Network


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

Hotspot


Dive into the research topics where Julie Adam is active.

Publication


Featured researches published by Julie Adam.


Cancer Cell | 2011

Renal Cyst Formation in Fh1-Deficient Mice Is Independent of the Hif/Phd Pathway: Roles for Fumarate in KEAP1 Succination and Nrf2 Signaling

Julie Adam; Emine Hatipoglu; Linda O'Flaherty; Nicola Ternette; Natasha Sahgal; Helen Lockstone; Dilair Baban; Emma Nye; Gordon Stamp; Kathryn Wolhuter; Marcus Stevens; R. Fischer; Peter Carmeliet; Patrick H. Maxwell; Christopher W. Pugh; Norma Frizzell; Tomoyoshi Soga; Benedikt M. Kessler; Mona El-Bahrawy; Peter J. Ratcliffe; Patrick J. Pollard

Summary The Krebs cycle enzyme fumarate hydratase (FH) is a human tumor suppressor whose inactivation is associated with the development of leiomyomata, renal cysts, and tumors. It has been proposed that activation of hypoxia inducible factor (HIF) by fumarate-mediated inhibition of HIF prolyl hydroxylases drives oncogenesis. Using a mouse model, we provide genetic evidence that Fh1-associated cyst formation is Hif independent, as is striking upregulation of antioxidant signaling pathways revealed by gene expression profiling. Mechanistic analysis revealed that fumarate modifies cysteine residues within the Kelch-like ECH-associated protein 1 (KEAP1), abrogating its ability to repress the Nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated antioxidant response pathway, suggesting a role for Nrf2 dysregulation in FH-associated cysts and tumors.


Nature | 2011

Haem oxygenase is synthetically lethal with the tumour suppressor fumarate hydratase

Christian Frezza; Liang Zheng; Ori Folger; Kartik N. Rajagopalan; Elaine D. MacKenzie; Livnat Jerby; Massimo Micaroni; Barbara Chaneton; Julie Adam; Ann Hedley; Gabriela Kalna; Ian Tomlinson; Patrick J. Pollard; Watson Dg; Ralph J. DeBerardinis; Tomer Shlomi; Eytan Ruppin; Eyal Gottlieb

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid cycle (TCA cycle) that catalyses the hydration of fumarate into malate. Germline mutations of FH are responsible for hereditary leiomyomatosis and renal-cell cancer (HLRCC). It has previously been demonstrated that the absence of FH leads to the accumulation of fumarate, which activates hypoxia-inducible factors (HIFs) at normal oxygen tensions. However, so far no mechanism that explains the ability of cells to survive without a functional TCA cycle has been provided. Here we use newly characterized genetically modified kidney mouse cells in which Fh1 has been deleted, and apply a newly developed computer model of the metabolism of these cells to predict and experimentally validate a linear metabolic pathway beginning with glutamine uptake and ending with bilirubin excretion from Fh1-deficient cells. This pathway, which involves the biosynthesis and degradation of haem, enables Fh1-deficient cells to use the accumulated TCA cycle metabolites and permits partial mitochondrial NADH production. We predicted and confirmed that targeting this pathway would render Fh1-deficient cells non-viable, while sparing wild-type Fh1-containing cells. This work goes beyond identifying a metabolic pathway that is induced in Fh1-deficient cells to demonstrate that inhibition of haem oxygenation is synthetically lethal when combined with Fh1 deficiency, providing a new potential target for treating HLRCC patients.


The Journal of Pathology | 2011

Aberrant succination of proteins in fumarate hydratase-deficient mice and HLRCC patients is a robust biomarker of mutation status

Chiara Bardella; Mona El-Bahrawy; Norma Frizzell; Julie Adam; Nicola Ternette; Emine Hatipoglu; Kimberley Howarth; Linda O'Flaherty; Ian S. Roberts; Gareth D. H. Turner; Jennifer M. Taylor; Konstantinos Giaslakiotis; Valentine M. Macaulay; Adrian L. Harris; Ashish Chandra; Heli J. Lehtonen; Virpi Launonen; Lauri A. Aaltonen; Christopher W. Pugh; Radu Mihai; David C. Trudgian; Benedikt M. Kessler; John W. Baynes; Peter J. Ratcliffe; Ian Tomlinson; Patrick J. Pollard

Germline mutations in the FH gene encoding the Krebs cycle enzyme fumarate hydratase predispose to hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome. FH‐deficient cells and tissues accumulate high levels of fumarate, which may act as an oncometabolite and contribute to tumourigenesis. A recently proposed role for fumarate in the covalent modification of cysteine residues to S‐(2‐succinyl) cysteine (2SC) (termed protein succination) prompted us to assess 2SC levels in our existing models of HLRCC. Herein, using a previously characterized antibody against 2SC, we show that genetic ablation of FH causes high levels of protein succination. We next hypothesized that immunohistochemistry for 2SC would serve as a metabolic biomarker for the in situ detection of FH‐deficient tissues. Robust detection of 2SC was observed in Fh1 (murine FH)‐deficient renal cysts and in a retrospective series of HLRCC tumours (n = 16) with established FH mutations. Importantly, 2SC was undetectable in normal tissues (n = 200) and tumour types not associated with HLRCC (n = 1342). In a prospective evaluation of cases referred for genetic testing for HLRCC, the presence of 2SC‐modified proteins (2SCP) correctly predicted genetic alterations in FH in every case. In two series of unselected type II papillary renal cancer (PRCC), prospectively analysed by 2SCP staining followed by genetic analysis, the biomarker accurately identified previously unsuspected FH mutations (2/33 and 1/36). The investigation of whether metabolites in other tumour types produce protein modification signature(s) that can be assayed using similar strategies will be of interest in future studies of cancer. Copyright


Human Molecular Genetics | 2010

Dysregulation of hypoxia pathways in fumarate hydratase-deficient cells is independent of defective mitochondrial metabolism

Linda O'Flaherty; Julie Adam; Lisa C. Heather; Alexander V. Zhdanov; Yuen-Li Chung; Melroy X. Miranda; Joanne Croft; S. E. Olpin; Kieran Clarke; Christopher W. Pugh; John R. Griffiths; Dmitri B. Papkovsky; Houman Ashrafian; Peter J. Ratcliffe; Patrick J. Pollard

Mutations in the gene encoding the Krebs cycle enzyme fumarate hydratase (FH) predispose to hereditary leiomyomatosis and renal cell cancer in affected individuals. FH-associated neoplasia is characterized by defective mitochondrial function and by upregulation of transcriptional pathways mediated by hypoxia-inducible factor (HIF), although whether and by what means these processes are linked has been disputed. We analysed the HIF pathway in Fh1-/- mouse embryonic fibroblasts (MEFs), in FH-defective neoplastic tissues and in Fh1-/- MEFs re-expressing either wild-type or an extra-mitochondrial restricted form of FH. These experiments demonstrated that upregulation of HIF-1alpha occurs as a direct consequence of FH inactivation. Fh1-/- cells accumulated intracellular fumarate and manifested severe impairment of HIF prolyl but not asparaginyl hydroxylation which was corrected by provision of exogenous 2-oxoglutarate (2-OG). Re-expression of the extra-mitochondrial form of FH in Fh1-/- cells was sufficient to reduce intracellular fumarate and to correct dysregulation of the HIF pathway completely, even in cells that remained profoundly defective in mitochondrial energy metabolism. The findings indicate that upregulation of HIF-1alpha arises from competitive inhibition of the 2-OG-dependent HIF hydroxylases by fumarate and not from disruption of mitochondrial energy metabolism.


Cell Reports | 2013

Inhibition of Mitochondrial Aconitase by Succination in Fumarate Hydratase Deficiency

Nicola Ternette; Ming Yang; Mahima Laroyia; Mitsuhiro Kitagawa; Linda O’Flaherty; Kathryn Wolhulter; Kaori Igarashi; Kaori Saito; Keiko Kato; R. Fischer; Alexandre Berquand; Benedikt M. Kessler; Terry Lappin; Norma Frizzell; Tomoyoshi Soga; Julie Adam; Patrick J. Pollard

Summary The gene encoding the Krebs cycle enzyme fumarate hydratase (FH) is mutated in hereditary leiomyomatosis and renal cell cancer (HLRCC). Loss of FH activity causes accumulation of intracellular fumarate, which can directly modify cysteine residues to form 2-succinocysteine through succination. We undertook a proteomic-based screen in cells and renal cysts from Fh1 (murine FH)-deficient mice and identified 94 protein succination targets. Notably, we identified the succination of three cysteine residues in mitochondrial Aconitase2 (ACO2) crucial for iron-sulfur cluster binding. We show that fumarate exerts a dose-dependent inhibition of ACO2 activity, which correlates with increased succination as determined by mass spectrometry, possibly by interfering with iron chelation. Importantly, we show that aconitase activity is impaired in FH-deficient cells. Our data provide evidence that succination, resulting from FH deficiency, targets and potentially alters the function of multiple proteins and may contribute to the dysregulated metabolism observed in HLRCC.


Oncogene | 2014

Rare insights into cancer biology.

Julie Adam; Ming Yang; Tomoyoshi Soga; Patrick J. Pollard

Cancer-associated mutations have been identified in the metabolic genes succinate dehydrogenase (SDH), fumarate hydratase (FH) and isocitrate dehydrogenase (IDH), advancing and challenging our understanding of cellular function and disease mechanisms and providing direct links between dysregulated metabolism and cancer. Some striking parallels exist in the cellular consequences of the genetic mutations within this triad of cancer syndromes, including accumulation of oncometabolites and competitive inhibition of 2-oxoglutarate-dependent dioxygenases, particularly, hypoxia-inducible factor (HIF) prolyl hydroxylases, JmjC domain-containing histone demethylases (part of the JMJD family) and the ten-eleven translocation (TET) family of 5methyl cytosine (5mC) DNA hydroxylases. These lead to activation of HIF-dependent oncogenic pathways and inhibition of histone and DNA demethylation. Mutations in FH, resulting in loss of enzyme activity, predispose affected individuals to a rare cancer, hereditary leiomyomatosis and renal cell cancer (HLRCC), characterised by benign smooth muscle cutaneous and uterine tumours (leiomyomata) and an aggressive form of collecting duct and type 2 papillary renal cancer. Interestingly, loss of FH activity results in the accumulation of high levels of fumarate that can lead to the non-enzymatic modification of cysteine residues in multiple proteins (succination) and in some cases to their disrupted function. Here we consider that the study of rare diseases such as HLRCC, combining analyses of human tumours and cell lines with in vitro and in vivo murine models has provided novel insights into cancer biology associated with dysregulated metabolism and represents a useful paradigm for cancer research.


Frontiers in Oncology | 2012

The emerging role of fumarate as an oncometabolite

Ming Yang; Tomoyoshi Soga; Patrick J. Pollard; Julie Adam

The drive to understand how altered cellular metabolism and cancer are linked has caused a paradigm shift in the focus of cancer research. The discovery of a mutated metabolic enzyme, isocitrate dehydrogenase 1, that leads to accumulation of the oncometabolite 2-hydroxyglutarate, provided significant direct evidence that dysfunctional metabolism plays an important role in oncogenesis. Striking parallels exist with the Krebs cycle enzyme fumarate hydratase (FH), a tumor suppressor, whose mutation is associated with the development of leiomyomata, renal cysts, and tumors. Loss of FH enzymatic activity results in accumulation of intracellular fumarate which has been proposed to act as a competitive inhibitor of 2-oxoglutarate-dependent oxygenases including the hypoxia-inducible factor (HIF) hydroxylases, thus activating oncogenic HIF pathways. Interestingly, our studies have questioned the role of HIF and have highlighted other candidate mechanisms, in particular the non-enzymatic modification of cysteine residues (succination) that could lead to disruption or loss of protein functions, dysfunctional cell metabolism and cell signaling. Here, we discuss the evidence for proposing fumarate as an onco-metabolite.


PLOS ONE | 2011

Human AlkB Homologue 5 Is a Nuclear 2-Oxoglutarate Dependent Oxygenase and a Direct Target of Hypoxia-Inducible Factor 1α (HIF-1α)

Armin Thalhammer; Zuzana Bencokova; Rachel Poole; Christoph Loenarz; Julie Adam; Linda O'Flaherty; Johannes Schödel; David R. Mole; Konstantinos Giaslakiotis; Christopher J. Schofield; Ester M. Hammond; Peter J. Ratcliffe; Patrick J. Pollard

Human 2-oxoglutarate oxygenases catalyse a range of biological oxidations including the demethylation of histone and nucleic acid substrates and the hydroxylation of proteins and small molecules. Some of these processes are centrally involved in regulation of cellular responses to hypoxia. The ALKBH proteins are a sub-family of 2OG oxygenases that are defined by homology to the Escherichia coli DNA-methylation repair enzyme AlkB. Here we report evidence that ALKBH5 is probably unique amongst the ALKBH genes in being a direct transcriptional target of hypoxia inducible factor-1 (HIF-1) and is induced by hypoxia in a range of cell types. We show that purified recombinant ALKBH5 is a bona fide 2OG oxygenase that catalyses the decarboxylation of 2OG but appears to have different prime substrate requirements from those so far defined for other ALKBH family members. Our findings define a new class of HIF-transcriptional target gene and suggest that ALKBH5 may have a role in the regulation of cellular responses to hypoxia.


Proceedings of the National Academy of Sciences of the United States of America | 2014

OGFOD1 catalyzes prolyl hydroxylation of RPS23 and is involved in translation control and stress granule formation

Rachelle S. Singleton; Phebee Liu-Yi; Fabio Formenti; Wei Ge; Rok Sekirnik; R. Fischer; Julie Adam; Patrick J. Pollard; Alexander Wolf; Armin Thalhammer; Christoph Loenarz; Emily Flashman; Atsushi Yamamoto; Mathew L. Coleman; Benedikt M. Kessler; Pablo Wappner; Christopher J. Schofield; Peter J. Ratcliffe; Matthew E. Cockman

Significance Members of the 2-oxoglutarate (2OG)-dependent oxygenase superfamily catalyze a range of important biological oxidations. Structurally informed bioinformatic predictions suggest that the human genome encodes as yet unassigned members of the superfamily. We describe work demonstrating that 2OG and Fe(II)-dependent oxygenase domain-containing protein 1 (OGFOD1) is a protein hydroxylase that modifies the small ribosomal subunit protein RPS23 at a conserved prolyl residue in the ribosome-decoding center and that suppression or deletion of OGFOD1 is associated with the activation of translational stress pathways. Together with studies of homologous genes in flies and yeast described in accompanying manuscripts, the work identifies a unique function for 2OG oxygenase-catalyzed hydroxylation in ribosome biology. 2-Oxoglutarate (2OG) and Fe(II)-dependent oxygenase domain-containing protein 1 (OGFOD1) is predicted to be a conserved 2OG oxygenase, the catalytic domain of which is related to hypoxia-inducible factor prolyl hydroxylases. OGFOD1 homologs in yeast are implicated in diverse cellular functions ranging from oxygen-dependent regulation of sterol response genes (Ofd1, Schizosaccharomyces pombe) to translation termination/mRNA polyadenylation (Tpa1p, Saccharomyces cerevisiae). However, neither the biochemical activity of OGFOD1 nor the identity of its substrate has been defined. Here we show that OGFOD1 is a prolyl hydroxylase that catalyzes the posttranslational hydroxylation of a highly conserved residue (Pro-62) in the small ribosomal protein S23 (RPS23). Unusually OGFOD1 retained a high affinity for, and forms a stable complex with, the hydroxylated RPS23 substrate. Knockdown or inactivation of OGFOD1 caused a cell type-dependent induction of stress granules, translational arrest, and growth impairment in a manner complemented by wild-type but not inactive OGFOD1. The work identifies a human prolyl hydroxylase with a role in translational regulation.


Cell Reports | 2013

A Role for Cytosolic Fumarate Hydratase in Urea Cycle Metabolism and Renal Neoplasia

Julie Adam; Ming Yang; Christina Bauerschmidt; Mitsuhiro Kitagawa; Linda O’Flaherty; Pratheesh Maheswaran; Gizem Özkan; Natasha Sahgal; Dilair Baban; Keiko Kato; Kaori Saito; Keiko Iino; Kaori Igarashi; Michael R.L. Stratford; Christopher W. Pugh; Daniel A. Tennant; Christian Ludwig; Benjamin Davies; Peter J. Ratcliffe; Mona El-Bahrawy; Houman Ashrafian; Tomoyoshi Soga; Patrick J. Pollard

Summary The identification of mutated metabolic enzymes in hereditary cancer syndromes has established a direct link between metabolic dysregulation and cancer. Mutations in the Krebs cycle enzyme, fumarate hydratase (FH), predispose affected individuals to leiomyomas, renal cysts, and cancers, though the respective pathogenic roles of mitochondrial and cytosolic FH isoforms remain undefined. On the basis of comprehensive metabolomic analyses, we demonstrate that FH1-deficient cells and tissues exhibit defects in the urea cycle/arginine metabolism. Remarkably, transgenic re-expression of cytosolic FH ameliorated both renal cyst development and urea cycle defects associated with renal-specific FH1 deletion in mice. Furthermore, acute arginine depletion significantly reduced the viability of FH1-deficient cells in comparison to controls. Our findings highlight the importance of extramitochondrial metabolic pathways in FH-associated oncogenesis and the urea cycle/arginine metabolism as a potential therapeutic target.

Collaboration


Dive into the Julie Adam's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian Tomlinson

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Norma Frizzell

University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge