Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Elife ; 92020 07 06.
Article in English | MEDLINE | ID: mdl-32628111

ABSTRACT

Inhibition of WEE1 kinase by AZD1775 has shown promising results in clinical cancer trials, but markers predicting AZD1775 response are lacking. Here we analysed AZD1775 response in a panel of human breast cancer (BC) cell lines by global proteome/transcriptome profiling and identified two groups of basal-like BC (BLBCs): 'PTEN low' BLBCs were highly sensitive to AZD1775 and failed to recover following removal of AZD1775, while 'PTEN high' BLBCs recovered. AZD1775 induced phosphorylation of DNA-PK, protecting cells from replication-associated DNA damage and promoting cellular recovery. Deletion of DNA-PK or PTEN, or inhibition of DNA-PK sensitized recovering BLBCs to AZD1775 by abrogating replication arrest, allowing replication despite DNA damage. This was linked to reduced CHK1 activation, increased cyclin E levels and apoptosis. In conclusion, we identified PTEN and DNA-PK as essential regulators of replication checkpoint arrest in response to AZD1775 and defined PTEN as a promising biomarker for efficient WEE1 cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle Proteins/genetics , DNA-Activated Protein Kinase/genetics , PTEN Phosphohydrolase/genetics , Protein-Tyrosine Kinases/genetics , Pyrazoles/pharmacology , Pyrimidinones/pharmacology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA-Activated Protein Kinase/metabolism , Female , Gene Expression Profiling , Humans , PTEN Phosphohydrolase/metabolism , Protein-Tyrosine Kinases/metabolism , Proteome
2.
Nat Commun ; 11(1): 394, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959748

ABSTRACT

Ionising radiation (IR) is a recognised carcinogen responsible for cancer development in patients previously treated using radiotherapy, and in individuals exposed as a result of accidents at nuclear energy plants. However, the mutational signatures induced by distinct types and doses of radiation are unknown. Here, we analyse the genetic architecture of mammary tumours, lymphomas and sarcomas induced by high (56Fe-ions) or low (gamma) energy radiation in mice carrying Trp53 loss of function alleles. In mammary tumours, high-energy radiation is associated with induction of focal structural variants, leading to genomic instability and Met amplification. Gamma-radiation is linked to large-scale structural variants and a point mutation signature associated with oxidative stress. The genomic architecture of carcinomas, sarcomas and lymphomas arising in the same animals are significantly different. Our study illustrates the complex interactions between radiation quality, germline Trp53 deficiency and tissue/cell of origin in shaping the genomic landscape of IR-induced tumours.


Subject(s)
Carcinogenesis/radiation effects , Genomic Instability/radiation effects , Neoplasms, Radiation-Induced/genetics , Radiation Injuries, Experimental/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis/genetics , DNA Damage/radiation effects , DNA Mutational Analysis , Dose-Response Relationship, Radiation , Female , Gene Amplification/radiation effects , Germ-Line Mutation , Humans , Male , Mice , Mice, Knockout , Neoplasms, Radiation-Induced/pathology , Point Mutation/radiation effects , Proto-Oncogene Proteins c-met/genetics , Radiation Injuries, Experimental/pathology , Whole Genome Sequencing
3.
Int J Cancer ; 146(4): 1125-1138, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31219615

ABSTRACT

Increased expression of GLI1, the main Hedgehog signalling pathway effector, is related to unfavourable prognosis and progressive disease of certain breast cancer subtypes. We used conditional transgenic mice induced to overexpress GLI1 in the mammary epithelium either alone or in combination with deletion of one Trp53 allele to address the role of elevated GLI1 expression in breast tumour initiation and progression. Induced GLI1 expression facilitates mammary gland tumour formation and this was further increased upon heterozygous deletion of Trp53. The GLI1-induced primary tumours were of different murine molecular subtypes, including Normal-likeEx , Class8Ex , Claudin-LowEx and Erbb2-likeEx . The gene expression profiles of some of the tumours correlated well with the PAM50 subtypes for human breast cancer. Whole-exome sequencing revealed somatic mutation profiles with only little overlap between the primary tumours. Orthotopically serially transplanted GLI1-induced tumours maintained the main morphological characteristics of the primary tumours for ≥10 generations. Independent of Trp53 status and molecular subtype, the serially transplanted GLI1-induced tumours were able to grow both in the absence of transgenic GLI1 expression and in the presence of the GLI1 inhibitor GANT61. These data suggest that elevated GLI1 expression has a determinant role in tumour initiation; however, additional genetic events are required for tumour progression.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Zinc Finger Protein GLI1/genetics , Animals , Female , Gene Expression , Genetic Heterogeneity , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Neoplasm Transplantation , Zinc Finger Protein GLI1/biosynthesis
4.
Nat Commun ; 10(1): 1600, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30962452

ABSTRACT

In the preceding decades, molecular characterization has revolutionized breast cancer (BC) research and therapeutic approaches. Presented herein, an unbiased analysis of breast tumor proteomes, inclusive of 9995 proteins quantified across all tumors, for the first time recapitulates BC subtypes. Additionally, poor-prognosis basal-like and luminal B tumors are further subdivided by immune component infiltration, suggesting the current classification is incomplete. Proteome-based networks distinguish functional protein modules for breast tumor groups, with co-expression of EGFR and MET marking ductal carcinoma in situ regions of normal-like tumors and lending to a more accurate classification of this poorly defined subtype. Genes included within prognostic mRNA panels have significantly higher than average mRNA-protein correlations, and gene copy number alterations are dampened at the protein-level; underscoring the value of proteome quantification for prognostication and phenotypic classification. Furthermore, protein products mapping to non-coding genomic regions are identified; highlighting a potential new class of tumor-specific immunotherapeutic targets.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Protein Interaction Maps , Proteome/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/immunology , DNA Copy Number Variations , Datasets as Topic , Female , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Proteogenomics/methods , Proteome/genetics , Proteome/immunology , RNA, Messenger/metabolism
5.
Mol Cell ; 73(1): 166-182.e7, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30609389

ABSTRACT

Subcellular localization is a main determinant of protein function; however, a global view of cellular proteome organization remains relatively unexplored. We have developed a robust mass spectrometry-based analysis pipeline to generate a proteome-wide view of subcellular localization for proteins mapping to 12,418 individual genes across five cell lines. Based on more than 83,000 unique classifications and correlation profiling, we investigate the effect of alternative splicing and protein domains on localization, complex member co-localization, cell-type-specific localization, as well as protein relocalization after growth factor inhibition. Our analysis provides information about the cellular architecture and complexity of the spatial organization of the proteome; we show that the majority of proteins have a single main subcellular location, that alternative splicing rarely affects subcellular location, and that cell types are best distinguished by expression of proteins exposed to the surrounding environment. The resource is freely accessible via www.subcellbarcode.org.


Subject(s)
Chromatography, Liquid , Mass Spectrometry , Proteins/metabolism , Proteome , Proteomics/methods , Subcellular Fractions/metabolism , Biomarkers/metabolism , Cell Fractionation , Computational Biology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gefitinib/pharmacology , Humans , Isoelectric Focusing , MCF-7 Cells , Protein Kinase Inhibitors/pharmacology , Protein Transport , Proteins/antagonists & inhibitors , Proteins/classification , Proteins/genetics , Reproducibility of Results , Subcellular Fractions/classification , Subcellular Fractions/drug effects
6.
Cell Rep ; 20(9): 2238-2250, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28854371

ABSTRACT

Human immune systems are variable, and immune responses are often unpredictable. Systems-level analyses offer increased power to sort patients on the basis of coordinated changes across immune cells and proteins. Allogeneic stem cell transplantation is a well-established form of immunotherapy whereby a donor immune system induces a graft-versus-leukemia response. This fails when the donor immune system regenerates improperly, leaving the patient susceptible to infections and leukemia relapse. We present a systems-level analysis by mass cytometry and serum profiling in 26 patients sampled 1, 2, 3, 6, and 12 months after transplantation. Using a combination of machine learning and topological data analyses, we show that global immune signatures associated with clinical outcome can be revealed, even when patients are few and heterogeneous. This high-resolution systems immune monitoring approach holds the potential for improving the development and evaluation of immunotherapies in the future.


Subject(s)
Flow Cytometry , Hematopoietic Stem Cell Transplantation/adverse effects , Leukemia/immunology , Leukemia/therapy , Statistics as Topic , Acute Disease , Blood Proteins/metabolism , Bone Marrow Transplantation , Cytomegalovirus/physiology , Graft vs Host Disease/immunology , Humans , Leukemia/blood , Lymphocytes/metabolism , Transplantation, Homologous , Treatment Outcome
7.
Acta Orthop ; 88(1): 35-40, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27892748

ABSTRACT

Background and purpose - Porous tantalum cups have been introduced as an alternative to various reinforcement rings in revision hip surgery. We hypothesized that porous tantalum cups would be superior to Müller acetabular roof reinforcement rings (MARRs) in revision hip surgery with re-revision for aseptic loosening as the primary outcome measure. Patients and methods - 207 hips operated with either a porous tantalum cup (TM cup, n = 111) or a MARR (n = 96) at index procedure were identified in our local arthroplasty register. Acetabular defects were classified according to Paprosky. There were 96 men and 111 women with a median age of 71 (35-95) years, presenting acetabular defect size type I in 39 cases, IIA in 22, IIB in 27, IIC in 43, IIIA in 32, and IIIB in 37 cases. Analysis of medical records identified all patients with subsequent re-revision and reasons for re-revisions. Kaplan-Meier survival functions were used to estimate implant survival. Results - With re-revision for aseptic loosening as the endpoint, the 6-year unadjusted cumulative survival was 97% (95% CI: 94-100) for TM cups and 96% (CI: 92-100) for MARR (p = 0.6). Using re-revision for any reason as the endpoint, 6-year survival was 87% (CI: 81-94) for TM cups and 95% (CI: 90-99) for MARR (p = 0.06). The main reason for re-revision in the TM group was dislocation (n = 10), followed by loosening (n = 3), whereas the main reason for re-revision in the MARR group was aseptic loosening (n = 8). Duration of the index procedure and perioperative blood loss were lower in the TM group. Interpretation - Both TM and MARR lead to good 6-year results in acetabular revision surgery. The methods differ in their respective failure mechanisms. We conclude that TM cups are a valuable treatment option in acetabular revision surgery, but the reasons underlying dislocations after the use of TM cups must be analyzed further.


Subject(s)
Acetabulum/surgery , Arthroplasty, Replacement, Hip/instrumentation , Hip Prosthesis , Registries , Tantalum , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Male , Middle Aged , Porosity , Postoperative Complications , Prosthesis Design , Reoperation , Retrospective Studies , Treatment Outcome
8.
Proc Natl Acad Sci U S A ; 113(38): E5618-27, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27608497

ABSTRACT

Vascular pericytes, an important cellular component in the tumor microenvironment, are often associated with tumor vasculatures, and their functions in cancer invasion and metastasis are poorly understood. Here we show that PDGF-BB induces pericyte-fibroblast transition (PFT), which significantly contributes to tumor invasion and metastasis. Gain- and loss-of-function experiments demonstrate that PDGF-BB-PDGFRß signaling promotes PFT both in vitro and in in vivo tumors. Genome-wide expression analysis indicates that PDGF-BB-activated pericytes acquire mesenchymal progenitor features. Pharmacological inhibition and genetic deletion of PDGFRß ablate the PDGF-BB-induced PFT. Genetic tracing of pericytes with two independent mouse strains, TN-AP-CreERT2:R26R-tdTomato and NG2-CreERT2:R26R-tdTomato, shows that PFT cells gain stromal fibroblast and myofibroblast markers in tumors. Importantly, coimplantation of PFT cells with less-invasive tumor cells in mice markedly promotes tumor dissemination and invasion, leading to an increased number of circulating tumor cells and metastasis. Our findings reveal a mechanism of vascular pericytes in PDGF-BB-promoted cancer invasion and metastasis by inducing PFT, and thus targeting PFT may offer a new treatment option of cancer metastasis.


Subject(s)
Carcinoma, Renal Cell/genetics , Pericytes/metabolism , Proto-Oncogene Proteins c-sis/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Animals , Becaplermin , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Mice, Knockout , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Pericytes/pathology , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
10.
Nat Commun ; 7: 12321, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27492255

ABSTRACT

A role for Hedgehog (Hh) signalling in the development of colorectal cancer (CRC) has been proposed. In CRC and other solid tumours, Hh ligands are upregulated; however, a specific Hh antagonist provided no benefit in a clinical trial. Here we use Hh reporter mice to show that downstream Hh activity is unexpectedly diminished in a mouse model of colitis-associated colon cancer, and that downstream Hh signalling is restricted to the stroma. Functionally, stroma-specific Hh activation in mice markedly reduces the tumour load and blocks progression of advanced neoplasms, partly via the modulation of BMP signalling and restriction of the colonic stem cell signature. By contrast, attenuated Hh signalling accelerates colonic tumourigenesis. In human CRC, downstream Hh activity is similarly reduced and canonical Hh signalling remains predominantly paracrine. Our results suggest that diminished downstream Hh signalling enhances CRC development, and that stromal Hh activation can act as a colonic tumour suppressor.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Signal Transduction , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Azoxymethane , Bone Morphogenetic Proteins/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Integrases/metabolism , Mice, Inbred C57BL , Recombination, Genetic/genetics , Stromal Cells/metabolism , Stromal Cells/pathology , Transcription, Genetic , Tumor Burden
11.
Mol Cell ; 59(6): 984-97, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26321255

ABSTRACT

Transcriptionally active and inactive chromatin domains tend to segregate into separate sub-nuclear compartments to maintain stable expression patterns. However, here we uncovered an inter-chromosomal network connecting active loci enriched in circadian genes to repressed lamina-associated domains (LADs). The interactome is regulated by PARP1 and its co-factor CTCF. They not only mediate chromatin fiber interactions but also promote the recruitment of circadian genes to the lamina. Synchronization of the circadian rhythm by serum shock induces oscillations in PARP1-CTCF interactions, which is accompanied by oscillating recruitment of circadian loci to the lamina, followed by the acquisition of repressive H3K9me2 marks and transcriptional attenuation. Furthermore, depletion of H3K9me2/3, inhibition of PARP activity by olaparib, or downregulation of PARP1 or CTCF expression counteracts both recruitment to the envelope and circadian transcription. PARP1- and CTCF-regulated contacts between circadian loci and the repressive chromatin environment at the lamina therefore mediate circadian transcriptional plasticity.


Subject(s)
Chromatin/genetics , Human Embryonic Stem Cells/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Adaptor Proteins, Signal Transducing , CCCTC-Binding Factor , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin Immunoprecipitation , Circadian Rhythm , Embryoid Bodies/enzymology , Epistasis, Genetic , Gene Expression Regulation , Gene Regulatory Networks , HCT116 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Lamina/metabolism , Poly (ADP-Ribose) Polymerase-1 , Protein Binding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
12.
BMC Cancer ; 15: 311, 2015 Apr 25.
Article in English | MEDLINE | ID: mdl-25907971

ABSTRACT

BACKGROUND: Deregulated Notch signaling is linked to a variety of tumors and it is therefore important to learn more about the frequency and distribution of Notch mutations in a tumor context. METHODS: In this report, we use data from the recently developed Cancer Cell Line Encyclopedia to assess the frequency and distribution of Notch mutations in a large panel of cancer cell lines in silico. RESULTS: Our results show that the mutation frequency of Notch receptor and ligand genes is at par with that for established oncogenes and higher than for a set of house-keeping genes. Mutations were found across all four Notch receptor genes, but with notable differences between protein domains, mutations were for example more prevalent in the regions encoding the LNR and PEST domains in the Notch intracellular domain. Furthermore, an in silico estimation of functional impact showed that deleterious mutations cluster to the ligand-binding and the intracellular domains of NOTCH1. For most cell line groups, the mutation frequency of Notch genes is higher than in associated primary tumors. CONCLUSIONS: Our results shed new light on the spectrum of Notch mutations after in vitro culturing of tumor cells. The higher mutation frequency in tumor cell lines indicates that Notch mutations are associated with a growth advantage in vitro, and thus may be considered to be driver mutations in a tumor cell line context.


Subject(s)
Gene Frequency , Receptors, Notch/genetics , Cell Line, Tumor , DNA Copy Number Variations , DNA Mutational Analysis , Genes, Essential , Humans , Mutation, Missense
13.
Oncotarget ; 6(14): 12297-309, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25909218

ABSTRACT

Melanoma is currently divided on a genetic level according to mutational status. However, this classification does not optimally predict prognosis. In prior studies, we have defined gene expression phenotypes (high-immune, pigmentation, proliferative and normal-like), which are predictive of survival outcome as well as informative of biology. Herein, we employed a population-based metastatic melanoma cohort and external cohorts to determine the prognostic and predictive significance of the gene expression phenotypes. We performed expression profiling on 214 cutaneous melanoma tumors and found an increased risk of developing distant metastases in the pigmentation (HR, 1.9; 95% CI, 1.05-3.28; P=0.03) and proliferative (HR, 2.8; 95% CI, 1.43-5.57; P=0.003) groups as compared to the high-immune response group. Further genetic characterization of melanomas using targeted deep-sequencing revealed similar mutational patterns across these phenotypes. We also used publicly available expression profiling data from melanoma patients treated with targeted or vaccine therapy in order to determine if our signatures predicted therapeutic response. In patients receiving targeted therapy, melanomas resistant to targeted therapy were enriched in the MITF-low proliferative subtype as compared to pre-treatment biopsies (P=0.02). In summary, the melanoma gene expression phenotypes are highly predictive of survival outcome and can further help to discriminate patients responding to targeted therapy.


Subject(s)
Melanoma/genetics , Skin Neoplasms/genetics , Transcriptome , Aged , Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Cohort Studies , DNA Mutational Analysis , Female , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Molecular Targeted Therapy/methods , Phenotype , Prognosis , Proportional Hazards Models , Skin Neoplasms/mortality , Skin Neoplasms/pathology
14.
Nature ; 517(7535): 489-92, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25363767

ABSTRACT

Next-generation sequencing of human tumours has refined our understanding of the mutational processes operative in cancer initiation and progression, yet major questions remain regarding the factors that induce driver mutations and the processes that shape mutation selection during tumorigenesis. Here we performed whole-exome sequencing on adenomas from three mouse models of non-small-cell lung cancer, which were induced either by exposure to carcinogens (methyl-nitrosourea (MNU) and urethane) or by genetic activation of Kras (Kras(LA2)). Although the MNU-induced tumours carried exactly the same initiating mutation in Kras as seen in the Kras(LA2) model (G12D), MNU tumours had an average of 192 non-synonymous, somatic single-nucleotide variants, compared with only six in tumours from the Kras(LA2) model. By contrast, the Kras(LA2) tumours exhibited a significantly higher level of aneuploidy and copy number alterations compared with the carcinogen-induced tumours, suggesting that carcinogen-induced and genetically engineered models lead to tumour development through different routes. The wild-type allele of Kras has been shown to act as a tumour suppressor in mouse models of non-small-cell lung cancer. We demonstrate that urethane-induced tumours from wild-type mice carry mostly (94%) Kras Q61R mutations, whereas those from Kras heterozygous animals carry mostly (92%) Kras Q61L mutations, indicating a major role for germline Kras status in mutation selection during initiation. The exome-wide mutation spectra in carcinogen-induced tumours overwhelmingly display signatures of the initiating carcinogen, while adenocarcinomas acquire additional C > T mutations at CpG sites. These data provide a basis for understanding results from human tumour genome sequencing, which has identified two broad categories of tumours based on the relative frequency of single-nucleotide variations and copy number alterations, and underline the importance of carcinogen models for understanding the complex mutation spectra seen in human cancers.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Genes, ras/genetics , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Mutation/genetics , Oncogene Protein p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma/chemically induced , Adenocarcinoma/genetics , Animals , Carcinogens/toxicity , Carcinoma, Non-Small-Cell Lung/chemically induced , Carcinoma, Non-Small-Cell Lung/genetics , DNA Copy Number Variations/genetics , Disease Progression , Female , Genomic Instability/genetics , Germ-Line Mutation/genetics , Humans , Male , Methylnitrosourea/toxicity , Mice , Models, Genetic , Point Mutation/genetics , Urethane/toxicity
15.
EMBO Mol Med ; 5(7): 1067-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23776131

ABSTRACT

SCF (Skp1/Cul1/F-box) ubiquitin ligases act as master regulators of cellular homeostasis by targeting key proteins for ubiquitylation. Here, we identified a hitherto uncharacterized F-box protein, FBXO28 that controls MYC-dependent transcription by non-proteolytic ubiquitylation. SCF(FBXO28) activity and stability are regulated during the cell cycle by CDK1/2-mediated phosphorylation of FBXO28, which is required for its efficient ubiquitylation of MYC and downsteam enhancement of the MYC pathway. Depletion of FBXO28 or overexpression of an F-box mutant unable to support MYC ubiquitylation results in an impairment of MYC-driven transcription, transformation and tumourigenesis. Finally, in human breast cancer, high FBXO28 expression and phosphorylation are strong and independent predictors of poor outcome. In conclusion, our data suggest that SCF(FBXO28) plays an important role in transmitting CDK activity to MYC function during the cell cycle, emphasizing the CDK-FBXO28-MYC axis as a potential molecular drug target in MYC-driven cancers, including breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast/pathology , CDC2 Protein Kinase/metabolism , Cyclin-Dependent Kinase 2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Amino Acid Sequence , Breast/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Phosphorylation , Prognosis , Promoter Regions, Genetic , Proteolysis , SKP Cullin F-Box Protein Ligases/analysis , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction , Survival Analysis , Transcriptional Activation , Ubiquitination
16.
Proc Natl Acad Sci U S A ; 110(18): 7312-7, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23589885

ABSTRACT

Hypoxia, or low oxygen tension, is a major regulator of tumor development and aggressiveness. However, how cancer cells adapt to hypoxia and communicate with their surrounding microenvironment during tumor development remain important questions. Here, we show that secreted vesicles with exosome characteristics mediate hypoxia-dependent intercellular signaling of the highly malignant brain tumor glioblastoma multiforme (GBM). In vitro hypoxia experiments with glioma cells and studies with patient materials reveal the enrichment in exosomes of hypoxia-regulated mRNAs and proteins (e.g., matrix metalloproteinases, IL-8, PDGFs, caveolin 1, and lysyl oxidase), several of which were associated with poor glioma patient prognosis. We show that exosomes derived from GBM cells grown at hypoxic compared with normoxic conditions are potent inducers of angiogenesis ex vivo and in vitro through phenotypic modulation of endothelial cells. Interestingly, endothelial cells were programmed by GBM cell-derived hypoxic exosomes to secrete several potent growth factors and cytokines and to stimulate pericyte PI3K/AKT signaling activation and migration. Moreover, exosomes derived from hypoxic compared with normoxic conditions showed increased autocrine, promigratory activation of GBM cells. These findings were correlated with significantly enhanced induction by hypoxic compared with normoxic exosomes of tumor vascularization, pericyte vessel coverage, GBM cell proliferation, as well as decreased tumor hypoxia in a mouse xenograft model. We conclude that the proteome and mRNA profiles of exosome vesicles closely reflect the oxygenation status of donor glioma cells and patient tumors, and that the exosomal pathway constitutes a potentially targetable driver of hypoxia-dependent intercellular signaling during tumor development.


Subject(s)
Blood Vessels/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Exosomes/metabolism , Glioma/blood supply , Glioma/pathology , Animals , Autocrine Communication , Brain Neoplasms/genetics , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , Mice , Mice, SCID , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Paracrine Communication , Pericytes/metabolism , Pericytes/pathology , Proteome/metabolism , Signal Transduction/genetics , Tissue Donors , Transcriptome/genetics , Xenograft Model Antitumor Assays
17.
Clin Cancer Res ; 19(6): 1353-62, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23339125

ABSTRACT

PURPOSE: Ionizing radiation is a well-established carcinogen in rodent models and a risk factor associated with human cancer. We developed a mouse model that captures radiation effects on host biology by transplanting unirradiated Trp53-null mammary tissue to sham or irradiated hosts. Gene expression profiles of tumors that arose in irradiated mice are distinct from those that arose in naïve hosts. We asked whether expression metaprofiles could discern radiation-preceded human cancer or be informative in sporadic breast cancers. EXPERIMENTAL DESIGN: Affymetrix microarray gene expression data from 56 Trp53-null mammary tumors were used to define gene profiles and a centroid that discriminates tumors arising in irradiated hosts. These were applied to publicly available human cancer datasets. RESULTS: Host irradiation induces a metaprofile consisting of gene modules representing stem cells, cell motility, macrophages, and autophagy. Human orthologs of the host irradiation metaprofile discriminated between radiation-preceded and sporadic human thyroid cancers. An irradiated host centroid was strongly associated with estrogen receptor-negative breast cancer. When applied to sporadic human breast cancers, the irradiated host metaprofile strongly associated with basal-like and claudin-low breast cancer intrinsic subtypes. Comparing host irradiation in the context of TGF-ß levels showed that inflammation was robustly associated with claudin-low tumors. CONCLUSIONS: Detection of radiation-preceded human cancer by the irradiated host metaprofile raises possibilities of assessing human cancer etiology. Moreover, the association of the irradiated host metaprofiles with estrogen receptor-negative status and claudin-low subtype suggests that host processes similar to those induced by radiation underlie sporadic cancers.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Mammary Neoplasms, Animal/genetics , Tumor Suppressor Protein p53/genetics , Animals , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Female , Humans , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/pathology , Mice , Radiation, Ionizing , Transcriptome/radiation effects , Tumor Microenvironment/genetics , Tumor Microenvironment/radiation effects , Tumor Suppressor Protein p53/metabolism
18.
Breast Cancer Res ; 14(4): R113, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22839103

ABSTRACT

INTRODUCTION: Gene expression data derived from clinical cancer specimens provide an opportunity to characterize cancer-specific transcriptional programs. Here, we present an analysis delineating a correlation-based gene expression landscape of breast cancer that identifies modules with strong associations to breast cancer-specific and general tumor biology. METHODS: Modules of highly connected genes were extracted from a gene co-expression network that was constructed based on Pearson correlation, and module activities were then calculated using a pathway activity score. Functional annotations of modules were experimentally validated with an siRNA cell spot microarray system using the KPL-4 breast cancer cell line, and by using gene expression data from functional studies. Modules were derived using gene expression data representing 1,608 breast cancer samples and validated in data sets representing 971 independent breast cancer samples as well as 1,231 samples from other cancer forms. RESULTS: The initial co-expression network analysis resulted in the characterization of eight tightly regulated gene modules. Cell cycle genes were divided into two transcriptional programs, and experimental validation using an siRNA screen showed different functional roles for these programs during proliferation. The division of the two programs was found to act as a marker for tumor protein p53 (TP53) gene status in luminal breast cancer, with the two programs being separated only in luminal tumors with functional p53 (encoded by TP53). Moreover, a module containing fibroblast and stroma-related genes was highly expressed in fibroblasts, but was also up-regulated by overexpression of epithelial-mesenchymal transition factors such as transforming growth factor beta 1 (TGF-beta1) and Snail in immortalized human mammary epithelial cells. Strikingly, the stroma transcriptional program related to less malignant tumors for luminal disease and aggressive lymph node positive disease among basal-like tumors. CONCLUSIONS: We have derived a robust gene expression landscape of breast cancer that reflects known subtypes as well as heterogeneity within these subtypes. By applying the modules to TP53-mutated samples we shed light on the biological consequences of non-functional p53 in otherwise low-proliferating luminal breast cancer. Furthermore, as in the case of the stroma module, we show that the biological and clinical interpretation of a set of co-regulated genes is subtype-dependent.


Subject(s)
Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p53/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/genetics , Cluster Analysis , Datasets as Topic , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Stromal Cells/metabolism , Transcriptome
19.
PLoS One ; 6(8): e23057, 2011.
Article in English | MEDLINE | ID: mdl-21826227

ABSTRACT

BACKGROUND: Despite recent progress, therapy for metastatic clear cell renal cell carcinoma (CCRCC) is still inadequate. Dysregulated Notch signaling in CCRCC contributes to tumor growth, but the full spectrum of downstream processes regulated by Notch in this tumor form is unknown. METHODOLOGY/PRINCIPAL FINDINGS: We show that inhibition of endogenous Notch signaling modulates TGF-ß dependent gene regulation in CCRCC cells. Analysis of gene expression data representing 176 CCRCCs showed that elevated TGF-ß pathway activity correlated significantly with shortened disease specific survival (log-rank test, p = 0.006) and patients with metastatic disease showed a significantly elevated TGF-ß signaling activity (two-sided Student's t-test, p = 0.044). Inhibition of Notch signaling led to attenuation of both basal and TGF-ß1 induced TGF-ß signaling in CCRCC cells, including an extensive set of genes known to be involved in migration and invasion. Functional analyses revealed that Notch inhibition decreased the migratory and invasive capacity of CCRCC cells. CONCLUSION: An extensive cross-talk between the Notch and TGF-ß signaling cascades is present in CCRCC and the functional properties of these two pathways are associated with the aggressiveness of this disease.


Subject(s)
Carcinoma, Renal Cell/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Benzamides/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Dioxoles/pharmacology , Dipeptides/pharmacology , Humans , Receptor, Notch1/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1/pharmacology
20.
Nucleic Acids Res ; 39(20): e136, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21835775

ABSTRACT

While a growing body of evidence implicates regulatory miRNA modules in various aspects of human disease and development, insights into specific miRNA function remain limited. Here, we present an innovative approach to elucidate tissue-specific miRNA functions that goes beyond miRNA target prediction and expression correlation. This approach is based on a multi-level integration of corresponding miRNA and mRNA gene expression levels, miRNA target prediction, transcription factor target prediction and mechanistic models of gene network regulation. Predicted miRNA functions were either validated experimentally or compared to published data. The predicted miRNA functions are accessible in the miRNA bodymap, an interactive online compendium and mining tool of high-dimensional newly generated and published miRNA expression profiles. The miRNA bodymap enables prioritization of candidate miRNAs based on their expression pattern or functional annotation across tissue or disease subgroup. The miRNA bodymap project provides users with a single one-stop data-mining solution and has great potential to become a community resource.


Subject(s)
MicroRNAs/metabolism , Software , Animals , Cell Line, Tumor , Data Mining , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Genomics , Humans , Mice , Models, Genetic , Molecular Sequence Annotation , RNA, Messenger/metabolism , Rats , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...