Network


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

Hotspot


Dive into the research topics where Cristina Gamell is active.

Publication


Featured researches published by Cristina Gamell.


FEBS Letters | 2014

PML tumour suppression and beyond: Therapeutic implications

Cristina Gamell; Piotr Jan Paul; Ygal Haupt; Sue Haupt

Recognition of the tumour suppressive capacity of the Promyelocytic Leukemia protein (PML) has emerged beyond its identification through APL, to a broad spectrum of tumors. This ability has chiefly been linked to its role as a core component of dynamic structures termed PML Nuclear Bodies (PML‐NBs). In response to a variety of stresses, key factors and their molecular modifiers are recruited to PML‐NBs, where activating modifications are facilitated, leading to a cellular stress response. PML was also found to perform anti‐tumourigenic functions through cytoplasmic activities. Surprisingly, important recent research defined growth promoting capabilities of PML, which significantly challenges the notion of a ‘classic’ tumour suppressor. Through metabolic reprogramming, PML can afford a selective advantage for tumor cells in certain settings. The multiple forms in which PML exists are the likely explanation of this functional diversity. This behavioral ambiguity however raises a significant challenge to the design of strategies to therapeutically target PML. In this review we discuss this change of paradigm in the PML field and its ramifications, particularly for tailoring cancer therapies.


Cell Death and Disease | 2015

Targeting Mdmx to treat breast cancers with wild-type p53

Susan Haupt; D Buckley; J-Mb Pang; J Panimaya; Piotr Jan Paul; Cristina Gamell; Elena A. Takano; Y Ying Lee; S Hiddingh; T-M Rogers; Amina Teunisse; Marco J. Herold; J-C Marine; Stephen B. Fox; Aart G. Jochemsen; Ygal Haupt

The function of the tumor suppressor p53 is universally compromised in cancers. It is the most frequently mutated gene in human cancers (reviewed). In cases where p53 is not mutated, alternative regulatory pathways inactivate its tumor suppressive functions. This is primarily achieved through elevation in the expression of the key inhibitors of p53: Mdm2 or Mdmx (also called Mdm4) (reviewed). In breast cancer (BrCa), the frequency of p53 mutations varies markedly between the different subtypes, with basal-like BrCas bearing a high frequency of p53 mutations, whereas luminal BrCas generally express wild-type (wt) p53. Here we show that Mdmx is unexpectedly highly expressed in normal breast epithelial cells and its expression is further elevated in most luminal BrCas, whereas p53 expression is generally low, consistent with wt p53 status. Inducible knockdown (KD) of Mdmx in luminal BrCa MCF-7 cells impedes the growth of these cells in culture, in a p53-dependent manner. Importantly, KD of Mdmx in orthotopic xenograft transplants resulted in growth inhibition associated with prolonged survival, both in a preventative model and also in a treatment model. Growth impediment in response to Mdmx KD was associated with cellular senescence. The growth inhibitory capacity of Mdmx KD was recapitulated in an additional luminal BrCa cell line MPE600, which expresses wt p53. Further, the growth inhibitory capacity of Mdmx KD was also demonstrated in the wt p53 basal-like cell line SKBR7 line. These results identify Mdmx growth dependency in wt p53 expressing BrCas, across a range of subtypes. Based on our findings, we propose that Mdmx targeting is an attractive strategy for treating BrCas harboring wt p53.


Redox biology | 2018

Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis

Shashank Masaldan; Sharnel A.S. Clatworthy; Cristina Gamell; Peter M. Meggyesy; Antonia-Tonia Rigopoulos; Sue Haupt; Ygal Haupt; Delphine Denoyer; Paul A. Adlard; Ashley I. Bush; Michael A. Cater

Cellular senescence is characterised by the irreversible arrest of proliferation, a pro-inflammatory secretory phenotype and evasion of programmed cell death mechanisms. We report that senescence alters cellular iron acquisition and storage and also impedes iron-mediated cell death pathways. Senescent cells, regardless of stimuli (irradiation, replicative or oncogenic), accumulate vast amounts of intracellular iron (up to 30-fold) with concomitant changes in the levels of iron homeostasis proteins. For instance, ferritin (iron storage) levels provided a robust biomarker of cellular senescence, for associated iron accumulation and for resistance to iron-induced toxicity. Cellular senescence preceded iron accumulation and was not perturbed by sustained iron chelation (deferiprone). Iron accumulation in senescent cells was driven by impaired ferritinophagy, a lysosomal process that promotes ferritin degradation and ferroptosis. Lysosomal dysfunction in senescent cells was confirmed through several markers, including the build-up of microtubule-associated protein light chain 3 (LC3-II) in autophagosomes. Impaired ferritin degradation explains the iron accumulation phenotype of senescent cells, whereby iron is effectively trapped in ferritin creating a perceived cellular deficiency. Accordingly, senescent cells were highly resistant to ferroptosis. Promoting ferritin degradation by using the autophagy activator rapamycin averted the iron accumulation phenotype of senescent cells, preventing the increase of TfR1, ferritin and intracellular iron, but failed to re-sensitize these cells to ferroptosis. Finally, the enrichment of senescent cells in mouse ageing hepatic tissue was found to accompany iron accumulation, an elevation in ferritin and mirrored our observations using cultured senescent cells.


The Journal of Pathology | 2017

MDM4 is a rational target for treating breast cancers with mutant p53

Panimaya Jeffreena Miranda; Daniel Buckley; Dinesh Raghu; Jia-Min B. Pang; Elena A. Takano; Reshma Vijayakumaran; Amina Teunisse; Atara Posner; Tahlia Procter; Marco J. Herold; Cristina Gamell; Jean-Christophe Marine; Stephen B. Fox; Aart G. Jochemsen; Sue Haupt; Ygal Haupt

Mutation of the key tumour suppressor p53 defines a transition in the progression towards aggressive and metastatic breast cancer (BC) with the poorest outcome. Specifically, the p53 mutation frequency exceeds 50% in triple‐negative BC. Key regulators of mutant p53 that facilitate its oncogenic functions are potential therapeutic targets. We report here that the MDM4 protein is frequently abundant in the context of mutant p53 in basal‐like BC samples. Importantly, we show that MDM4 plays a critical role in the proliferation of these BC cells. We demonstrate that conditional knockdown (KD) of MDM4 provokes growth inhibition across a range of BC subtypes with mutant p53, including luminal, Her2+ and triple‐negative BCs. In vivo, MDM4 was shown to be crucial for the establishment and progression of tumours. This growth inhibition was mediated, at least in part, by the cell cycle inhibitor p27. Depletion of p27 together with MDM4 KD led to recovery of the proliferative capacity of cells that were growth‐inhibited by MDM4 KD alone. Consistently, we identified low levels of p27 expression in basal‐like tumours corresponding to high levels of MDM4 and p53. This predicts a signature for a subset of tumours that may be amenable to therapies targeted towards MDM4 and mutant p53. The therapeutic potential of MDM4 as a target in BC with mutant p53 was shown in vitro by use of a small‐molecule inhibitor. Overall, our study supports MDM4 as a novel therapeutic target for BC expressing mutant p53. Copyright


Science Signaling | 2017

Reduced abundance of the E3 ubiquitin ligase E6AP contributes to decreased expression of the INK4/ARF locus in non–small cell lung cancer

Cristina Gamell; Twishi Gulati; Yaara Levav-Cohen; Richard J. Young; Hongdo Do; Pat Pilling; Elena A. Takano; Neil Watkins; Stephen B. Fox; Prudence A. Russell; Doron Ginsberg; Brendon J. Monahan; Gavin Wright; Alexander Dobrovic; Sue Haupt; Ben Solomon; Ygal Haupt

E6AP exhibits tumor suppressor activity that may help stratify NSCLC patients. INK4/ARF repression in lung cancer by loss of E6AP The abundance of the cell cycle decelerator p16INK4a, encoded at the INK4/ARF locus, is decreased in various cancers. Gamell et al. found that the absence of p16INK4a in some patients is explained by the lack of the E3 ubiquitin ligase E6AP. E6AP bound to and inhibited the activity of transcription factor E2F1; a decrease in E6AP abundance enabled E2F1-mediated expression of the cell cycle promoter CDC6, which encodes a transcriptional regulator that represses INK4/ARF expression. The findings identify a tumor-suppressive role for E6AP in lung cancer and suggest that targeting the E2F1-CDC6 pathway might slow tumor growth in some NSCLC patients. The tumor suppressor p16INK4a, one protein encoded by the INK4/ARF locus, is frequently absent in multiple cancers, including non–small cell lung cancer (NSCLC). Whereas increased methylation of the encoding gene (CDKN2A) accounts for its loss in a third of patients, no molecular explanation exists for the remainder. We unraveled an alternative mechanism for the silencing of the INK4/ARF locus involving the E3 ubiquitin ligase and transcriptional cofactor E6AP (also known as UBE3A). We found that the expression of three tumor suppressor genes encoded in the INK4/ARF locus (p15INK4b, p16INK4a, and p19ARF) was decreased in E6AP−/− mouse embryo fibroblasts. E6AP induced the expression of the INK4/ARF locus at the transcriptional level by inhibiting CDC6 transcription, a gene encoding a key repressor of the locus. Luciferase assays revealed that E6AP inhibited CDC6 expression by reducing its E2F1-dependent transcription. Chromatin immunoprecipitation analysis indicated that E6AP reduced the amount of E2F1 at the CDC6 promoter. In a subset of NSCLC samples, an E6AP-low/CDC6-high/p16INK4a-low protein abundance profile correlated with low methylation of the gene encoding p16INK4a (CDKN2A) and poor patient prognosis. These findings define a previously unrecognized tumor-suppressive role for E6AP in NSCLC, reveal an alternative silencing mechanism of the INK4/ARF locus, and reveal E6AP as a potential prognostic marker in NSCLC.


Oncogene | 2016

Restoration of tumor suppression in prostate cancer by targeting the E3 ligase E6AP

Piotr Jan Paul; Dinesh Raghu; Ai-Leen Chan; Twishi Gulati; Luke S. Lambeth; Eriko Takano; Marco J. Herold; Jim Hagekyriakou; Robert L. Vessella; Clare G Fedele; Mark Shackleton; Elizabeth D. Williams; Stephen B. Fox; Scott Williams; Susan Haupt; Cristina Gamell; Ygal Haupt

Restoration of tumor suppression is an attractive onco-therapeutic approach. It is particularly relevant when a tumor suppressor is excessively degraded by an overactive oncogenic E3 ligase. We previously discovered that the E6-associated protein (E6AP; as classified in the human papilloma virus context) is an E3 ligase that has an important role in the cellular stress response, and it directly targets the tumor-suppressor promyelocytic leukemia protein (PML) for proteasomal degradation. In this study, we have examined the role of the E6AP–PML axis in prostate cancer (PC). We show that knockdown (KD) of E6AP expression attenuates growth of PC cell lines in vitro. We validated this finding in vivo using cell line xenografts, patient-derived xenografts and mouse genetics. We found that KD of E6AP attenuates cancer cell growth by promoting cellular senescence in vivo, which correlates with restoration of tumor suppression by PML. In addition, we show that KD of E6AP sensitizes cells to radiation-induced death. Overall, our findings demonstrate a role for E6AP in the promotion of PC and support E6AP targeting as a novel approach for PC treatment, either alone or in combination with radiation.


The Prostate | 2018

Exploring the oncoproteomic response of human prostate cancer to therapeutic radiation using data-independent acquisition (DIA) mass spectrometry

Simon P. Keam; Twishi Gulati; Cristina Gamell; Franco Caramia; Cheng Huang; Ralf B. Schittenhelm; Oded Kleifeld; Paul Neeson; Ygal Haupt; Scott Williams

The development of radioresistance in prostate cancer (PCa) is an important clinical issue and is still largely uninformed by personalized molecular characteristics. The aim of this study was to establish a platform that describes the early oncoproteomic response of human prostate tissue to radiation therapy (RT) using a prospective human tissue cohort.


Oncotarget | 2017

E6AP promotes prostate cancer by reducing p27 expression

Dinesh Raghu; Piotr Jan Paul; Twishi Gulati; Siddhartha Deb; Christine Khoo; Andrea Russo; Enzo Gallo; Giovanni Blandino; Ai-Leen Chan; Elena A. Takano; Shahneen Sandhu; Stephen B. Fox; Scott Williams; Sue Haupt; Cristina Gamell; Ygal Haupt

Prostate cancer (PC) is the most common cancer in men. Elevated levels of E3 ligase, E6-Associated Protein (E6AP) were previously linked to PC, consistent with increased protein expression in a subset of PC patients. In cancers, irregular E3 ligase activity drives proteasomal degradation of tumor suppressor proteins. Accordingly, E3 ligase inhibitors define a rational therapy to restore tumor suppression. The relevant tumor suppressors targeted by E6AP in PC are yet to be fully identified. In this study we show that p27, a key cell cycle regulator, is a target of E6AP in PC. Down regulation of E6AP increases p27 expression and enhances its nuclear accumulation in PC. We demonstrate that E6AP regulates p27 expression by inhibiting its transcription in an E2F1-dependent manner. Concomitant knockdown of E6AP and p27 partially restores PC cell growth, supporting the contribution of p27 to the overall effect of E6AP on prostate tumorigenesis. Overall, we unravelled the E6AP-p27 axis as a new promoter of PC, exposing an attractive target for therapy through the restoration of tumor suppression.


Redox biology | 2018

Copper accumulation in senescent cells: Interplay between copper transporters and impaired autophagy

Shashank Masaldan; Sharnel A.S. Clatworthy; Cristina Gamell; Zoe M. Smith; Paul S. Francis; Delphine Denoyer; Peter M. Meggyesy; Sharon La Fontaine; Michael A. Cater

Cellular senescence is characterized by irreversible growth arrest incurred through either replicative exhaustion or by pro-oncogenic cellular stressors (radioactivity, oxidative stress, oncogenic activation). The enrichment of senescent cells in tissues with age has been associated with tissue dyshomeostasis and age-related pathologies including cancers, neurodegenerative disorders (e.g. Alzheimers, Parkinsons, etc.) and metabolic disorders (e.g. diabetes). We identified copper accumulation as being a universal feature of senescent cells [mouse embryonic fibroblasts (MEF), human prostate epithelial cells and human diploid fibroblasts] in vitro. Elevated copper in senescent MEFs was accompanied by elevated levels of high-affinity copper uptake protein 1 (Ctr1), diminished levels of copper-transporting ATPase 1 (Atp7a) (copper export) and enhanced antioxidant defence reflected by elevated levels of glutathione (GSH), superoxide dismutase 1 (SOD1) and glutaredoxin 1 (Grx1). The levels of intracellular copper were further increased in senescent MEFs cultured in copper supplemented medium and in senescent Mottled Brindled (Mobr) MEFs lacking functional Atp7a. Finally, we demonstrated that the restoration/preservation of autophagic-lysosomal degradation in senescent MEFs following rapamycin treatment correlated with attenuation of copper accumulation in these cells despite a further decrease in Atp7a levels. This study for the first time establishes a link between Atp7a and the autophagic-lysosomal pathway, and a requirement for both to effect efficient copper export. Such a connection between cellular autophagy and copper homeostasis is significant, as both have emerged as important facets of age-associated degenerative disease.


Radiation and Environmental Biophysics | 2018

Biodosimetric transcriptional and proteomic changes are conserved in irradiated human tissue

Simon P. Keam; Twishi Gulati; Cristina Gamell; Franco Caramia; Gisela Mir Arnau; Cheng Huang; Ralf B. Schittenhelm; Oded Kleifeld; Paul Neeson; Scott Williams; Ygal Haupt

Transcriptional dosimetry is an emergent field of radiobiology aimed at developing robust methods for detecting and quantifying absorbed doses using radiation-induced fluctuations in gene expression. A combination of RNA sequencing, array-based and quantitative PCR transcriptomics in cellular, murine and various ex vivo human models has led to a comprehensive description of a fundamental set of genes with demonstrable dosimetric qualities. However, these are yet to be validated in human tissue due to the scarcity of in situ-irradiated source material. This represents a major hurdle to the continued development of transcriptional dosimetry. In this study, we present a novel evaluation of a previously reported set of dosimetric genes in human tissue exposed to a large therapeutic dose of radiation. To do this, we evaluated the quantitative changes of a set of dosimetric transcripts consisting of FDXR, BAX, BCL2, CDKN1A, DDB2, BBC3, GADD45A, GDF15, MDM2, SERPINE1, TNFRSF10B, PLK3, SESN2 and VWCE in guided pre- and post-radiation (2 weeks) prostate cancer biopsies from seven patients. We confirmed the prolonged dose-responsivity of most of these transcripts in in situ-irradiated tissue. BCL2, GDF15, and to some extent TNFRSF10B, were markedly unreliable single markers of radiation exposure. Nevertheless, as a full set, these genes reliably segregated non-irradiated and irradiated tissues and predicted radiation absorption on a patient-specific basis. We also confirmed changes in the translated protein product for a small subset of these dosimeters. This study provides the first confirmatory evidence of an existing dosimetric gene set in less-accessible tissues—ensuring peripheral responses reflect tissue-specific effects. Further work will be required to determine if these changes are conserved in different tissue types, post-radiation times and doses.

Collaboration


Dive into the Cristina Gamell's collaboration.

Top Co-Authors

Avatar

Ygal Haupt

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Sue Haupt

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Twishi Gulati

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Piotr Jan Paul

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Scott Williams

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Stephen B. Fox

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Dinesh Raghu

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Elena A. Takano

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Franco Caramia

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Simon P. Keam

Peter MacCallum Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge