Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Pathol ; 258(3): 289-299, 2022 11.
Article in English | MEDLINE | ID: mdl-36106661

ABSTRACT

R-spondins (RSPOs) are influential signaling molecules that promote the Wnt/ß-catenin pathway and self-renewal of stem cells. Currently, RSPOs are emerging as clinically relevant oncogenes, being linked to cancer development in multiple organs. Although this has instigated the rapid development and testing of therapeutic antibodies targeting RSPOs, functional evidence that RSPO causally drives cancer has focused primarily on the intestinal tract. Here, we assess the oncogenic capacity of RSPO in breast cancer in a direct fashion by generating and characterizing a novel mouse model with conditional Rspo3 expression in the mammary gland. We also address the prevalence of RSPO gene alterations in breast cancer patients. We found that a quarter of breast cancer patients harbor RSPO2/RSPO3 copy number amplifications, which are associated with lack of steroid hormone receptor expression and reduced patient survival. Foremost, we demonstrate the causal oncogenic capacity of RSPO3 in the breast, as conditional Rspo3 overexpression consistently drives the development of mammary adenocarcinomas in our novel Rspo3 breast cancer model. RSPO3-driven mammary tumors typically show poor differentiation, areas of epithelial-to-mesenchymal transition, and metastatic potential. Given the reported interplay in the Wnt/ß-catenin pathway, we comparatively analyzed RSPO3-driven mouse mammary tumors versus classical WNT1-driven analogues. This revealed that RSPO3-driven tumors are distinct, as the poorly differentiated tumor morphology and metastatic potential were observed in RSPO3-driven tumorigenesis exclusively, further substantiated by differentiating gene expression profiles. Co-expression of Rspo3 and Wnt1 transduced mammary tumors with a mixed phenotype harboring morphological features characteristic of both transgenes. In summary, we report that a quarter of breast cancer patients harbor RSPO2/RSPO3 copy number gains, and these patients have a worse prognosis, whilst providing in vivo evidence that RSPO3 drives poorly differentiated invasive breast cancer in mice. Herewith, we establish RSPO3 as a driver of breast cancer with clinical relevance, proposing RSPO3 as a novel candidate target for therapy in breast cancer. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Mammary Neoplasms, Animal , beta Catenin , Animals , Carcinogenesis/genetics , Hormones , Mice , Oncogenes , Steroids , Thrombospondins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
2.
Gut ; 66(6): 1095-1105, 2017 06.
Article in English | MEDLINE | ID: mdl-27511199

ABSTRACT

OBJECTIVE: The gross majority of colorectal cancer cases results from aberrant Wnt/ß-catenin signalling through adenomatous polyposis coli (APC) or CTNNB1 mutations. However, a subset of human colon tumours harbour, mutually exclusive with APC and CTNNB1 mutations, gene fusions in RSPO2 or RSPO3, leading to enhanced expression of these R-spondin genes. This suggested that RSPO activation can substitute for the most common mutations as an alternative driver for intestinal cancer. Involvement of RSPO3 in tumour growth was recently shown in RSPO3-fusion-positive xenograft models. The current study determines the extent into which solely a gain in RSPO3 actually functions as a driver of intestinal cancer in a direct, causal fashion, and addresses the in vivo activities of RSPO3 in parallel. DESIGN: We generated a conditional Rspo3 transgenic mouse model in which the Rspo3 transgene is expressed upon Cre activity. Cre is provided by cross-breeding with Lgr5-GFP-CreERT2 mice. RESULTS: Upon in vivo Rspo3 expression, mice rapidly developed extensive hyperplastic, adenomatous and adenocarcinomatous lesions throughout the intestine. RSPO3 induced the expansion of Lgr5+ stem cells, Paneth cells, non-Paneth cell label-retaining cells and Lgr4+ cells, thus promoting both intestinal stem cell and niche compartments. Wnt/ß-catenin signalling was modestly increased upon Rspo3 expression and mutant Kras synergised with Rspo3 in hyperplastic growth. CONCLUSIONS: We provide in vivo evidence that RSPO3 stimulates the crypt stem cell and niche compartments and drives rapid intestinal tumorigenesis. This establishes RSPO3 as a potent driver of intestinal cancer and proposes RSPO3 as a candidate target for therapy in patients with colorectal cancer harbouring RSPO3 fusions.


Subject(s)
Adenocarcinoma/genetics , Adenoma/genetics , Carcinogenesis/genetics , Intestinal Neoplasms/genetics , Intestines/pathology , Paneth Cells/pathology , Stem Cells/pathology , Thrombospondins/genetics , Thrombospondins/metabolism , Adenocarcinoma/pathology , Adenoma/pathology , Animals , Cell Enlargement , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression , Hyperplasia/genetics , Hyperplasia/pathology , Intestinal Mucosa/metabolism , Intestinal Neoplasms/pathology , Mice , Mice, Transgenic , Mutation , Organoids/pathology , Paneth Cells/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/analysis , Stem Cells/chemistry , Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
3.
Nat Genet ; 39(6): 759-69, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17468756

ABSTRACT

We performed a high-throughput retroviral insertional mutagenesis screen in mouse mammary tumor virus (MMTV)-induced mammary tumors and identified 33 common insertion sites, of which 17 genes were previously not known to be associated with mammary cancer and 13 had not previously been linked to cancer in general. Although members of the Wnt and fibroblast growth factors (Fgf) families were frequently tagged, our exhaustive screening for MMTV insertion sites uncovered a new repertoire of candidate breast cancer oncogenes. We validated one of these genes, Rspo3, as an oncogene by overexpression in a p53-deficient mammary epithelial cell line. The human orthologs of the candidate oncogenes were frequently deregulated in human breast cancers and associated with several tumor parameters. Computational analysis of all MMTV-tagged genes uncovered specific gene families not previously associated with cancer and showed a significant overrepresentation of protein domains and signaling pathways mainly associated with development and growth factor signaling. Comparison of all tagged genes in MMTV and Moloney murine leukemia virus-induced malignancies showed that both viruses target mostly different genes that act predominantly in distinct pathways.


Subject(s)
Genes, Neoplasm/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse/genetics , Multigene Family/genetics , Mutagenesis, Insertional/genetics , Signal Transduction , Animals , Cell Transformation, Neoplastic , Epithelium/metabolism , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Oncogenic Viruses/genetics , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Virus Integration
4.
Oncogene ; 37(12): 1594-1609, 2018 03.
Article in English | MEDLINE | ID: mdl-29326437

ABSTRACT

Personalized medicine for cancer patients requires a deep understanding of the underlying genetics that drive cancer and the subsequent identification of predictive biomarkers. To discover new genes and pathways contributing to oncogenesis and therapy resistance in HER2+ breast cancer, we performed Mouse Mammary Tumor Virus (MMTV)-induced insertional mutagenesis screens in ErbB2/cNeu-transgenic mouse models. The screens revealed 34 common integration sites (CIS) in mammary tumors of MMTV-infected mice, highlighting loci with multiple independent MMTV integrations in which potential oncogenes are activated, most of which had never been reported as MMTV CIS. The CIS most strongly associated with the ErbB2-transgenic genotype was the locus containing Eras (ES cell-expressed Ras), a constitutively active RAS-family GTPase. We show that upon expression, Eras acts as a potent oncogenic driver through hyperactivation of the PI3K/AKT pathway, in contrast to other RAS proteins that signal primarily via the MAPK/ERK pathway and require upstream activation or activating mutations to induce signaling. We additionally show that ERAS synergistically enhances HER2-induced tumorigenesis and, in this role, can functionally replace ERBB3/HER3 by acting as a more powerful activator of PI3K/AKT signaling. Although previously reported as pseudogene in humans, we observed ERAS RNA and protein expression in a substantial subset of human primary breast carcinomas. Importantly, we show that ERAS induces primary resistance to the widely used HER2-targeting drugs Trastuzumab (Herceptin) and Lapatinib (Tykerb/Tyverb) in vivo, and is involved in acquired resistance via selective upregulation during treatment in vitro, indicating that ERAS may serve as a novel clinical biomarker for PI3K/AKT pathway hyperactivation and HER2-targeted therapy resistance.


Subject(s)
Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , Mammary Neoplasms, Experimental/pathology , Mutagenesis, Insertional/physiology , Oncogene Protein p21(ras)/physiology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cells, Cultured , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Oncogene Protein p21(ras)/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
5.
Nat Commun ; 7: 13567, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27876799

ABSTRACT

In search of oncogenic drivers and mechanisms affecting therapy resistance in breast cancer, we identified Irs4, a poorly studied member of the insulin receptor substrate (IRS) family, as a mammary oncogene by insertional mutagenesis. Whereas normally silent in the postnatal mammary gland, IRS4 is found to be highly expressed in a subset of breast cancers. We show that Irs4 expression in mammary epithelial cells induces constitutive PI3K/AKT pathway hyperactivation, insulin/IGF1-independent cell proliferation, anchorage-independent growth and in vivo tumorigenesis. The constitutive PI3K/AKT pathway hyperactivation by IRS4 is unique to the IRS family and we identify the lack of a SHP2-binding domain in IRS4 as the molecular basis of this feature. Finally, we show that IRS4 and ERBB2/HER2 synergistically induce tumorigenesis and that IRS4-expression confers resistance to HER2-targeted therapy. Taken together, our findings present the cellular and molecular mechanisms of IRS4-induced tumorigenesis and establish IRS4 as an oncogenic driver and biomarker for therapy resistance in breast cancer.


Subject(s)
Insulin Receptor Substrate Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation , Cells, Cultured , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Insulin Receptor Substrate Proteins/genetics , Mammary Tumor Virus, Mouse/physiology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Receptor, ErbB-2/genetics
6.
Oncogene ; 23(36): 6047-55, 2004 Aug 12.
Article in English | MEDLINE | ID: mdl-15208658

ABSTRACT

Mouse mammary tumor virus (MMTV) infection causes a high incidence of murine mammary carcinomas by insertion of its proviral DNA in the genome of mammary epithelial cells. Retroviral insertion can activate flanking proto-oncogenes by a process called insertional mutagenesis. By sequencing the DNA adjacent to MMTV proviral insertions in mammary tumors from BALB/c mice infected with C3H-MMTV, we have found a common MMTV insertion site in the Fgf10 locus. RT-PCR studies showed that Fgf10 is expressed only in those tumors harboring a MMTV proviral insertion in this locus, suggesting that Fgf10 is a proto-oncogene. The oncogenicity of Fgf10 was evaluated in vivo by subcutaneous transplantation of retrovirally transduced HC11 mammary epithelial cells into BALB/c mice. Highly vascularized invasive subcutaneous tumors developed indicating that Fgf10 can act as an oncogene. A survey of primary human breast carcinomas revealed strongly elevated Fgf10 mRNA levels in approximately 10% of the tumors tested, suggesting that Fgf10 may also be involved in oncogenicity of a subset of human breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Fibroblast Growth Factors/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/genetics , Mutagenesis, Insertional , Oncogenes , Animals , Breast Neoplasms/genetics , Carcinoma/genetics , Cell Division , Cell Line, Tumor , Female , Fibroblast Growth Factor 10 , Fibroblast Growth Factors/metabolism , Gene Expression , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Proto-Oncogene Mas , RNA, Messenger/metabolism , Tumor Cells, Cultured , Wnt Proteins
7.
PLoS One ; 8(5): e62113, 2013.
Article in English | MEDLINE | ID: mdl-23690930

ABSTRACT

Cancer develops through a multistep process in which normal cells progress to malignant tumors via the evolution of their genomes as a result of the acquisition of mutations in cancer driver genes. The number, identity and mode of action of cancer driver genes, and how they contribute to tumor evolution is largely unknown. This study deployed the Mouse Mammary Tumor Virus (MMTV) as an insertional mutagen to find both the driver genes and the networks in which they function. Using deep insertion site sequencing we identified around 31000 retroviral integration sites in 604 MMTV-induced mammary tumors from mice with mammary gland-specific deletion of Trp53, Pten heterozygous knockout mice, or wildtype strains. We identified 18 known common integration sites (CISs) and 12 previously unknown CISs marking new candidate cancer genes. Members of the Wnt, Fgf, Fgfr, Rspo and Pdgfr gene families were commonly mutated in a mutually exclusive fashion. The sequence data we generated yielded also information on the clonality of insertions in individual tumors, allowing us to develop a data-driven model of MMTV-induced tumor development. Insertional mutations near Wnt and Fgf genes mark the earliest "initiating" events in MMTV induced tumorigenesis, whereas Fgfr genes are targeted later during tumor progression. Our data shows that insertional mutagenesis can be used to discover the mutational networks, the timing of mutations, and the genes that initiate and drive tumor evolution.


Subject(s)
Gene Regulatory Networks/genetics , Genes, Neoplasm/genetics , High-Throughput Nucleotide Sequencing , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/physiology , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Disease Progression , Female , Genotype , Humans , Mammary Neoplasms, Experimental/virology , Mice , Mutagenesis, Insertional , PTEN Phosphohydrolase/genetics , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL