. O'Donoghue; Matthias Ernst

Aberrant activation of the SRC family kinase hematopoietic cell kinase (HCK) triggers hematological malignancies as a tumor cell-intrinsic oncogene. Here we find that high HCK levels correlate with reduced survival of colorectal cancer patients. Likewise, increased Hck activity in mice promotes the growth of endogenous colonic malignancies and of human colorectal cancer cell xenografts. Furthermore, tumor-associated macrophages of the corresponding tumors show a pronounced alternatively activated endotype, which occurs independently of mature lymphocytes or of Stat6-dependent Th2 cytokine signaling. Accordingly, pharmacological inhibition or genetic reduction of Hck activity suppresses alternative activation of tumor-associated macrophages and the growth of colon cancer xenografts. Thus, Hck may serve as a promising therapeutic target for solid malignancies.


Oncotarget | 2017

The angiotensin receptor blocker, Losartan, inhibits mammary tumor development and progression to invasive carcinoma

Rhiannon Coulson; Seng H. Liew; Angela A. Connelly; Nicholas S. Yee; Siddhartha Deb; Beena Kumar; Ana Cristina Vargas; Sandra A. O’Toole; Adam C. Parslow; Ashleigh R. Poh; Tracy Putoczki; Riley J. Morrow; Mariah G. Alorro; Kyren A. Lazarus; Evie F.W. Yeap; Kelly L. Walton; Craig A. Harrison; Natalie J. Hannan; Amee J. George; Colin Clyne; Matthias Ernst; Andrew M. Allen; Ashwini L. Chand

Drugs that target the Renin-Angiotensin System (RAS) have recently come into focus for their potential utility as cancer treatments. The use of Angiotensin Receptor Blockers (ARBs) and Angiotensin-Converting Enzyme (ACE) Inhibitors (ACEIs) to manage hypertension in cancer patients is correlated with improved survival outcomes for renal, prostate, breast and small cell lung cancer. Previous studies demonstrate that the Angiotensin Receptor Type I (AT1R) is linked to breast cancer pathogenesis, with unbiased analysis of gene-expression studies identifying significant up-regulation of AGTR1, the gene encoding AT1R in ER+ve/HER2−ve tumors correlating with poor prognosis. However, there is no evidence, so far, of the functional contribution of AT1R to breast tumorigenesis. We explored the potential therapeutic benefit of ARB in a carcinogen-induced mouse model of breast cancer and clarified the mechanisms associated with its success. Mammary tumors were induced with 7,12-dimethylbenz[α]antracene (DMBA) and medroxyprogesterone acetate (MPA) in female wild type mice and the effects of the ARB, Losartan treatment assessed in a preventative setting (n = 15 per group). Tumor histopathology was characterised by immunohistochemistry, real-time qPCR to detect gene expression signatures, and tumor cytokine levels measured with quantitative bioplex assays. AT1R was detected with radiolabelled ligand binding assays in fresh frozen tumor samples. We showed that therapeutic inhibition of AT1R, with Losartan, resulted in a significant reduction in tumor burden; and no mammary tumor incidence in 20% of animals. We observed a significant reduction in tumor progression from DCIS to invasive cancer with Losartan treatment. This was associated with reduced tumor cell proliferation and a significant reduction in IL-6, pSTAT3 and TNFα levels. Analysis of tumor immune cell infiltrates, however, demonstrated no significant differences in the recruitment of lymphocytes or tumour-associated macrophages in Losartan or vehicle-treated mammary tumors. Analysis of AT1R expression with radiolabelled ligand binding assays in human breast cancer biopsies showed high AT1R levels in 30% of invasive ductal carcinomas analysed. Furthermore, analysis of the TCGA database identified that high AT1R expression to be associated with luminal breast cancer subtype. Our in vivo data and analysis of human invasive ductal carcinoma samples identify the AT1R is a potential therapeutic target in breast cancer, with the availability of a range of well-tolerated inhibitors currently used in clinics. We describe a novel signalling pathway critical in breast tumorigenesis, that may provide new therapeutic avenues to complement current treatments.


Journal of Gastroenterology and Hepatology | 2016

Mouse models for gastric cancer: Matching models to biological questions

Ashleigh R. Poh; Robert J.J. O'Donoghue; Matthias Ernst; Tracy Putoczki

Gastric cancer is the third leading cause of cancer‐related mortality worldwide. This is in part due to the asymptomatic nature of the disease, which often results in late‐stage diagnosis, at which point there are limited treatment options. Even when treated successfully, gastric cancer patients have a high risk of tumor recurrence and acquired drug resistance. It is vital to gain a better understanding of the molecular mechanisms underlying gastric cancer pathogenesis to facilitate the design of new‐targeted therapies that may improve patient survival. A number of chemically and genetically engineered mouse models of gastric cancer have provided significant insight into the contribution of genetic and environmental factors to disease onset and progression. This review outlines the strengths and limitations of current mouse models of gastric cancer and their relevance to the pre‐clinical development of new therapeutics.


Cancer Research | 2016

Abstract 536: Excessive HCK kinase activity in the tumor stroma promotes colorectal cancer progression

Matthias Ernst; Ashleigh R. Poh; Frederic Masson; Tracy Putoczki; Robert O’Donoghue

Excessive activation of the myeloid-specific Src-family kinase Hematopoietic Cell Kinase (HCK) acts as a tumor intrinsic oncogene by promoting proliferation and survival of immune cells, and confers a poor prognosis for leukemia. However, the role of HCK in the tumor stroma of solid malignancies remains unexplored. We analyzed the expression level of HCK in matched biopsies from sporadic colorectal cancer patients and observed elevated HCK phosphorylation in tumors compared to unaffected colons. Analysis of corresponding RNAseq data revealed a striking correlation between tumors with high HCK gene expression and a gene signature indicative of a tumor-promoting alternatively-activated macrophage (AAM) endotype. To functionally assess this observation, we exploited HckCA mice [Ernst et al., J Exp Med 2002] that express a constitutively active form of Hck as a knockin mutation, and subjected these mice to a chemically-induced model of sporadic colorectal cancer. HckCA mice developed more and larger tumors compared to wild-type (WT) animals, and this was associated with a significant bias towards CD206+ AAMs in tumors of HckCA mice without affecting the total number of tumor-associated leukocyte and lymphocytes. Likewise, adoptive bone-marrow transfer experiments revealed enhanced tumor formation and AAM differentiation in WT mice reconstituted with HckCA bone marrow, with a reciprocal decrease of these parameters in HckCA mice reconstituted with WT marrow. Finally, pharmacologic targeting of the catalytic activity of Hck with a small molecule inhibitor significantly reduced tumor progression and impaired AAM polarisation. Collectively, our findings suggest that excessive Hck activity in the tumor stroma promotes the progression of solid cancers by modulating the endotype of tumor-associated myeloid cells. Therefore, Hck represents a rational therapeutic target for macrophage re-education in solid cancers by limiting polarization of tumor-promoting AAM. Citation Format: Matthias Ernst, Ashleigh Poh, Frederic Masson, Tracy Putoczki, Robert O’Donoghue. Excessive HCK kinase activity in the tumor stroma promotes colorectal cancer progression. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 536.


Oncotarget | 2015

Hematopoietic cell kinase (HCK) as a therapeutic target in immune and cancer cells

Ashleigh R. Poh; Robert J.J. O'Donoghue; Matthias Ernst


Archive | 2017

Targeting H(i)ck education for cancer therapy

Matthias Ernst; Robert J.J. O'Donoghue; Ashleigh R. Poh

Collaboration


Dive into the Ashleigh R. Poh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tracy Putoczki

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Adele Preaudet

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Robert J.J. O'Donoghue

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Michael Buchert

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Robert O’Donoghue

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guillaume Lessene

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge