Network


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

Hotspot


Dive into the research topics where Jessamy Tiffen is active.

Publication


Featured researches published by Jessamy Tiffen.


Nature Genetics | 2014

POT1 loss-of-function variants predispose to familial melanoma

Carla Daniela Robles-Espinoza; Mark Harland; Andrew J. Ramsay; Lauren G. Aoude; Zhihao Ding; Karen A. Pooley; Antonia L. Pritchard; Jessamy Tiffen; Mia Petljak; Jane M. Palmer; Judith Symmons; Peter Johansson; Mitchell S. Stark; Michael Gartside; Helen Snowden; Grant W. Montgomery; Nicholas G. Martin; Jimmy Z. Liu; Jiyeon Choi; Matthew Makowski; Kevin M. Brown; Alison M. Dunning; Thomas M. Keane; Carlos López-Otín; Nelleke A. Gruis; Nicholas K. Hayward; D. Timothy Bishop; Julia Newton-Bishop; David J. Adams

Deleterious germline variants in CDKN2A account for around 40% of familial melanoma cases, and rare variants in CDK4, BRCA2, BAP1 and the promoter of TERT have also been linked to the disease. Here we set out to identify new high-penetrance susceptibility genes by sequencing 184 melanoma cases from 105 pedigrees recruited in the UK, The Netherlands and Australia that were negative for variants in known predisposition genes. We identified families where melanoma cosegregates with loss-of-function variants in the protection of telomeres 1 gene (POT1), with a proportion of family members presenting with an early age of onset and multiple primary tumors. We show that these variants either affect POT1 mRNA splicing or alter key residues in the highly conserved oligonucleotide/oligosaccharide-binding (OB) domains of POT1, disrupting protein-telomere binding and leading to increased telomere length. These findings suggest that POT1 variants predispose to melanoma formation via a direct effect on telomeres.


International Journal of Cancer | 2014

Targeting glutamine transport to suppress melanoma cell growth

Qian Wang; Kimberley A. Beaumont; Nicholas J. Otte; Josep Font; Charles G. Bailey; Michelle van Geldermalsen; Danae M. Sharp; Jessamy Tiffen; Renae M. Ryan; Mika Jormakka; Nikolas K. Haass; John E.J. Rasko; Jeff Holst

Amino acids, especially leucine and glutamine, are important for tumor cell growth, survival and metabolism. A range of different transporters deliver each specific amino acid into cells, some of which are increased in cancer. These amino acids consequently activate the mTORC1 pathway and drive cell cycle progression. The leucine transporter LAT1/4F2hc heterodimer assembles as part of a large complex with the glutamine transporter ASCT2 to transport amino acids. In this study, we show that the expression of LAT1 and ASCT2 is significantly increased in human melanoma samples and is present in both BRAFWT (C8161 and WM852) and BRAFV600E mutant (1205Lu and 451Lu) melanoma cell lines. While inhibition of LAT1 by BCH did not suppress melanoma cell growth, the ASCT2 inhibitor BenSer significantly reduced both leucine and glutamine transport in melanoma cells, leading to inhibition of mTORC1 signaling. Cell proliferation and cell cycle progression were significantly reduced in the presence of BenSer in melanoma cells in 2D and 3D cell culture. This included reduced expression of the cell cycle regulators CDK1 and UBE2C. The importance of ASCT2 expression in melanoma was confirmed by shRNA knockdown, which inhibited glutamine uptake, mTORC1 signaling and cell proliferation. Taken together, our study demonstrates that ASCT2‐mediated glutamine transport is a potential therapeutic target for both BRAFWT and BRAFV600E melanoma.


Journal of the National Cancer Institute | 2013

Targeting Amino Acid Transport in Metastatic Castration-Resistant Prostate Cancer: Effects on Cell Cycle, Cell Growth, and Tumor Development

Qian Wang; Jessamy Tiffen; Charles G. Bailey; Melanie Lehman; William Ritchie; Ladan Fazli; Cynthia Metierre; Yue Feng; Estelle Li; Martin Gleave; Grant Buchanan; Colleen C. Nelson; John E.J. Rasko; Jeff Holst

BACKGROUND L-type amino acid transporters (LATs) uptake neutral amino acids including L-leucine into cells, stimulating mammalian target of rapamycin complex 1 signaling and protein synthesis. LAT1 and LAT3 are overexpressed at different stages of prostate cancer, and they are responsible for increasing nutrients and stimulating cell growth. METHODS We examined LAT3 protein expression in human prostate cancer tissue microarrays. LAT function was inhibited using a leucine analog (BCH) in androgen-dependent and -independent environments, with gene expression analyzed by microarray. A PC-3 xenograft mouse model was used to study the effects of inhibiting LAT1 and LAT3 expression. Results were analyzed with the Mann-Whitney U or Fisher exact tests. All statistical tests were two-sided. RESULTS LAT3 protein was expressed at all stages of prostate cancer, with a statistically significant decrease in expression after 4-7 months of neoadjuvant hormone therapy (4-7 month mean = 1.571; 95% confidence interval = 1.155 to 1.987 vs 0 month = 2.098; 95% confidence interval = 1.962 to 2.235; P = .0187). Inhibition of LAT function led to activating transcription factor 4-mediated upregulation of amino acid transporters including ASCT1, ASCT2, and 4F2hc, all of which were also regulated via the androgen receptor. LAT inhibition suppressed M-phase cell cycle genes regulated by E2F family transcription factors including critical castration-resistant prostate cancer regulatory genes UBE2C, CDC20, and CDK1. In silico analysis of BCH-downregulated genes showed that 90.9% are statistically significantly upregulated in metastatic castration-resistant prostate cancer. Finally, LAT1 or LAT3 knockdown in xenografts inhibited tumor growth, cell cycle progression, and spontaneous metastasis in vivo. CONCLUSION Inhibition of LAT transporters may provide a novel therapeutic target in metastatic castration-resistant prostate cancer, via suppression of mammalian target of rapamycin complex 1 activity and M-phase cell cycle genes.


PLOS ONE | 2015

Inducible but not constitutive expression of PD-L1 in human melanoma cells is dependent on activation of NF-κB

Kavitha Gowrishankar; Dilini Gunatilake; Stuart J. Gallagher; Jessamy Tiffen; Helen Rizos; Peter Hersey

Monoclonal antibodies against immune checkpoint blockade have proven to be a major success in the treatment of melanoma. The programmed death receptor-1 ligand-1 (PD-L1) expression on melanoma cells is believed to have an inhibitory effect on T cell responses and to be an important escape mechanism from immune attack. Previous studies have shown that PD-L1 can be expressed constitutively or can be induced by IFN-γ secreted by infiltrating lymphocytes. In the present study we have investigated the mechanism underlying these two modes of PD-L1 expression in melanoma cells including cells that had acquired resistance to the BRAF inhibitor vemurafenib. PD-L1 expression was examined by flow cytometry and immunoblotting. Specific inhibitors and siRNA knockdown approaches were used to examine the roles of the RAF/ MEK, PI3K, NF-κB, STAT3 and AP1/ c-Jun pathways. IFN-γ inducible expression of PD-L1 was dependent on NF-κB as shown by inhibition with BMS-345541, an inhibitor of IκB and the BET protein inhibitor I-BET151, as well as by siRNA knockdown of NF-κB subunits. We were unable to implicate the BRAF/MEK pathway as major regulators in PD-L1 expression on vemurafenib resistant cells. Similarly the PI3K/AKT pathway and the transcription factors STAT3 and c-Jun had only minor roles in IFN-γ induced expression of PD-L1. The mechanism underlying constitutive expression remains unresolved. We suggest these results have significance in selection of treatments that can be used in combination with monoclonal antibodies against PD1, to enhance their effectiveness and to reduce inhibitory effects melanoma cells have against cytotoxic T cell activity.


Molecular Cancer | 2010

Luciferase expression and bioluminescence does not affect tumor cell growth in vitro or in vivo

Jessamy Tiffen; Charles G. Bailey; Cynthia Ng; John E.J. Rasko; Jeff Holst

Live animal imaging is becoming an increasingly common technique for accurate and quantitative assessment of tumor burden over time. Bioluminescence imaging systems rely on a bioluminescent signal from tumor cells, typically generated from expression of the firefly luciferase gene. However, previous reports have suggested that either a high level of luciferase or the resultant light reaction produced upon addition of D-luciferin substrate can have a negative influence on tumor cell growth. To address this issue, we designed an expression vector that allows simultaneous fluorescence and luminescence imaging. Using fluorescence activated cell sorting (FACS), we generated clonal cell populations from a human breast cancer (MCF-7) and a mouse melanoma (B16-F10) cell line that stably expressed different levels of luciferase. We then compared the growth capabilities of these clones in vitro by MTT proliferation assay and in vivo by bioluminescence imaging of tumor growth in live mice. Surprisingly, we found that neither the amount of luciferase nor biophotonic activity was sufficient to inhibit tumor cell growth, in vitro or in vivo. These results suggest that luciferase toxicity is not a necessary consideration when designing bioluminescence experiments, and therefore our approach can be used to rapidly generate high levels of luciferase expression for sensitive imaging experiments.


Nature Genetics | 2014

Inactivating CUX1 mutations promote tumorigenesis

Chi C. Wong; Inigo Martincorena; Alistair G. Rust; Mamunur Rashid; Constantine Alifrangis; Ludmil B. Alexandrov; Jessamy Tiffen; Christina Kober; Anthony R. Green; Charlie E. Massie; Jyoti Nangalia; Stella Lempidaki; Hartmut Döhner; Konstanze Döhner; Sarah Bray; Ultan McDermott; Elli Papaemmanuil; Peter J. Campbell; David J. Adams

A major challenge in cancer genetics is to determine which low-frequency somatic mutations are drivers of tumorigenesis. Here we interrogate the genomes of 7,651 diverse human cancers and find inactivating mutations in the homeodomain transcription factor gene CUX1 (cut-like homeobox 1) in ∼1–5% of various tumors. Meta-analysis of CUX1 mutational status in 2,519 cases of myeloid malignancies reveals disruptive mutations associated with poor survival, highlighting the clinical significance of CUX1 loss. In parallel, we validate CUX1 as a bona fide tumor suppressor using mouse transposon-mediated insertional mutagenesis and Drosophila cancer models. We demonstrate that CUX1 deficiency activates phosphoinositide 3-kinase (PI3K) signaling through direct transcriptional downregulation of the PI3K inhibitor PIK3IP1 (phosphoinositide-3-kinase interacting protein 1), leading to increased tumor growth and susceptibility to PI3K-AKT inhibition. Thus, our complementary approaches identify CUX1 as a pan-driver of tumorigenesis and uncover a potential strategy for treating CUX1-mutant tumors.


Pigment Cell & Melanoma Research | 2014

Control of NF‐kB activity in human melanoma by bromodomain and extra‐terminal protein inhibitor I‐BET151

Stuart J. Gallagher; Branka Mijatov; Dilini Gunatilake; Kavitha Gowrishankar; Jessamy Tiffen; Wilmott James; Lei Jin; Gulietta M. Pupo; Carleen Cullinane; Grant A. McArthur; Peter J. Tummino; Helen Rizos; Peter Hersey

The transcription factor NF‐kappaB (NF‐kB) is a key regulator of cytokine and chemokine production in melanoma and is responsible for symptoms such as anorexia, fatigue, and weight loss. In addition, NF‐kB is believed to contribute to progression of the disease by upregulation of cell cycle and anti‐apoptotic genes and to contribute to resistance against targeted therapies and immunotherapy. In this study, we have examined the ability of the bromodomain and extra‐terminal (BET) protein inhibitor I‐BET151 to inhibit NF‐kB in melanoma cells. We show that I‐BET151 is a potent, selective inhibitor of a number of NF‐kB target genes involved in induction of inflammation and cell cycle regulation and downregulates production of cytokines such as IL‐6 and IL‐8. SiRNA studies indicate that BRD2 is the main BET protein involved in regulation of NF‐kB and that I‐BET151 caused transcriptional downregulation of the NF‐kB subunit p105/p50. These results suggest that BET inhibitors may have an important role in treatment of melanoma where activation of NF‐kB may have a key pathogenic role.


International Journal of Cancer | 2013

The cancer-testis antigen BORIS phenocopies the tumor suppressor CTCF in normal and neoplastic cells

Jessamy Tiffen; Charles G. Bailey; Amy D. Marshall; Cynthia Metierre; Yue Feng; Qian Wang; Sarah Watson; Jeff Holst; John E.J. Rasko

BORIS and CTCF are paralogous, multivalent 11‐zinc finger transcription factors that play important roles in organizing higher‐order chromatin architecture. BORIS is a cancer‐testis antigen with a poorly defined function in cancer, although it has been hypothesized to exhibit oncogenic properties. CTCF, however, has been postulated as a candidate tumor suppressor. We collated the genetic lesions in BORIS and CTCF from multiple cancers identified using high‐throughput genomics. In BORIS, nonsense and missense mutations are evenly distributed. In CTCF, recurrent mutations are mostly clustered in the conserved zinc finger domain and at residues critical for contacting DNA and zinc ion co‐ordination. Three missense mutations are common to both proteins. We used an inducible lentivector to express wildtype BORIS or CTCF in primary cells and cancer cell lines in order to define their functional differences. Both BORIS and CTCF caused a significant decrease in cell proliferation and clonogenic capacity, without alteration of specific cell cycle phases. Both BORIS and CTCF conferred protective effects in primary cells and some cancer cells during UV damage‐induced apoptosis. Using a bioluminescent MCF‐7 orthotopic breast cancer model in vivo, we demonstrated that CTCF and BORIS suppressed breast cancer growth. These findings provide further evidence that CTCF behaves as a tumor suppressor, and show BORIS has a similar growth inhibitory effect in vitro and in vivo. Hence, acquired zinc finger mutations may disrupt these functions, thereby contributing to tumor growth and development.


Oncotarget | 2015

Targeting activating mutations of EZH2 leads to potent cell growth inhibition in human melanoma by derepression of tumor suppressor genes

Jessamy Tiffen; Dilini Gunatilake; Stuart J. Gallagher; Kavitha Gowrishankar; Anja Heinemann; Carleen Cullinane; Ken Dutton-Regester; Gulietta M. Pupo; Dario Strbenac; Jean Y. Yang; Jason Madore; Graham J. Mann; Nicholas K. Hayward; Grant A. McArthur; Fabian V. Filipp; Peter Hersey

The epigenetic modifier EZH2 is part of the polycomb repressive complex that suppresses gene expression via histone methylation. Activating mutations in EZH2 are found in a subset of melanoma that contributes to disease progression by inactivating tumor suppressor genes. In this study we have targeted EZH2 with a specific inhibitor (GSK126) or depleted EZH2 protein by stable shRNA knockdown. We show that inhibition of EZH2 has potent effects on the growth of both wild-type and EZH2 mutant human melanoma in vitro particularly in cell lines harboring the EZH2Y646 activating mutation. This was associated with cell cycle arrest, reduced proliferative capacity in both 2D and 3D culture systems, and induction of apoptosis. The latter was caspase independent and mediated by the release of apoptosis inducing factor (AIFM1) from mitochondria. Gene expression arrays showed that several well characterized tumor suppressor genes were reactivated by EZH2 inhibition. This included activating transcription factor 3 (ATF3) that was validated as an EZH2 target gene by ChIP-qPCR. These results emphasize a critical role for EZH2 in the proliferation and viability of melanoma and highlight the potential for targeted therapy against EZH2 in treatment of patients with melanoma.


The Journal of Pathology | 2016

Adenoma development in familial adenomatous polyposis and MUTYH-associated polyposis: somatic landscape and driver genes

Mamunur Rashid; Andrej Fischer; Cathy H Wilson; Jessamy Tiffen; Alistair G. Rust; Philip Stevens; Shelley Idziaszczyk; Julie Helen Maynard; Geraint T. Williams; Ville Mustonen; Julian Roy Sampson; David J. Adams

Familial adenomatous polyposis (FAP) and MUTYH‐associated polyposis (MAP) are inherited disorders associated with multiple colorectal adenomas that lead to a very high risk of colorectal cancer. The somatic mutations that drive adenoma development in these conditions have not been investigated comprehensively. In this study we performed analysis of paired colorectal adenoma and normal tissue DNA from individuals with FAP or MAP, sequencing 14 adenoma whole exomes (eight MAP, six FAP), 55 adenoma targeted exomes (33 MAP, 22 FAP) and germline DNA from each patient, and a further 63 adenomas by capillary sequencing (41 FAP, 22 MAP). With these data we examined the profile of mutated genes, the mutational signatures and the somatic mutation rates, observing significant diversity in the constellations of mutated driver genes in different adenomas, and loss‐of‐function mutations in WTX (9%; p < 9.99e‐06), a gene implicated in regulation of the WNT pathway and p53 acetylation. These data extend our understanding of the early events in colorectal tumourigenesis in the polyposis syndromes.

Collaboration


Dive into the Jessamy Tiffen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dilini Gunatilake

Kolling Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John E.J. Rasko

Royal Prince Alfred Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carleen Cullinane

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Grant A. McArthur

Peter MacCallum Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge