Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Cell ; 72(1): 37-47.e4, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30197296

RESUMEN

Adenomatous polyposis coli (APC) and Axin are core components of the ß-catenin destruction complex. How APC's function is regulated and whether Wnt signaling influences the direct APC-Axin interaction to inhibit the ß-catenin destruction complex is not clear. Through a CRISPR screen of ß-catenin stability, we have identified ICAT, a polypeptide previously known to block ß-catenin-TCF interaction, as a natural inhibitor of APC. ICAT blocks ß-catenin-APC interaction and prevents ß-catenin-mediated APC-Axin interaction, enhancing stabilization of ß-catenin in cells harboring truncated APC or stimulated with Wnt, but not in cells deprived of a Wnt signal. Using ICAT as a tool to disengage ß-catenin-mediated APC-Axin interaction, we demonstrate that Wnt quickly inhibits the direct interaction between APC and Axin. Our study highlights an important scaffolding function of ß-catenin in the assembly of the destruction complex and suggests Wnt-inhibited APC-Axin interaction as a mechanism of Wnt-dependent inhibition of the destruction complex.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Dominios y Motivos de Interacción de Proteínas/genética , beta Catenina/genética , Proteínas Adaptadoras Transductoras de Señales , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína Axina/genética , Humanos , Estabilidad Proteica , Proteína 1 Similar al Factor de Transcripción 7/genética , Vía de Señalización Wnt/genética
2.
Proc Natl Acad Sci U S A ; 116(41): 20672-20678, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31551264

RESUMEN

Radiation-induced gastrointestinal syndrome (RIGS) is a limiting factor for therapeutic abdominopelvic radiation and is predicted to be a major source of morbidity in the event of a nuclear accident or radiological terrorism. In this study, we developed an in vivo mouse-modeling platform that enables spatial and temporal manipulation of potential RIGS targets in mice following whole-abdomen irradiation without the confounding effects of concomitant hematopoietic syndrome that occur following whole-body irradiation. We then tested the utility of this platform to explore the effects of transient Wnt pathway activation on intestinal regeneration and animal recovery following induction of RIGS. Our results demonstrate that intestinal epithelial suppression of adenomatous polyposis coli (Apc) mitigates RIGS lethality in vivo after lethal ionizing radiation injury-induced intestinal epithelial damage. These results highlight the potential of short-term Wnt agonism as a therapeutic target and establish a platform to evaluate other strategies to stimulate intestinal regeneration after ionizing radiation damage.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Enfermedades Gastrointestinales/prevención & control , Intestinos/citología , Traumatismos Experimentales por Radiación/prevención & control , Regeneración , Irradiación Corporal Total/efectos adversos , Proteínas Wnt/metabolismo , Animales , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/metabolismo , Intestinos/efectos de la radiación , Ratones , ARN Interferente Pequeño , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/metabolismo , Síndrome
3.
Molecules ; 26(4)2021 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-33670371

RESUMEN

Metastasis is the major cause of death in colorectal cancer and it has been proven that inhibiting an interaction between adenomatous polyposis coli (APC) and Rho guanine nucleotide exchange factor 4 (Asef) efficaciously restrain metastasis. However, current inhibitors cannot achieve a satisfying effect in vivo and need to be optimized. In the present study, we applied molecular dynamics (MD) simulations and extensive analyses to apo and holo APC systems in order to reveal the inhibitor mechanism in detail and provide insights into optimization. MD simulations suggested that apo APC takes on a broad array of conformations and inhibitors stabilize conformation selectively. Representative structures in trajectories show specific APC-ligand interactions, explaining the different binding process. The stability and dynamic properties of systems elucidate the inherent factors of the conformation selection mechanism. Binding free energy analysis quantitatively confirms key interface residues and guide optimization. This study elucidates the conformation selection mechanism in APC-Asef inhibition and provides insights into peptide-based drug design.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Neoplasias Colorrectales/tratamiento farmacológico , Péptidos/química , Proteína de la Poliposis Adenomatosa del Colon/química , Proteína de la Poliposis Adenomatosa del Colon/genética , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Ligandos , Simulación de Dinámica Molecular , Metástasis de la Neoplasia , Péptidos/antagonistas & inhibidores , Unión Proteica/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido Rho/química , Factores de Intercambio de Guanina Nucleótido Rho/genética
4.
Bioorg Chem ; 99: 103838, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32334194

RESUMEN

A series of novel pyrazoline derivatives containing methyl-1H-indole moiety were discovered as potential inhibitors for blocking APC-Asef interactions. The top hit Q19 suggested potency of inhibiting APC-Asef interactions and attractive preference for human-sourced colorectal cells. It was already comparable with the previous representative and the positive control Regorafenib before further pharmacokinetic optimization. The introduction of methyl-1H-indole moiety realized the Mitochondrial affection thus might connect the impact on the protein-interaction level with the apoptosis events. The molecular docking simulation inferred that bringing trifluoromethyl groups seemed a promising approach for causing more key interactions such as H-bonds. This work raised referable information for further discovery of inhibitors for blocking APC-Asef interactions.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Antineoplásicos/farmacología , Descubrimiento de Drogas , Indoles/farmacología , Pirazoles/farmacología , Proteína de la Poliposis Adenomatosa del Colon/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indoles/química , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estructura Molecular , Unión Proteica/efectos de los fármacos , Pirazoles/síntesis química , Pirazoles/química , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido Rho/química , Relación Estructura-Actividad
5.
Int J Mol Sci ; 21(24)2020 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-33348563

RESUMEN

Colorectal cancer (CRC) is one of the leading causes of cancer deaths worldwide. The initiation and progression of CRC is a multi-step process that proceeds via precursor lesions to carcinoma, with each stage characterized by its distinct molecular and tissue microenvironment changes. Precursor lesions of CRC, aberrant crypt foci, and adenoma exhibit drastic changes in genetic, transcriptomic, and proteomic profiles compared to normal tissue. The identification of these changes is essential and provides further validation as an initiator or promoter of CRC and, more so, as lesion-specific druggable molecular targets for the precision chemoprevention of CRC. Mutated/dysregulated signaling (adenomatous polyposis coli, ß-catenin, epidermal growth factor receptor, V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS), tumor protein53, Akt, etc.), inflammatory (cyclooxygenase-2, microsomal prostaglandin E synthase-1, inducible nitric oxide synthase, and other pro-inflammatory mediators), and metabolic/growth factor (fatty acid synthase, ß-Hydroxy ß-methylglutaryl-CoA reductase, and ornithine decarboxylase) related targets are some of the well-characterized molecular targets in the precision chemoprevention of CRC. In this review, we discuss precursor-lesion specific targets of CRC and the current status of pre-clinical studies regarding clinical interventions and combinations for better efficacy and safety toward future precision clinical chemoprevention. In addition, we provide a brief discussion on the usefulness of secondary precision chemopreventive targets for tertiary precision chemoprevention to improve the disease-free and overall survival of advanced stage CRC patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/prevención & control , Terapia Molecular Dirigida/métodos , Medicina de Precisión/métodos , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Modelos Animales de Enfermedad , Ácido Graso Sintasas/antagonistas & inhibidores , Ácido Graso Sintasas/metabolismo , Humanos , Ratones
6.
Dev Growth Differ ; 60(1): 76-81, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29266402

RESUMEN

Planarians have become widely recognized as one of the major animal models for regeneration studies in invertebrates. To induce RNA interference (RNAi) by feeding in planarians, the widely accepted protocol is one in which animals undergo two or three feedings of food containing double-stranded RNA (dsRNA) plus visible food coloring (e.g., blood) for confirmation of feeding by individual animals. However, one possible problem is that incorporated food coloring is often retained within the gut for several days, which makes it difficult to confirm the success of each round of dsRNA feeding based on the difference of the color density within the gut before and after feeding. As a consequence, the difference of appetite levels among individuals undergoing dsRNA feeding leads to phenotypic variability among them due to insufficient knockdown. In our attempts to overcome this problem, we have developed a novel method for achieving robust confirmation of the success of dsRNA feeding in individuals fed multiple times by means of including a combination of three different colored chalks (pink, yellow and blue) as food coloring. Notably, we found that this method is superior to the conventional method for positively marking individuals that actively consumed the dsRNA-containing food during four times of once-daily feeding. Using these selected animals, we obtained stable and sufficiently strong RNAi-induced phenotypes. We termed this improved multi-colored chalk-spiked method of feeding RNAi "Candi" and propose its benefits for gene function analysis in planarians.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Carbonato de Calcio/farmacología , Colorantes de Alimentos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Planarias/fisiología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Carbonato de Calcio/química , Sistema Digestivo/metabolismo , Métodos de Alimentación , Colorantes de Alimentos/química , Fenotipo , Planarias/genética , Planarias/crecimiento & desarrollo , ARN Bicatenario , Regeneración
7.
Biochem Biophys Res Commun ; 488(2): 418-424, 2017 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-28506830

RESUMEN

Gut microbiota is critical for maintaining body immune homeostasis and thus affects tumor growth and therapeutic efficiency. Here, we investigated the link between microbiota and tumorgenesis in a mice model of subcutaneous melanoma cell transplantation, and explored the underlying mechanism. We found disruption of gut microbiota by pretreating mice with antibiotics promote tumor growth and remodeling the immune compartment within the primary tumor. Indeed, gut microbial dysbiosis reduced the infiltrated mature antigen-presenting cells of tumor, together with lower levels of co-stimulators, such as CD80, CD86 and MHCII, as well as defective Th1 cytokines, including IFNγ, TNFα, IL12p40, and IL12p35. Meantime, splenic APCs displayed blunted ability in triggering T cell proliferation and IFNγ secretion. However, oral administration of LPS restored the immune surveillance effects and thus inhibited tumor growth in the antibiotics induced gut microbiota dysbiosis group. Taken together, these data highly supported that antibiotics induced gut microbiota dysbiosis promotes tumor initiation, while LPS supplementation would restore the effective immune surveillance and repress tumor initiation.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Antibacterianos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Melanoma/tratamiento farmacológico , Proteína de la Poliposis Adenomatosa del Colon/inmunología , Administración Oral , Animales , Antibacterianos/administración & dosificación , Antibacterianos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal/inmunología , Inyecciones Subcutáneas , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Melanoma/inmunología , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Células TH1/efectos de los fármacos , Células TH1/inmunología
8.
Carcinogenesis ; 34(4): 750-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23239745

RESUMEN

Mitotic progression of mammalian cells is tightly regulated by the E3 ubiquitin ligase anaphase promoting complex (APC)/C. Deregulation of APC/C is frequently observed in cancer cells and is suggested to contribute to chromosome instability and cancer predisposition. In this study, we identified Daxx as a novel APC/C inhibitor frequently overexpressed in prostate cancer. Daxx interacts with the APC/C coactivators Cdc20 and Cdh1 in vivo, with the binding of Cdc20 dependent on the consensus destruction boxes near the N-terminal of the Daxx protein. Ectopic expression of Daxx, but not the D-box deleted mutant (DaxxΔD-box), inhibited the degradation of APC/Cdc20 and APC/Cdh1 substrates, leading to a transient delay in mitotic progression. Daxx is frequently upregulated in prostate cancer tissues; the expression level positively correlated with the Gleason score and disease metastasis (P = 0.027 and 0.032, respectively). Furthermore, ectopic expression of Daxx in a non-malignant prostate epithelial cell line induced polyploidy under mitotic stress. Our data suggest that Daxx may function as a novel APC/C inhibitor, which promotes chromosome instability during prostate cancer development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Cadherinas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica , Mitosis , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Antígenos CD , Proteínas Cdc20 , Ciclo Celular , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proteínas Co-Represoras , Células HEK293 , Células HeLa , Humanos , Masculino , Chaperonas Moleculares , Mutación , Clasificación del Tumor , Metástasis de la Neoplasia , Proteínas Nucleares/genética , Interferencia de ARN , ARN Interferente Pequeño
9.
Carcinogenesis ; 34(2): 277-86, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23129580

RESUMEN

We previously reported that quinacrine (QC) has anticancer activity against breast cancer cells. Here, we examine the mechanism of action of QC and its ability to inhibit Wnt-TCF signaling in two independent breast cancer cell lines. QC altered Wnt-TCF signaling components by increasing the levels of adenomatous polyposis coli (APC), DAB2, GSK-3ß and axin and decreasing the levels of ß-catenin, p-GSK3ß (ser 9) and CK1. QC also reduced the activity of the Wnt transcription factor TCF/LEF and its downstream targets cyclin D1 and c-MYC. Using a luciferase-based Wnt-TCF transcription factor assay, it was shown that APC levels were inversely associated with TCF/LEF activity. Induction of apoptosis and DNA damage was observed after treatment with QC, which was associated with increased expression of APC. The effects induced by QC depend on APC because the inhibition of Wnt-TCF signaling by QC is lost in APC-knockdown cells, and consequently, the extent of apoptosis and DNA damage caused by QC is reduced compared with parental cells. Because we previously showed that QC inhibits topoisomerase, we examined the effect of another topoisomerase inhibitor, etoposide, on Wnt signaling. Interestingly, etoposide treatment also reduced TCF/LEF activity, ß-catenin and cyclin D1 levels commensurate with induction of DNA damage and apoptosis. Lycopene, a plant-derived antioxidant, synergistically increased QC activity and inhibited Wnt-TCF signaling in cancer cells without affecting the MCF-10A normal breast cell line. Collectively, the data suggest that QC-mediated Wnt-TCF signal inhibition depends on APC and that the addition of lycopene synergistically increases QC anticancer activity.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Carotenoides/farmacología , Quinacrina/farmacología , Transducción de Señal/efectos de los fármacos , Factor 1 de Transcripción de Linfocitos T/antagonistas & inhibidores , Proteínas Wnt/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/genética , Anticarcinógenos/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular , Proliferación Celular , Ensayo Cometa , Ciclina D1/metabolismo , Sinergismo Farmacológico , Etopósido/farmacología , Femenino , Citometría de Flujo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Licopeno , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Factor 1 de Transcripción de Linfocitos T/metabolismo , Factores de Transcripción TCF , Transactivadores/metabolismo , Transcripción Genética , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
10.
Proc Natl Acad Sci U S A ; 107(43): 18517-22, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20937854

RESUMEN

Microtubules (MTs) contribute to key processes during cell motility, including the regulation of focal adhesion turnover and the establishment and maintenance of cell orientation. It was previously demonstrated that the ErbB2 receptor tyrosine kinase regulated MT outgrowth to the cell cortex via a complex including Memo, the GTPase RhoA, and the formin mDia1. But the mechanism that linked this signaling module to MTs remained undefined. We report that ErbB2-induced repression of glycogen synthase kinase-3 (GSK3) activity, mediated by Memo and mDia1, is required for MT capture and stabilization. Memo-dependent inhibition of GSK3 allows the relocalization of APC (adenomatous polyposis coli) and cytoplasmic linker-associated protein 2 (CLASP2), known MT-associated proteins, to the plasma membrane and ruffles. Peripheral microtubule extension also requires expression of the plus-end binding protein EB1 and its recently described interactor, the spectraplakin ACF7. In fact, in migrating cells, ACF7 localizes to the plasma membrane and ruffles, in a Memo-, GSK3-, and APC-dependent manner. Finally, we demonstrate that ACF7 targeting to the plasma membrane is both required and sufficient for MT capture downstream of ErbB2. This function of ACF7 does not require its recently described ATPase activity. By defining the signaling pathway by which ErbB2 allows MT capture and stabilization at the cell leading edge, we provide insights into the mechanism underlying cell motility and steering.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Microfilamentos/metabolismo , Microtúbulos/metabolismo , Receptor ErbB-2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Membrana Celular/metabolismo , Femenino , Adhesiones Focales/metabolismo , Forminas , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Microfilamentos/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Proteínas de Hierro no Heme/antagonistas & inhibidores , Proteínas de Hierro no Heme/genética , Proteínas de Hierro no Heme/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal
11.
PLoS Biol ; 6(3): e50, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18318601

RESUMEN

In response to misaligned sister chromatids during mitosis, the spindle checkpoint protein Mad2 inhibits the anaphase-promoting complex or cyclosome (APC/C) through binding to its mitotic activator Cdc20, thus delaying anaphase onset. Mad1, an upstream regulator of Mad2, forms a tight core complex with Mad2 and facilitates Mad2 binding to Cdc20. In the absence of its binding proteins, free Mad2 has two natively folded conformers, termed N1-Mad2/open-Mad2 (O-Mad2) and N2-Mad2/closed Mad2 (C-Mad2), with C-Mad2 being more active in APC/C(Cdc20) inhibition. Here, we show that whereas O-Mad2 is monomeric, C-Mad2 forms either symmetric C-Mad2-C-Mad2 (C-C) or asymmetric O-Mad2-C-Mad2 (O-C) dimers. We also report the crystal structure of the symmetric C-C Mad2 dimer, revealing the basis for the ability of unliganded C-Mad2, but not O-Mad2 or liganded C-Mad2, to form symmetric dimers. A Mad2 mutant that predominantly forms the C-C dimer is functional in vitro and in living cells. Finally, the Mad1-Mad2 core complex facilitates the conversion of O-Mad2 to C-Mad2 in vitro. Collectively, our results establish the existence of a symmetric Mad2 dimer and provide insights into Mad1-assisted conformational activation of Mad2 in the spindle checkpoint.


Asunto(s)
Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Huso Acromático/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas Cdc20 , Proteínas de Ciclo Celular/genética , Cristalografía por Rayos X , Dimerización , Células HeLa , Humanos , Proteínas Mad2 , Modelos Biológicos , Modelos Moleculares , Conformación Molecular , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Conformación Proteica , Proteínas Represoras/genética , Relación Estructura-Actividad
12.
PLoS One ; 16(8): e0255738, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34370741

RESUMEN

Adenomatous Polyposis Coli (APC) is lost in approximately 70% of sporadic breast cancers, with an inclination towards triple negative breast cancer (TNBC). TNBC is treated with traditional chemotherapy, such as paclitaxel (PTX); however, tumors often develop drug resistance. We previously created APC knockdown cells (APC shRNA1) using the human TNBC cells, MDA-MB-157, and showed that APC loss induces PTX resistance. To understand the mechanisms behind APC-mediated PTX response, we performed cell cycle analysis and analyzed cell cycle related proteins. Cell cycle analysis indicated increased G2/M population in both PTX-treated APC shRNA1 and parental cells, suggesting that APC expression does not alter PTX-induced G2/M arrest. We further studied the subcellular localization of the G2/M transition proteins, cyclin B1 and CDK1. The APC shRNA1 cells had increased CDK1, which was preferentially localized to the cytoplasm, and increased baseline CDK6. RNA-sequencing was performed to gain a global understanding of changes downstream of APC loss and identified a broad mis-regulation of cell cycle-related genes in APC shRNA1 cells. Our studies are the first to show an interaction between APC and taxane response in breast cancer. The implications include designing combination therapy to re-sensitize APC-mutant breast cancers to taxanes using the specific cell cycle alterations.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína Quinasa CDC2/genética , Metaplasia/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Ciclina B1/genética , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Metaplasia/genética , Metaplasia/patología , Paclitaxel/efectos adversos , Paclitaxel/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
13.
Cancer Res ; 66(18): 9245-51, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16982769

RESUMEN

Inhibitors of histone deacetylases (HDAC) inhibit malignant cell growth and induce apoptosis through unknown mechanisms. Here, we report that the expression status of adenomatous polyposis coli (APC) protein determines the relative sensitivity of colon cancer cells to HDAC inhibitor-induced apoptosis. HCA-7 cells (expressing wild-type beta-catenin and APC proteins) are more sensitive to apoptosis induced by HDAC inhibitors valproic acid (VPA) and suberoylanilide hydroxamic acid than SW620 or HT-29 cells (both expressing mutant APC). When wild-type APC protein was expressed using an inducible expression system, HT-29 cells became sensitive to apoptosis in response to VPA. Conversely, knocking down of endogenous APC protein by small interfering RNA (siRNA) blocked VPA-induced apoptosis in HCA-7 cells. APC mediated VPA-induced apoptosis through down-regulation of survivin. The level of survivin protein decreased in HCA-7 and HT-29/APC cells, but not in SW620 and HT-29/beta-Gal cells after VPA treatment. Whereas knocking down of survivin by siRNA sensitized SW620 cells to VPA-induced apoptosis, overexpression of survivin blocked VPA-induced apoptosis in HCA-7 cells. Down-regulation of survivin transcription occurred through changes in GSK-3beta/beta-catenin/Tcf-4 signaling molecules. VPA also induced proteasome-mediated degradation of survivin protein in HCA-7 cells. Furthermore, we have shown that APC mutation-mediated resistance to apoptosis can be overcome by cotreatment with Flavopiridol, which promotes survivin degradation. These results suggest that APC is a critical determinant of HDAC inhibitor-induced apoptosis in colon cancer cells and survivin is a potential target to enhance apoptotic response to HDAC inhibitors.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Apoptosis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Ácido Valproico/farmacología , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/biosíntesis , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HT29 , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , ARN Interferente Pequeño/genética , Survivin , Factores de Transcripción TCF/metabolismo , Proteína 2 Similar al Factor de Transcripción 7 , Transfección , Vorinostat , beta Catenina/metabolismo
14.
J Med Chem ; 61(17): 8017-8028, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30095910

RESUMEN

In colorectal cancer, adenomatous polyposis coli (APC) interacts with Rho guanine-nucleotide-exchange factor 4 (Asef), thereby stimulating aberrant colorectal-cancer-cell migration. Consequently, the APC-Asef interaction represents a promising therapeutic target for mitigating colorectal-cancer migration. In this study, we adopted the rational-design strategy involving the introduction of intramolecular hydrogen bonds and optimization of the lipophilic substituents to improve the binding affinities of peptides, leading to the discovery of MAI-400, the best inhibitor of the APC-Asef interaction known to date ( Kd = 0.012 µM, IC50 = 0.25 µM). Comprehensive evaluation of MAI-400 by biochemical and biophysical assays revealed the formation and effect of an intramolecular hydrogen bond. A cell-based assay showed MAI-400 efficiently blocking the APC-Asef interaction in a dose-dependent manner. Therefore, our study provides a best-in-class inhibitor, MAI-400, based on the rational drug design and structural validation, that can effectively inhibit the APC-Asef interaction.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Diseño de Fármacos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Cristalografía por Rayos X , Células HEK293 , Humanos , Conformación Proteica , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores
15.
Cancer Res ; 78(16): 4658-4670, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29871936

RESUMEN

Macropinocytosis has emerged as an important pathway of protein acquisition in cancer cells, particularly in tumors with activated Ras such as pancreatic and colon cancer. Macropinocytosis is also the route of entry of Bacillus Calmette-Guerin (BCG) and other microbial therapies of cancer. Despite this important role in tumor biology and therapy, the full mechanisms by which cancer cells can activate macropinocytosis remain incompletely defined. Using BCG uptake to assay macropinocytosis, we executed a genome-wide shRNA screen for macropinocytosis activators and identified Wnt pathway activation as a strong driver of macropinocytosis. Wnt-driven macropinocytosis was downstream of the ß-catenin-dependent canonical Wnt pathway, was PAK1 dependent, and supported albumin-dependent growth in Ras-WT cells. In cells with activated Ras-dependent macropinocytosis, pharmacologic or genetic inhibition of Wnt signaling suppressed macropinocytosis. In a mouse model of Wnt-driven colonic hyperplasia via APC silencing, Wnt-activated macropinocytosis stimulated uptake of luminal microbiota, a process reversed by topical pharmacologic inhibition of macropinocytosis. Our findings indicate that Wnt pathway activation drives macropinocytosis in cancer, and its inhibition could provide a therapeutic vulnerability in Wnt-driven intestinal polyposis and cancers with Wnt activation.Significance: The Wnt pathway drives macropinocytosis in cancer cells, thereby contributing to cancer growth in nutrient-deficient conditions and, in the context of colon cancer, to the early phases of oncogenesis. Cancer Res; 78(16); 4658-70. ©2018 AACR.


Asunto(s)
Neoplasias/tratamiento farmacológico , Pinocitosis/genética , Vía de Señalización Wnt/genética , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Silenciador del Gen , Genoma Humano/genética , Humanos , Ratones , Mycobacterium bovis/genética , Neoplasias/genética , Neoplasias/patología , ARN Interferente Pequeño/genética , beta Catenina/genética
16.
Cell Rep ; 21(1): 181-194, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28978472

RESUMEN

Adenomatous polyposis coli (APC) is a polarity regulator and tumor suppressor associated with familial adenomatous polyposis and colorectal cancer development. Although extensively studied in epithelial transformation, the effect of APC on T lymphocyte activation remains poorly defined. We found that APC ensures T cell receptor-triggered activation through Nuclear Factor of Activated T cells (NFAT), since APC is necessary for NFAT's nuclear localization in a microtubule-dependent fashion and for NFAT-driven transcription leading to cytokine gene expression. Interestingly, NFAT forms clusters juxtaposed with microtubules. Ultimately, mouse Apc deficiency reduces the presence of NFAT in the nucleus of intestinal regulatory T cells (Tregs) and impairs Treg differentiation and the acquisition of a suppressive phenotype, which is characterized by the production of the anti-inflammatory cytokine IL-10. These findings suggest a dual role for APC mutations in colorectal cancer development, where mutations drive the initiation of epithelial neoplasms and also reduce Treg-mediated suppression of the detrimental inflammation that enhances cancer growth.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/genética , Regulación Neoplásica de la Expresión Génica , Microtúbulos/inmunología , Factores de Transcripción NFATC/genética , Linfocitos T Reguladores/inmunología , Poliposis Adenomatosa del Colon/inmunología , Poliposis Adenomatosa del Colon/patología , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/inmunología , Animales , Diferenciación Celular , Línea Celular Tumoral , Células HCT116 , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Células Jurkat , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microtúbulos/ultraestructura , Factores de Transcripción NFATC/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Linfocitos T Reguladores/patología
17.
Cancer Res ; 64(8): 2692-8, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15087381

RESUMEN

As an important regulator in Wnt-signaling pathway, the APC gene is involved in apoptosis and cell cycle arrest. The loss of APC function is observed in most familial adenomatous polyposis-associated and sporadic colorectal cancer. APC gene is frequently inactivated by DNA mutations. However, hypermethylation in APC gene promoter was also observed in different cancers. In this study, by analyzing the methylation status of APC promoter in 22 colorectal cancer cell lines with different APC expression levels, we identified Regions A and B in the promoter, where the methylation of CpG sites was invariably correlated with the loss of gene expression. By nuclease accessibility assay, we also observed a correlation between the closed chromatin conformation in APC promoter and loss of gene expression. When the nonexpressing cell lines were treated with a DNA methyltransferase inhibitor, 5-Aza-2'-Deoxycytidine, the APC expression in these cells was induced, CpG sites were demethylated, and closed chromatin conformation was opened. However, when these cell lines were treated with a histone deacetylase inhibitor, Trichostatin A, no significant changes in APC expression, methylation status, and chromatin conformation were observed. Using transient transfection assay, a CCAAT box located in Region B was identified, which was involved in up-regulation of APC expression. Methylation of CpG sites around the CCAAT box resulted in a significant inhibition in the gene expression. The specific binding of a transcription factor CCAAT-binding factor (CBF) to the CCAAT box was determined by electrophoretic mobility shift analysis. The binding was inhibited after CpG sites close to the CCAAT box were methylated, indicating that DNA methylation can silence gene expression through interfering with the binding of transcription factors to the promoter. The biological function of CBF in APC gene regulation was further indicated by the decrease of luciferase activities in cells cotransfected with a plasmid carrying APC promoter/luciferase gene and a plasmid expressing dominant negative CBF mutant. In summary, methylation of CpG sites around CCAAT box in APC promoter inhibits the gene expression by changing the chromatin conformation and interfering with the binding of transcription factor CBF to CCAAT box.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Azacitidina/análogos & derivados , Factor de Unión a CCAAT/metabolismo , Cromatina/fisiología , Metilación de ADN , Regulación Neoplásica de la Expresión Génica/fisiología , Genes APC/fisiología , Proteína de la Poliposis Adenomatosa del Colon/biosíntesis , Proteína de la Poliposis Adenomatosa del Colon/genética , Azacitidina/farmacología , Línea Celular Tumoral , Cromatina/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Islas de CpG/fisiología , Decitabina , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes APC/efectos de los fármacos , Humanos , Luciferasas/biosíntesis , Luciferasas/genética , Luciferasas/metabolismo , Regiones Promotoras Genéticas , Conformación Proteica , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transfección , Regulación hacia Arriba
18.
Oncotarget ; 7(7): 7761-72, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26760960

RESUMEN

Toll-like receptors (TLRs)/NF-κB activation stimulated by lipopolysaccharide (LPS) was associated with diverse biological response in colon cancer, but the underlying mechanism was largely unknown. In the current study, we reported cell proliferation was elevated in adenomatous polyposis coli (APC) mutated- and APC knockdown cell lines, while the proliferation was inhibited in APC wild-type cell lines. Besides, in vivo experiments showed that LPS promoted APC knockdown tumor growth while inhibited proliferation of APC wild type. Further study confirmed that activation of TLRs/NF-κB signaling pathway by LPS cross regulated with APC/GSK-3ß/ß-catenin pathway, which were depend on APC status of cell lines. Taken together, APC genotypes play a key role in LPS induced different colon cancer biological response by cross-regulating ß-catenin and NF-κB, which may provide a novel strategy for carcinogenesis prevention.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Neoplasias del Colon/patología , Mutación/genética , Receptor Toll-Like 4/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Apoptosis , Western Blotting , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Genotipo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Técnicas para Inmunoenzimas , Lipopolisacáridos/farmacología , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
19.
Sci Rep ; 6: 31986, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27558955

RESUMEN

Molecular hydrogen (H2) is effective for many diseases. However, molecular bases of H2 have not been fully elucidated. Cumulative evidence indicates that H2 acts as a gaseous signal modulator. We found that H2 suppresses activated Wnt/ß-catenin signaling by promoting phosphorylation and degradation οf ß-catenin. Either complete inhibition of GSK3 or mutations at CK1- and GSK3-phosphorylation sites of ß-catenin abolished the suppressive effect of H2. H2 did not increase GSK3-mediated phosphorylation of glycogen synthase, indicating that H2 has no direct effect on GSK3 itself. Knock-down of adenomatous polyposis coli (APC) or Axin1, which form the ß-catenin degradation complex, minimized the suppressive effect of H2 on ß-catenin accumulation. Accordingly, the effect of H2 requires CK1/GSK3-phosphorylation sites of ß-catenin, as well as the ß-catenin degradation complex comprised of CK1, GSK3, APC, and Axin1. We additionally found that H2 reduces the activation of Wnt/ß-catenin signaling in human osteoarthritis chondrocytes. Oral intake of H2 water tended to ameliorate cartilage degradation in a surgery-induced rat osteoarthritis model through attenuating ß-catenin accumulation. We first demonstrate that H2 suppresses abnormally activated Wnt/ß-catenin signaling, which accounts for the protective roles of H2 in a fraction of diseases.


Asunto(s)
Hidrógeno/farmacología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Proteína Axina/metabolismo , Quinasa de la Caseína I/metabolismo , Línea Celular , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Gases/química , Glucógeno Sintasa Quinasa 3/metabolismo , Células HCT116 , Células HT29 , Células HeLa , Humanos , Hidrógeno/química , Leupeptinas/farmacología , Cloruro de Litio/farmacología , Masculino , Microscopía Fluorescente , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/veterinaria , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción SOX9/metabolismo , Agua/química , Proteína Wnt3A/metabolismo
20.
Int J Biochem Cell Biol ; 70: 39-47, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26556314

RESUMEN

The adenomatous polyposis coli (APC) tumor suppressor is multi-functional. APC is known to localize at the centrosome, and in mitotic cells contributes to formation of the mitotic spindle. To test whether APC contributes to nascent microtubule (MT) growth at interphase centrosomes, we employed MT regrowth assays in U2OS cells to measure MT assembly before and after nocodazole treatment and release. We showed that siRNA knockdown of full-length APC delayed both initial MT aster formation and MT elongation/regrowth. In contrast, APC-mutant SW480 cancer cells displayed a defect in MT regrowth that was unaffected by APC knockdown, but which was rescued by reconstitution of full-length APC. Our findings identify APC as a positive regulator of centrosome MT initial assembly and suggest that this process is disrupted by cancer mutations. We confirmed that full-length APC associates with the MT-nucleation factor γ-tubulin, and found that the APC cancer-truncated form (1-1309) also bound to γ-tubulin through APC amino acids 1-453. While binding to γ-tubulin may help target APC to the site of MT nucleation complexes, additional C-terminal sequences of APC are required to stimulate and stabilize MT growth.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Centrosoma/metabolismo , Células Epiteliales/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Sitios de Unión , Línea Celular Tumoral , Centrosoma/efectos de los fármacos , Centrosoma/ultraestructura , Células Epiteliales/efectos de los fármacos , Células Epiteliales/ultraestructura , Regulación de la Expresión Génica , Genes Reporteros , Prueba de Complementación Genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interfase/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura , Mitosis/efectos de los fármacos , Mutación , Nocodazol/farmacología , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Tubulina (Proteína)/genética , Moduladores de Tubulina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA