Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 152(1-2): 25-38, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23273993

ABSTRACT

Cell-type plasticity within a tumor has recently been suggested to cause a bidirectional conversion between tumor-initiating stem cells and nonstem cells triggered by an inflammatory stroma. NF-κB represents a key transcription factor within the inflammatory tumor microenvironment. However, NF-κB's function in tumor-initiating cells has not been examined yet. Using a genetic model of intestinal epithelial cell (IEC)-restricted constitutive Wnt-activation, which comprises the most common event in the initiation of colon cancer, we demonstrate that NF-κB modulates Wnt signaling and show that IEC-specific ablation of RelA/p65 retards crypt stem cell expansion. In contrast, elevated NF-κB signaling enhances Wnt activation and induces dedifferentiation of nonstem cells that acquire tumor-initiating capacity. Thus, our data support the concept of bidirectional conversion and highlight the importance of inflammatory signaling for dedifferentiation and generation of tumor-initiating cells in vivo.


Subject(s)
Cell Dedifferentiation , Cell Transformation, Neoplastic , Colonic Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Colon/pathology , Epithelial Cells/pathology , Female , Humans , Male , Mice , NF-kappa B/metabolism , Wnt Signaling Pathway
2.
Gastroenterology ; 159(1): 183-199, 2020 07.
Article in English | MEDLINE | ID: mdl-32179094

ABSTRACT

BACKGROUND & AIMS: Intestinal epithelial homeostasis depends on a tightly regulated balance between intestinal epithelial cell (IEC) death and proliferation. While the disruption of several IEC death regulating factors result in intestinal inflammation, the loss of the anti-apoptotic BCL2 family members BCL2 and BCL2L1 has no effect on intestinal homeostasis in mice. We investigated the functions of the antiapoptotic protein MCL1, another member of the BCL2 family, in intestinal homeostasis in mice. METHODS: We generated mice with IEC-specific disruption of Mcl1 (Mcl1ΔIEC mice) or tamoxifen-inducible IEC-specific disruption of Mcl1 (i-Mcl1ΔIEC mice); these mice and mice with full-length Mcl1 (controls) were raised under normal or germ-free conditions. Mice were analyzed by endoscopy and for intestinal epithelial barrier permeability. Intestinal tissues were analyzed by histology, in situ hybridization, proliferation assays, and immunoblots. Levels of calprotectin, a marker of intestinal inflammation, were measured in intestinal tissues and feces. RESULTS: Mcl1ΔIEC mice spontaneously developed apoptotic enterocolopathy, characterized by increased IEC apoptosis, hyperproliferative crypts, epithelial barrier dysfunction, and chronic inflammation. Loss of MCL1 retained intestinal crypts in a hyperproliferated state and prevented the differentiation of intestinal stem cells. Proliferation of intestinal stem cells in MCL1-deficient mice required WNT signaling and was associated with DNA damage accumulation. By 1 year of age, Mcl1ΔIEC mice developed intestinal tumors with morphologic and genetic features of human adenomas and carcinomas. Germ-free housing of Mcl1ΔIEC mice reduced markers of microbiota-induced intestinal inflammation but not tumor development. CONCLUSION: The antiapoptotic protein MCL1, a member of the BCL2 family, is required for maintenance of intestinal homeostasis and prevention of carcinogenesis in mice. Loss of MCL1 results in development of intestinal carcinomas, even under germ-free conditions, and therefore does not involve microbe-induced chronic inflammation. Mcl1ΔIEC mice might be used to study apoptotic enterocolopathy and inflammatory bowel diseases.


Subject(s)
Carcinoma/pathology , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinoma/diagnosis , Carcinoma/genetics , Disease Models, Animal , Endoscopy , Epithelial Cells/pathology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/diagnostic imaging , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/genetics , Mice , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/genetics
3.
Front Cell Dev Biol ; 11: 1272730, 2023.
Article in English | MEDLINE | ID: mdl-37886398

ABSTRACT

Cellular plasticity defines the capacity of cells to adopt distinct identities during development, tissue homeostasis and regeneration. Dynamic fluctuations between different states, within or across lineages, are regulated by changes in chromatin accessibility and in gene expression. When deregulated, cellular plasticity can contribute to cancer initiation and progression. Cancer cells are remarkably plastic which contributes to phenotypic and functional heterogeneity within tumours as well as resistance to targeted therapies. It is for these reasons that the scientific community has become increasingly interested in understanding the molecular mechanisms governing cancer cell plasticity. The purpose of this mini-review is to discuss different examples of cellular plasticity associated with metaplasia and epithelial-mesenchymal transition with a focus on therapy resistance.

4.
Nat Commun ; 14(1): 5211, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37626054

ABSTRACT

The molecular basis of disease progression from UV-induced precancerous actinic keratosis (AK) to malignant invasive cutaneous squamous cell carcinoma (cSCC) and potentially lethal metastatic disease remains unclear. DNA sequencing studies have revealed a massive mutational burden but have yet to illuminate mechanisms of disease progression. Here we perform RNAseq transcriptomic profiling of 110 patient samples representing normal sun-exposed skin, AK, primary and metastatic cSCC and reveal a disease continuum from a differentiated to a progenitor-like state. This is accompanied by the orchestrated suppression of master regulators of epidermal differentiation, dynamic modulation of the epidermal differentiation complex, remodelling of the immune landscape and an increase in the preponderance of tumour specific keratinocytes. Comparative systems analysis of human cSCC coupled with the generation of genetically engineered murine models reveal that combinatorial sequential inactivation of the tumour suppressor genes Tgfbr2, Trp53, and Notch1 coupled with activation of Ras signalling progressively drives cSCC progression along a differentiated to progenitor axis. Taken together we provide a comprehensive map of the cSCC disease continuum and reveal potentially actionable events that promote and accompany disease progression.


Subject(s)
Carcinoma, Squamous Cell , Keratosis, Actinic , Skin Neoplasms , Humans , Animals , Mice , Carcinoma, Squamous Cell/genetics , Skin Neoplasms/genetics , Cell Differentiation , Disease Progression , Gene Expression Profiling
5.
Gastroenterology ; 140(1): 297-309, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20951698

ABSTRACT

BACKGROUND & AIMS: The limited clinical response observed in many patients with colorectal cancer may be related to the presence of chemoresistant colorectal cancer stem cells (CRC-SCs). Bone morphogenetic protein 4 (BMP4) promotes the differentiation of normal colonic stem cells. We investigated whether BMP4 might be used to induce differentiation of CRC-SCs and for therapeutic purposes. METHODS: CRC-SCs were isolated from 25 tumor samples based on expression of CD133 or using a selection culture medium. BMP4 expression and activity on CRC-SCs were evaluated in vitro; progeny of the stem cells were evaluated by immunofluorescence, immunoblot, and flow cytometry analyses. The potential therapeutic effect of BMP4 was assessed in immunocompromised mice after injection of CRC-SCs that responded to chemotherapy (n = 4) or that did not (n = 2). RESULTS: CRC-SCs did not express BMP4 whereas differentiated cells did. Recombinant BMP4 promoted differentiation and apoptosis of CRC-SCs in 12 of 15 independent experiments; this effect did not depend on Small Mothers against decapentaplegic (Smad)4 expression level or microsatellite stability. BMP4 activated the canonical and noncanonical BMP signaling pathways, including phosphoInositide 3-kinase (PI3K) and PKB (protein kinase B)/AKT. Mutations in PI3K or loss of Phosphatase and Tensin homolog (PTEN) in Smad4-defective tumors made CRC-SCs unresponsive to BMP4. Administration of BMP4 to immunocompromised mice with tumors that arose from CRC-SCs increased the antitumor effects of 5-fluorouracil and oxaliplatin. CONCLUSIONS: BMP4 promotes terminal differentiation, apoptosis, and chemosensitization of CRC-SCs in tumors that do not have simultaneous mutations in Smad4 and constitutive activation of PI3K. BMP4 might be developed as a therapeutic agent against cancer stem cells in advanced colorectal tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Morphogenetic Protein 4/therapeutic use , Colorectal Neoplasms/drug therapy , Fluorouracil/therapeutic use , Neoplastic Stem Cells/drug effects , Organoplatinum Compounds/therapeutic use , AC133 Antigen , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation , Cells, Cultured , Colorectal Neoplasms/pathology , Female , Glycoproteins/metabolism , Humans , Male , Mice , Microsatellite Instability , Middle Aged , Mutation , Oxaliplatin , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Peptides/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Smad4 Protein/metabolism
6.
Nat Commun ; 13(1): 2791, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589755

ABSTRACT

Tumour cell plasticity is a major barrier to the efficacy of targeted cancer therapies but the mechanisms that mediate it are poorly understood. Here, we identify dysregulated RNA splicing as a key driver of tumour cell dedifferentiation in colorectal cancer (CRC). We find that Apc-deficient CRC cells have dysregulated RNA splicing machinery and exhibit global rewiring of RNA splicing. We show that the splicing factor SRSF1 controls the plasticity of tumour cells by controlling Kras splicing and is required for CRC invasion in a mouse model of carcinogenesis. SRSF1 expression maintains stemness in human CRC organoids and correlates with cancer stem cell marker expression in human tumours. Crucially, partial genetic downregulation of Srsf1 does not detrimentally affect normal tissue homeostasis, demonstrating that tumour cell plasticity can be differentially targeted. Thus, our findings link dysregulation of the RNA splicing machinery and control of tumour cell plasticity.


Subject(s)
Cell Plasticity , Colorectal Neoplasms , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Plasticity/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Mice , RNA Splicing/genetics , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism
7.
Nat Commun ; 13(1): 7551, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36477656

ABSTRACT

The pro-tumourigenic role of epithelial TGFß signalling in colorectal cancer (CRC) is controversial. Here, we identify a cohort of born to be bad early-stage (T1) colorectal tumours, with aggressive features and a propensity to disseminate early, that are characterised by high epithelial cell-intrinsic TGFß signalling. In the presence of concurrent Apc and Kras mutations, activation of epithelial TGFß signalling rampantly accelerates tumourigenesis and share transcriptional signatures with those of the born to be bad T1 human tumours and predicts recurrence in stage II CRC. Mechanistically, epithelial TGFß signalling induces a growth-promoting EGFR-signalling module that synergises with mutant APC and KRAS to drive MAPK signalling that re-sensitise tumour cells to MEK and/or EGFR inhibitors. Together, we identify epithelial TGFß signalling both as a determinant of early dissemination and a potential therapeutic vulnerability of CRC's with born to be bad traits.


Subject(s)
Apoptosis , Transforming Growth Factor beta , Humans , Apoptosis/genetics
8.
Cell Death Dis ; 12(10): 873, 2021 09 25.
Article in English | MEDLINE | ID: mdl-34564693

ABSTRACT

RAC1B is a tumour-related alternative splice isoform of the small GTPase RAC1, found overexpressed in a large number of tumour types. Building evidence suggests it promotes tumour progression but compelling in vivo evidence, demonstrating a role in driving tumour invasion, is currently lacking. In the present study, we have overexpressed RAC1B in a colorectal cancer mouse model with potential invasive properties. Interestingly, RAC1B overexpression did not trigger tumour invasion, rather it led to an acceleration of tumour initiation and reduced mouse survival. By modelling early stages of adenoma initiation we observed a reduced apoptotic rate in RAC1B overexpressing tumours, suggesting protection from apoptosis as a mediator of this phenotype. RAC1B overexpressing tumours displayed attenuated TGFß signalling and functional analysis in ex vivo organoid cultures demonstrated that RAC1B negatively modulates TGFß signalling and confers resistance to TGFß-driven cell death. This work defines a novel mechanism by which early adenoma cells can overcome the cytostatic and cytotoxic effects of TGFß signalling and characterises a new oncogenic function of RAC1B in vivo.


Subject(s)
Apoptosis , Carcinogenesis/metabolism , Carcinogenesis/pathology , Intestines/pathology , Transforming Growth Factor beta/metabolism , rac1 GTP-Binding Protein/metabolism , Adenoma/pathology , Adenomatous Polyposis Coli/metabolism , Animals , Carcinogenesis/genetics , Disease Models, Animal , Down-Regulation , Gene Expression Regulation, Neoplastic , Mice , Models, Biological , Signal Transduction , Survival Analysis , Tumor Suppressor Protein p53/metabolism
9.
Cell Mol Gastroenterol Hepatol ; 11(2): 465-489, 2021.
Article in English | MEDLINE | ID: mdl-32971322

ABSTRACT

BACKGROUND & AIMS: Aspirin reduces colorectal cancer (CRC) incidence and mortality. Understanding the biology responsible for this protective effect is key to developing biomarker-led approaches for rational clinical use. Wnt signaling drives CRC development from initiation to progression through regulation of epithelial-mesenchymal transition (EMT) and cancer stem cell populations. Here, we investigated whether aspirin can rescue these proinvasive phenotypes associated with CRC progression in Wnt-driven human and mouse intestinal organoids. METHODS: We evaluated aspirin-mediated effects on phenotype and stem cell markers in intestinal organoids derived from mouse (ApcMin/+ and Apcflox/flox) and human familial adenomatous polyposis patients. CRC cell lines (HCT116 and Colo205) were used to study effects on motility, invasion, Wnt signaling, and EMT. RESULTS: Aspirin rescues the Wnt-driven cystic organoid phenotype by promoting budding in mouse and human Apc deficient organoids, which is paralleled by decreased stem cell marker expression. Aspirin-mediated Wnt inhibition in ApcMin/+ mice is associated with EMT inhibition and decreased cell migration, invasion, and motility in CRC cell lines. Chemical Wnt activation induces EMT and stem-like alterations in CRC cells, which are rescued by aspirin. Aspirin increases expression of the Wnt antagonist Dickkopf-1 in CRC cells and organoids derived from familial adenomatous polyposis patients, which contributes to EMT and cancer stem cell inhibition. CONCLUSIONS: We provide evidence of phenotypic biomarkers of response to aspirin with an increased epithelial and reduced stem-like state mediated by an increase in Dickkopf-1. This highlights a novel mechanism of aspirin-mediated Wnt inhibition and potential phenotypic and molecular biomarkers for trials.


Subject(s)
Adenomatous Polyposis Coli/drug therapy , Aspirin/pharmacology , Intercellular Signaling Peptides and Proteins/agonists , Intestinal Mucosa/drug effects , Wnt Signaling Pathway/drug effects , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Aspirin/therapeutic use , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Intravital Microscopy , Male , Mice , Mice, Transgenic , Organoids/drug effects , Organoids/pathology , Primary Cell Culture
10.
Nat Commun ; 12(1): 2335, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879799

ABSTRACT

Current therapeutic options for treating colorectal cancer have little clinical efficacy and acquired resistance during treatment is common, even following patient stratification. Understanding the mechanisms that promote therapy resistance may lead to the development of novel therapeutic options that complement existing treatments and improve patient outcome. Here, we identify RAC1B as an important mediator of colorectal tumourigenesis and a potential target for enhancing the efficacy of EGFR inhibitor treatment. We find that high RAC1B expression in human colorectal cancer is associated with aggressive disease and poor prognosis and deletion of Rac1b in a mouse colorectal cancer model reduces tumourigenesis. We demonstrate that RAC1B interacts with, and is required for efficient activation of the EGFR signalling pathway. Moreover, RAC1B inhibition sensitises cetuximab resistant human tumour organoids to the effects of EGFR inhibition, outlining a potential therapeutic target for improving the clinical efficacy of EGFR inhibitors in colorectal cancer.


Subject(s)
Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Carcinogenesis , Cell Line, Tumor , Cetuximab/pharmacology , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/deficiency , Neuropeptides/genetics , Neuropeptides/metabolism , Signal Transduction , Up-Regulation , Wnt Signaling Pathway , rac1 GTP-Binding Protein/deficiency , rac1 GTP-Binding Protein/genetics
11.
Nat Commun ; 11(1): 499, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980649

ABSTRACT

Protein-protein-interaction networks (PPINs) organize fundamental biological processes, but how oncogenic mutations impact these interactions and their functions at a network-level scale is poorly understood. Here, we analyze how a common oncogenic KRAS mutation (KRASG13D) affects PPIN structure and function of the Epidermal Growth Factor Receptor (EGFR) network in colorectal cancer (CRC) cells. Mapping >6000 PPIs shows that this network is extensively rewired in cells expressing transforming levels of KRASG13D (mtKRAS). The factors driving PPIN rewiring are multifactorial including changes in protein expression and phosphorylation. Mathematical modelling also suggests that the binding dynamics of low and high affinity KRAS interactors contribute to rewiring. PPIN rewiring substantially alters the composition of protein complexes, signal flow, transcriptional regulation, and cellular phenotype. These changes are validated by targeted and global experimental analysis. Importantly, genetic alterations in the most extensively rewired PPIN nodes occur frequently in CRC and are prognostic of poor patient outcomes.


Subject(s)
Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , ErbB Receptors/metabolism , Mutation/genetics , Protein Interaction Maps , Proto-Oncogene Proteins p21(ras)/genetics , Cell Line, Tumor , Humans , Phosphorylation , Prognosis , Survival Analysis , bcl-Associated Death Protein/metabolism
13.
Cell Death Differ ; 24(10): 1681-1693, 2017 10.
Article in English | MEDLINE | ID: mdl-28622298

ABSTRACT

Recent studies have suggested increased plasticity of differentiated cells within the intestine to act both as intestinal stem cells (ISCs) and tumour-initiating cells. However, little is known of the processes that regulate this plasticity. Our previous work has shown that activating mutations of Kras or the NF-κB pathway can drive dedifferentiation of intestinal cells lacking Apc. To investigate this process further, we profiled both cells undergoing dedifferentiation in vitro and tumours generated from these cells in vivo by gene expression analysis. Remarkably, no clear differences were observed in the tumours; however, during dedifferentiation in vitro we found a marked upregulation of TGFß signalling, a pathway commonly mutated in colorectal cancer (CRC). Genetic inactivation of TGFß type 1 receptor (Tgfbr1/Alk5) enhanced the ability of KrasG12D/+ mutation to drive dedifferentiation and markedly accelerated tumourigenesis. Mechanistically this is associated with a marked activation of MAPK signalling. Tumourigenesis from differentiated compartments is potently inhibited by MEK inhibition. Taken together, we show that tumours arising in differentiated compartments will be exposed to different suppressive signals, for example, TGFß and blockade of these makes tumourigenesis more efficient from this compartment.


Subject(s)
Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , MAP Kinase Signaling System , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Proliferation/genetics , Genes, ras/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , NF-kappa B/metabolism , Transforming Growth Factor beta/genetics
14.
EMBO Mol Med ; 9(2): 181-197, 2017 02.
Article in English | MEDLINE | ID: mdl-28003334

ABSTRACT

Cancer genome sequencing projects have identified hundreds of genetic alterations, often at low frequencies, raising questions as to their functional relevance. One exemplar gene is HUWE1, which has been found to be mutated in numerous studies. However, due to the large size of this gene and a lack of functional analysis of identified mutations, their significance to carcinogenesis is unclear. To determine the importance of HUWE1, we chose to examine its function in colorectal cancer, where it is mutated in up to 15 per cent of tumours. Modelling of identified mutations showed that they inactivate the E3 ubiquitin ligase activity of HUWE1. Genetic deletion of Huwe1 rapidly accelerated tumourigenic in mice carrying loss of the intestinal tumour suppressor gene Apc, with a dramatic increase in tumour initiation. Mechanistically, this phenotype was driven by increased MYC and rapid DNA damage accumulation leading to loss of the second copy of Apc The increased levels of DNA damage sensitised Huwe1-deficient tumours to DNA-damaging agents and to deletion of the anti-apoptotic protein MCL1. Taken together, these data identify HUWE1 as a bona fide tumour suppressor gene in the intestinal epithelium and suggest a potential vulnerability of HUWE1-mutated tumours to DNA-damaging agents and inhibitors of anti-apoptotic proteins.


Subject(s)
Carcinogenesis , Colorectal Neoplasms/pathology , DNA Damage , Genes, Tumor Suppressor , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , Gene Deletion , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases/genetics
15.
Nat Commun ; 7: 12493, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27558455

ABSTRACT

Melanoma patients treated with oncogenic BRAF inhibitors can develop cutaneous squamous cell carcinoma (cSCC) within weeks of treatment, driven by paradoxical RAS/RAF/MAPK pathway activation. Here we identify frequent TGFBR1 and TGFBR2 mutations in human vemurafenib-induced skin lesions and in sporadic cSCC. Functional analysis reveals these mutations ablate canonical TGFß Smad signalling, which is localized to bulge stem cells in both normal human and murine skin. MAPK pathway hyperactivation (through Braf(V600E) or Kras(G12D) knockin) and TGFß signalling ablation (through Tgfbr1 deletion) in LGR5(+ve) stem cells enables rapid cSCC development in the mouse. Mutation of Tp53 (which is commonly mutated in sporadic cSCC) coupled with Tgfbr1 deletion in LGR5(+ve) cells also results in cSCC development. These findings indicate that LGR5(+ve) stem cells may act as cells of origin for cSCC, and that RAS/RAF/MAPK pathway hyperactivation or Tp53 mutation, coupled with loss of TGFß signalling, are driving events of skin tumorigenesis.


Subject(s)
Antineoplastic Agents/adverse effects , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/genetics , Melanoma/drug therapy , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Animals , Biopsy , Carcinogenesis/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , DNA Mutational Analysis/methods , Female , Humans , Indoles/adverse effects , Male , Mice , Mice, Inbred Strains , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction/drug effects , Skin Neoplasms/pathology , Stem Cells , Sulfonamides/adverse effects , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/genetics , Vemurafenib , Exome Sequencing
16.
Cell Rep ; 12(6): 1019-31, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26235622

ABSTRACT

An understanding of the mechanisms determining MYC's transcriptional and proliferation-promoting activities in vivo could facilitate approaches for MYC targeting. However, post-translational mechanisms that control MYC function in vivo are poorly understood. Here, we demonstrate that MYC phosphorylation at serine 62 enhances MYC accumulation on Lamin A/C-associated nuclear structures and that the protein phosphatase 2A (PP2A) inhibitor protein CIP2A is required for this process. CIP2A is also critical for serum-induced MYC phosphorylation and for MYC-elicited proliferation induction in vitro. Complementary transgenic approaches and an intestinal regeneration model further demonstrated the in vivo importance of CIP2A and serine 62 phosphorylation for MYC activity upon DNA damage. However, targeting of CIP2A did not influence the normal function of intestinal crypt cells. These data underline the importance of nuclear organization in the regulation of MYC phosphorylation, leading to an in vivo demonstration of a strategy for inhibiting MYC activity without detrimental physiological effects.


Subject(s)
Autoantigens/metabolism , Cell Nucleus/metabolism , Lamin Type A/metabolism , Membrane Proteins/metabolism , Animals , Autoantigens/genetics , Cell Nucleus/genetics , Gene Expression Regulation, Neoplastic/genetics , Immunohistochemistry , Lamin Type A/genetics , Membrane Proteins/genetics , Mice , Models, Biological , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
17.
Oncol Rep ; 10(5): 1257-63, 2003.
Article in English | MEDLINE | ID: mdl-12883690

ABSTRACT

The gene Nm23, which encodes for a nucleoside diphosphate kinase, has been defined as a metastasis-suppressor gene because of the inverse correlation between its expression and the metastatic capacity of the tumor cells. For colorectal cancer, however, the findings are equivocal. The aim of our study was to assess, in 160 patients undergoing surgery for colorectal cancer (CRC), the expression of the Nm23-H1 protein and to evaluate its possible associations with traditional clinicopathologic variables, with DNA-ploidy and proliferative activity (S-phase fraction, SPF), and with disease-free and overall survival of patients. Nm23-H1 expressions were evaluated on paraffin-embedded tissue by immunohistochemistry; DNA-ploidy and SPF on frozen tissue by flow-cytometric analysis. The median follow-up time in our study group was 71 months (range 34-115 months). No association was observed between Nm23-H1 protein expression and clinicopathological variables, S-phase fraction and DNA-ploidy. Furthermore, no significant differences were observed in the survival of patients with either moderate or strong Nm23-H1 expression. The major significant predictors for both disease relapse and death were advanced Dukes' stage, DNA aneuploid tumors and high SPF, while lymphohematic invasion was the only independent factor for relapse and non-curative resection for death. Our results indicate that Nm23-H1 activity is tissue-specific and that in CRCs the expression of the protein is not associated with tumor progression and patient prognosis, although further studies are required in order to throw more light on the possible clinical significance of the overexpression of the protein Nm23-H1 in such tumors.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Nucleoside-Diphosphate Kinase , Protein Biosynthesis , Cell Division , Cytoplasm/metabolism , Disease Progression , Disease-Free Survival , Exons , Flow Cytometry , Humans , Immunohistochemistry , Models, Genetic , NM23 Nucleoside Diphosphate Kinases , Ploidies , Prognosis , S Phase , Time Factors
18.
Cell Rep ; 8(6): 1957-1973, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25242332

ABSTRACT

The Wnt pathway is abnormally activated in the majority of colorectal cancers, and significant knowledge has been gained in understanding its role in tumor initiation. However, the mechanisms of metastatic outgrowth in colorectal cancer remain a major challenge. We report that autophagy-dependent metabolic adaptation and survival of metastatic colorectal cancer cells is regulated by the target of oncogenic Wnt signaling, homeobox transcription factor PROX1, expressed by a subpopulation of colon cancer progenitor/stem cells. We identify direct PROX1 target genes and show that repression of a pro-apoptotic member of the BCL2 family, BCL2L15, is important for survival of PROX1(+) cells under metabolic stress. PROX1 inactivation after the establishment of metastases prevented further growth of lesions. Furthermore, autophagy inhibition efficiently targeted metastatic PROX1(+) cells, suggesting a potential therapeutic approach. These data identify PROX1 as a key regulator of the transcriptional network contributing to metastases outgrowth in colorectal cancer.


Subject(s)
Colonic Neoplasms/pathology , Homeodomain Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Wnt Proteins/metabolism , Animals , Apoptosis/drug effects , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/toxicity , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymph Nodes/pathology , Lymphatic Metastasis , Mice , Mice, Inbred NOD , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Stress, Physiological , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Wnt Signaling Pathway
19.
Methods Mol Biol ; 1012: 237-48, 2013.
Article in English | MEDLINE | ID: mdl-24006069

ABSTRACT

Within the intestinal epithelium, c-Myc has been characterized as a target of ß-catenin-TCF signalling (He et al., Science 281:1509-1512, 1998). Given the most commonly mutated tumor suppressor gene within colorectal cancer (CRC) is the APC (Adenomatous Polyposis Coli) gene, a negative regulator of ß-catenin-TCF signalling (Korinek et al., Science 275:1784-1787, 1997), loss of APC leads to Myc deregulation in the vast majority of CRC. This probably explains the numerous studies investigating c-Myc function within the intestinal epithelium. These have shown that c-Myc inhibition or deletion in the adult intestine results in proliferative defects (Muncan et al., Mol Cell Biol 26:8418-8426, 2006; Soucek et al., Nature 455:679-683, 2008). Importantly, intestinal enterocytes are able to survive in the absence of c-Myc which has allowed us (and others) to test the role of c-Myc in intestinal regeneration and tumorigenesis. Remarkably c-Myc deletion suppresses all the phenotypes of the Apc tumor suppressor gene loss and stops intestinal regeneration (Ashton et al., Dev Cell 19:259-269, 2010; Sansom et al., Oncogene 29:2585-2590, 2007). This suggests a clear therapeutic rationale for targeting c-Myc in CRC. Moreover haploinsufficiency for c-Myc in this tissue also reduces intestinal tumorigenesis (Athineos and Sansom, Oncogene 29:2585-2590, 2010; Yekkala and Baudino, Mol Cancer Res 5:1296-1303, 2007), and overexpression of c-Myc affects tissue homeostasis (Finch et al., Mol Cell Biol 29:5306-5315, 2009; Murphy et al., Cancer Cell 14:447-457, 2008). In this chapter we will provide an overview of our current laboratory protocols to characterize c-Myc function in intestinal homeostasis, regeneration, and tumorigenesis in vivo and in vitro.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Homeostasis/genetics , Intestinal Mucosa/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Regeneration/genetics , Cell Culture Techniques , Enterocytes/metabolism , Enterocytes/pathology , Humans , Intestinal Mucosa/pathology , Intestines/pathology , Spheroids, Cellular , Tissue Culture Techniques , Tumor Cells, Cultured
20.
Cell Stem Cell ; 12(6): 761-73, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23665120

ABSTRACT

The Adenomatous Polyposis Coli (APC) gene is mutated in the majority of colorectal cancers (CRCs). Loss of APC leads to constitutively active WNT signaling, hyperproliferation, and tumorigenesis. Identification of pathways that facilitate tumorigenesis after APC loss is important for therapeutic development. Here, we show that RAC1 is a critical mediator of tumorigenesis after APC loss. We find that RAC1 is required for expansion of the LGR5 intestinal stem cell (ISC) signature, progenitor hyperproliferation, and transformation. Mechanistically, RAC1-driven ROS and NF-κB signaling mediate these processes. Together, these data highlight that ROS production and NF-κB activation triggered by RAC1 are critical events in CRC initiation.


Subject(s)
Colorectal Neoplasms/pathology , Intestine, Small/cytology , NF-kappa B/metabolism , Neuropeptides/metabolism , Reactive Oxygen Species/metabolism , Stem Cells/cytology , Wnt Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Proliferation , Colorectal Neoplasms/metabolism , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL