Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cell ; 157(5): 1175-88, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24813849

ABSTRACT

Upon ligand binding, RIPK1 is recruited to tumor necrosis factor receptor superfamily (TNFRSF) and Toll-like receptor (TLR) complexes promoting prosurvival and inflammatory signaling. RIPK1 also directly regulates caspase-8-mediated apoptosis or, if caspase-8 activity is blocked, RIPK3-MLKL-dependent necroptosis. We show that C57BL/6 Ripk1(-/-) mice die at birth of systemic inflammation that was not transferable by the hematopoietic compartment. However, Ripk1(-/-) progenitors failed to engraft lethally irradiated hosts properly. Blocking TNF reversed this defect in emergency hematopoiesis but, surprisingly, Tnfr1 deficiency did not prevent inflammation in Ripk1(-/-) neonates. Deletion of Ripk3 or Mlkl, but not Casp8, prevented extracellular release of the necroptotic DAMP, IL-33, and reduced Myd88-dependent inflammation. Reduced inflammation in the Ripk1(-/-)Ripk3(-/-), Ripk1(-/-)Mlkl(-/-), and Ripk1(-/-)Myd88(-/-) mice prevented neonatal lethality, but only Ripk1(-/-)Ripk3(-/-)Casp8(-/-) mice survived past weaning. These results reveal a key function for RIPK1 in inhibiting necroptosis and, thereby, a role in limiting, not only promoting, inflammation.


Subject(s)
Genes, Lethal , Hematopoiesis , Inflammation/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Caspase 8/metabolism , Cell Death , Liver/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factors/metabolism
2.
Int J Mol Sci ; 21(11)2020 05 30.
Article in English | MEDLINE | ID: mdl-32486243

ABSTRACT

The Wnt signaling pathway is evolutionarily conserved, regulating both embryonic development and maintaining adult tissue homeostasis. Wnt signaling controls several fundamental cell functions, including proliferation, differentiation, migration, and stemness. It therefore plays an important role in the epithelial homeostasis and regeneration of the gastrointestinal tract. Often, both hypo- or hyper-activation of the pathway due to genetic, epigenetic, or receptor/ligand alterations are seen in many solid cancers, such as breast, colorectal, gastric, and prostate. Gastric cancer (GC) is the fourth commonest cause of cancer worldwide and is the second leading cause of cancer-related death annually. Although the number of new diagnoses has declined over recent decades, prognosis remains poor, with only 15% surviving to five years. Geographical differences in clinicopathological features are also apparent, with epidemiological and genetic studies revealing GC to be a highly heterogeneous disease with phenotypic diversity as a result of etiological factors. The molecular heterogeneity associated with GC dictates that a single 'one size fits all' approach to management is unlikely to be successful. Wnt pathway dysregulation has been observed in approximately 50% of GC tumors and may offer a novel therapeutic target for patients who would otherwise have a poor outcome. This mini review will highlight some recent discoveries involving Wnt signaling in GC.


Subject(s)
Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Frizzled Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Ligands , Mice , Mutation , Neoplasm Metastasis , beta Catenin/metabolism
3.
J Pathol ; 245(3): 270-282, 2018 07.
Article in English | MEDLINE | ID: mdl-29603746

ABSTRACT

Epigenetic regulation plays a key role in the link between inflammation and cancer. Here we examine Mbd2, which mediates epigenetic transcriptional silencing by binding to methylated DNA. In separate studies the Mbd2-/- mouse has been shown (1) to be resistant to intestinal tumourigenesis and (2) to have an enhanced inflammatory/immune response, observations that are inconsistent with the links between inflammation and cancer. To clarify its role in tumourigenesis and inflammation, we used constitutive and conditional models of Mbd2 deletion to explore its epithelial and non-epithelial roles in the intestine. Using a conditional model, we found that suppression of intestinal tumourigenesis is due primarily to the absence of Mbd2 within the epithelia. Next, we demonstrated, using the DSS colitis model, that non-epithelial roles of Mbd2 are key in preventing the transition from acute to tumour-promoting chronic inflammation. Combining models revealed that prior to inflammation the altered Mbd2-/- immune response plays a role in intestinal tumour suppression. However, following inflammation the intestine converts from tumour suppressive to tumour promoting. To summarise, in the intestine the normal function of Mbd2 is exploited by cancer cells to enable tumourigenesis, while in the immune system it plays a key role in preventing tumour-enabling inflammation. Which role is dominant depends on the inflammation status of the intestine. As environmental interactions within the intestine can alter DNA methylation patterns, we propose that Mbd2 plays a key role in determining whether these interactions are anti- or pro-tumourigenic and this makes it a useful new epigenetic model for inflammation-associated carcinogenesis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colitis/metabolism , DNA-Binding Proteins/metabolism , Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Animals , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , DNA Methylation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dextran Sulfate , Disease Models, Animal , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, APC , Intestinal Mucosa/pathology , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Mice, Knockout , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/metabolism , Th2 Cells/pathology
4.
Dev Dyn ; 247(3): 521-530, 2018 03.
Article in English | MEDLINE | ID: mdl-28560804

ABSTRACT

BACKGROUND: Metastasis underlies most colorectal cancer mortality. Cancer cells spread through the body as single cells or small clusters of cells that have an invasive, mesenchymal, nonproliferative phenotype. At the secondary site, they revert to a proliferative "tumor constructing" epithelial phenotype to rebuild a tumor. We previously developed a unique in vitro three-dimensional model, called LIM1863-Mph, which faithfully recapitulates these reversible transitions that underpin colorectal cancer metastasis. Wnt signaling plays a key role in these transitions and is initiated by the coupling of extracellular Wnt to Frizzled (FZD). Using the LIM1863-Mph model system we demonstrated that the Wnt receptor FZD7 is necessary for mesenchymal to epithelial transition (MET). Here we investigate the role of Wnt in MET. RESULTS: Wnt secretion is dependent on palmitoylation by Porcupine (PORC). A PORC inhibitor (IWP2) that prevents Wnt secretion, blocked the epithelial transition of mesenchymal LIM1863-Mph cells. Wnt gene array analysis identified several Wnts that are upregulated in epithelial compared with mesenchymal LIM1863-Mph cells, suggesting these ligands in MET. Wnt2B was the most abundant differentially expressed Wnt gene. Indeed, recombinant Wnt2B could overcome the IWP2-mediated block in epithelial transition of mesenchymal LIM1863-Mph cells. CONCLUSIONS: Wnt2B co-operates with Frizzled7 to mediate MET in colorectal cancer. Developmental Dynamics 247:521-530, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Frizzled Receptors/metabolism , Glycoproteins/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Humans , Models, Biological , Wnt Proteins/physiology
5.
Carcinogenesis ; 38(10): 953-965, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28981588

ABSTRACT

Over the past 20 years, huge advances have been made in modelling human diseases such as cancer using genetically modified mice. Accurate in vivo models are essential to examine the complex interaction between cancer cells, surrounding stromal cells, tumour-associated inflammatory cells, fibroblast and blood vessels, and to recapitulate all the steps involved in metastasis. Elucidating these interactions in vitro has inherent limitations, and thus animal models are a powerful tool to enable researchers to gain insight into the complex interactions between signalling pathways and different cells types. This review will focus on how advances in in vivo models have shed light on many aspects of cancer biology including the identification of oncogenes, tumour suppressors and stem cells, epigenetics, cell death and context dependent cell signalling.


Subject(s)
Epithelium/pathology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Neoplasms, Experimental/pathology , Animals , Apoptosis , CRISPR-Cas Systems , Disease Models, Animal , Genes, Tumor Suppressor , Humans , Intestinal Neoplasms/genetics , Methylation , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/pathology , Oncogenes , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology
6.
Mol Cancer ; 14: 169, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26376988

ABSTRACT

BACKGROUND: The establishment and maintenance of polarity is vital for embryonic development and loss of polarity is a frequent characteristic of epithelial cancers, however the underlying molecular mechanisms remain unclear. Here, we identify a novel role for the polarity protein Scrib as a mediator of epidermal permeability barrier acquisition, skeletal morphogenesis, and as a potent tumor suppressor in cutaneous carcinogenesis. METHODS: To explore the role of Scrib during epidermal development, we compared the permeability of toluidine blue dye in wild-type, Scrib heterozygous and Scrib KO embryonic epidermis at E16.5, E17.5 and E18.5. Mouse embryos were stained with alcian blue and alizarin red for skeletal analysis. To establish whether Scrib plays a tumor suppressive role during skin tumorigenesis and/or progression, we evaluated an autochthonous mouse model of skin carcinogenesis in the context of Scrib loss. We utilised Cre-LoxP technology to conditionally deplete Scrib in adult epidermis, since Scrib KO embryos are neonatal lethal. RESULTS: We establish that Scrib perturbs keratinocyte maturation during embryonic development, causing impaired epidermal barrier formation, and that Scrib is required for skeletal morphogenesis in mice. Analysis of conditional transgenic mice deficient for Scrib specifically within the epidermis revealed no skin pathologies, indicating that Scrib is dispensable for normal adult epidermal homeostasis. Nevertheless, bi-allelic loss of Scrib significantly enhanced tumor multiplicity and progression in an autochthonous model of epidermal carcinogenesis in vivo, demonstrating Scrib is an epidermal tumor suppressor. Mechanistically, we show that apoptosis is the critical effector of Scrib tumor suppressor activity during skin carcinogenesis and provide new insight into the function of polarity proteins during DNA damage repair. CONCLUSIONS: For the first time, we provide genetic evidence of a unique link between skin carcinogenesis and loss of the epithelial polarity regulator Scrib, emphasizing that Scrib exerts a wide-spread tumor suppressive function in epithelia.


Subject(s)
Carcinogenesis/genetics , Epidermis/growth & development , Intracellular Signaling Peptides and Proteins/genetics , Skin Neoplasms/genetics , Animals , Carcinogenesis/pathology , Cell Differentiation/genetics , Cell Polarity/genetics , Disease Models, Animal , Embryo, Mammalian , Epidermis/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genes, Tumor Suppressor , Humans , Integrases/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Knockout , Skin Neoplasms/pathology
7.
Biochem J ; 458(2): 251-8, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24320611

ABSTRACT

PIK3CA, the gene encoding the p110α catalytic subunit of PI3K (phosphoinositide 3-kinase), is mutated in approximately 20% of sporadic CRCs (colorectal cancers), but the role of these mutations in the pathogenesis of CRC remains unclear. In the present study we used a novel mouse model to investigate the role of the Pik3caH1047R mutation, the most common PIK3CA mutation in CRC, during the development and progression of intestinal cancer. Our results demonstrate that Pik3caH1047R, when expressed at physiological levels, is insufficient to initiate intestinal tumorigenesis; however, in the context of Apc (adenomatous polyposis coli) loss, which is observed in 80% of CRCs and by itself results in benign intestinal adenomas, the Pik3caH1047R mutation promotes the development of highly aggressive and invasive adenocarcinomas in both the small and large intestines. The results of the present study show that an activating Pik3ca mutation can act in tandem with Apc loss to drive the progression of gastrointestinal cancer and thus this disease may be susceptible to therapeutic targeting using PI3K pathway inhibitors.


Subject(s)
Adenocarcinoma/genetics , Adenomatous Polyposis Coli Protein/deficiency , Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/genetics , Mutation/genetics , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Class I Phosphatidylinositol 3-Kinases , Disease Progression , Female , Gene Knock-In Techniques , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/biosynthesis
8.
Cells ; 13(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38667288

ABSTRACT

As the treatment landscape for prostate cancer gradually evolves, the frequency of treatment-induced neuroendocrine prostate cancer (NEPC) and double-negative prostate cancer (DNPC) that is deficient for androgen receptor (AR) and neuroendocrine (NE) markers has increased. These prostate cancer subtypes are typically refractory to AR-directed therapies and exhibit poor clinical outcomes. Only a small range of NEPC/DNPC models exist, limiting our molecular understanding of this disease and hindering our ability to perform preclinical trials exploring novel therapies to treat NEPC/DNPC that are urgently needed in the clinic. Here, we report the development of the CU-PC01 PDX model that represents AR-negative mCRPC with PTEN/RB/PSMA loss and CTNN1B/TP53/BRCA2 genetic variants. The CU-PC01 model lacks classic NE markers, with only focal and/or weak expression of chromogranin A, INSM1 and CD56. Collectively, these findings are most consistent with a DNPC phenotype. Ex vivo and in vivo preclinical studies revealed that CU-PC01 PDX tumours are resistant to mCRPC standard-of-care treatments enzalutamide and docetaxel, mirroring the donor patient's treatment response. Furthermore, short-term CU-PC01 tumour explant cultures indicate this model is initially sensitive to PARP inhibition with olaparib. Thus, the CU-PC01 PDX model provides a valuable opportunity to study AR-negative mCRPC biology and to discover new treatment avenues for this hard-to-treat disease.


Subject(s)
Piperazines , Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Male , Humans , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Mice , Xenograft Model Antitumor Assays , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , Neoplasm Metastasis , Nitriles/pharmacology , Disease Models, Animal , Benzamides/pharmacology , Phthalazines/pharmacology , Phthalazines/therapeutic use
9.
Nature ; 446(7136): 676-9, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17377531

ABSTRACT

The APC gene encodes the adenomatous polyposis coli tumour suppressor protein, germline mutation of which characterizes familial adenomatous polyposis (FAP), an autosomal intestinal cancer syndrome. Inactivation of APC is also recognized as the key early event in the development of sporadic colorectal cancers, and its loss results in constitutive activity of the beta-catenin-Tcf4 transcription complex. The proto-oncogene c-MYC has been identified as a target of the Wnt pathway in colorectal cancer cells in vitro, in normal crypts in vivo and in intestinal epithelial cells acutely transformed on in vivo deletion of the APC gene; however, the significance of this is unclear. Therefore, to elucidate the role Myc has in the intestine after Apc loss, we have simultaneously deleted both Apc and Myc in the adult murine small intestine. Here we show that loss of Myc rescued the phenotypes of perturbed differentiation, migration, proliferation and apoptosis, which occur on deletion of Apc. Remarkably, this rescue occurred in the presence of high levels of nuclear beta-catenin. Array analysis revealed that Myc is required for the majority of Wnt target gene activation following Apc loss. These data establish Myc as the critical mediator of the early stages of neoplasia following Apc loss.


Subject(s)
Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Gene Deletion , Genes, APC , Genes, myc/genetics , Intestine, Small/metabolism , Proto-Oncogene Proteins c-myc/deficiency , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Enterocytes/metabolism , Intestine, Small/cytology , Male , Mice , Phenotype , Proto-Oncogene Proteins c-myc/genetics
10.
Biomolecules ; 12(2)2022 02 15.
Article in English | MEDLINE | ID: mdl-35204808

ABSTRACT

Aberrant activation of the Wnt pathway is emerging as a frequent event during prostate cancer that can facilitate tumor formation, progression, and therapeutic resistance. Recent discoveries indicate that targeting the Wnt pathway to treat prostate cancer may be efficacious. However, the functional consequence of activating the Wnt pathway during the different stages of prostate cancer progression remains unclear. Preclinical work investigating the efficacy of targeting Wnt signaling for the treatment of prostate cancer, both in primary and metastatic lesions, and improving our molecular understanding of treatment responses is crucial to identifying effective treatment strategies and biomarkers that help guide treatment decisions and improve patient care. In this review, we outline the type of genetic alterations that lead to activated Wnt signaling in prostate cancer, highlight the range of laboratory models used to study the role of Wnt genetic drivers in prostate cancer, and discuss new mechanistic insights into how the Wnt cascade facilitates prostate cancer growth, metastasis, and drug resistance.


Subject(s)
Prostatic Neoplasms , Wnt Signaling Pathway , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
11.
Elife ; 112022 08 30.
Article in English | MEDLINE | ID: mdl-36040316

ABSTRACT

The Wnt/ß-catenin signalling pathway regulates multiple cellular processes during development and many diseases, including cell proliferation, migration, and differentiation. Despite their hydrophobic nature, Wnt proteins exert their function over long distances to induce paracrine signalling. Recent studies have identified several factors involved in Wnt secretion; however, our understanding of how Wnt ligands are transported between cells to interact with their cognate receptors is still debated. Here, we demonstrate that gastric cancer cells utilise cytonemes to transport Wnt3 intercellularly to promote proliferation and cell survival. Furthermore, we identify the membrane-bound scaffolding protein Flotillin-2 (Flot2), frequently overexpressed in gastric cancer, as a modulator of these cytonemes. Together with the Wnt co-receptor and cytoneme initiator Ror2, Flot2 determines the number and length of Wnt3 cytonemes in gastric cancer. Finally, we show that Flotillins are also necessary for Wnt8a cytonemes during zebrafish embryogenesis, suggesting a conserved mechanism for Flotillin-mediated Wnt transport on cytonemes in development and disease.


Subject(s)
Stomach Neoplasms , Zebrafish , Animals , Embryonic Development , Wnt Proteins/physiology , Wnt Signaling Pathway
12.
Gastroenterology ; 136(7): 2316-2324.e1-3, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19268669

ABSTRACT

BACKGROUND AND AIMS: The Wnt pathway has previously been shown to play a role in hepatic zonation. Herein, we have explored the role of 3 key components (Apc, beta-catenin, and c-Myc) of the Wnt pathway in the zonation of ammonia metabolizing enzymes. METHODS: Conditional deletion of Apc, beta-catenin, and c-Myc was induced in the livers of mice and the expression of periportal and perivenous hepatocyte markers was determined by polymerase chain reaction, Western blotting, and immunohistochemical techniques. RESULTS: Under normal circumstances, the urea cycle enzyme carbamoylphosphate synthetase I (CPS I) is present in the periportal, intermediate, and the first few layers of the perivenous zone. In contrast, glutamine synthetase (GS)--and nuclear beta-catenin--is expressed in a complementary fashion in the last 1-2 cell layers of the perivenous zone. Conditional loss of Apc resulted in the expression of nuclear beta-catenin and GS in most hepatocytes irrespective of zone. Induction of GS in hepatocytes outside the normal perivenous zone was accompanied by a reduction in the expression of CPS I. Deletion of beta-catenin induces a loss of GS and a complementary increase in expression of CPS I irrespective of whether Apc is present. Remarkably, deletion of c-Myc did not perturb the pattern of zonation. CONCLUSIONS: It has been shown that the Wnt pathway is key to imposing the pattern of zonation within the liver. Herein we have addressed the relevance of 3 major Wnt pathway components and show critically that the zonation is c-Myc independent but beta-catenin dependent.


Subject(s)
Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cells, Cultured , Gene Deletion , Gene Expression Regulation , Hepatocytes/enzymology , Hepatocytes/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Models, Animal , Proto-Oncogene Proteins c-myc/genetics , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Signal Transduction/physiology , Wnt Proteins/genetics , beta Catenin/genetics
13.
Cell Stem Cell ; 27(4): 646-662.e7, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32693086

ABSTRACT

Epidermal growth factor (EGF) maintains intestinal stem cell (ISC) proliferation and is a key component of organoid growth media yet is dispensable for intestinal homeostasis, suggesting roles for multiple EGF family ligands in ISC function. Here, we identified neuregulin 1 (NRG1) as a key EGF family ligand that drives tissue repair following injury. NRG1, but not EGF, is upregulated upon damage and is expressed in mesenchymal stromal cells, macrophages, and Paneth cells. NRG1 deletion reduces proliferation in intestinal crypts and compromises regeneration capacity. NRG1 robustly stimulates proliferation in crypts and induces budding in organoids, in part through elevated and sustained activation of mitogen-activated protein kinase (MAPK) and AKT. Consistently, NRG1 treatment induces a proliferative gene signature and promotes organoid formation from progenitor cells and enhances regeneration following injury. These data suggest mesenchymal-derived NRG1 is a potent mediator of tissue regeneration and may inform the development of therapies for enhancing intestinal repair after injury.


Subject(s)
Intestines , Neuregulin-1 , Cell Proliferation , Epithelium , Paneth Cells
14.
Mol Cell Biol ; 26(22): 8418-26, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16954380

ABSTRACT

Inhibition of the mutationally activated Wnt cascade in colorectal cancer cell lines induces a rapid G1 arrest and subsequent differentiation. This arrest can be overcome by maintaining expression of a single Tcf4 target gene, the proto-oncogene c-Myc. Since colorectal cancer cells share many molecular characteristics with proliferative crypt progenitors, we have assessed the physiological role of c-Myc in adult crypts by conditional gene deletion. c-Myc-deficient crypts are lost within weeks and replaced by c-Myc-proficient crypts through a fission process of crypts that have escaped gene deletion. Although c-Myc(-/-) crypt cells remain in the cell cycle, they are on average much smaller than wild-type cells, cycle slower, and divide at a smaller cell size. c-Myc appears essential for crypt progenitor cells to provide the necessary biosynthetic capacity to successfully progress through the cell cycle.


Subject(s)
Colorectal Neoplasms/metabolism , Genes, myc , Intestines/pathology , Proto-Oncogene Proteins c-myc/genetics , TCF Transcription Factors/genetics , Wnt Proteins/genetics , Animals , Apoptosis , Cell Count , Colorectal Neoplasms/pathology , Epithelium/physiology , Gene Targeting , Integrases/genetics , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mitosis , Transcription Factor 7-Like 2 Protein
15.
Cancer Res ; 79(23): 5901-5906, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31431458

ABSTRACT

In the March 1 issue of Cancer Research, we identified the Wnt receptor Fzd7 as an attractive therapeutic target for the treatment of gastric cancer. In summary, we showed that pharmacological inhibition of Wnt receptors, or genetic deletion of Fzd7, blocks the initiation and growth of gastric tumors. Inhibiting Fzd receptors, specifically Fzd7, inhibits the growth of gastric cancer cells even in the presence of adenomatous polyposis coli (Apc) mutation. Apc is located in the cytoplasm downstream of Fzd7 in the Wnt signaling cascade and APC mutations activate Wnt/ß-catenin signaling, therefore, this result seems counterintuitive. Here, we analyze this result in greater detail in the context of current knowledge of Wnt signaling and discuss the wider implications of this aspect of Wnt signaling in other cancers.


Subject(s)
Genetic Heterogeneity , Neoplasms/pathology , Wnt Signaling Pathway/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Frizzled Receptors/antagonists & inhibitors , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/genetics , Intracellular Space/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Wnt Signaling Pathway/drug effects
16.
Methods Mol Biol ; 1576: 123-133, 2019.
Article in English | MEDLINE | ID: mdl-27704362

ABSTRACT

The discovery of Lgr5 as a marker of adult stem cells meant that stem cell populations could be purified and studied in isolation. Importantly, when cultured under the appropriate conditions these stem cells form organoids in tissue culture that retain many features of the tissue of origin. The organoid cultures are accessible to genetic and biochemical manipulation, bridging the gap between in vivo mouse models and conventional tissue culture. Here we describe robust protocols to establish organoids from gastrointestinal tissues (stomach, intestine, liver) and Cre-recombinase mediated gene manipulation in vitro.


Subject(s)
Adult Stem Cells/cytology , Cell Separation/methods , Gene Deletion , Integrases/metabolism , Intestines/cytology , Liver/cytology , Organoids/cytology , Stomach/cytology , Adult Stem Cells/metabolism , Animals , Cell Culture Techniques/methods , Cells, Cultured , Liver/metabolism , Mice , Organoids/metabolism , Receptors, G-Protein-Coupled/metabolism
17.
Cancer Res ; 79(5): 970-981, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30622113

ABSTRACT

A subset of patients with gastric cancer have mutations in genes that participate in or regulate Wnt signaling at the level of ligand (Wnt) receptor (Fzd) binding. Moreover, increased Fzd expression is associated with poor clinical outcome. Despite these findings, there are no in vivo studies investigating the potential of targeting Wnt receptors for treating gastric cancer, and the specific Wnt receptor transmitting oncogenic Wnt signaling in gastric cancer is unknown. Here, we use inhibitors of Wnt/Fzd (OMP-18R5/vantictumab) and conditional gene deletion to test the therapeutic potential of targeting Wnt signaling in preclinical models of intestinal-type gastric cancer and ex vivo organoid cultures. Pharmacologic targeting of Fzd inhibited the growth of gastric adenomas in vivo. We identified Fzd7 to be the predominant Wnt receptor responsible for transmitting Wnt signaling in human gastric cancer cells and mouse models of gastric cancer, whereby Fzd7-deficient cells were retained in gastric adenomas but were unable to respond to Wnt signals and consequently failed to proliferate. Genetic deletion of Fzd7 or treatment with vantictumab was sufficient to inhibit the growth of gastric adenomas with or without mutations to Apc. Vantictumab is currently in phase Ib clinical trials for advanced pancreatic, lung, and breast cancer. Our data extend the scope of patients that may benefit from this therapeutic approach as we demonstrate that this drug will be effective in treating patients with gastric cancer regardless of APC mutation status. SIGNIFICANCE: The Wnt receptor Fzd7 plays an essential role in gastric tumorigenesis irrespective of Apc mutation status, therefore targeting Wnt/Fzd7 may be of therapeutic benefit to patients with gastric cancer.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Frizzled Receptors/metabolism , Stomach Neoplasms/metabolism , Wnt Signaling Pathway , Adenomatous Polyposis Coli Protein/metabolism , Animals , Carcinogenesis , Cell Growth Processes/physiology , Cell Line, Tumor , Frizzled Receptors/antagonists & inhibitors , Frizzled Receptors/genetics , Gene Deletion , Gene Knockdown Techniques , Heterografts , Humans , Mice , Mutation , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
18.
Genes (Basel) ; 9(4)2018 Mar 23.
Article in English | MEDLINE | ID: mdl-29570681

ABSTRACT

Wnt signalling regulates several cellular functions including proliferation, differentiation, apoptosis and migration, and is critical for embryonic development. Stem cells are defined by their ability for self-renewal and the ability to be able to give rise to differentiated progeny. Consequently, they are essential for the homeostasis of many organs including the gastrointestinal tract. This review will describe the huge advances in our understanding of how stem cell functions in the gastrointestinal tract are regulated by Wnt signalling, including how deregulated Wnt signalling can hijack these functions to transform cells and lead to cancer.

19.
Prog Mol Biol Transl Sci ; 153: 299-319, 2018 01.
Article in English | MEDLINE | ID: mdl-29389521

ABSTRACT

The Wnt pathway is at the heart of organoid technology, which is set to revolutionize the cancer field. We can now predetermine a patient's response to any given anticancer therapy by exposing tumor organoids established from the patient's own tumor. This cutting-edge biomedical platform translates to patients being treated with the correct drug at the correct dose from the outset, a truly personalized and precise medical approach. A high throughput drug screen on organoids also allows drugs to be tested in limitless combinations. More recently, the tumor cells that are resistant to the therapy given to a patient were selected in culture using the patient's organoids. The resistant tumor organoids were then screened empirically to identify drugs that will kill the resistant cells. This information allows diagnosis in real-time to either prevent tumor recurrence or effectively treat the recurring tumor. Furthermore, the ability to culture stem cell-derived epithelium as organoids has enabled us to begin to understand how a stem cell becomes a cancer cell or to pin-point the genetic alteration that underlies a given genetic syndrome. Here we summarize these advances and the central role of Wnt signaling, and identify the next challenges for organoid technology.


Subject(s)
Neoplasms/therapy , Organ Culture Techniques/methods , Organoids/cytology , Precision Medicine , Regenerative Medicine , Wnt Proteins/metabolism , Wnt Signaling Pathway , Cell Proliferation , Humans , Neoplasms/metabolism , Neoplasms/pathology , Organoids/physiology
20.
Cancer Discov ; 8(6): 764-779, 2018 06.
Article in English | MEDLINE | ID: mdl-29581176

ABSTRACT

Genetic alterations that potentiate PI3K signaling are frequent in prostate cancer, yet how different genetic drivers of the PI3K cascade contribute to prostate cancer is unclear. Here, we report PIK3CA mutation/amplification correlates with poor survival of patients with prostate cancer. To interrogate the requirement of different PI3K genetic drivers in prostate cancer, we employed a genetic approach to mutate Pik3ca in mouse prostate epithelium. We show Pik3caH1047R mutation causes p110α-dependent invasive prostate carcinoma in vivo Furthermore, we report that PIK3CA mutation and PTEN loss coexist in patients with prostate cancer and can cooperate in vivo to accelerate disease progression via AKT-mTORC1/2 hyperactivation. Contrasting single mutants that slowly acquire castration-resistant prostate cancer (CRPC), concomitant Pik3ca mutation and Pten loss caused de novo CRPC. Thus, Pik3ca mutation and Pten deletion are not functionally redundant. Our findings indicate that PIK3CA mutation is an attractive prognostic indicator for prostate cancer that may cooperate with PTEN loss to facilitate CRPC in patients.Significance: We show PIK3CA mutation correlates with poor prostate cancer prognosis and causes prostate cancer in mice. Moreover, PIK3CA mutation and PTEN loss coexist in prostate cancer and can cooperate in vivo to accelerate tumorigenesis and facilitate CRPC. Delineating this synergistic relationship may present new therapeutic/prognostic approaches to overcome castration/PI3K-AKT-mTORC1/2 inhibitor resistance. Cancer Discov; 8(6); 764-79. ©2018 AACR.See related commentary by Triscott and Rubin, p. 682This article is highlighted in the In This Issue feature, p. 663.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Mutation , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Animals , Cell Line, Tumor , Disease Progression , Gene Amplification , Gene Deletion , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasms, Experimental , Prognosis , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL