Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cancer Causes Control ; 34(12): 1123-1132, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37505316

RESUMEN

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.


Asunto(s)
Neoplasias , Investigadores , Humanos , Investigadores/educación , Proyectos de Investigación , Neoplasias/terapia , Relaciones Comunidad-Institución
2.
Biochemistry ; 59(41): 4039-4050, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32941008

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/química , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , beta Catenina/metabolismo , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Sitios de Unión , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Mutación/genética , Fosforilación , Unión Proteica , beta Catenina/química , beta Catenina/genética
3.
Exp Physiol ; 105(12): 2154-2167, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33150708

RESUMEN

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.


Asunto(s)
Poliposis Adenomatosa del Colon/patología , Colon/patología , Células Caliciformes/patología , Inflamación/patología , Poliposis Adenomatosa del Colon/metabolismo , Animales , Células Cultivadas , Colon/metabolismo , Chaperón BiP del Retículo Endoplásmico , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Células Caliciformes/metabolismo , Humanos , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mucina 2/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Transducción de Señal/fisiología
4.
J Cell Sci ; 130(4): 805-813, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28082422

RESUMEN

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.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Eliminación de Gen , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Animales , Recuento de Células , Proliferación Celular , Pólipos del Colon/metabolismo , Pólipos del Colon/patología , Modelos Animales de Enfermedad , Epitelio/metabolismo , Epitelio/patología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores Notch/metabolismo , Células Madre/metabolismo , Vía de Señalización Wnt
5.
BMC Cancer ; 18(1): 809, 2018 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-30097032

RESUMEN

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.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Gosipol/análogos & derivados , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Productos Biológicos/administración & dosificación , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Gosipol/administración & dosificación , Humanos , Liposomas/administración & dosificación , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Carcinogenesis ; 35(8): 1881-90, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24894865

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/patología , Colitis/complicaciones , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Inflamación/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Apoptosis , Azoximetano/toxicidad , Western Blotting , Carcinógenos/toxicidad , Núcleo Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Colitis/inducido químicamente , Colitis/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Ciclooxigenasa 2 , Sulfato de Dextran/toxicidad , Inflamación/etiología , Inflamación/metabolismo , Ratones , Mutación/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , beta Catenina/genética , beta Catenina/metabolismo
7.
Biochim Biophys Acta ; 1836(1): 80-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23333833

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Poliposis Adenomatosa del Colon/etiología , Modelos Animales de Enfermedad , Poliposis Adenomatosa del Colon/patología , Animales , Genotipo , Humanos , Ratones , Fenotipo , Ratas
10.
J Biol Chem ; 286(7): 4946-50, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21199875

RESUMEN

Loss of tumor suppressor adenomatous polyposis coli (APC) is thought to initiate the majority of all colorectal cancers. The predominant theory of colorectal carcinogenesis implicates stem cells as the initiating cells. However, relatively little is known about the function of APC in governing the homeostasis of normal intestinal stem cells. Here, we identify a novel double-negative feedback loop between APC and a translation inhibitor protein, Musashi1 (MSI1), in cultured human colonocytes. We show APC as a key factor in MSI1 regulation through Wnt signaling and identify APC mRNA as a novel target of translational inhibition by MSI1. We propose that APC/MSI1 interactions maintain homeostatic balance in the intestinal epithelium.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Colon/metabolismo , Mucosa Intestinal/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Biosíntesis de Proteínas/fisiología , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/fisiología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Colon/citología , Humanos , Mucosa Intestinal/citología , Ratones , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
11.
Mol Biol Cell ; 32(1): 28-44, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33175598

RESUMEN

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.


Asunto(s)
Crecimiento y Desarrollo , Homeostasis , Intestinos/crecimiento & desarrollo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas Cdc20/metabolismo , Proliferación Celular , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Transgénicos , Microvellosidades/metabolismo , Modelos Animales , Especificidad de Órganos , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Serrate-Jagged/metabolismo , Transgenes , Regulación hacia Arriba
12.
Sci Rep ; 10(1): 2957, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32076059

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Complejo de Señalización de la Axina/metabolismo , Células Epiteliales/metabolismo , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína Axina/metabolismo , Colon/citología , Células HCT116 , Humanos , Mutación , Vía de Señalización Wnt
13.
Cancers (Basel) ; 12(8)2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32751567

RESUMEN

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.

14.
J Cell Biol ; 219(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32597939

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/metabolismo , Microtúbulos/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Movimiento Celular/fisiología , Células Cultivadas , Conos de Crecimiento/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Cancers (Basel) ; 12(8)2020 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-32784494

RESUMEN

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.

16.
BMC Cell Biol ; 10: 75, 2009 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-19845967

RESUMEN

BACKGROUND: As a key player in suppression of colon tumorigenesis, Adenomatous Polyposis Coli (APC) has been widely studied to determine its cellular functions. However, inconsistencies of commercially available APC antibodies have limited the exploration of APC function. APC is implicated in spindle formation by direct interactions with tubulin and microtubule-binding protein EB1. APC also interacts with the actin cytoskeleton to regulate cell polarity. Until now, interaction of APC with the third cytoskeletal element, intermediate filaments, has remained unexamined. RESULTS: We generated an APC antibody (APC-M2 pAb) raised against the 15 amino acid repeat region, and verified its reliability in applications including immunoprecipitation, immunoblotting, and immunofluorescence in cultured cells and tissue. Utilizing this APC-M2 pAb, we immunoprecipitated endogenous APC and its binding proteins from colon epithelial cells expressing wild-type APC. Using Liquid Chromatography Tandem Mass Spectrometry (LC-MS/MS), we identified 42 proteins in complex with APC, including beta-catenin and intermediate filament (IF) proteins lamin B1 and keratin 81. Association of lamin B1 with APC in cultured cells and human colonic tissue was verified by co-immunoprecipitation and colocalization. APC also colocalized with keratins and remained associated with IF proteins throughout a sequential extraction procedure. CONCLUSION: We introduce a versatile APC antibody that is useful for cell/tissue immunostaining, immunoblotting and immunoprecipitation. We also present evidence for interactions between APC and IFs, independent of actin filaments and microtubules. Our results suggest that APC associates with all three major components of the cytoskeleton, thus expanding potential roles for APC in the regulation of cytoskeletal integrity.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Anticuerpos/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Anticuerpos/inmunología , Línea Celular , Cromatografía Líquida de Alta Presión , Colon/metabolismo , Colon/patología , Células Epiteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Inmunoprecipitación , Proteínas de Filamentos Intermediarios/análisis , Queratinas/análisis , Queratinas/metabolismo , Lamina Tipo B/análisis , Lamina Tipo B/metabolismo , Interferencia de ARN , Espectrometría de Masas en Tándem , beta Catenina/metabolismo
17.
Adv Exp Med Biol ; 656: 13-29, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19928349

RESUMEN

Mutational inactivation of the tumor suppressor gene APC (Adenomatous polyposis coli) is thought to be an initiating step in the progression of the vast majority ofcolorectal cancers. Attempts to understand APC function have revealed more than a dozen binding partners as well as several subcellular localizations including at cell-cell junctions, associated with microtubules at the leading edge of migrating cells, at the apical membrane, in the cytoplasm and in the nucleus. The present chapter focuses on APC localization and functions in the nucleus. APC contains two classical nuclear localization signals, with a third domain that can enhance nuclear import. Along with two sets of nuclear export signals, the nuclear localization signals enable the large APC protein to shuttle between the nucleus and cytoplasm. Nuclear APC can oppose beta-catenin-mediated transcription. This down-regulation of nuclear beta-catenin activity by APC most likely involves nuclear sequestration of beta-catenin from the transcription complex as well as interaction of APC with transcription corepressor CtBP. Additional nuclear binding partners for APC include transcription factor activator protein AP-2alpha, nuclear export factor Crm1, protein tyrosine phosphatase PTP-BL and perhaps DNA itself. Interaction of APC with polymerase beta and PCNA, suggests a role for APC in DNA repair. The observation that increases in the cytoplasmic distribution of APC correlate with colon cancer progression suggests that disruption of these nuclear functions of APC plays an important role in cancer progression. APC prevalence in the cytoplasm of quiescent cells points to a potential function for nuclear APC in control of cell proliferation. Clear definition of APC's nuclear function(s) will expand the possibilities for early colorectal cancer diagnostics and therapeutics targeted to APC.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Núcleo Celular/metabolismo , Transporte Activo de Núcleo Celular , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Citoplasma/metabolismo , Genes APC , Humanos , Señales de Exportación Nuclear , Proteínas Nucleares/metabolismo , Fosforilación , beta Catenina/metabolismo
19.
Mol Biol Cell ; 28(5): 587-599, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28057765

RESUMEN

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.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Dineínas Citoplasmáticas/metabolismo , Insulina/metabolismo , Animales , Línea Celular , Citoplasma/metabolismo , Complicaciones de la Diabetes/metabolismo , Femenino , Genes APC/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Unión Proteica , Transducción de Señal , beta Catenina/metabolismo
20.
Oncotarget ; 6(14): 12558-73, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25940441

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Animales , Western Blotting , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Genes Supresores de Tumor , Células HCT116 , Xenoinjertos , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA