Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Semin Cell Dev Biol ; 150-151: 28-34, 2023 12.
Article in English | MEDLINE | ID: mdl-37095033

ABSTRACT

Mutations in the gene encoding the Adenomatous polyposis coli protein (APC) were discovered as driver mutations in colorectal cancers almost 30 years ago. Since then, the importance of APC in normal tissue homeostasis has been confirmed in a plethora of other (model) organisms spanning a large evolutionary space. APC is a multifunctional protein, with roles as a key scaffold protein in complexes involved in diverse signalling pathways, most prominently the Wnt signalling pathway. APC is also a cytoskeletal regulator with direct and indirect links to and impacts on all three major cytoskeletal networks. Correspondingly, a wide range of APC binding partners have been identified. Mutations in APC are extremely strongly associated with colorectal cancers, particularly those that result in the production of truncated proteins and the loss of significant regions from the remaining protein. Understanding the complement of its role in health and disease requires knowing the relationship between and regulation of its diverse functions and interactions. This in turn requires understanding its structural and biochemical features. Here we set out to provide a brief overview of the roles and function of APC and then explore its conservation and structure using the extensive sequence data, which is now available, and spans a broad range of taxonomy. This revealed conservation of APC across taxonomy and new relationships between different APC protein families.


Subject(s)
Adenomatous Polyposis Coli Protein , Adenomatous Polyposis Coli , Humans , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Mutation , Cytoskeleton/metabolism , Wnt Signaling Pathway/genetics
2.
PLoS Biol ; 17(11): e3000540, 2019 11.
Article in English | MEDLINE | ID: mdl-31770366

ABSTRACT

Interleukin-22 (IL-22) is a critical immune defence cytokine that maintains intestinal homeostasis and promotes wound healing and tissue regeneration, which can support the growth of colorectal tumours. Mutations in the adenomatous polyposis coli gene (Apc) are a major driver of familial colorectal cancers (CRCs). How IL-22 contributes to APC-mediated tumorigenesis is poorly understood. To investigate IL-22 signalling in wild-type (WT) and APC-mutant cells, we performed RNA sequencing (RNAseq) of IL-22-treated murine small intestinal epithelial organoids. In WT epithelia, antimicrobial defence and cellular stress response pathways were most strongly induced by IL-22. Surprisingly, although IL-22 activates signal transducer and activator of transcription 3 (STAT3) in APC-mutant cells, STAT3 target genes were not induced. Our analyses revealed that ApcMin/Min cells are resistant to IL-22 due to reduced expression of the IL-22 receptor, and increased expression of inhibitors of STAT3, particularly histone deacetylases (HDACs). We further show that IL-22 increases DNA damage and genomic instability, which can accelerate cellular transition from heterozygosity (ApcMin/+) to homozygosity (ApcMin/Min) to drive tumour formation. Our data reveal an unexpected role for IL-22 in promoting early tumorigenesis while excluding a function for IL-22 in transformed epithelial cells.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Epithelial Cells/metabolism , Interleukins/metabolism , Adenomatous Polyposis Coli/genetics , Animals , Carcinogenesis/genetics , Colorectal Neoplasms/metabolism , Cytokines/metabolism , Female , Interleukins/genetics , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Intestines/physiology , Male , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , Sequence Analysis, RNA/methods , Signal Transduction , Interleukin-22
3.
PLoS Biol ; 14(6): e1002491, 2016 06.
Article in English | MEDLINE | ID: mdl-27348469

ABSTRACT

The crypts of the intestinal epithelium house the stem cells that ensure the continual renewal of the epithelial cells that line the intestinal tract. Crypt number increases by a process called crypt fission, the division of a single crypt into two daughter crypts. Fission drives normal tissue growth and maintenance. Correspondingly, it becomes less frequent in adulthood. Importantly, fission is reactivated to drive adenoma growth. The mechanisms governing fission are poorly understood. However, only by knowing how normal fission operates can cancer-associated changes be elucidated. We studied normal fission in tissue in three dimensions using high-resolution imaging and used intestinal organoids to identify underlying mechanisms. We discovered that both the number and relative position of Paneth cells and Lgr5+ cells are important for fission. Furthermore, the higher stiffness and increased adhesion of Paneth cells are involved in determining the site of fission. Formation of a cluster of Lgr5+ cells between at least two Paneth-cell-rich domains establishes the site for the upward invagination that initiates fission.


Subject(s)
Intestinal Mucosa/cytology , Paneth Cells/cytology , Receptors, G-Protein-Coupled/metabolism , Stem Cell Niche , Stem Cells/cytology , Age Factors , Animals , Cell Adhesion , Cell Count , Cell Division , Cell Proliferation , Integrin beta4/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Models, Biological , Organoids/cytology , Organoids/metabolism , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Stem Cells/metabolism
4.
Bull Math Biol ; 80(2): 335-359, 2018 02.
Article in English | MEDLINE | ID: mdl-29234982

ABSTRACT

Crypt fission is an in vivo tissue deformation process that is involved in both intestinal homeostasis and colorectal tumourigenesis. Despite its importance, the mechanics underlying crypt fission are currently poorly understood. Recent experimental development of organoids, organ-like buds cultured from crypt stem cells in vitro, has shown promise in shedding light on crypt fission. Drawing inspiration from observations of organoid growth and fission in vivo, we develop a computational model of a deformable epithelial tissue layer. Results from in silico experiments show the stiffness of cells and the proportions of cell subpopulations affect the nature of deformation in the epithelial layer. In particular, we find that increasing the proportion of stiffer cells in the layer increases the likelihood of crypt fission occurring. This is in agreement with and helps explain recent experimental work.


Subject(s)
Intestinal Mucosa/anatomy & histology , Models, Biological , Animals , Biomechanical Phenomena , Cell Death , Cell Proliferation , Cell Size , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Computer Simulation , Homeostasis , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Mathematical Concepts , Models, Anatomic
5.
Bioessays ; 36(9): 818-26, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24943963

ABSTRACT

Regulation of microtubule (MT) dynamics is essential for many cellular processes, but the machinery that controls MT dynamics remains poorly understood. MT plus-end tracking proteins (+TIPs) are a set of MT-associated proteins that dynamically track growing MT ends and are uniquely positioned to govern MT dynamics. +TIPs associate with each other in a complex array of inter- and intra-molecular interactions known as the "+TIP network." Why do so many +TIPs bind to other +TIPs? Typical answers include the ideas that these interactions localize proteins where they are needed, deliver proteins to the cortex, and/or create regulatory pathways. We propose an additional and more mechanistic hypothesis: that +TIPs bind each other to create a superstructure that promotes MT assembly by constraining the structural fluctuations of the MT tip, thus acting as a polymerization chaperone.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Animals , Humans , Molecular Chaperones/metabolism , Protein Interaction Maps , Protein Multimerization
6.
Curr Opin Cell Biol ; 20(2): 186-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18359618

ABSTRACT

The adenomatous polyposis coli (Apc) protein participates in many of the fundamental cellular processes that govern epithelial tissues: Apc is directly involved in regulating the availability of beta-catenin for transcriptional de-repression of Tcf/LEF transcription factors, it contributes to the stability of microtubules in interphase and mitosis, and has an impact on the dynamics of F-actin. Thus Apc contributes directly and/or indirectly to proliferation, differentiation, migration, and apoptosis. This particular multifunctionality can explain why disruption of Apc is especially detrimental for the epithelium of the gut, where Apc mutations are common in most cancers. We summarise recent data that shed light on the molecular mechanisms involved in the different functions of Apc.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Physiological Phenomena , Epithelium/metabolism , Animals , Apoptosis , Cell Nucleus/metabolism , Humans , Wnt Proteins/metabolism
7.
J Cell Sci ; 125(Pt 4): 887-95, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22399804

ABSTRACT

Colorectal cancers commonly carry truncation mutations in the adenomatous polyposis coli (APC) gene. The APC protein contributes to the stabilization of microtubules. Consistently, microtubules in cells lacking APC depolymerize more readily in response to microtubule-destabilizing drugs. This raises the possibility that such agents are suitable for treatment of APC-deficient cancers. However, APC-deficient cells have a compromised spindle assembly checkpoint, which renders them less sensitive to killing by microtubule poisons whose toxicity relies on the induction of prolonged mitotic arrest. Here, we describe the novel discovery that the clinically used microtubule-depolymerizing drug vinorelbine (Navelbine) kills APC-deficient cells in culture and in intestinal tissue more effectively than it kills wild-type cells. This is due to the ability of vinorelbine to kill cells in interphase independently of mitotic arrest. Consistent with a role for p53 in cell death in interphase, depletion of p53 renders cells less sensitive to vinorelbine, but only in the presence of wild-type APC. The pro-apoptotic protein BIM (also known as BCL2L11) is recruited to mitochondria in response to vinorelbine, where it can inhibit the anti-apoptotic protein BCL2, suggesting that BIM mediates vinorelbine-induced cell death. This recruitment of BIM is enhanced in cells lacking APC. Consistently, BIM depletion dampens the selective effect of vinorelbine on these cells. Our findings reveal that vinorelbine is a potential therapeutic agent for colorectal cancer, but they also illustrate the importance of the APC tumour suppressor status when predicting therapeutic efficacy.


Subject(s)
Adenomatous Polyposis Coli Protein/deficiency , Microtubules/drug effects , Mitosis/drug effects , Vinblastine/analogs & derivatives , Adenoma/drug therapy , Adenoma/genetics , Adenomatous Polyposis Coli Protein/genetics , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Cycle/physiology , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Humans , Interphase/drug effects , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/deficiency , Vinblastine/pharmacology , Vinorelbine
8.
PLoS Comput Biol ; 8(5): e1002515, 2012.
Article in English | MEDLINE | ID: mdl-22654652

ABSTRACT

The role of the basement membrane is vital in maintaining the integrity and structure of an epithelial layer, acting as both a mechanical support and forming the physical interface between epithelial cells and the surrounding connective tissue. The function of this membrane is explored here in the context of the epithelial monolayer that lines the colonic crypt, test-tube shaped invaginations that punctuate the lining of the intestine and coordinate a regular turnover of cells to replenish the epithelial layer every few days. To investigate the consequence of genetic mutations that perturb the system dynamics and can lead to colorectal cancer, it must be possible to track the emerging tissue level changes that arise in the crypt. To that end, a theoretical crypt model with a realistic, deformable geometry is required. A new discrete crypt model is presented, which focuses on the interaction between cell- and tissue-level behaviour, while incorporating key subcellular components. The model contains a novel description of the role of the surrounding tissue and musculature, based upon experimental observations of the tissue structure of the crypt, which are also reported. A two-dimensional (2D) cross-sectional geometry is considered, and the shape of the crypt is allowed to evolve and deform. Simulation results reveal how the shape of the crypt may contribute mechanically to the asymmetric division events typically associated with the stem cells at the base. The model predicts that epithelial cell migration may arise due to feedback between cell loss at the crypt collar and density-dependent cell division, an hypothesis which can be investigated in a wet lab. This work forms the basis for investigation of the deformation of the crypt structure that can occur due to proliferation of cells exhibiting mutant phenotypes, experiments that would not be possible in vivo or in vitro.


Subject(s)
Basement Membrane/cytology , Basement Membrane/physiology , Cell Communication/physiology , Colon/cytology , Colon/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Models, Biological , Animals , Computer Simulation , Humans
9.
PLoS Genet ; 6(1): e1000816, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20084116

ABSTRACT

Contributions of null and hypomorphic alleles of Apc in mice produce both developmental and pathophysiological phenotypes. To ascribe the resulting genotype-to-phenotype relationship unambiguously to the Wnt/beta-catenin pathway, we challenged the allele combinations by genetically restricting intracellular beta-catenin expression in the corresponding compound mutant mice. Subsequent evaluation of the extent of resulting Tcf4-reporter activity in mouse embryo fibroblasts enabled genetic measurement of Wnt/beta-catenin signaling in the form of an allelic series of mouse mutants. Different permissive Wnt signaling thresholds appear to be required for the embryonic development of head structures, adult intestinal polyposis, hepatocellular carcinomas, liver zonation, and the development of natural killer cells. Furthermore, we identify a homozygous Apc allele combination with Wnt/beta-catenin signaling capacity similar to that in the germline of the Apc(min) mice, where somatic Apc loss-of-heterozygosity triggers intestinal polyposis, to distinguish whether co-morbidities in Apc(min) mice arise independently of intestinal tumorigenesis. Together, the present genotype-phenotype analysis suggests tissue-specific response levels for the Wnt/beta-catenin pathway that regulate both physiological and pathophysiological conditions.


Subject(s)
Mice/genetics , Mice/metabolism , Signal Transduction , beta Catenin/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Female , Fibroblasts/metabolism , Intestinal Mucosa/metabolism , Intestines/embryology , Intestines/growth & development , Liver/embryology , Liver/growth & development , Liver/metabolism , Male , Mice/embryology , Mice/growth & development , Mice, Inbred C57BL , Mice, Knockout , Wnt Proteins , Wnt3 Protein , beta Catenin/genetics
10.
J Cell Sci ; 123(Pt 5): 736-46, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20144988

ABSTRACT

Mutations in the tumour suppressor Adenomatous polyposis coli (Apc) initiate most sporadic colorectal cancers. Apc is implicated in regulating microtubule (MT) dynamics in interphase and mitosis. However, little is known about the underlying mechanism or regulation of this Apc function. We identified importin-beta as a binding partner of Apc that regulates its effect on MTs. Apc binds importin-beta in vitro and in Xenopus egg extracts, and RanGTP inhibits this interaction. The armadillo-like repeat domain of importin-beta binds to the middle of Apc, where it can compete with beta-catenin. In addition, two independent sites in the C terminus of Apc bind the N-terminal region of importin-beta. Binding to importin-beta reduces the ability of Apc to assemble and bundle MTs in vitro and to promote assembly of microtubule asters in Xenopus egg extracts, but does not affect the binding of Apc to MTs or to EB1. Depletion of Apc decreases the formation of cold-stable spindles in Xenopus egg extracts. Importantly, the ability of purified Apc to rescue this phenotype was reduced when it was constitutively bound to importin-beta. Thus, importin-beta binds to Apc and negatively regulates the MT-assembly and spindle-promoting activity of Apc in a Ran-regulatable manner.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Microtubules/metabolism , Xenopus Proteins/metabolism , beta Karyopherins/metabolism , ran GTP-Binding Protein/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Binding Sites/genetics , Binding Sites/physiology , Immunoprecipitation , Microtubule-Associated Proteins/metabolism , Protein Binding/genetics , Protein Binding/physiology , Xenopus , Xenopus Proteins/genetics , beta Catenin/metabolism
11.
J Cell Biol ; 176(2): 183-95, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17227893

ABSTRACT

Mutations in the adenomatous polyposis coli (APC) tumor suppressor gene initiate a majority of colorectal cancers. Acquisition of chromosomal instability is an early event in these tumors. We provide evidence that the loss of APC leads to a partial loss of interkinetochore tension at metaphase and alters mitotic progression. Furthermore, we show that inhibition of APC in U2OS cells compromises the mitotic spindle checkpoint. This is accompanied by a decrease in the association of the checkpoint proteins Bub1 and BubR1 with kinetochores. Additionally, APC depletion reduced apoptosis. As expected from this combination of defects, tetraploidy and polyploidy are consequences of APC inhibition in vitro and in vivo. The removal of APC produced the same defects in HCT116 cells that have constitutively active beta-catenin. These data show that the loss of APC immediately induces chromosomal instability as a result of a combination of mitotic and apoptotic defects. We suggest that these defects amplify each other to increase the incidence of tetra- and polyploidy in early stages of tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli Protein/deficiency , Apoptosis/physiology , Mitosis/physiology , Polyploidy , Adenomatous Polyposis Coli Protein/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , Cyclin B/metabolism , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , HCT116 Cells , Histones/analysis , Humans , Intestinal Mucosa/metabolism , Intestines/chemistry , Intestines/pathology , Mice , Mice, Transgenic , Mitosis/drug effects , Mitosis/genetics , Models, Biological , Nocodazole/pharmacology , Paclitaxel/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , RNA, Small Interfering/genetics , Spindle Apparatus/metabolism , Staurosporine/pharmacology , beta Catenin/analysis , beta Catenin/metabolism
13.
Mol Biol Cell ; 18(3): 910-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17192415

ABSTRACT

Most sporadic colorectal tumors carry truncation mutations in the adenomatous polyposis coli (APC) gene. The APC protein is involved in many processes that govern gut tissue. In addition to its involvement in the regulation of beta-catenin, APC is a cytoskeletal regulator with direct and indirect effects on microtubules. Cancer-related truncation mutations lack direct and indirect binding sites for microtubules in APC, suggesting that loss of this function contributes to defects in APC-mutant cells. In this study, we show that loss of APC results in disappearance of cellular protrusions and decreased cell migration. These changes are accompanied by a decrease in overall microtubule stability and also by a decrease in posttranslationally modified microtubules in the cell periphery particularly the migrating edge. Consistent with the ability of APC to affect cell shape, the overexpression of APC in cells can induce cellular protrusions. These data demonstrate that cell migration and microtubule stability are linked to APC status, thereby revealing a weakness in APC-deficient cells with potential therapeutic implications.


Subject(s)
Adenomatous Polyposis Coli Protein/deficiency , Cell Movement , Microtubules/metabolism , Acetylation , Adenomatous Polyposis Coli Protein/chemistry , Cell Line, Tumor , Cell Shape , Cell Surface Extensions/metabolism , Fibroblasts/cytology , Humans
14.
J Cell Biol ; 157(6): 1041-8, 2002 Jun 10.
Article in English | MEDLINE | ID: mdl-12058019

ABSTRACT

Loss of full-length adenomatous polyposis coli (APC) protein correlates with the development of colon cancers in familial and sporadic cases. In addition to its role in regulating beta-catenin levels in the Wnt signaling pathway, the APC protein is implicated in regulating cytoskeletal organization. APC stabilizes microtubules in vivo and in vitro, and this may play a role in cell migration (Näthke, I.S., C.L. Adams, P. Polakis, J.H. Sellin, and W.J. Nelson. 1996. J. Cell Biol. 134:165-179; Mimori-Kiyosue, Y., N. Shiina, and S. Tsukita. 2000. J. Cell Biol. 148:505-517; Zumbrunn, J., K. Inoshita, A.A. Hyman, and I.S. Näthke. 2001. Curr. Biol. 11:44-49) and in the attachment of microtubules to kinetochores during mitosis (Fodde, R., J. Kuipers, C. Rosenberg, R. Smits, M. Kielman, C. Gaspar, J.H. van Es, C. Breukel, J. Wiegant, R.H. Giles, and H. Clevers. 2001. Nat. Cell Biol. 3:433-438; Kaplan, K.B., A. Burds, J.R. Swedlow, S.S. Bekir, P.K. Sorger, and I.S. Näthke. 2001. Nat. Cell Biol. 3:429-432). The localization of endogenous APC protein is complex: actin- and microtubule-dependent pools of APC have been identified in cultured cells (Näthke et al., 1996; Mimori-Kiyosue et al., 2000; Reinacher-Schick, A., and B.M. Gumbiner. 2001. J. Cell Biol. 152:491-502; Rosin-Arbesfeld, R., G. Ihrke, and M. Bienz. 2001. EMBO J. 20:5929-5939). However, the localization of APC in tissues has not been identified at high resolution. Here, we show that in fully polarized epithelial cells from the inner ear, endogenous APC protein associates with the plus ends of microtubules located at the basal plasma membrane. Consistent with a role for APC in supporting the cytoskeletal organization of epithelial cells in vivo, the number of microtubules is significantly reduced in apico-basal arrays of microtubule bundles isolated from mice heterozygous for APC.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cochlea/metabolism , Microtubules/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/ultrastructure , Animals , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Polarity , Cochlea/cytology , Cochlea/ultrastructure , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Guinea Pigs , Heterozygote , Humans , Mice , Mice, Mutant Strains , Microtubules/genetics , Microtubules/ultrastructure , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure
15.
Mol Biol Cell ; 17(5): 2331-45, 2006 May.
Article in English | MEDLINE | ID: mdl-16525027

ABSTRACT

In interphase cells, the adenomatous polyposis coli (APC) protein accumulates on a small subset of microtubules (MTs) in cell protrusions, suggesting that APC may regulate the dynamics of these MTs. We comicroinjected a nonperturbing fluorescently labeled monoclonal antibody and labeled tubulin to simultaneously visualize dynamics of endogenous APC and MTs in living cells. MTs decorated with APC spent more time growing and had a decreased catastrophe frequency compared with non-APC-decorated MTs. Endogenous APC associated briefly with shortening MTs. To determine the relationship between APC and its binding partner EB1, we monitored EB1-green fluorescent protein and endogenous APC concomitantly in living cells. Only a small fraction of EB1 colocalized with APC at any one time. APC-deficient cells and EB1 small interfering RNA showed that EB1 and APC localized at MT ends independently. Depletion of EB1 did not change the growth-stabilizing effects of APC on MT plus ends. In addition, APC remained bound to MTs stabilized with low nocodazole, whereas EB1 did not. Thus, we demonstrate that the association of endogenous APC with MT ends correlates directly with their increased growth stability, that this can occur independently of its association with EB1, and that APC and EB1 can associate with MT plus ends by distinct mechanisms.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Surface Extensions/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Adenomatous Polyposis Coli Protein/analysis , Adenomatous Polyposis Coli Protein/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Surface Extensions/chemistry , Dogs , Humans , Mice , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/genetics , Microtubules/chemistry
16.
Mol Biol Cell ; 15(6): 2978-91, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15075372

ABSTRACT

Mutations in the adenomatous polyposis coli (APC) protein occur early in colon cancer and correlate with chromosomal instability. Here, we show that depletion of APC from cystostatic factor (CSF) Xenopus extracts leads to a decrease in microtubule density and changes in tubulin distribution in spindles and asters formed in such extracts. Addition of full-length APC protein or a large, N-terminally truncated APC fragment to APC-depleted extracts restored normal spindle morphology and the intact microtubule-binding site of APC was necessary for this rescue. These data indicate that the APC protein plays a role in the formation of spindles that is dependent on its effect on microtubules. Spindles formed in cycled extracts were not sensitive to APC depletion. In CSF extracts, spindles predominantly formed from aster-like intermediates, whereas in cycled extracts chromatin was the major site of initial microtubule polymerization. These data suggest that APC is important for centrosomally driven spindle formation, which was confirmed by our finding that APC depletion reduced the size of asters nucleated from isolated centrosomes. We propose that lack of microtubule binding in cancer-associated mutations of APC may contribute to defects in the assembly of mitotic spindles and lead to missegregation of chromosomes.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Biological Factors/metabolism , Cell Extracts/chemistry , Spindle Apparatus/metabolism , Xenopus/metabolism , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Animals , Meiosis , Microtubules/metabolism , Mutation , Protein Binding , Tubulin/metabolism
17.
Cancer Res ; 65(12): 5195-204, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958564

ABSTRACT

Truncation mutations in the adenomatous polyposis coli (APC) gene are responsible for familial and sporadic colorectal cancer. APC is a large, multifunctional protein involved in cell migration, proliferation, and differentiation. Dominant effects that have been attributed to the NH2-terminal fragments of APC expressed in tumors may result from loss of functions due to lack of COOH-terminal regions or gain of functions due to fewer regulatory interactions. Resolving this issue and determining how structural changes contribute to the multiple functions of the APC protein requires knowledge about the structural organization of the APC molecule. To this end, we used limited proteolysis to distinguish regions of the molecule with limited structure from those that form well-folded domains. We discovered that the NH2-terminal region of APC was most resistant to proteolytic degradation, whereas middle and COOH-terminal regions were significantly more sensitive. Binding of APC to microtubules protected COOH-terminal regions of APC against proteolysis, consistent with the idea that this region of the molecule becomes ordered when bound to microtubules. Furthermore, interactions between the NH2- and COOH-terminal domains of APC were identified in vitro and in vivo, suggesting that NH2-terminal fragments of APC may be regulated by interactions with COOH-terminal domains. Indeed, expressing COOH-terminal APC fragments in tumor cells resulted in changes in the protein interactions of endogenous NH2-terminal fragments in these cells. Thus, the dominant function of NH2-terminal APC fragments found in tumor cells could be explained by loss of this regulation in tumors where COOH-terminal domains are missing.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Peptide Fragments/metabolism , Cell Line, Tumor , HeLa Cells , Humans , Immunoprecipitation , Microtubules/metabolism , Peptide Hydrolases/metabolism , Phosphorylation , Protein Conformation , Protein Structure, Tertiary
18.
Biotechniques ; 41(2): 199-208, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16925022

ABSTRACT

Biomedical research and drug development increasingly involve the extraction of quantitative data from digital microscope images, such as those obtained using fluorescence microscopy. Here, we describe a novel approach for both managing and analyzing such images. The Open Microscopy Environment (OME) is a sophisticated open-source scientific image management database that coordinates the organization, storage, and analysis of the large volumes of image data typically generated by modern imaging methods. We describe FindSpots, a powerful image-analysis package integrated in OME that will be of use to those who wish to identify and measure objects within microscope images or time-lapse movies. The algorithm used in FindSpots is in fact only one of many possible segmentation (object detection) algorithms, and the underlying data model used by OME to capture and store its results can also be used to store results from other segmentation algorithms. In this report, we illustrate how image segmentation can be achieved in OME using one such implementation of a segmentation algorithm, and how this output subsequently can be displayed graphically or processed numerically using a spreadsheet.


Subject(s)
Computational Biology , Image Interpretation, Computer-Assisted , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Microscopy, Fluorescence , Algorithms , Animals , Database Management Systems , Databases, Factual , Fibroblasts/metabolism , Fluorescent Dyes , Indoles , Information Storage and Retrieval , Kinetochores/metabolism , Mice , Protein Kinases/analysis , Protein Serine-Threonine Kinases , Reproducibility of Results , User-Computer Interface
20.
Philos Trans R Soc Lond B Biol Sci ; 368(1629): 20130014, 2013.
Article in English | MEDLINE | ID: mdl-24062584

ABSTRACT

Cell and tissue polarity are tightly coupled and are vital for normal tissue homeostasis. Changes in cellular and tissue organization are common to even early stages of disease, particularly cancer. The digestive tract is the site of the second most common cause of cancer deaths in the developed world. Tumours in this tissue arise in an epithelium that has a number of axes of cell and tissue polarity. Changes in cell and tissue polarity in response to genetic changes that are known to underpin disease progression provide clues about the link between molecular-, cellular- and tissue-based mechanisms that accompany cancer. Mutations in adenomatous polyposis coli (APC) are common to most colorectal cancers in humans and are sufficient to cause tumours in mouse intestine. Tissue organoids mimic many features of whole tissue and permit identifying changes at different times after inactivation of APC. Using gut organoids, we show that tissue polarity is lost very early during cancer progression, whereas cell polarity, at least apical-basal polarity, is maintained and changes only at later stages. These observations reflect the situation in tumours and validate tissue organoids as a useful system to investigate the relationship between cell polarity and tissue organization.


Subject(s)
Adenomatous Polyposis Coli/physiopathology , Carcinogenesis/pathology , Cell Polarity/physiology , Gastrointestinal Tract/cytology , Organoids/cytology , Animals , Cell Adhesion/physiology , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Mice , Mice, Inbred C57BL , Organoids/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL