Network


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

Hotspot


Dive into the research topics where Rachel A. Ridgway is active.

Publication


Featured researches published by Rachel A. Ridgway.


Nature | 2009

Crypt stem cells as the cells-of-origin of intestinal cancer

Nick Barker; Rachel A. Ridgway; Johan H. van Es; Marc van de Wetering; Harry Begthel; Maaike van den Born; Esther Danenberg; Alan Richard Clarke; Owen J. Sansom; Hans Clevers

Intestinal cancer is initiated by Wnt-pathway-activating mutations in genes such as adenomatous polyposis coli (APC). As in most cancers, the cell of origin has remained elusive. In a previously established Lgr5 (leucine-rich-repeat containing G-protein-coupled receptor 5) knockin mouse model, a tamoxifen-inducible Cre recombinase is expressed in long-lived intestinal stem cells. Here we show that deletion of Apc in these stem cells leads to their transformation within days. Transformed stem cells remain located at crypt bottoms, while fuelling a growing microadenoma. These microadenomas show unimpeded growth and develop into macroscopic adenomas within 3-5weeks. The distribution of Lgr5+ cells within stem-cell-derived adenomas indicates that a stem cell/progenitor cell hierarchy is maintained in early neoplastic lesions. When Apc is deleted in short-lived transit-amplifying cells using a different cre mouse, the growth of the induced microadenomas rapidly stalls. Even after 30weeks, large adenomas are very rare in these mice. We conclude that stem-cell-specific loss of Apc results in progressively growing neoplasia.


Nature Medicine | 2012

Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease

Luke Boulter; Olivier Govaere; Tom Bird; Sorina Radulescu; Antonella Pellicoro; Rachel A. Ridgway; Sang Soo Seo; Bart Spee; Nico van Rooijen; Owen J. Sansom; John P. Iredale; Sally Lowell; Tania Roskams; Stuart J. Forbes

During chronic injury, regeneration of the adult liver becomes impaired. In this context bipotent Hepatic Progenitor Cells (HPCs) become activated and can regenerate both cholangiocytes and hepatocytes. Notch and Wnt signalling during hepatic ontogeny are described, but their roles in HPC mediated liver regeneration are unclear. Here we show in human diseased liver and murine models of the ductular reaction with biliary and hepatocyte regeneration that Notch and Wnt signalling direct HPC specification within the activated myofibroblasts and macrophages HPC niche. During biliary regeneration, Numb is downregulated in HPCs, Jagged1 promotes biliary specification within HPCs. During hepatocyte regeneration, macrophage derived canonical Wnt signalling maintains Numb within HPCs, and Notch signalling is reduced promoting hepatocyte specification. This dominant Wnt state is stimulated through engulfment of hepatocyte debris by niche macrophages and can directly influence the HPCs. Macrophage Wnt3a expression in turn facilitates hepatocyte regeneration – thus exemplifying a novel positive feedback mechanism in adult parenchymal regeneration.During chronic injury a population of bipotent hepatic progenitor cells (HPCs) become activated to regenerate both cholangiocytes and hepatocytes. Here we show in human diseased liver and mouse models of the ductular reaction that Notch and Wnt signaling direct specification of HPCs via their interactions with activated myofibroblasts or macrophages. In particular, we found that during biliary regeneration, expression of Jagged 1 (a Notch ligand) by myofibroblasts promoted Notch signaling in HPCs and thus their biliary specification to cholangiocytes. Alternatively, during hepatocyte regeneration, macrophage engulfment of hepatocyte debris induced Wnt3a expression. This resulted in canonical Wnt signaling in nearby HPCs, thus maintaining expression of Numb (a cell fate determinant) within these cells and the promotion of their specification to hepatocytes. By these two pathways adult parenchymal regeneration during chronic liver injury is promoted.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer

Jennifer P. Morton; Paul Timpson; Saadia A. Karim; Rachel A. Ridgway; Dimitris Athineos; Brendan Doyle; Nigel B. Jamieson; Karin A. Oien; Andrew M. Lowy; Valerie G. Brunton; Margaret C. Frame; T.R. Jeffry Evans; Owen J. Sansom

TP53 mutation occurs in 50–75% of human pancreatic ductal adenocarcinomas (PDAC) following an initiating activating mutation in the KRAS gene. These p53 mutations frequently result in expression of a stable protein, p53R175H, rather than complete loss of protein expression. In this study we elucidate the functions of mutant p53 (Trp53R172H), compared to knockout p53 (Trp53fl), in a mouse model of PDAC. First we find that although KrasG12D is one of the major oncogenic drivers of PDAC, most KrasG12D-expressing pancreatic cells are selectively lost from the tissue, and those that remain form premalignant lesions. Loss, or mutation, of Trp53 allows retention of the KrasG12D-expressing cells and drives rapid progression of these premalignant lesions to PDAC. This progression is consistent with failed growth arrest and/or senescence of premalignant lesions, since a mutant of p53, p53R172P, which can still induce p21 and cell cycle arrest, is resistant to PDAC formation. Second, we find that despite similar kinetics of primary tumor formation, mutant p53R172H, as compared with genetic loss of p53, specifically promotes metastasis. Moreover, only mutant p53R172H-expressing tumor cells exhibit invasive activity in an in vitro assay. Importantly, in human PDAC, p53 accumulation significantly correlates with lymph node metastasis. In summary, by using ‘knock-in’ mutations of Trp53 we have identified two critical acquired functions of a stably expressed mutant form of p53 that drive PDAC; first, an escape from KrasG12D-induced senescence/growth arrest and second, the promotion of metastasis.


Nature | 2013

p53 status determines the role of autophagy in pancreatic tumour development

Mathias Rosenfeldt; Jim O'Prey; Jennifer P. Morton; Colin Nixon; Gillian M. Mackay; Agata Mrowinska; Amy Au; Taranjit Singh Rai; Liang Zheng; Rachel A. Ridgway; Peter D. Adams; Kurt I. Anderson; Eyal Gottlieb; Owen J. Sansom; Kevin M. Ryan

Macroautophagy (hereafter referred to as autophagy) is a process in which organelles termed autophagosomes deliver cytoplasmic constituents to lysosomes for degradation. Autophagy has a major role in cellular homeostasis and has been implicated in various forms of human disease. The role of autophagy in cancer seems to be complex, with reports indicating both pro-tumorigenic and tumour-suppressive roles. Here we show, in a humanized genetically-modified mouse model of pancreatic ductal adenocarcinoma (PDAC), that autophagy’s role in tumour development is intrinsically connected to the status of the tumour suppressor p53. Mice with pancreases containing an activated oncogenic allele of Kras (also called Ki-Ras)—the most common mutational event in PDAC—develop a small number of pre-cancerous lesions that stochastically develop into PDAC over time. However, mice also lacking the essential autophagy genes Atg5 or Atg7 accumulate low-grade, pre-malignant pancreatic intraepithelial neoplasia lesions, but progression to high-grade pancreatic intraepithelial neoplasias and PDAC is blocked. In marked contrast, in mice containing oncogenic Kras and lacking p53, loss of autophagy no longer blocks tumour progression, but actually accelerates tumour onset, with metabolic analysis revealing enhanced glucose uptake and enrichment of anabolic pathways, which can fuel tumour growth. These findings provide considerable insight into the role of autophagy in cancer and have important implications for autophagy inhibition in cancer therapy. In this regard, we also show that treatment of mice with the autophagy inhibitor hydroxychloroquine, which is currently being used in several clinical trials, significantly accelerates tumour formation in mice containing oncogenic Kras but lacking p53.


Cell Stem Cell | 2010

Spindle Orientation Bias in Gut Epithelial Stem Cell Compartments Is Lost in Precancerous Tissue

Aaron Quyn; Paul L. Appleton; Francis A. Carey; Robert Steele; Nick Barker; Hans Clevers; Rachel A. Ridgway; Owen J. Sansom; Inke S. Näthke

The importance of asymmetric divisions for stem cell function and maintenance is well established in the developing nervous system and the skin; however, its role in gut epithelium and its importance for tumorigenesis is still debated. We demonstrate alignment of mitotic spindles perpendicular to the apical surface specifically in the stem cell compartments of mouse and human intestine and colon. This orientation correlates with the asymmetric retention of label-retaining DNA. Both the preference for perpendicular spindle alignment and asymmetric label retention are lost in precancerous tissue heterozygous for the adenomatous polyposis coli tumor suppressor (Apc). This loss correlates with cell shape changes specifically in the stem cell compartment. Our data suggest that loss of asymmetric division in stem cells might contribute to the oncogenic effect of Apc mutations in gut epithelium.


Cell Stem Cell | 2013

ROS Production and NF-κB Activation Triggered by RAC1 Facilitate WNT-Driven Intestinal Stem Cell Proliferation and Colorectal Cancer Initiation

Kevin Myant; Patrizia Cammareri; Ewan J. McGhee; Rachel A. Ridgway; David J. Huels; Julia B. Cordero; Sarah Schwitalla; Gabriela Kalna; Erinn-Lee Ogg; Dimitris Athineos; Paul Timpson; Marcos Vidal; Graeme I. Murray; Florian R. Greten; Kurt I. Anderson; Owen J. Sansom

Summary The Adenomatous Polyposis Coli (APC) gene is mutated in the majority of colorectal cancers (CRCs). Loss of APC leads to constitutively active WNT signaling, hyperproliferation, and tumorigenesis. Identification of pathways that facilitate tumorigenesis after APC loss is important for therapeutic development. Here, we show that RAC1 is a critical mediator of tumorigenesis after APC loss. We find that RAC1 is required for expansion of the LGR5 intestinal stem cell (ISC) signature, progenitor hyperproliferation, and transformation. Mechanistically, RAC1-driven ROS and NF-κB signaling mediate these processes. Together, these data highlight that ROS production and NF-κB activation triggered by RAC1 are critical events in CRC initiation.


Nature Cell Biology | 2015

Hepatic progenitor cells of biliary origin with liver repopulation capacity

Wei-Yu Lu; Tom Bird; Luke Boulter; Atsunori Tsuchiya; Alicia M. Cole; Trevor Hay; Rachel Guest; Davina Wojtacha; Tak Yung Man; Alison C. MacKinnon; Rachel A. Ridgway; Timothy Kendall; Michael Williams; Thomas Jamieson; Alex Raven; David C. Hay; John P. Iredale; Alan Richard Clarke; Owen J. Sansom; Stuart J. Forbes

Hepatocytes and cholangiocytes self-renew following liver injury. Following severe injury hepatocytes are increasingly senescent, but whether hepatic progenitor cells (HPCs) then contribute to liver regeneration is unclear. Here, we describe a mouse model where the E3 ubiquitin ligase Mdm2 is inducibly deleted in more than 98% of hepatocytes, causing apoptosis, necrosis and senescence with nearly all hepatocytes expressing p21. This results in florid HPC activation, which is necessary for survival, followed by complete, functional liver reconstitution. HPCs isolated from genetically normal mice, using cell surface markers, were highly expandable and phenotypically stable in vitro. These HPCs were transplanted into adult mouse livers where hepatocyte Mdm2 was repeatedly deleted, creating a non-competitive repopulation assay. Transplanted HPCs contributed significantly to restoration of liver parenchyma, regenerating hepatocytes and biliary epithelia, highlighting their in vivo lineage potency. HPCs are therefore a potential future alternative to hepatocyte or liver transplantation for liver disease.


Cancer Cell | 2014

MicroRNA-135b Promotes Cancer Progression by Acting as a Downstream Effector of Oncogenic Pathways in Colon Cancer

Nicola Valeri; Chiara Braconi; Pierluigi Gasparini; Claudio Murgia; Andrea Lampis; Viola Paulus-Hock; Jonathan R. Hart; Lynn Ueno; Sergei I. Grivennikov; Francesca Lovat; Alessio Paone; Luciano Cascione; Khlea M. Sumani; Angelo Veronese; Muller Fabbri; Stefania Carasi; Hansjuerg Alder; Giovanni Lanza; Roberta Gafà; Mary P. Moyer; Rachel A. Ridgway; Julia B. Cordero; Gerard J. Nuovo; Wendy L. Frankel; Massimo Rugge; Matteo Fassan; Joanna Groden; Peter K. Vogt; Michael Karin; Owen J. Sansom

Summary MicroRNA deregulation is frequent in human colorectal cancers (CRCs), but little is known as to whether it represents a bystander event or actually drives tumor progression in vivo. We show that miR-135b overexpression is triggered in mice and humans by APC loss, PTEN/PI3K pathway deregulation, and SRC overexpression and promotes tumor transformation and progression. We show that miR-135b upregulation is common in sporadic and inflammatory bowel disease-associated human CRCs and correlates with tumor stage and poor clinical outcome. Inhibition of miR-135b in CRC mouse models reduces tumor growth by controlling genes involved in proliferation, invasion, and apoptosis. We identify miR-135b as a key downsteam effector of oncogenic pathways and a potential target for CRC treatment.


Developmental Cell | 2010

Focal Adhesion Kinase Is Required for Intestinal Regeneration and Tumorigenesis Downstream of Wnt/c-Myc Signaling

Gabrielle H. Ashton; Jennifer P. Morton; Kevin Myant; Toby J. Phesse; Rachel A. Ridgway; Victoria Marsh; Julie A. Wilkins; Dimitris Athineos; Vanesa Muncan; Richard Kemp; Kristi L. Neufeld; Hans Clevers; Valerie G. Brunton; Douglas J. Winton; Xiaoyan Wang; Rosalie C. Sears; Alan Richard Clarke; Margaret C. Frame; Owen J. Sansom

The intestinal epithelium has a remarkable capacity to regenerate after injury and DNA damage. Here, we show that the integrin effector protein Focal Adhesion Kinase (FAK) is dispensable for normal intestinal homeostasis and DNA damage signaling, but is essential for intestinal regeneration following DNA damage. Given Wnt/c-Myc signaling is activated following intestinal regeneration, we investigated the functional importance of FAK following deletion of the Apc tumor suppressor protein within the intestinal epithelium. Following Apc loss, FAK expression increased in a c-Myc-dependent manner. Codeletion of Apc and Fak strongly reduced proliferation normally induced following Apc loss, and this was associated with reduced levels of phospho-Akt and suppression of intestinal tumorigenesis in Apc heterozygous mice. Thus, FAK is required downstream of Wnt Signaling, for Akt/mTOR activation, intestinal regeneration, and tumorigenesis. Importantly, this work suggests that FAK inhibitors may suppress tumorigenesis in patients at high risk of developing colorectal cancer.


Nature | 2015

mTORC1 mediated translational elongation limits intestinal tumour initiation and growth

William J. Faller; Thomas J. Jackson; John R. P. Knight; Rachel A. Ridgway; Thomas Jamieson; Saadia A. Karim; Carolyn Jones; Sorina Radulescu; David J. Huels; Kevin Myant; Kate Dudek; Helen A. Casey; Alessandro Scopelliti; Julia B. Cordero; Marcos Vidal; Mario Pende; Alexey G. Ryazanov; Nahum Sonenberg; Oded Meyuhas; Michael N. Hall; Martin Bushell; Anne E. Willis; Owen J. Sansom

Inactivation of APC is a strongly predisposing event in the development of colorectal cancer, prompting the search for vulnerabilities specific to cells that have lost APC function. Signalling through the mTOR pathway is known to be required for epithelial cell proliferation and tumour growth, and the current paradigm suggests that a critical function of mTOR activity is to upregulate translational initiation through phosphorylation of 4EBP1 (refs 6, 7). This model predicts that the mTOR inhibitor rapamycin, which does not efficiently inhibit 4EBP1 (ref. 8), would be ineffective in limiting cancer progression in APC-deficient lesions. Here we show in mice that mTOR complex 1 (mTORC1) activity is absolutely required for the proliferation of Apc-deficient (but not wild-type) enterocytes, revealing an unexpected opportunity for therapeutic intervention. Although APC-deficient cells show the expected increases in protein synthesis, our study reveals that it is translation elongation, and not initiation, which is the rate-limiting component. Mechanistically, mTORC1-mediated inhibition of eEF2 kinase is required for the proliferation of APC-deficient cells. Importantly, treatment of established APC-deficient adenomas with rapamycin (which can target eEF2 through the mTORC1–S6K–eEF2K axis) causes tumour cells to undergo growth arrest and differentiation. Taken together, our data suggest that inhibition of translation elongation using existing, clinically approved drugs, such as the rapalogs, would provide clear therapeutic benefit for patients at high risk of developing colorectal cancer.

Collaboration


Dive into the Rachel A. Ridgway's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nick Barker

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar

Hans Clevers

Royal Netherlands Academy of Arts and Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sorina Radulescu

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge