Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cancer Causes Control ; 34(12): 1123-1132, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37505316

ABSTRACT

PURPOSE: There is an increasing awareness of the importance of patient engagement in cancer research, but many basic and translational researchers have never been trained to do so. To address this unmet need, a 1-year patient engagement training program for researchers was developed. METHODS: Eleven researchers and eleven paired research advocates participated. This program, designed for virtual delivery, included 3 didactic modules focused on (1) Community Outreach and Engagement principles and methods, (2) Communication skills, and (3) Team Science. This was followed by longitudinal projects to be completed by the researcher/advocate pairs, including learning about the research project, and co-authoring abstracts, manuscripts and grant proposals. Monthly group meetings allowed pairs to share their experiences. The program culminated in the pairs creating and presenting oral abstracts for the University of Kansas Cancer Center's Annual Research Symposium. RESULTS: All participants indicated that the modules had a positive impact on their ability to collaborate in research. Both researcher self-evaluations and patient advocate evaluations of their researcher partner showed an improvement in researcher communication competency. Results from the Patient Engagement in Research Scale showed that advocates were highly engaged. Within 1 year after program completion, participating pairs have completed four abstracts and 9 grant proposals. CONCLUSION: The program will be modified based on participant feedback, and can be adapted for future cohorts if an increased number of sessions per month and shortened program duration are desired. The program's virtual format allows scalability across institutions to potentially benefit large cohorts of researchers.


Subject(s)
Neoplasms , Research Personnel , Humans , Research Personnel/education , Research Design , Neoplasms/therapy , Community-Institutional Relations
3.
Mol Biol Cell ; 32(1): 28-44, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33175598

ABSTRACT

Evolutionarily conserved RNA-binding protein Musashi1 (Msi1) can regulate developmentally relevant genes. Here we report the generation and characterization of a mouse model that allows inducible Msi1 overexpression in a temporal and tissue-specific manner. We show that ubiquitous Msi1 induction in ∼5-wk-old mice delays overall growth, alters organ-to-body proportions, and causes premature death. Msi1-overexpressing mice had shortened intestines, diminished intestinal epithelial cell (IEC) proliferation, and decreased growth of small intestine villi and colon crypts. Although Lgr5-positive intestinal stem cell numbers remained constant in Msi1-overexpressing tissue, an observed reduction in Cdc20 expression provided a potential mechanism underlying the intestinal growth defects. We further demonstrated that Msi1 overexpression affects IEC differentiation in a region-specific manner, with ileum tissue being influenced the most. Ilea of mutant mice displayed increased expression of enterocyte markers, but reduced expression of the goblet cell marker Mucin2 and fewer Paneth cells. A higher hairy and enhancer of split 1:mouse atonal homolog 1 ratio in ilea from Msi1-overexpressing mice implicated Notch signaling in inducing enterocyte differentiation. Together, this work implicates Msi1 in mouse postnatal development of multiple organs, with Notch signaling alterations contributing to intestinal defects. This new mouse model will be a useful tool to further elucidate the role of Msi1 in other tissue settings.


Subject(s)
Growth and Development , Homeostasis , Intestines/growth & development , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Cdc20 Proteins/metabolism , Cell Proliferation , Intestinal Mucosa/metabolism , Membrane Proteins/metabolism , Mice, Transgenic , Microvilli/metabolism , Models, Animal , Organ Specificity , Receptors, G-Protein-Coupled/metabolism , Serrate-Jagged Proteins/metabolism , Transgenes , Up-Regulation
4.
Exp Physiol ; 105(12): 2154-2167, 2020 12.
Article in English | MEDLINE | ID: mdl-33150708

ABSTRACT

NEW FINDINGS: What is the central question of this study? What is the localization and distribution pattern of adenomatous polyposis coli (APC) in intestinal epithelial cells? Does this distribution change in different regions of the colon or in the condition of inflammation? What is the main finding and its importance? Colonic epithelia from mice and humans contain a subset of goblet cells displaying high APC levels. The number of APChigh goblet cells increases in inflamed tissue, which also displays increased GRP78, indicating potential stress from mucin production. In cultured human colon cells, expression of interleukin 1 pathway components (inducers of MUC2 expression) is reduced upon APC depletion raising the potential for APC participation in an inflammatory response. ABSTRACT: Adenomatous polyposis coli (APC) serves as a gatekeeper of intestinal homeostasis by promoting cellular differentiation and maintaining crypt architecture. Although appreciated as a critical colon tumour suppressor, roles for APC in disease states such as inflammation have yet to be fully delineated. This study aimed to characterize the localization of APC protein in gastrointestinal tissues from human patients with active inflammatory bowel disease and mice with dextran sodium sulfate (DSS)-induced colitis. Fluorescence immunohistochemistry revealed a subset of goblet cells with elevated Apc staining intensity in the small intestines and proximal/medial colons of mice. Upon induction of colitis with DSS, these 'APChigh ' goblet cells remained in the proximal and medial colon, but now were also observed in the distal colon. This phenotype was recapitulated in humans, with APChigh goblet cells observed only in the descending colons of patients with active ulcerative colitis. In cultured human colon cells derived from normal tissue, APC depletion reduced expression of mRNAs encoding the interleukin 1 (IL1) signalling pathway components IL1ß and interleukin-1 receptor (IL1R), known regulators of Muc2 expression. Treating cancer cells lacking wild-type APC with IL1ß, or induction of full-length APC in these cells led to increases in IL1R and MUC2 expression. Combining IL1ß treatment with APC induction led to an increase of MUC2 expression greater than expected for additive affects, suggesting that APC sensitizes cells to IL1 signalling. These findings suggest that APC has novel roles in maintaining proper goblet cell function, thus providing further evidence for APC as an important factor in intestinal tissue homeostasis and disease.


Subject(s)
Adenomatous Polyposis Coli/pathology , Colon/pathology , Goblet Cells/pathology , Inflammation/pathology , Adenomatous Polyposis Coli/metabolism , Animals , Cells, Cultured , Colon/metabolism , Endoplasmic Reticulum Chaperone BiP , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Goblet Cells/metabolism , Humans , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mucin-2/metabolism , Receptors, Interleukin-1 Type I/metabolism , Signal Transduction/physiology
5.
Biochemistry ; 59(41): 4039-4050, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32941008

ABSTRACT

The tumor suppressor Adenomatous polyposis coli (APC) is a large, multidomain protein with many identified cellular functions. The best characterized role of APC is to scaffold a protein complex that negatively regulates Wnt signaling via ß-catenin destruction. This destruction is mediated by ß-catenin binding to centrally located 15- and 20-amino acid repeat regions of APC. More than 80% of cancers of the colon and rectum present with an APC mutation. Most carcinomas with mutant APC express a truncated APC protein that retains the ∼200-amino acid long' 15-amino acid repeat region'. This study demonstrates that the 15-amino acid repeat region of APC is intrinsically disordered. We investigated the backbone dynamics in the presence of ß-catenin and predicted residues that may contribute to transient secondary features. This study reveals that the 15-amino acid region of APC retains flexibility upon binding ß-catenin and that APC does not have a single, observable "highest-affinity" binding site for ß-catenin. This flexibility potentially allows ß-catenin to be more readily captured by APC and then remain accessible to other elements of the destruction complex for subsequent processing.


Subject(s)
Adenomatous Polyposis Coli Protein/chemistry , Adenomatous Polyposis Coli Protein/metabolism , beta Catenin/metabolism , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli Protein/genetics , Binding Sites , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Mutation/genetics , Phosphorylation , Protein Binding , beta Catenin/chemistry , beta Catenin/genetics
6.
Cancers (Basel) ; 12(8)2020 Aug 08.
Article in English | MEDLINE | ID: mdl-32784494

ABSTRACT

RNA-binding protein Musashi-1 (MSI1) is a key regulator of several stem cell populations. MSI1 is involved in tumor proliferation and maintenance, and it regulates target mRNAs at the translational level. The known mRNA targets of MSI1 include Numb, APC, and P21WAF-1, key regulators of Notch/Wnt signaling and cell cycle progression, respectively. In this study, we aim to identify small molecule inhibitors of MSI1-mRNA interactions, which could block the growth of cancer cells with high levels of MSI1. Using a fluorescence polarization (FP) assay, we screened small molecules from several chemical libraries for those that disrupt the binding of MSI1 to its consensus RNA. One cluster of hit compounds is the derivatives of secondary metabolites from Aspergillus nidulans. One of the top hits, Aza-9, from this cluster was further validated by surface plasmon resonance and nuclear magnetic resonance spectroscopy, which demonstrated that Aza-9 binds directly to MSI1, and the binding is at the RNA binding pocket. We also show that Aza-9 binds to Musashi-2 (MSI2) as well. To test whether Aza-9 has anti-cancer potential, we used liposomes to facilitate Aza-9 cellular uptake. Aza-9-liposome inhibits proliferation, induces apoptosis and autophagy, and down-regulates Notch and Wnt signaling in colon cancer cell lines. In conclusion, we identified a series of potential lead compounds for inhibiting MSI1/2 function, while establishing a framework for identifying small molecule inhibitors of RNA binding proteins using FP-based screening methodology.

7.
Cancers (Basel) ; 12(8)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751567

ABSTRACT

The Wnt/ß-catenin signaling pathway is deregulated in nearly all colorectal cancers (CRCs), predominantly through mutation of the tumor suppressor Adenomatous Polyposis Coli (APC). APC mutation is thought to allow a "just-right" amount of Wnt pathway activation by fine-tuning ß-catenin levels. While at a much lower frequency, mutations that result in a ß-catenin that is compromised for degradation occur in a subset of human CRCs. Here, we investigate whether one such "stabilized" ß-catenin responds to regulatory stimuli, thus allowing ß-catenin levels conducive for tumor formation. We utilize cells harboring a single mutant allele encoding Ser45-deleted ß-catenin (ß-catΔS45) to test the effects of Wnt3a treatment or APC-depletion on ß-catΔS45 regulation and activity. We find that APC and ß-catΔS45 retain interaction with Wnt receptors. Unexpectedly, ß-catΔS45 accumulates and activates TOPflash reporter upon Wnt treatment or APC-depletion, but only accumulates in the nucleus upon APC loss. Finally, we find that ß-catenin phosphorylation at GSK-3ß sites and proteasomal degradation continue to occur in the absence of Ser45. Our results expand the current understanding of Wnt/ß-catenin signaling and provide an example of a ß-catenin mutation that maintains some ability to respond to Wnt, a possible key to establishing ß-catenin activity that is "just-right" for tumorigenesis.

8.
J Cell Biol ; 219(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32597939

ABSTRACT

Cell migration is driven by pushing and pulling activities of the actin cytoskeleton, but migration directionality is largely controlled by microtubules. This function of microtubules is especially critical for neuron navigation. However, the underlying mechanisms are poorly understood. Here we show that branched actin filament networks, the main pushing machinery in cells, grow directly from microtubule tips toward the leading edge in growth cones of hippocampal neurons. Adenomatous polyposis coli (APC), a protein with both tumor suppressor and cytoskeletal functions, concentrates at the microtubule-branched network interface, whereas APC knockdown nearly eliminates branched actin in growth cones and prevents growth cone recovery after repellent-induced collapse. Conversely, encounters of dynamic APC-positive microtubule tips with the cell edge induce local actin-rich protrusions. Together, we reveal a novel mechanism of cell navigation involving APC-dependent assembly of branched actin networks on microtubule tips.


Subject(s)
Actins/metabolism , Adenomatous Polyposis Coli Protein/metabolism , Adenomatous Polyposis Coli/metabolism , Microtubules/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Movement/physiology , Cells, Cultured , Growth Cones/metabolism , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Rats , Rats, Sprague-Dawley
9.
Sci Rep ; 10(1): 2957, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32076059

ABSTRACT

Wnt/ß-catenin signaling is essential for intestinal homeostasis and is aberrantly activated in most colorectal cancers (CRC) through mutation of the tumor suppressor Adenomatous Polyposis Coli (APC). APC is an essential component of a cytoplasmic protein complex that targets ß-catenin for destruction. Following Wnt ligand presentation, this complex is inhibited. However, a role for APC in this inhibition has not been shown. Here, we utilized Wnt3a-beads to locally activate Wnt co-receptors. In response, the endogenous ß-catenin destruction complex reoriented toward the local Wnt cue in CRC cells with full-length APC, but not if APC was truncated or depleted. Non-transformed human colon epithelial cells displayed similar Wnt-induced destruction complex localization which appeared to be dependent on APC and less so on Axin. Our results expand the current model of Wnt/ß-catenin signaling such that in response to Wnt, the ß-catenin destruction complex: (1) maintains composition and binding to ß-catenin, (2) moves toward the plasma membrane, and (3) requires full-length APC for this relocalization.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Axin Signaling Complex/metabolism , Epithelial Cells/metabolism , Wnt3A Protein/metabolism , beta Catenin/metabolism , Adenomatous Polyposis Coli Protein/genetics , Axin Protein/metabolism , Colon/cytology , HCT116 Cells , Humans , Mutation , Wnt Signaling Pathway
10.
BMC Cancer ; 18(1): 809, 2018 Aug 10.
Article in English | MEDLINE | ID: mdl-30097032

ABSTRACT

BACKGROUND: The Musashi (MSI) family of RNA-binding proteins is best known for the role in post-transcriptional regulation of target mRNAs. Elevated MSI1 levels in a variety of human cancer are associated with up-regulation of Notch/Wnt signaling. MSI1 binds to and negatively regulates translation of Numb and APC (adenomatous polyposis coli), negative regulators of Notch and Wnt signaling respectively. METHODS: Previously, we have shown that the natural product (-)-gossypol as the first known small molecule inhibitor of MSI1 that down-regulates Notch/Wnt signaling and inhibits tumor xenograft growth in vivo. Using a fluorescence polarization (FP) competition assay, we identified gossypolone (Gn) with a > 20-fold increase in Ki value compared to (-)-gossypol. We validated Gn binding to MSI1 using surface plasmon resonance, nuclear magnetic resonance, and cellular thermal shift assay, and tested the effects of Gn on colon cancer cells and colon cancer DLD-1 xenografts in nude mice. RESULTS: In colon cancer cells, Gn reduced Notch/Wnt signaling and induced apoptosis. Compared to (-)-gossypol, the same concentration of Gn is less active in all the cell assays tested. To increase Gn bioavailability, we used PEGylated liposomes in our in vivo studies. Gn-lip via tail vein injection inhibited the growth of human colon cancer DLD-1 xenografts in nude mice, as compared to the untreated control (P < 0.01, n = 10). CONCLUSION: Our data suggest that PEGylation improved the bioavailability of Gn as well as achieved tumor-targeted delivery and controlled release of Gn, which enhanced its overall biocompatibility and drug efficacy in vivo. This provides proof of concept for the development of Gn-lip as a molecular therapy for colon cancer with MSI1/MSI2 overexpression.


Subject(s)
Colonic Neoplasms/drug therapy , Gossypol/analogs & derivatives , Nerve Tissue Proteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Biological Products/administration & dosage , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gossypol/administration & dosage , Humans , Liposomes/administration & dosage , Mice , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
13.
J Cell Sci ; 130(4): 805-813, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28082422

ABSTRACT

Therapeutic strategies based on a specific oncogenic target are better justified when elimination of that particular oncogene reduces tumorigenesis in a model organism. One such oncogene, Musashi-1 (Msi-1), regulates translation of target mRNAs and is implicated in promoting tumorigenesis in the colon and other tissues. Msi-1 targets include the tumor suppressor adenomatous polyposis coli (Apc), a Wnt pathway antagonist lost in ∼80% of all colorectal cancers. Cell culture experiments have established that Msi-1 is a Wnt target, thus positioning Msi-1 and Apc as mutual antagonists in a mutually repressive feedback loop. Here, we report that intestines from mice lacking Msi-1 display aberrant Apc and Msi-1 mutually repressive feedback, reduced Wnt and Notch signaling, decreased proliferation, and changes in stem cell populations, features predicted to suppress tumorigenesis. Indeed, mice with germline Apc mutations (ApcMin ) or with the Apc1322T truncation mutation have a dramatic reduction in intestinal polyp number when Msi-1 is deleted. Taken together, these results provide genetic evidence that Msi-1 contributes to intestinal tumorigenesis driven by Apc loss, and validate the pursuit of Msi-1 inhibitors as chemo-prevention agents to reduce tumor burden.


Subject(s)
Adenomatous Polyposis Coli/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Gene Deletion , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Animals , Cell Count , Cell Proliferation , Colonic Polyps/metabolism , Colonic Polyps/pathology , Disease Models, Animal , Epithelium/metabolism , Epithelium/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway
14.
Mol Biol Cell ; 28(5): 587-599, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28057765

ABSTRACT

Diabetes is linked to an increased risk for colorectal cancer, but the mechanistic underpinnings of this clinically important effect are unclear. Here we describe an interaction between the microtubule motor cytoplasmic dynein, the adenomatous polyposis coli tumor suppressor protein (APC), and glycogen synthase kinase-3ß (GSK-3ß), which could shed light on this issue. GSK-3ß is perhaps best known for glycogen regulation, being inhibited downstream in an insulin-signaling pathway. However, the kinase is also important in many other processes. Mutations in APC that disrupt the regulation of ß-catenin by GSK-3ß cause colorectal cancer in humans. Of interest, both APC and GSK-3ß interact with microtubules and cellular membranes. We recently demonstrated that dynein is a GSK-3ß substrate and that inhibition of GSK-3ß promotes dynein-dependent transport. We now report that dynein stimulation in intestinal cells in response to acute insulin exposure (or GSK-3ß inhibition) is blocked by tumor-promoting isoforms of APC that reduce an interaction between wild-type APC and dynein. We propose that under normal conditions, insulin decreases dynein binding to APC to stimulate minus end-directed transport, which could modulate endocytic and secretory systems in intestinal cells. Mutations in APC likely impair the ability to respond appropriately to insulin signaling. This is exciting because it has the potential to be a contributing factor in the development of colorectal cancer in patients with diabetes.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Colorectal Neoplasms/metabolism , Cytoplasmic Dyneins/metabolism , Insulin/metabolism , Animals , Cell Line , Cytoplasm/metabolism , Diabetes Complications/metabolism , Female , Genes, APC/physiology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Protein Binding , Signal Transduction , beta Catenin/metabolism
15.
Oncotarget ; 6(14): 12558-73, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25940441

ABSTRACT

Stem cell marker, Musashi-1 (MSI1) is over-expressed in many cancer types; however the molecular mechanisms involved in MSI1 over-expression are not well understood. We investigated the microRNA (miRNA) regulation of MSI1 and the implications this regulation plays in colorectal cancer. MicroRNA miR-137 was identified as a MSI1-targeting microRNA by immunoblotting and luciferase reporter assays. MSI1 protein was found to be highly expressed in 79% of primary rectal tumors (n=146), while miR-137 expression was decreased in 84% of the rectal tumor tissues (n=68) compared to paired normal mucosal samples. In addition to reduced MSI1 protein, exogenous expression of miR-137 inhibited cell growth, colony formation, and tumorsphere growth of colon cancer cells. Finally, in vivo studies demonstrated that induction of miR-137 can decrease growth of human colon cancer xenografts. Our results demonstrate that miR-137 acts as a tumor-suppressive miRNA in colorectal cancers and negatively regulates oncogenic MSI1.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Animals , Blotting, Western , Colorectal Neoplasms/pathology , Disease Progression , Genes, Tumor Suppressor , HCT116 Cells , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Nude , Nerve Tissue Proteins/biosynthesis , RNA-Binding Proteins/biosynthesis , Real-Time Polymerase Chain Reaction , Tissue Array Analysis
16.
Mol Oncol ; 9(7): 1406-20, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25933687

ABSTRACT

Musashi-1 (MSI1) is an RNA-binding protein that acts as a translation activator or repressor of target mRNAs. The best-characterized MSI1 target is Numb mRNA, whose encoded protein negatively regulates Notch signaling. Additional MSI1 targets include the mRNAs for the tumor suppressor protein APC that regulates Wnt signaling and the cyclin-dependent kinase inhibitor P21(WAF-1). We hypothesized that increased expression of NUMB, P21 and APC, through inhibition of MSI1 RNA-binding activity might be an effective way to simultaneously downregulate Wnt and Notch signaling, thus blocking the growth of a broad range of cancer cells. We used a fluorescence polarization assay to screen for small molecules that disrupt the binding of MSI1 to its consensus RNA binding site. One of the top hits was (-)-gossypol (Ki = 476 ± 273 nM), a natural product from cottonseed, known to have potent anti-tumor activity and which has recently completed Phase IIb clinical trials for prostate cancer. Surface plasmon resonance and nuclear magnetic resonance studies demonstrate a direct interaction of (-)-gossypol with the RNA binding pocket of MSI1. We further showed that (-)-gossypol reduces Notch/Wnt signaling in several colon cancer cell lines having high levels of MSI1, with reduced SURVIVIN expression and increased apoptosis/autophagy. Finally, we showed that orally administered (-)-gossypol inhibits colon cancer growth in a mouse xenograft model. Our study identifies (-)-gossypol as a potential small molecule inhibitor of MSI1-RNA interaction, and suggests that inhibition of MSI1's RNA binding activity may be an effective anti-cancer strategy.


Subject(s)
Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Gossypol/pharmacology , Nerve Tissue Proteins/drug effects , RNA-Binding Proteins/drug effects , Animals , Cell Line , Cell Line, Tumor , Colonic Neoplasms/metabolism , Female , Humans , Mice , Mice, Nude , Nerve Tissue Proteins/metabolism , Protein Binding , RNA-Binding Proteins/metabolism , Xenograft Model Antitumor Assays
17.
Carcinogenesis ; 35(8): 1881-90, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24894865

ABSTRACT

Mutation of tumor suppressor adenomatous polyposis coli (APC) initiates most colorectal cancers and chronic colitis increases risk. APC is a nucleo-cytoplasmic shuttling protein, best known for antagonizing Wnt signaling by forming a cytoplasmic complex that marks ß-catenin for degradation. Using our unique mouse model with compromised nuclear Apc import (Apc(mNLS)), we show that Apc(mNLS/mNLS) mice have increased susceptibility to tumorigenesis induced with azoxymethane (AOM) and dextran sodium sulfate (DSS). The AOM-DSS-induced colon adenoma histopathology, proliferation, apoptosis, stem cell number and ß-catenin and Kras mutation spectra were similar in Apc(mNLS/mNLS) and Apc(+/+) mice. However, AOM-DSS-treated Apc(mNLS/mNLS) mice showed more weight loss, more lymphoid follicles and edema, and increased colon shortening than treated Apc(+/+) mice, indicating a colitis predisposition. To test this directly, we induced acute colitis with a 7 day DSS treatment followed by 5 days of recovery. Compared with Apc(+/+) mice, DSS-treated Apc(mNLS/mNLS) mice developed more severe colitis based on clinical grade and histopathology. Apc(mNLS/mNLS) mice also had higher lymphocytic infiltration and reduced expression of stem cell markers, suggesting an increased propensity for chronic inflammation. Moreover, colons from DSS-treated Apc(mNLS/mNLS) mice showed fewer goblet cells and reduced Muc2 expression. Even in untreated Apc(mNLS/mNLS) mice, there were significantly fewer goblet cells in jejuna, and a modest decrease in colonocyte Muc2 expression compared with Apc(+/+) mice. Colonocytes from untreated Apc(mNLS/mNLS) mice also showed increased expression of inflammatory mediators cyclooxygenase-2 (Cox-2) and macrophage inflammatory protein-2 (MIP-2). These findings reveal novel functions for nuclear Apc in goblet cell differentiation and protection against inflammation-induced colon tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/pathology , Colitis/complications , Colorectal Neoplasms/pathology , Disease Models, Animal , Inflammation/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Apoptosis , Azoxymethane/toxicity , Blotting, Western , Carcinogens/toxicity , Cell Nucleus/genetics , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Colitis/chemically induced , Colitis/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Cyclooxygenase 2 , Dextran Sulfate/toxicity , Inflammation/etiology , Inflammation/metabolism , Mice , Mutation/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
18.
Genetics ; 197(4): 1365-76, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24913681

ABSTRACT

Tumorigenicity studies often employ outbred nude mice, in the absence of direct evidence that this mixed genetic background will negatively affect experimental outcome. Here we show that outbred nude mice carry two different alleles of Pla2g2a, a genetic modifier of intestinal tumorigenesis in mice. Here, we identify previous unreported linked polymorphisms in the promoter, noncoding and coding sequences of Pla2g2a and show that outbred nude mice from different commercial providers are heterogeneous for this polymorphic Pla2g2a allele. This heterogeneity even extends to mice obtained from a single commercial provider, which display mixed Pla2g2a genotypes. Notably, we demonstrated that the polymorphic Pla2g2a allele affects orthotopic xenograft establishment of human colon cancer cells in outbred nude mice. This finding establishes a non-cell-autonomous role for Pla2g2a in suppressing intestinal tumorigenesis. Using in vitro reporter assays and pharmacological inhibitors, we show promoter polymorphisms and nonsense-mediated RNA decay (NMD) as underlying mechanisms that lead to low Pla2g2a mRNA levels in tumor-sensitive mice. Together, this study provides mechanistic insight regarding Pla2g2a polymorphisms and demonstrates a non-cell-autonomous role for Pla2g2a in suppressing tumors. Moreover, our direct demonstration that mixed genetic backgrounds of outbred nude mice can significantly affect baseline tumorigenicity cautions against future use of outbred mice for tumor xenograft studies.


Subject(s)
Carcinogenesis/genetics , Group II Phospholipases A2/genetics , Polymorphism, Genetic , Xenograft Model Antitumor Assays/methods , Alleles , Animals , Cloning, Molecular , Genotype , Group II Phospholipases A2/metabolism , HCT116 Cells , Humans , Intestines/pathology , Mice , Mice, Nude , Nonsense Mediated mRNA Decay , Plasmids/genetics , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Cancer Res ; 73(8): 2389-99, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23580574

ABSTRACT

Adenomatous polyposis coli (APC) is best known for its crucial role in colorectal cancer suppression. Rodent models with various Apc mutations have enabled experimental validation of different Apc functions in tumors and normal tissues. Since the development of the first mouse model with a germline Apc mutation in the early 1990s, 20 other Apc mouse and rat models have been generated. This article compares and contrasts currently available Apc rodent models with particular emphasis on providing potential explanations for their reported variation in three areas: (i) intestinal polyp multiplicity, (ii) intestinal polyp distribution, and (iii) extraintestinal phenotypes.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Germ-Line Mutation , Phenotype , Animals , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Genetic Association Studies , Humans , Intestinal Polyps/genetics , Intestinal Polyps/pathology , Mice , Rats , Rodentia
20.
Biochim Biophys Acta ; 1836(1): 80-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23333833

ABSTRACT

Mutation of tumor suppressor gene adenomatous polyposis coli (APC) is an initiating step in most colon cancers. This review summarizes Apc models in mice and rats, with particular concentration on those most recently developed, phenotypic variation among different models, and genotype/phenotype correlations.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Adenomatous Polyposis Coli/etiology , Disease Models, Animal , Adenomatous Polyposis Coli/pathology , Animals , Genotype , Humans , Mice , Phenotype , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...