Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Genome Res ; 25(2): 201-12, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25452314

ABSTRACT

Mechanisms to coordinate programs of highly transcribed genes required for cellular homeostasis and growth are unclear. Upstream binding transcription factor (UBTF, also called UBF) is thought to function exclusively in RNA polymerase I (Pol I)-specific transcription of the ribosomal genes. Here, we report that the two isoforms of UBTF (UBTF1/2) are also enriched at highly expressed Pol II-transcribed genes throughout the mouse genome. Further analysis of UBTF1/2 DNA binding in immortalized human epithelial cells and their isogenically matched transformed counterparts reveals an additional repertoire of UBTF1/2-bound genes involved in the regulation of cell cycle checkpoints and DNA damage response. As proof of a functional role for UBTF1/2 in regulating Pol II transcription, we demonstrate that UBTF1/2 is required for recruiting Pol II to the highly transcribed histone gene clusters and for their optimal expression. Intriguingly, lack of UBTF1/2 does not affect chromatin marks or nucleosome density at histone genes. Instead, it results in increased accessibility of the histone promoters and transcribed regions to micrococcal nuclease, implicating UBTF1/2 in mediating DNA accessibility. Unexpectedly, UBTF2, which does not function in Pol I transcription, is sufficient to regulate histone gene expression in the absence of UBTF1. Moreover, depletion of UBTF1/2 and subsequent reduction in histone gene expression is associated with DNA damage and genomic instability independent of Pol I transcription. Thus, we have uncovered a novel role for UBTF1 and UBTF2 in maintaining genome stability through coordinating the expression of highly transcribed Pol I (UBTF1 activity) and Pol II genes (UBTF2 activity).


Subject(s)
Gene Expression Regulation , Genomic Instability , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Polymerase II/genetics , RNA Polymerase I/genetics , Transcription, Genetic , Animals , Binding Sites , Cell Line, Transformed , Chromatin/metabolism , Chromatin Immunoprecipitation , Computational Biology , DNA Damage , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Histones/genetics , Humans , Mice , Multigene Family , NIH 3T3 Cells , Nucleosomes/metabolism , Pol1 Transcription Initiation Complex Proteins/genetics , Protein Binding , Transcription Initiation Site
2.
J Pathol ; 242(3): 273-283, 2017 07.
Article in English | MEDLINE | ID: mdl-28369925

ABSTRACT

Hereditary endocrine neoplasias, including phaeochromocytoma/paraganglioma and medullary thyroid cancer, are caused by autosomal dominant mutations in several familial cancer genes. A common feature of these diseases is the presentation of multiple primary tumours, or multifocal disease representing independent tumour clones that have arisen from the same initiating genetic lesion, but have undergone independent clonal evolution. Such tumours provide an opportunity to discover common cooperative changes required for tumourigenesis, while controlling for the genetic background of the individual. We performed genomic analysis of synchronous and metachronous tumours from five patients bearing germline mutations in the genes SDHB, RET, and MAX. Using whole exome sequencing and high-density single-nucleotide polymorphism arrays, we analysed two to four primary tumours from each patient. We also applied multi-region sampling, to assess intratumoural heterogeneity and clonal evolution, in two cases involving paraganglioma and medullary thyroid cancer, respectively. Heterogeneous patterns of genomic change existed between synchronous or metachronous tumours, with evidence of branching evolution. We observed striking examples of evolutionary convergence involving the same rare somatic copy-number events in synchronous primary phaeochromocytoma/paraganglioma. Convergent events also occurred during clonal evolution of metastatic medullary thyroid cancer. These observations suggest that genetic or epigenetic changes acquired early within precursor cells, or pre-existing within the genetic background of the individual, create contingencies that determine the evolutionary trajectory of the tumour. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Genes, Neoplasm/genetics , Germ-Line Mutation/genetics , Neoplasms, Multiple Primary/genetics , Neoplasms, Second Primary/genetics , Neuroendocrine Tumors/genetics , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Adult , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Exome/genetics , Female , Humans , Loss of Heterozygosity , Male , Neoplasms, Multiple Primary/pathology , Neoplasms, Second Primary/pathology , Neuroendocrine Tumors/pathology , Paraganglioma/genetics , Pheochromocytoma/genetics , Pheochromocytoma/pathology , Polymorphism, Single Nucleotide/genetics , Positron Emission Tomography Computed Tomography , Proto-Oncogene Proteins c-ret/genetics , Succinate Dehydrogenase/genetics , Young Adult
3.
BMC Bioinformatics ; 18(1): 555, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29246107

ABSTRACT

BACKGROUND: High throughput sequencing requires bioinformatics pipelines to process large volumes of data into meaningful variants that can be translated into a clinical report. These pipelines often suffer from a number of shortcomings: they lack robustness and have many components written in multiple languages, each with a variety of resource requirements. Pipeline components must be linked together with a workflow system to achieve the processing of FASTQ files through to a VCF file of variants. Crafting these pipelines requires considerable bioinformatics and IT skills beyond the reach of many clinical laboratories. RESULTS: Here we present Canary, a single program that can be run on a laptop, which takes FASTQ files from amplicon assays through to an annotated VCF file ready for clinical analysis. Canary can be installed and run with a single command using Docker containerization or run as a single JAR file on a wide range of platforms. Although it is a single utility, Canary performs all the functions present in more complex and unwieldy pipelines. All variants identified by Canary are 3' shifted and represented in their most parsimonious form to provide a consistent nomenclature, irrespective of sequencing variation. Further, proximate in-phase variants are represented as a single HGVS 'delins' variant. This allows for correct nomenclature and consequences to be ascribed to complex multi-nucleotide polymorphisms (MNPs), which are otherwise difficult to represent and interpret. Variants can also be annotated with hundreds of attributes sourced from MyVariant.info to give up to date details on pathogenicity, population statistics and in-silico predictors. CONCLUSIONS: Canary has been used at the Peter MacCallum Cancer Centre in Melbourne for the last 2 years for the processing of clinical sequencing data. By encapsulating clinical features in a single, easily installed executable, Canary makes sequencing more accessible to all pathology laboratories. Canary is available for download as source or a Docker image at https://github.com/PapenfussLab/Canary under a GPL-3.0 License.


Subject(s)
Computational Biology/methods , High-Throughput Nucleotide Sequencing/methods , Software , Databases, Genetic , Genetic Variation , Humans
4.
PLoS Genet ; 9(7): e1003627, 2013.
Article in English | MEDLINE | ID: mdl-23874226

ABSTRACT

The capacity of tumour cells to maintain continual overgrowth potential has been linked to the commandeering of normal self-renewal pathways. Using an epithelial cancer model in Drosophila melanogaster, we carried out an overexpression screen for oncogenes capable of cooperating with the loss of the epithelial apico-basal cell polarity regulator, scribbled (scrib), and identified the cell fate regulator, Abrupt, a BTB-zinc finger protein. Abrupt overexpression alone is insufficient to transform cells, but in cooperation with scrib loss of function, Abrupt promotes the formation of massive tumours in the eye/antennal disc. The steroid hormone receptor coactivator, Taiman (a homologue of SRC3/AIB1), is known to associate with Abrupt, and Taiman overexpression also drives tumour formation in cooperation with the loss of Scrib. Expression arrays and ChIP-Seq indicates that Abrupt overexpression represses a large number of genes, including steroid hormone-response genes and multiple cell fate regulators, thereby maintaining cells within an epithelial progenitor-like state. The progenitor-like state is characterised by the failure to express the conserved Eyes absent/Dachshund regulatory complex in the eye disc, and in the antennal disc by the failure to express cell fate regulators that define the temporal elaboration of the appendage along the proximo-distal axis downstream of Distalless. Loss of scrib promotes cooperation with Abrupt through impaired Hippo signalling, which is required and sufficient for cooperative overgrowth with Abrupt, and JNK (Jun kinase) signalling, which is required for tumour cell migration/invasion but not overgrowth. These results thus identify a novel cooperating oncogene, identify mammalian family members of which are also known oncogenes, and demonstrate that epithelial tumours in Drosophila can be characterised by the maintenance of a progenitor-like state.


Subject(s)
Carcinogenesis , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , MAP Kinase Signaling System/genetics , Neoplasms, Glandular and Epithelial/genetics , Nuclear Proteins/genetics , Animals , Cell Proliferation , Disease Models, Animal , Drosophila Proteins/metabolism , Eye Neoplasms/genetics , Eye Neoplasms/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Glandular and Epithelial/pathology , Nuclear Proteins/metabolism , Oncogene Protein p65(gag-jun)/genetics , Oncogene Protein p65(gag-jun)/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
5.
PLoS Genet ; 8(9): e1002894, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028338

ABSTRACT

Despite intensive efforts using linkage and candidate gene approaches, the genetic etiology for the majority of families with a multi-generational breast cancer predisposition is unknown. In this study, we used whole-exome sequencing of thirty-three individuals from 15 breast cancer families to identify potential predisposing genes. Our analysis identified families with heterozygous, deleterious mutations in the DNA repair genes FANCC and BLM, which are responsible for the autosomal recessive disorders Fanconi Anemia and Bloom syndrome. In total, screening of all exons in these genes in 438 breast cancer families identified three with truncating mutations in FANCC and two with truncating mutations in BLM. Additional screening of FANCC mutation hotspot exons identified one pathogenic mutation among an additional 957 breast cancer families. Importantly, none of the deleterious mutations were identified among 464 healthy controls and are not reported in the 1,000 Genomes data. Given the rarity of Fanconi Anemia and Bloom syndrome disorders among Caucasian populations, the finding of multiple deleterious mutations in these critical DNA repair genes among high-risk breast cancer families is intriguing and suggestive of a predisposing role. Our data demonstrate the utility of intra-family exome-sequencing approaches to uncover cancer predisposition genes, but highlight the major challenge of definitively validating candidates where the incidence of sporadic disease is high, germline mutations are not fully penetrant, and individual predisposition genes may only account for a tiny proportion of breast cancer families.


Subject(s)
Breast Neoplasms/genetics , Fanconi Anemia Complementation Group C Protein/genetics , RecQ Helicases/genetics , Sequence Deletion/genetics , Alleles , Exome/genetics , Exons , Fanconi Anemia Complementation Group C Protein/metabolism , Female , Genetic Predisposition to Disease , Genome, Human , Humans , Pedigree , Polymorphism, Genetic , RecQ Helicases/metabolism , Sequence Analysis, DNA
6.
Nucleic Acids Res ; 39(8): 3267-81, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21177653

ABSTRACT

Loss of c-MYC is required for downregulation of ribosomal RNA (rRNA) gene (rDNA) transcription by RNA Polymerase I (Pol I) during granulocyte differentiation. Here, we demonstrate a robust reduction of Pol I loading onto rDNA that along with a depletion of the MYC target gene upstream binding factor (UBF) and a switch from epigenetically active to silent rDNA accompanies this MYC reduction. We hypothesized that MYC may coordinate these mechanisms via direct regulation of multiple components of the Pol I transcription apparatus. Using gene expression arrays we identified a 'regulon' of Pol I factors that are both downregulated during differentiation and reinduced in differentiated granulocytes upon activation of the MYC-ER transgene. This regulon includes the novel c-MYC target genes RRN3 and POLR1B. Although enforced MYC expression during granulocyte differentiation was sufficient to increase the number of active rRNA genes, its activation in terminally differentiated cells did not alter the active to inactive gene ratio despite increased rDNA transcription. Thus, c-MYC dynamically controls rDNA transcription during granulocytic differentiation through the orchestrated transcriptional regulation of core Pol I factors and epigenetic modulation of number of active rRNA genes.


Subject(s)
DNA Polymerase I/metabolism , Genes, rRNA , Granulocytes/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Cell Differentiation , Cell Line , Chromatin Assembly and Disassembly , DNA, Ribosomal/metabolism , Gene Expression Profiling , Granulocytes/cytology , Granulocytes/enzymology , Neutrophils/metabolism , Pol1 Transcription Initiation Complex Proteins/biosynthesis , Pol1 Transcription Initiation Complex Proteins/genetics , Transcription, Genetic
7.
J Clin Oncol ; 40(18): 2036-2047, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35263119

ABSTRACT

PURPOSE: Tubo-ovarian cancer (TOC) is a sentinel cancer for BRCA1 and BRCA2 pathogenic variants (PVs). Identification of a PV in the first member of a family at increased genetic risk (the proband) provides opportunities for cancer prevention in other at-risk family members. Although Australian testing rates are now high, PVs in patients with TOC whose diagnosis predated revised testing guidelines might have been missed. We assessed the feasibility of detecting PVs in this population to enable genetic risk reduction in relatives. PATIENTS AND METHODS: In this pilot study, deceased probands were ascertained from research cohort studies, identification by a relative, and gynecologic oncology clinics. DNA was extracted from archival tissue or stored blood for panel sequencing of 10 risk-associated genes. Testing of deceased probands ascertained through clinic records was performed with a consent waiver. RESULTS: We identified 85 PVs in 84 of 787 (11%) probands. Familial contacts of 39 of 60 (65%) deceased probands with an identified recipient (60 of 84; 71%) have received a written notification of results, with follow-up verbal contact made in 85% (33 of 39). A minority of families (n = 4) were already aware of the PV. For many (29 of 33; 88%), the genetic result provided new information and referral to a genetic service was accepted in most cases (66%; 19 of 29). Those who declined referral (4 of 29) were all male next of kin whose family member had died more than 10 years before. CONCLUSION: We overcame ethical and logistic challenges to demonstrate that retrospective genetic testing to identify PVs in previously untested deceased probands with TOC is feasible. Understanding reasons for a family member's decision to accept or decline a referral will be important for guiding future TRACEBACK projects.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , Australia , Breast Neoplasms/genetics , Carcinoma, Ovarian Epithelial/genetics , Family , Female , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Male , Ovarian Neoplasms/genetics , Ovarian Neoplasms/prevention & control , Pilot Projects , Retrospective Studies
8.
Phys Eng Sci Med ; 44(4): 1213-1219, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34505991

ABSTRACT

Volumetric medical imaging lacks a standardised coordinate geometry which links image frame-of-reference to specific anatomical regions. This results in an inability to locate anatomy in medical images without visual assessment and precludes a variety of image analysis tasks which could benefit from a standardised, machine-readable coordinate system. In this work, a proposed geometric system that scales based on patient size is described and applied to a variety of cases in computed tomography imaging. Subsequently, a convolutional neural network is trained to associate axial slice CT image appearance with the standardised coordinate value along the patient superior-inferior axis. The trained neural network showed an accuracy of ± 12 mm in the ability to predict per-slice reference location and was relatively stable across all annotated regions ranging from brain to thighs. A version of the trained model along with scripts to perform network training in other applications are made available. Finally, a selection of potential use applications are illustrated including organ localisation, image registration initialisation, and scan length determination for auditing diagnostic reference levels.


Subject(s)
Image Processing, Computer-Assisted , Neural Networks, Computer , Humans , Tomography, X-Ray Computed
9.
Nat Commun ; 12(1): 1434, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664264

ABSTRACT

Although melanoma is initiated by acquisition of point mutations and limited focal copy number alterations in melanocytes-of-origin, the nature of genetic changes that characterise lethal metastatic disease is poorly understood. Here, we analyze the evolution of human melanoma progressing from early to late disease in 13 patients by sampling their tumours at multiple sites and times. Whole exome and genome sequencing data from 88 tumour samples reveals only limited gain of point mutations generally, with net mutational loss in some metastases. In contrast, melanoma evolution is dominated by whole genome doubling and large-scale aneuploidy, in which widespread loss of heterozygosity sculpts the burden of point mutations, neoantigens and structural variants even in treatment-naïve and primary cutaneous melanomas in some patients. These results imply that dysregulation of genomic integrity is a key driver of selective clonal advantage during melanoma progression.


Subject(s)
Aneuploidy , DNA Copy Number Variations/genetics , Genome, Human/genetics , Melanoma/genetics , Skin Neoplasms/genetics , Disease Progression , Exome/genetics , Humans , INDEL Mutation/genetics , Melanocytes/pathology , Point Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Exome Sequencing , Whole Genome Sequencing , Melanoma, Cutaneous Malignant
11.
Sci Rep ; 9(1): 6426, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015508

ABSTRACT

Next Generation Sequencing is now routinely used in the practice of diagnostic pathology to detect clinically relevant somatic and germline sequence variations in patient samples. However, clinical assessment of copy number variations (CNVs) and large-scale structural variations (SVs) is still challenging. While tools exist to estimate both, their results are typically presented separately in tables or static plots which can be difficult to read and are unable to show the context needed for clinical interpretation and reporting. We have addressed this problem with CNspector, a multi-scale interactive browser that shows CNVs in the context of other relevant genomic features to enable fast and effective clinical reporting. We illustrate the utility of CNspector at different genomic scales across a variety of sample types in a range of case studies. We show how CNspector can be used for diagnosis and reporting of exon-level deletions, focal gene-level amplifications, chromosome and chromosome arm level amplifications/deletions and in complex genomic rearrangements. CNspector is a web-based clinical variant browser tailored to the clinical application of next generation sequencing for CNV assessment. We have demonstrated the utility of this interactive software in typical applications across a range of tissue types and disease contexts encountered in the context of diagnostic pathology. CNspector is written in R and the source code is available for download under the GPL3 Licence from https://github.com/PapenfussLab/CNspector . A server running CNspector loaded with the figures from this paper can be accessed at https://shiny.wehi.edu.au/jmarkham/CNspector/index.html .


Subject(s)
Basal Cell Nevus Syndrome/diagnosis , Carcinoma, Basal Cell/diagnosis , DNA Copy Number Variations , High-Throughput Nucleotide Sequencing/methods , Web Browser , Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/pathology , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Chromosome Deletion , Chromosome Duplication , Exons , Genome, Human , Humans , Internet , Sequence Analysis, DNA
12.
Genome Med ; 9(1): 38, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28438193

ABSTRACT

BACKGROUND: The increasing affordability of DNA sequencing has allowed it to be widely deployed in pathology laboratories. However, this has exposed many issues with the analysis and reporting of variants for clinical diagnostic use. Implementing a high-throughput sequencing (NGS) clinical reporting system requires a diverse combination of capabilities, statistical methods to identify variants, global variant databases, a validated bioinformatics pipeline, an auditable laboratory workflow, reproducible clinical assays and quality control monitoring throughout. These capabilities must be packaged in software that integrates the disparate components into a useable system. RESULTS: To meet these needs, we developed a web-based application, PathOS, which takes variant data from a patient sample through to a clinical report. PathOS has been used operationally in the Peter MacCallum Cancer Centre for two years for the analysis, curation and reporting of genetic tests for cancer patients, as well as the curation of large-scale research studies. PathOS has also been deployed in cloud environments allowing multiple institutions to use separate, secure and customisable instances of the system. Increasingly, the bottleneck of variant curation is limiting the adoption of clinical sequencing for molecular diagnostics. PathOS is focused on providing clinical variant curators and pathology laboratories with a decision support system needed for personalised medicine. While the genesis of PathOS has been within cancer molecular diagnostics, the system is applicable to NGS clinical reporting generally. CONCLUSIONS: The widespread availability of genomic sequencers has highlighted the limited availability of software to support clinical decision-making in molecular pathology. PathOS is a system that has been developed and refined in a hospital laboratory context to meet the needs of clinical diagnostics. The software is available as a set of Docker images and source code at https://github.com/PapenfussLab/PathOS .


Subject(s)
Clinical Laboratory Services , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , Software , Humans , Neoplasms/diagnosis , Precision Medicine , Sequence Analysis, DNA/methods
13.
PLoS One ; 10(7): e0132987, 2015.
Article in English | MEDLINE | ID: mdl-26207831

ABSTRACT

During tumorigenesis, pathways that promote the epithelial-to-mesenchymal transition (EMT) can both facilitate metastasis and endow tumor cells with cancer stem cell properties. To gain a greater understanding of how these properties are interlinked in cancers we used Drosophila epithelial tumor models, which are driven by orthologues of human oncogenes (activated alleles of Ras and Notch) in cooperation with the loss of the cell polarity regulator, scribbled (scrib). Within these tumors, both invasive, mesenchymal-like cell morphology and continual tumor overgrowth, are dependent upon Jun N-terminal kinase (JNK) activity. To identify JNK-dependent changes within the tumors we used a comparative microarray analysis to define a JNK gene signature common to both Ras and Notch-driven tumors. Amongst the JNK-dependent changes was a significant enrichment for BTB-Zinc Finger (ZF) domain genes, including chronologically inappropriate morphogenesis (chinmo). chinmo was upregulated by JNK within the tumors, and overexpression of chinmo with either RasV12 or Nintra was sufficient to promote JNK-independent epithelial tumor formation in the eye/antennal disc, and, in cooperation with RasV12, promote tumor formation in the adult midgut epithelium. Chinmo primes cells for oncogene-mediated transformation through blocking differentiation in the eye disc, and promoting an escargot-expressing stem or enteroblast cell state in the adult midgut. BTB-ZF genes are also required for Ras and Notch-driven overgrowth of scrib mutant tissue, since, although loss of chinmo alone did not significantly impede tumor development, when loss of chinmo was combined with loss of a functionally related BTB-ZF gene, abrupt, tumor overgrowth was significantly reduced. abrupt is not a JNK-induced gene, however, Abrupt is present in JNK-positive tumor cells, consistent with a JNK-associated oncogenic role. As some mammalian BTB-ZF proteins are also highly oncogenic, our work suggests that EMT-promoting signals in human cancers could similarly utilize networks of these proteins to promote cancer stem cell states.


Subject(s)
Carcinogenesis/genetics , Drosophila Proteins/physiology , Genes, ras/physiology , Oncogenes/physiology , Receptors, Notch/physiology , Zinc Fingers/genetics , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Microarray Analysis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Interaction Domains and Motifs/genetics
14.
PLoS One ; 9(3): e88950, 2014.
Article in English | MEDLINE | ID: mdl-24658684

ABSTRACT

Tumor invasion and metastasis involves complex remodeling of gene expression programs governing epithelial homeostasis. Mutational activation of the RAS-ERK is a frequent occurrence in many cancers and has been shown to drive overexpression of the AP-1 family transcription factor FRA1, a potent regulator of migration and invasion in a variety of tumor cell types. However, the nature of FRA1 transcriptional targets and the molecular pathways through which they promote tumor progression remain poorly understood. We found that FRA1 was strongly expressed in tumor cells at the invasive front of human colorectal cancers (CRCs), and that its depletion suppressed mesenchymal-like features in CRC cells in vitro. Genome-wide analysis of FRA1 chromatin occupancy and transcriptional regulation identified epithelial-mesenchymal transition (EMT)-related genes as a major class of direct FRA1 targets in CRC cells. Expression of the pro-mesenchymal subset of these genes predicted adverse outcomes in CRC patients, and involved FRA-1-dependent regulation and cooperation with TGFß signaling pathway. Our findings reveal an unexpectedly widespread and direct role for FRA1 in control of epithelial-mesenchymal plasticity in CRC cells, and suggest that FRA1 plays an important role in mediating cross talk between oncogenic RAS-ERK and TGFß signaling networks during tumor progression.


Subject(s)
Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Colorectal Neoplasms/genetics , Humans , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-fos , Signal Transduction , Transforming Growth Factor beta/metabolism
15.
BMC Med Genomics ; 7: 23, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24885028

ABSTRACT

BACKGROUND: Clinical specimens undergoing diagnostic molecular pathology testing are fixed in formalin due to the necessity for detailed morphological assessment. However, formalin fixation can cause major issues with molecular testing, as it causes DNA damage such as fragmentation and non-reproducible sequencing artefacts after PCR amplification. In the context of massively parallel sequencing (MPS), distinguishing true low frequency variants from sequencing artefacts remains challenging. The prevalence of formalin-induced DNA damage and its impact on molecular testing and clinical genomics remains poorly understood. METHODS: The Cancer 2015 study is a population-based cancer cohort used to assess the feasibility of mutational screening using MPS in cancer patients from Victoria, Australia. While blocks were formalin-fixed and paraffin-embedded in different anatomical pathology laboratories, they were centrally extracted for DNA utilising the same protocol, and run through the same MPS platform (Illumina TruSeq Amplicon Cancer Panel). The sequencing artefacts in the 1-10% and the 10-25% allele frequency ranges were assessed in 488 formalin-fixed tumours from the pilot phase of the Cancer 2015 cohort. All blocks were less than 2.5 years of age (mean 93 days). RESULTS: Consistent with the signature of DNA damage due to formalin fixation, many formalin-fixed samples displayed disproportionate levels of C>T/G>A changes in the 1-10% allele frequency range. Artefacts were less apparent in the 10-25% allele frequency range. Significantly, changes were inversely correlated with coverage indicating high levels of sequencing artefacts were associated with samples with low amounts of available amplifiable template due to fragmentation. The degree of fragmentation and sequencing artefacts differed between blocks sourced from different anatomical pathology laboratories. In a limited validation of potentially actionable low frequency mutations, a NRAS G12D mutation in a melanoma was shown to be a false positive. CONCLUSIONS: These findings indicate that DNA damage following formalin fixation remains a major challenge in laboratories working with MPS. Methodologies that assess, minimise or remove formalin-induced DNA damaged templates as part of MPS protocols will aid in the interpretation of genomic results leading to better patient outcomes.


Subject(s)
Artifacts , High-Throughput Nucleotide Sequencing/methods , Mutation/genetics , Neoplasms/genetics , Tissue Fixation , Cell Line, Tumor , DNA Fragmentation , False Positive Reactions , Formaldehyde , Humans , Paraffin Embedding , Polymerase Chain Reaction , Prospective Studies , Sequence Analysis, DNA , Templates, Genetic , Uracil-DNA Glycosidase/metabolism
16.
Cell Rep ; 7(4): 1009-19, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24813887

ABSTRACT

N-methyl-2-pyrrolidone (NMP) is a common solvent and drug vehicle. We discovered unexpected antineoplastic and immunomodulatory activity of NMP in a cMYC-driven myeloma model. Coincident to this, NMP was identified as an acetyllysine mimetic and candidate bromodomain ligand. Accordingly, NMP-treated cells demonstrated transcriptional overlap with BET-bromodomain inhibition, including downregulation of cMYC and IRF4. NMP's immunomodulatory activity occurred at sub-BET inhibitory concentrations, and, despite phenotypic similarities to lenalidomide, its antimyeloma activity was independent of the IMiD targets cereblon and Ikaros-1/3. Thus, low-affinity yet broad-spectrum bromodomain inhibition by NMP mediates biologically potent, cereblon-independent immunomodulation and at higher doses targets malignant cells directly via BET antagonism. These data reveal that NMP is a functional acetyllysine mimetic with pleotropic antimyeloma and immunomodulatory activities. Our studies highlight the potential therapeutic benefits of NMP, the consequences of current human NMP exposures, and the need for reassessment of scientific literature where NMP was used as an "inert" drug-delivery vehicle.


Subject(s)
Antineoplastic Agents/pharmacology , Immunologic Factors/pharmacology , Multiple Myeloma/drug therapy , Pyrrolidinones/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
17.
PLoS One ; 9(4): e95217, 2014.
Article in English | MEDLINE | ID: mdl-24752294

ABSTRACT

Targeted resequencing by massively parallel sequencing has become an effective and affordable way to survey small to large portions of the genome for genetic variation. Despite the rapid development in open source software for analysis of such data, the practical implementation of these tools through construction of sequencing analysis pipelines still remains a challenging and laborious activity, and a major hurdle for many small research and clinical laboratories. We developed TREVA (Targeted REsequencing Virtual Appliance), making pre-built pipelines immediately available as a virtual appliance. Based on virtual machine technologies, TREVA is a solution for rapid and efficient deployment of complex bioinformatics pipelines to laboratories of all sizes, enabling reproducible results. The analyses that are supported in TREVA include: somatic and germline single-nucleotide and insertion/deletion variant calling, copy number analysis, and cohort-based analyses such as pathway and significantly mutated genes analyses. TREVA is flexible and easy to use, and can be customised by Linux-based extensions if required. TREVA can also be deployed on the cloud (cloud computing), enabling instant access without investment overheads for additional hardware. TREVA is available at http://bioinformatics.petermac.org/treva/.


Subject(s)
Computational Biology/methods , Exome/genetics , Genome, Human/genetics , Sequence Analysis, DNA , User-Computer Interface , Animals , Humans , Melanoma/genetics , Mice , Mutation/genetics
18.
J Clin Invest ; 123(12): 5351-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24231354

ABSTRACT

Ionizing radiation (IR) and germline mutations in the retinoblastoma tumor suppressor gene (RB1) are the strongest risk factors for developing osteosarcoma. Recapitulating the human predisposition, we found that Rb1+/- mice exhibited accelerated development of IR-induced osteosarcoma, with a latency of 39 weeks. Initial exposure of osteoblasts to carcinogenic doses of IR in vitro and in vivo induced RB1-dependent senescence and the expression of a panel of proteins known as senescence-associated secretory phenotype (SASP), dominated by IL-6. RB1 expression closely correlated with that of the SASP cassette in human osteosarcomas, and low expression of both RB1 and the SASP genes was associated with poor prognosis. In vivo, IL-6 was required for IR-induced senescence, which elicited NKT cell infiltration and a host inflammatory response. Mice lacking IL-6 or NKT cells had accelerated development of IR-induced osteosarcomas. These data elucidate an important link between senescence, which is a cell-autonomous tumor suppressor response, and the activation of host-dependent cancer immunosurveillance. Our findings indicate that overcoming the immune response to senescence is a rate-limiting step in the formation of IR-induced osteosarcoma.


Subject(s)
Bone Neoplasms/immunology , Cellular Senescence/physiology , Natural Killer T-Cells/immunology , Neoplasms, Radiation-Induced/immunology , Osteosarcoma/immunology , Retinoblastoma Protein/physiology , Animals , Bone Neoplasms/etiology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Calcium Radioisotopes/toxicity , Cytokines/physiology , Genes, Retinoblastoma , Humans , Immunologic Surveillance , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-6/deficiency , Interleukin-6/physiology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Neoplasm Transplantation/immunology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Osteoblasts/pathology , Osteosarcoma/etiology , Osteosarcoma/genetics , Osteosarcoma/pathology , Phenotype , Prognosis , RNA Interference , Retinoblastoma Protein/antagonists & inhibitors
19.
Eur J Cancer ; 49(18): 3936-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24011934

ABSTRACT

BACKGROUND: Ovarian cancer is the major cause of death from gynaecological malignancy with a 5year survival of only ∼30% due to resistance to platinum and paclitaxel-based first line therapy. Dysregulation of the phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) and RAS/extracellular signal-regulated kinase (ERK) pathways is common in ovarian cancer, providing potential new targets for 2nd line therapy. METHODS: We determined the inhibition of proliferation of an extensive panel of ovarian cancer cell lines, encompassing all the major histotypes, by the dual PI3K/mTOR inhibitor PF-04691502 and a MEK inhibitor, PD-0325901. In addition, we analysed global gene expression, mutation status of key PI3K/mTOR and RAS/ERK pathway members and pathway activation to identify predictors of drug response. RESULTS: PF-04691502 inhibits proliferation of the majority of cell lines with potencies that correlate with the extent of pathway inhibition. Resistant cell lines were characterised by activation of the RAS/ERK pathway as indicated by differential gene expression profiles and pathway activity analysis. PD-0325901 suppressed growth of a subset of cell lines that were characterised by high basal RAS/ERK signalling. Strikingly, using PF-04691502 and PD-0325901 in combination resulted in synergistic growth inhibition in 5/6 of PF-04691502 resistant cell lines and two cell lines resistant to both single agents showed robust synergistic growth arrest. Xenograft studies confirm the utility of combination therapy to synergistically inhibit tumour growth of PF-04691502-resistant tumours in vivo. CONCLUSIONS: These studies identify dual targeted inhibitors of PI3K/mTOR in combination with inhibitors of RAS/ERK signalling as a potentially effective new approach to treating ovarian cancer.


Subject(s)
Benzamides/pharmacology , Cell Proliferation/drug effects , Diphenylamine/analogs & derivatives , Ovarian Neoplasms/drug therapy , Pyridones/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/administration & dosage , Cell Line, Tumor , Diphenylamine/administration & dosage , Diphenylamine/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pyridones/administration & dosage , Pyrimidines/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays , ras Proteins/genetics , ras Proteins/metabolism
20.
Clin Cancer Res ; 17(9): 3039-47, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21224373

ABSTRACT

PURPOSE: Patients presenting with locally advanced rectal cancer currently receive preoperative radiotherapy with or without chemotherapy. Although pathologic complete response is achieved for approximately 10% to 30% of patients, a proportion of patients derive no benefit from this therapy while being exposed to toxic side effects of treatment. Therefore, there is a strong need to identify patients who are unlikely to benefit from neoadjuvant therapy to help direct them toward alternate and ultimately more successful treatment options. EXPERIMENTAL DESIGN: In this study, we obtained expression profiles from pretreatment biopsies for 51 rectal cancer patients. All patients underwent preoperative chemoradiotherapy, followed by resection of the tumor 6 to 8 weeks posttreatment. Gene expression and response to treatment were correlated, and a supervised learning algorithm was used to generate an original predictive classifier and validate previously published classifiers. RESULTS: Novel predictive classifiers based on Mandard's tumor regression grade, metabolic response, TNM (tumor node metastasis) downstaging, and normal tissue expression profiles were generated. Because there were only 7 patients who had minimal treatment response (>80% residual tumor), expression profiles were used to predict good tumor response and outcome. These classifiers peaked at 82% sensitivity and 89% specificity; however, classifiers with the highest sensitivity had poor specificity, and vice versa. Validation of predictive classifiers from previously published reports was attempted using this cohort; however, sensitivity and specificity ranged from 21% to 70%. CONCLUSIONS: These results show that the clinical utility of microarrays in predictive medicine is not yet within reach for rectal cancer and alternatives to microarrays should be considered for predictive studies in rectal adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Gene Expression Profiling , Rectal Neoplasms/drug therapy , Rectal Neoplasms/radiotherapy , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Pharmacological/analysis , Biomarkers, Pharmacological/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Combined Modality Therapy , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Microarray Analysis , Middle Aged , Neoadjuvant Therapy , Prognosis , Rectal Neoplasms/diagnosis , Rectal Neoplasms/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL