Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(8): e0255738, 2021.
Article in English | MEDLINE | ID: mdl-34370741

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , CDC2 Protein Kinase/genetics , Metaplasia/drug therapy , Triple Negative Breast Neoplasms/drug therapy , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cyclin B1/genetics , Drug Resistance, Neoplasm/genetics , Female , Humans , Metaplasia/genetics , Metaplasia/pathology , Paclitaxel/adverse effects , Paclitaxel/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
2.
Molecules ; 26(4)2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33670371

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , Peptides/chemistry , Adenomatous Polyposis Coli Protein/chemistry , Adenomatous Polyposis Coli Protein/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Ligands , Molecular Dynamics Simulation , Neoplasm Metastasis , Peptides/antagonists & inhibitors , Protein Binding/drug effects , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/chemistry , Rho Guanine Nucleotide Exchange Factors/genetics
3.
Int J Mol Sci ; 21(24)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348563

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/prevention & control , Molecular Targeted Therapy/methods , Precision Medicine/methods , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/therapeutic use , Disease Models, Animal , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/metabolism , Humans , Mice
4.
Bioorg Chem ; 99: 103838, 2020 06.
Article in English | MEDLINE | ID: mdl-32334194

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Discovery , Indoles/pharmacology , Pyrazoles/pharmacology , Adenomatous Polyposis Coli Protein/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Protein Binding/drug effects , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/chemistry , Structure-Activity Relationship
5.
Proc Natl Acad Sci U S A ; 116(41): 20672-20678, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31551264

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Gastrointestinal Diseases/prevention & control , Intestines/cytology , Radiation Injuries, Experimental/prevention & control , Regeneration , Whole-Body Irradiation/adverse effects , Wnt Proteins/metabolism , Animals , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/metabolism , Intestines/radiation effects , Mice , RNA, Small Interfering , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Syndrome
6.
Mol Cell ; 72(1): 37-47.e4, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30197296

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Interaction Domains and Motifs/genetics , beta Catenin/genetics , Adaptor Proteins, Signal Transducing , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Axin Protein/genetics , Humans , Protein Stability , Transcription Factor 7-Like 1 Protein/genetics , Wnt Signaling Pathway/genetics
7.
J Med Chem ; 61(17): 8017-8028, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30095910

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Drug Design , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Interaction Domains and Motifs/drug effects , Crystallography, X-Ray , HEK293 Cells , Humans , Protein Conformation , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors
8.
Cancer Res ; 78(16): 4658-4670, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29871936

ABSTRACT

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.


Subject(s)
Neoplasms/drug therapy , Pinocytosis/genetics , Wnt Signaling Pathway/genetics , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Animals , Cell Line, Tumor , Gene Silencing , Genome, Human/genetics , Humans , Mice , Mycobacterium bovis/genetics , Neoplasms/genetics , Neoplasms/pathology , RNA, Small Interfering/genetics , beta Catenin/genetics
9.
Dev Growth Differ ; 60(1): 76-81, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29266402

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Calcium Carbonate/pharmacology , Food Coloring Agents/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Silencing , Planarians/physiology , Adenomatous Polyposis Coli Protein/genetics , Animals , Calcium Carbonate/chemistry , Digestive System/metabolism , Feeding Methods , Food Coloring Agents/chemistry , Phenotype , Planarians/genetics , Planarians/growth & development , RNA, Double-Stranded , Regeneration
10.
Cell Rep ; 21(1): 181-194, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978472

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/genetics , Gene Expression Regulation, Neoplastic , Microtubules/immunology , NFATC Transcription Factors/genetics , T-Lymphocytes, Regulatory/immunology , Adenomatous Polyposis Coli/immunology , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/immunology , Animals , Cell Differentiation , Cell Line, Tumor , HCT116 Cells , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Jurkat Cells , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/ultrastructure , NFATC Transcription Factors/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/pathology
11.
Biochem Biophys Res Commun ; 488(2): 418-424, 2017 06 24.
Article in English | MEDLINE | ID: mdl-28506830

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Melanoma/drug therapy , Adenomatous Polyposis Coli Protein/immunology , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/immunology , Disease Models, Animal , Female , Gastrointestinal Microbiome/immunology , Injections, Subcutaneous , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Th1 Cells/drug effects , Th1 Cells/immunology
12.
PLoS One ; 11(11): e0166422, 2016.
Article in English | MEDLINE | ID: mdl-27835699

ABSTRACT

Mutations in TGFBR2, a component of the transforming growth factor (TGF)-ß signaling pathway, occur in high-frequency microsatellite instability (MSI-H) colorectal cancer (CRC). In mouse models, Tgfbr2 inactivation in the intestinal epithelium accelerates the development of malignant intestinal tumors in combination with disruption of the Wnt-ß-catenin pathway. However, no studies have further identified the genes influenced by TGFBR2 inactivation following disruption of the Wnt-ß-catenin pathway. We previously described CDX2P-G19Cre;Apcflox/flox mice, which is stochastically null for Apc in the colon epithelium. In this study, we generated CDX2P-G19Cre;Apcflox/flox;Tgfbr2flox/flox mice, with simultaneous loss of Apc and Tgfbr2. These mice developed tumors, including adenocarcinoma in the proximal colon. We compared gene expression profiles between tumors of the two types of mice using microarray analysis. Our results showed that the expression of the murine homolog of GSDMC was significantly upregulated by 9.25-fold in tumors of CDX2P-G19Cre;Apcflox/flox;Tgfbr2flox/flox mice compared with those of CDX2P-G19Cre;Apcflox/flox mice. We then investigated the role of GSDMC in regulating CRC tumorigenesis. The silencing of GSDMC led to a significant reduction in the proliferation and tumorigenesis of CRC cell lines, whereas the overexpression of GSDMC enhanced cell proliferation. These results suggested that GSDMC functioned as an oncogene, promoting cell proliferation in colorectal carcinogenesis. In conclusion, combined inactivation of both Apc and Tgfbr2 in the colon epithelium of a CRC mouse model promoted development of adenocarcinoma in the proximal colon. Moreover, GSDMC was upregulated by TGFBR2 mutation in CRC and promoted tumor cell proliferation in CRC carcinogenesis, suggesting that GSDMC may be a promising therapeutic target.


Subject(s)
Adenocarcinoma/genetics , Adenomatous Polyposis Coli Protein/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/metabolism , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , Cell Line, Tumor , Cell Proliferation , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Microarray Analysis , Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/deficiency , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
13.
Sci Rep ; 6: 31986, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27558955

ABSTRACT

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.


Subject(s)
Hydrogen/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Axin Protein/metabolism , Casein Kinase I/metabolism , Cell Line , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Gases/chemistry , Glycogen Synthase Kinase 3/metabolism , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Hydrogen/chemistry , Leupeptins/pharmacology , Lithium Chloride/pharmacology , Male , Microscopy, Fluorescence , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/veterinary , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , SOX9 Transcription Factor/metabolism , Water/chemistry , Wnt3A Protein/metabolism
14.
DNA Repair (Amst) ; 45: 44-55, 2016 09.
Article in English | MEDLINE | ID: mdl-27334689

ABSTRACT

PARP inhibitors in combination with other agents are in clinical trial against cancer, but its effect on cancer stem cells (CSCs) is limited. CSCs are responsible for drug resistance, metastasis and cancer relapse due to high DNA repair capacity. Here, we present preclinical effects of Quinacrine (QC) with ABT-888, a PARP inhibitor, on highly metastatic breast cancer stem cells (mBCSCs). An increased level of Adenomatous polyposis coli (APC) was noted after treatment with ABT-888 in QC pre-treated mBCSCs cells. Increased APC physically interacts with PARP-1 and inhibits PARylation causing the non assembly of base excision repair (BER) multiprotein complex, resulting in an irreparable DNA damage and subsequent apoptosis. Knockdown of APC in mBCSCs inhibited DNA damage, increased BER and PARylation, reduces apoptosis while the over-expression of APC in BT20 (APC low expressing) cells reversed the effect. Thus, combination of QC and ABT-888 decreased mBCSCs growth by activating APC and inhibiting BER within the cells.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , DNA Repair/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Quinacrine/pharmacology , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Drug Combinations , Drug Evaluation, Preclinical , Drug Synergism , Female , Humans , Models, Biological , Neoplasm Metastasis , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Processing, Post-Translational/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
15.
Oncotarget ; 7(7): 7761-72, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26760960

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Colonic Neoplasms/pathology , Mutation/genetics , Toll-Like Receptor 4/metabolism , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Genotype , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Immunoenzyme Techniques , Lipopolysaccharides/pharmacology , Mice , Mice, Nude , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , beta Catenin/metabolism
16.
Int J Biochem Cell Biol ; 70: 39-47, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26556314

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Centrosome/metabolism , Epithelial Cells/metabolism , Microtubules/metabolism , Tubulin/metabolism , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/metabolism , Binding Sites , Cell Line, Tumor , Centrosome/drug effects , Centrosome/ultrastructure , Epithelial Cells/drug effects , Epithelial Cells/ultrastructure , Gene Expression Regulation , Genes, Reporter , Genetic Complementation Test , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Interphase/drug effects , Microtubules/drug effects , Microtubules/ultrastructure , Mitosis/drug effects , Mutation , Nocodazole/pharmacology , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tubulin/genetics , Tubulin Modulators/pharmacology
17.
Sci Rep ; 5: 18180, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26658992

ABSTRACT

The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase that regulates cell cycle progression in proliferating cells. To enter the S-phase, APC/C must be inactivated by phosphorylation of its cofactor, Cdh1. In post-mitotic cells such as neurons APC/C-Cdh1 complex is highly active and responsible for the continuous degradation of mitotic cyclins. However, the specific molecular pathway that determines neuronal cell cycle blockade in post-mitotic neurons is unknown. Here, we show that activation of glutamatergic receptors in rat cortical primary neurons endogenously triggers cyclin-dependent kinase-5 (Cdk5)-mediated phosphorylation of Cdh1 leading to its cytoplasmic accumulation and disassembly from the APC3 core protein, causing APC/C inactivation. Conversely, pharmacological or genetic inhibition of Cdk5 promotes Cdh1 ubiquitination and proteasomal degradation. Furthermore, we show that Cdk5-mediated phosphorylation and inactivation of Cdh1 leads to p27 depletion, which switches on the cyclin D1-cyclin-dependent kinase-4 (Cdk4)-retinoblastoma protein (pRb) pathway to allow the S-phase entry of neurons. However, neurons do not proceed through the cell cycle and die by apoptosis. These results indicate that APC/C-Cdh1 actively suppresses an aberrant cell cycle entry and death of neurons, highlighting its critical function in neuroprotection.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cadherins/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 5/metabolism , Neurons/metabolism , Retinoblastoma Protein/metabolism , S Phase , Signal Transduction , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Animals , Antigens, CD , Caspase 3/metabolism , Cell Cycle , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Glutamic Acid , Mitosis , Phosphorylation , Protein Stability , Rats
18.
FEBS J ; 281(3): 787-801, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24251807

ABSTRACT

The adenomatous polyposis coli (APC) membrane recruitment (Amer) family proteins Amer1/Wilms tumour gene on the X chromosome and Amer2 are binding partners of the APC tumour suppressor protein, and act as negative regulators in the Wnt signalling cascade. So far, nothing has been known about the third member of the family, Amer3. Here we show that Amer3 binds to the armadillo repeat domain of APC, similarly to Amer1 and Amer2. Amer3 also binds to the Wnt pathway regulator conductin/axin2. Furthermore, we identified Amer1 as binding partner of Amer3. Whereas Amer1 and Amer2 are linked to the plasma membrane by an N-terminal membrane localization domain, Amer3 lacks this domain. Amer3 localizes to the cytoplasm and nucleus of epithelial cells, and this is dependent on specific nuclear import and export sequences. Functionally, exogenous Amer3 enhances the expression of a ß-catenin/T-cell factor-dependent reporter gene, and knockdown of endogenous Amer3 reduces Wnt target gene expression in colorectal cancer cells. Thus, Amer3 acts as an activator of Wnt signalling, in contrast to Amer1 and Amer2, which are inhibitors, suggesting a nonredundant role of Amer proteins in the regulation of this pathway. Our data, together with those of previous studies, provide a comprehensive picture of similarities and differences within the Amer protein family.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenomatous Polyposis Coli Protein/metabolism , Adenomatous Polyposis Coli/metabolism , Cell Nucleus/metabolism , Colorectal Neoplasms/metabolism , Cytoplasm/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/chemistry , Adenomatous Polyposis Coli Protein/genetics , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/metabolism , Axin Protein/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , HEK293 Cells , Humans , Mutant Proteins , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ Specificity , Protein Interaction Domains and Motifs , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Sorting Signals , Protein Transport , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
19.
BMC Res Notes ; 6: 429, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24156781

ABSTRACT

BACKGROUND: The adenomatous polyposis coli (APC) tumour suppressor gene encodes a 2843 residue (310 kDa) protein. APC is a multifunctional protein involved in the regulation of ß-catenin/Wnt signalling, cytoskeletal dynamics and cell adhesion. APC mutations occur in most colorectal cancers and typically result in truncation of the C-terminal half of the protein. RESULTS: In order to investigate the biophysical properties of APC, we have generated a set of monoclonal antibodies which enable purification of recombinant forms of APC. Here we describe the characterisation of these anti-APC monoclonal antibodies (APC-NT) that specifically recognise endogenous APC both in solution and in fixed cells. Full-length APC(1-2843) and cancer-associated, truncated APC proteins, APC(1-1638) and APC(1-1311) were produced in Sf9 insect cells. CONCLUSIONS: Recombinant APC proteins were purified using a two-step affinity approach using our APC-NT antibodies. The purification of APC proteins provides the basis for detailed structure/function analyses of full-length, cancer-truncated and endogenous forms of the protein.


Subject(s)
Adenomatous Polyposis Coli Protein/isolation & purification , Antibodies, Monoclonal/biosynthesis , Chromatography, Affinity/methods , Recombinant Proteins/isolation & purification , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/chemistry , Adenomatous Polyposis Coli Protein/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigens/administration & dosage , Antigens/chemistry , Baculoviridae/genetics , Dogs , Gene Expression , Humans , Madin Darby Canine Kidney Cells , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sf9 Cells , Spodoptera
20.
Cell Calcium ; 54(3): 246-56, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23871111

ABSTRACT

The assembly of STIM1 protein puncta near endoplasmic reticulum-plasma membrane (ER-PM) junctions is required for optimal activation of store-operated channels (SOC). The mechanisms controlling the translocation of STIM1 puncta to ER-PM junctions remain largely unknown. In the present study, we have explored the role of the microtubule binding protein adenomatous polyposis coli (APC), on STIM1 puncta and store-operated calcium entry (SOCE). APC-depleted cells showed reduced STIM1 puncta near ER-PM junctions, instead puncta is found at the ER surrounding the cell nucleus. Reduced STIM1 puncta near ER-PM junctions in APC-depleted cells correlates with a strong inhibition of SOCE and diminished Orai whole-cell currents. Immunoprecipitation and confocal microscopy co-localization studies indicate that, upon depletion of the ER, STIM1 dissociates from EB1 and associates to APC. Deletion analysis identified an APC-binding domain in the carboxyl terminus of STIM1 (STIM1 650-685). These results together position APC as an important element in facilitating the translocation of STIM1 puncta near ER-PM junctions, which in turn is required for efficient SOCE and Orai activation upon depletion of the ER.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/genetics , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Membrane/drug effects , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , ORAI1 Protein , Patch-Clamp Techniques , Principal Component Analysis , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Stromal Interaction Molecule 1 , Thapsigargin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL