Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 14(10): 12116-12129, 2022 Mar 16.
Article in English | MEDLINE | ID: mdl-35238557

ABSTRACT

Here, we report a transparent, biodegradable, and cell-adhesive carrier that is securely coupled with the extracellular matrix (ECM) for corneal endothelial cell (CEC) transplantation. To fabricate a CEC carrier, poly(lactide-co-caprolactone) (PLCL) solution was poured onto the decellularized ECM (UMDM) derived from in vitro cultured umbilical cord blood-MSCs. Once completely dried, ECM-PLCL was then peeled off from the substrate. It was 20 µm thick, transparent, rich in fibronectin and collagen type IV, and easy to handle. Surface characterizations exhibited that ECM-PLCL was very rough (54.0 ± 4.50 nm) and uniformly covered in high density by ECM and retained a positive surface charge (65.2 ± 57.8 mV), as assessed via atomic force microscopy. Human CECs (B4G12) on the ECM-PLCL showed good cell attachment, with a cell density similar to the normal cornea. They could also maintain a cell phenotype, with nicely formed cell-cell junctions as assessed via ZO-1 and N-cadherin at 14 days. This was in sharp contrast to the CEC behaviors on the FNC-coated PLCL (positive control). A function-related marker, Na+/K+-ATPase, was also identified via western blot and immunofluorescence. In addition, primary rabbit CECs showed a normal shape and they could express structural and functional proteins on the ECM-PLCL. A simulation test confirmed that CECs loaded on the ECM-PLCL were successfully engrafted into the decellularized porcine corneal tissue, with a high engraftment level and cell viability. Moreover, ECM-PLCL transplantation into the anterior chamber of the rabbit eye for 8 weeks proved the maintenance of normal cornea properties. Taken together, this study demonstrates that our ECM-PLCL can be a promising cornea endothelium graft with an excellent ECM microenvironment for CECs.


Subject(s)
Extracellular Matrix , Mesenchymal Stem Cells , Animals , Cells, Cultured , Endothelial Cells/metabolism , Polymers/chemistry , Rabbits , Swine , Tissue Engineering
2.
Br J Cancer ; 124(3): 634-644, 2021 02.
Article in English | MEDLINE | ID: mdl-33071283

ABSTRACT

BACKGROUND: Most cancer cells employ the Warburg effect to support anabolic growth and tumorigenesis. Here, we discovered a key link between Warburg effect and aberrantly activated Wnt/ß-catenin signalling, especially by pathologically significant APC loss, in CRC. METHODS: Proteomic analyses were performed to evaluate the global effects of KYA1797K, Wnt/ß-catenin signalling inhibitor, on cellular proteins in CRC. The effects of APC-loss or Wnt ligand on the identified enzymes, PKM2 and LDHA, as well as Warburg effects were investigated. A linkage between activation of Wnt/ß-catenin signalling and cancer metabolism was analysed in tumour of Apcmin/+ mice and CRC patients. The roles of PKM2 in cancer metabolism, which depends on Wnt/ß-catenin signalling, were assessed in xenograft-tumours. RESULTS: By proteomic analysis, PKM2 and LDHA were identified as key molecules regulated by Wnt/ß-catenin signalling. APC-loss caused the increased expression of metabolic genes including PKM2 and LDHA, and increased glucose consumption and lactate secretion. Pathological significance of this linkage was indicated by increased expression of glycolytic genes with Wnt target genes in tumour of Apcmin/+ mice and CRC patients. Warburg effect and growth of xenografted tumours-induced by APC-mutated-CRC cells were suppressed by PKM2-depletion. CONCLUSIONS: The ß-catenin-PKM2 regulatory axis induced by APC loss activates the Warburg effect in CRC.


Subject(s)
Carrier Proteins/metabolism , Colorectal Neoplasms/metabolism , Genes, APC , L-Lactate Dehydrogenase/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Thyroid Hormones/metabolism , Warburg Effect, Oncologic , Wnt Signaling Pathway , Animals , Carrier Proteins/genetics , Colorectal Neoplasms/genetics , Heterografts , Humans , L-Lactate Dehydrogenase/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondrial Proton-Translocating ATPases/metabolism , Neoplasm Proteins/genetics , Proteomics , Thiazolidines/pharmacology , Thyroid Hormones/genetics , Tissue Array Analysis , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Thyroid Hormone-Binding Proteins
3.
Exp Mol Med ; 52(5): 832-842, 2020 05.
Article in English | MEDLINE | ID: mdl-32457491

ABSTRACT

Triple-negative breast cancer (TNBC) is a severe and heterogeneous disease that lacks an approved targeted therapy and has a poor clinical outcome to chemotherapy. Although the RAS-ERK signaling axis is rarely mutated in TNBC, ~50% of TNBCs show an increased copy number and overexpression of epidermal growth factor receptor (EGFR). However, EGFR-targeted therapies have offered no improvement in patient survival, underscoring the need to explore downstream targets, including RAS. We found that both ß-catenin and RAS, as well as epidermal growth factor receptor (EGFR), are overexpressed and correlated with one another in tumor tissues of TNBC patients. KYA1797K, an Axin-binding small molecule reducing ß-catenin and RAS expression via degradation and suppressing EGFR expression via transcriptional repression, inhibited the proliferation and the metastatic capability of stable cell lines as well as patient-derived cells (PDCs) established from TNBC patient tissues. KYA1797K also suppressed the stemness of 3D-cultured PDCs and xenografted tumors established by using residual tumors from TNBC patients and those established by the TNBC cell line. Targeting both the Wnt/ß-catenin and RAS-ERK pathways via small molecules simultaneously reducing the levels of ß-catenin, RAS, and EGFR could be a potential therapeutic approach for TNBC.


Subject(s)
Triple Negative Breast Neoplasms/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , ras Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Protein Stability/drug effects , Thiazolidines/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/etiology , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
Int J Cancer ; 146(10): 2877-2890, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31605540

ABSTRACT

Mutant KRAS provides a driving force for enhancement of cancer stem cells (CSCs) characteristics contributing transformation of colorectal cancer (CRC) cells harboring adenomatous polyposis coli (APC) mutations. Here, we identified the factors mediating the promotion of CSCs properties induced by KRAS mutation through microarray analyses of genes specifically induced in CRC spheroids harboring both KRAS and APC mutations. Among them, REG4 was identified as a key factor since CRISPR/Cas9-mediated knockout of REG4 most significantly affected the stem cell characteristics in which CSCs markers were effectively suppressed. We show that REG4 mediates promotion of CSCs properties via Wnt/ß-catenin signaling in various in vitro studies including tumor organoid systems. Furthermore, expression patterns of CSCs markers and REG4 correlated in intestinal tumors from Apcmin/+ /KrasG12D LA2 mice and in CRC patient tissues harboring both KRAS and APC mutations. The role of REG4 in the tumor-initiating capacity accompanied by enhancement of CSCs characteristics was also revealed by NSG mice xenograft system. Collectively, our study highlights the importance of REG4 in promoting CSCs properties induced by KRAS mutation, and provides a new therapeutic strategy for CRC harboring both APC and KRAS mutations.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Pancreatitis-Associated Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Transformation, Neoplastic/genetics , Heterografts , Humans , Mice , Neoplastic Stem Cells/metabolism , Wnt Signaling Pathway/physiology
5.
Exp Mol Med ; 51(9): 1-10, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31515471

ABSTRACT

Researchers have shown increased interest in determining what stimulates height. Currently, many children undergo precocious puberty, resulting in short stature due to premature closure of the growth plate. However, the current approach for height enhancement is limited to growth hormone treatment, which often results in side effects and clinical failure and is costly. Although recent studies have indicated the importance of paracrine signals in the growth plate for longitudinal bone growth, height-stimulating agents targeting the signaling pathways involved in growth plate maturation remain unavailable in the clinic. The Wnt/ß-catenin pathway plays a major role in the maturation of growth plate chondrocytes. In this study, by using an ex vivo tibial culture system, we identified indirubin-3'-oxime (I3O) as a compound capable of enhancing longitudinal bone growth. I3O promoted chondrocyte proliferation and differentiation via activation of the Wnt/ß-catenin pathway in vitro. Intraperitoneal injection of I3O in adolescent mice increased growth plate height along with incremental chondrocyte maturation. I3O promoted tibial growth without significant adverse effects on bone thickness and articular cartilage. Therefore, I3O could be a potential therapeutic agent for increasing height in children with growth retardation.


Subject(s)
Bone Development/drug effects , Chondrogenesis/drug effects , Glycogen Synthase Kinase 3 beta/genetics , Indoles/pharmacology , Oximes/pharmacology , Animals , Bone Development/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrocytes/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Growth Disorders/drug therapy , Growth Disorders/genetics , Growth Plate/drug effects , Humans , Mice , Osteogenesis/drug effects , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
6.
Life Sci Alliance ; 2(2)2019 04.
Article in English | MEDLINE | ID: mdl-30971423

ABSTRACT

Longitudinal bone growth ceases with growth plate senescence during puberty. However, the molecular mechanisms of this phenomenon are largely unexplored. Here, we examined Wnt-responsive genes before and after growth plate senescence and found that CXXC finger protein 5 (CXXC5), a negative regulator of the Wnt/ß-catenin pathway, was gradually elevated with reduction of Wnt/ß-catenin signaling during senescent changes of rodent growth plate. Cxxc5 -/- mice demonstrated delayed growth plate senescence and tibial elongation. As CXXC5 functions by interacting with dishevelled (DVL), we sought to identify small molecules capable of disrupting this interaction. In vitro screening assay monitoring CXXC5-DVL interaction revealed that several indirubin analogs were effective antagonists of this interaction. A functionally improved indirubin derivative, KY19382, elongated tibial length through delayed senescence and further activation of the growth plate in adolescent mice. Collectively, our findings reveal an important role for CXXC5 as a suppressor of longitudinal bone growth involving growth plate activity.


Subject(s)
Bone Development/physiology , DNA-Binding Proteins/metabolism , Growth Plate/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Dishevelled Proteins/metabolism , HEK293 Cells , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Transcription Factors/genetics , Transfection , Wnt Signaling Pathway , beta Catenin/metabolism
7.
Sci Rep ; 9(1): 648, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679620

ABSTRACT

The epidermal growth factor receptor (EGFR) inhibitors such as erlotinib and gefitinib are widely used for treatment of non-small cell lung cancer (NSCLC), but they have shown limited efficacy in an unselected population of patients. The KRAS mutations, which are identified in approximately 20% of NSCLC patients, have shown to be associated with the resistance to the EGFR tyrosine kinase inhibitors (TKIs). Currently, there is no clinically available targeted therapy which can effectively inhibit NSCLC tumors harboring KRAS mutations. This study aims to show the effectiveness of KYA1797K, a small molecule which revealed anti-cancer effect in colorectal cancer by destabilizing Ras via inhibiting the Wnt/ß-catenin pathway, for the treatment of KRAS-mutated NSCLC. While erlotinib fail to have anti-transforming effect in NSCLC cell lines harboring KRAS mutations, KYA1797K effectively inhibited the Ras-ERK pathway in KRAS-mutant NSCLC cell lines. As a result, KYA1797K treatment suppressed the growth and transformation of KRAS mutant NSCLC cells and also induced apoptosis. Furthermore, KYA1797K effectively inhibited Kras-driven tumorigenesis in the KrasLA2 mouse model by suppressing the Ras-ERK pathway. The destabilization of Ras via inhibition of the Wnt/ß-catenin pathway is a potential therapeutic strategy for KRAS-mutated NSCLC that is resistant to EGFR TKI.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Thiazolidines/pharmacology , ras Proteins/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , Mice , Protein Kinase Inhibitors/pharmacology , Protein Stability/drug effects , beta Catenin/metabolism
8.
Exp Mol Med ; 50(11): 1-12, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30459318

ABSTRACT

Drugs targeting the epidermal growth factor receptor (EGFR), such as cetuximab and panitumumab, have been prescribed for metastatic colorectal cancer (CRC), but patients harboring KRAS mutations are insensitive to them and do not have an alternative drug to overcome the problem. The levels of ß-catenin, EGFR, and RAS, especially mutant KRAS, are increased in CRC patient tissues due to mutations of adenomatous polyposis coli (APC), which occur in 90% of human CRCs. The increases in these proteins by APC loss synergistically promote tumorigenesis. Therefore, we tested KYA1797K, a recently identified small molecule that degrades both ß-catenin and Ras via GSK3ß activation, and its capability to suppress the cetuximab resistance of KRAS-mutated CRC cells. KYA1797K suppressed the growth of tumor xenografts induced by CRC cells as well as tumor organoids derived from CRC patients having both APC and KRAS mutations. Lowering the levels of both ß-catenin and RAS as well as EGFR via targeting the Wnt/ß-catenin pathway is a therapeutic strategy for controlling CRC and other types of cancer with aberrantly activated the Wnt/ß-catenin and EGFR-RAS pathways, including those with resistance to EGFR-targeting drugs attributed to KRAS mutations.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Line, Tumor , Cetuximab/therapeutic use , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice , Mice, Inbred C57BL , Mutation , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , Wnt Signaling Pathway , beta Catenin/metabolism
9.
EMBO Rep ; 19(12)2018 12.
Article in English | MEDLINE | ID: mdl-30413483

ABSTRACT

RAS proteins play critical roles in various cellular processes, including growth and transformation. RAS proteins are subjected to protein stability regulation via the Wnt/ß-catenin pathway, and glycogen synthase kinase 3 beta (GSK3ß) is a key player for the phosphorylation-dependent RAS degradation through proteasomes. GSK3ß-mediated RAS degradation does not occur in cells that express a nondegradable mutant (MT) ß-catenin. Here, we show that ß-catenin directly interacts with RAS at the α-interface region that contains the GSK3ß phosphorylation sites, threonine 144 and threonine 148 residues. Exposure of these sites by prior ß-catenin degradation is required for RAS degradation. The introduction of a peptide that blocks the ß-catenin-RAS interaction by binding to ß-catenin rescues the GSK3ß-mediated RAS degradation in colorectal cancer (CRC) cells that express MT ß-catenin. The coregulation of ß-catenin and RAS stabilities by the modulation of their interaction provides a mechanism for Wnt/ß-catenin and RAS-ERK pathway cross-talk and the synergistic transformation of CRC by both APC and KRAS mutations.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Proteolysis , Proto-Oncogene Proteins p21(ras)/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HEK293 Cells , Humans , Mice, Nude , Models, Biological , Models, Molecular , Mutation/genetics , Peptides/metabolism , Phosphorylation , Protein Binding , Protein Domains , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/chemistry , beta Catenin/genetics
10.
Oncotarget ; 8(70): 115596-115608, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29383184

ABSTRACT

Scaffold proteins of the mitogen activated protein kinase (MAPK) pathway recruit protein kinase cascades to confer context-specificity to cellular signaling. Varying concentrations of scaffold proteins determine different aspects of signaling outputs. However, regulatory mechanisms of scaffold proteins are poorly understood. Sur8, a scaffold protein in the Ras-MAPK pathway, is known to be involved in cell transformation and migration, and is increased in human colorectal cancer (CRC) patient tissue. Here we determine that regulation of Sur8 stability mediates transformation and migration of CRC cells. Fibroblast growth factor 2 (FGF2) is identified as an external regulator that stabilizes Sur8. Protein kinase C-alpha and -delta (PKCα/δ) are also identified as specific mediators of FGF2 regulation of Sur8 stability. PKCα/δ phosphorylate Sur8 at Thr-71 and Ser-297, respectively. This phosphorylation is essential for polyubiquitin-dependent degradation of Sur8. Sur8 mutations, which mimic phosphorylation by PKCα/δ and destabilized Sur8, suppress the FGF2-induced transformation and migration of CRC cells. The clinical relevance of Sur8 regulation by PKCα/δ is indicated by the inverse relationship between PKCα/δ and Sur8 expression in human CRC patient tissues. Overall, our findings demonstrate for the first time a regulatory mechanism of Sur8 stability involving cellular transformation and migration in CRC.

11.
Oncotarget ; 7(49): 81727-81740, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27835580

ABSTRACT

APC (80-90%) and K-Ras (40-50%) mutations frequently occur in human colorectal cancer (CRC) and these mutations cooperatively accelerate tumorigenesis including metastasis. In addition, both ß-catenin and Ras levels are highly increased in CRC, especially in metastatic CRC (mCRC). Therefore, targeting both the Wnt/ß-catenin and Ras pathways could be an ideal therapeutic approach for treating mCRC patients. In this study, we characterized the roles of KY1022, a small molecule that destabilizes both ß-catenin and Ras via targeting the Wnt/ß-catenin pathway, in inhibiting the cellular events, including EMT, an initial process of metastasis, and apoptosis. As shown by in vitro and in vivo studies using APCMin/+/K-RasG12DLA2 mice, KY1022 effectively suppressed the development of mCRC at an early stage of tumorigenesis. A small molecular approach degrading both ß-catenin and Ras via inhibition of the Wnt/ß-catenin signaling would be an ideal strategy for treatment of mCRC.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Colorectal Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/metabolism , Thiohydantoins/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Genes, APC , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neoplasm Invasiveness , Protein Stability , Proteolysis , Proto-Oncogene Proteins p21(ras)/genetics , Time Factors , beta Catenin/genetics
12.
BMB Rep ; 49(9): 455-6, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27470214

ABSTRACT

Mutations of APC and KRAS are frequently observed in human colorectal cancers (CRCs) and the Wnt/ß-catenin and Ras pathways are consequently activated in a significant proportion of CRC patients. Mutations in these two genes are also known to synergistically induce progression of CRCs. Through a series of studies, we have demonstrated that inhibition of the Wnt/ß-catenin signaling pathway negatively regulates Ras stability, therefore, Ras abundance is increased together with ß-catenin in both mice and human CRCs harboring adenomatous polyposis coli (APC) mutations. In a recent study, we identified KY1220, a small molecule that simultaneously degrades ß-catenin and Ras by inhibition of the Wnt/ß-catenin pathway, and obtained its derivative KYA1797K, which has improved activity and solubility. We found that KYA1797K binds the RGS domain of axin and enhances the binding affinity of ß-catenin or Ras with the ß-catenin destruction complex components, leading to simultaneous destabilization of ß-catenin and Ras via GSK3ß activation. By using both in vitro and in vivo studies, we showed that KYA1797K suppressed the growth of CRCs harboring APC and KRAS mutations through destabilization of ß-catenin and Ras. Therefore, our findings indicate that the simultaneous destabilization of ß-catenin and Ras via targeting axin may serve as an effective strategy for inhibition of CRCs. [BMB Reports 2016; 49(9): 455-456].


Subject(s)
Axin Protein/metabolism , beta Catenin/metabolism , ras Proteins/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Axin Protein/chemistry , Colorectal Neoplasms , Humans , Mice , Mutation , Protein Binding , Thiazolidines/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , ras Proteins/genetics
13.
Nat Chem Biol ; 12(8): 593-600, 2016 08.
Article in English | MEDLINE | ID: mdl-27294323

ABSTRACT

Both the Wnt/ß-catenin and Ras pathways are aberrantly activated in most human colorectal cancers (CRCs) and interact cooperatively in tumor promotion. Inhibition of these signaling may therefore be an ideal strategy for treating CRC. We identified KY1220, a compound that destabilizes both ß-catenin and Ras, via targeting the Wnt/ß-catenin pathway, and synthesized its derivative KYA1797K. KYA1797K bound directly to the regulators of G-protein signaling domain of axin, initiating ß-catenin and Ras degradation through enhancement of the ß-catenin destruction complex activating GSK3ß. KYA1797K effectively suppressed the growth of CRCs harboring APC and KRAS mutations, as shown by various in vitro studies and by in vivo studies using xenograft and transgenic mouse models of tumors induced by APC and KRAS mutations. Destabilization of both ß-catenin and Ras via targeting axin is a potential therapeutic strategy for treatment of CRC and other type cancers activated Wnt/ß-catenin and Ras pathways.


Subject(s)
Axin Protein/chemistry , Axin Protein/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RGS Proteins/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Thiohydantoins/pharmacology , beta Catenin/metabolism , Animals , Binding Sites , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genes, APC , Genes, ras , Humans , Mice , Mice, Transgenic , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Protein Stability/drug effects , Proto-Oncogene Proteins p21(ras)/genetics , RGS Proteins/metabolism , Thiohydantoins/chemical synthesis , Thiohydantoins/chemistry , Wnt Signaling Pathway/drug effects , beta Catenin/chemistry
14.
Phytother Res ; 30(5): 848-54, 2016 May.
Article in English | MEDLINE | ID: mdl-26929003

ABSTRACT

Polygonum aviculare L. is a member of the Polygonaceae family of plants, which has been known for its antioxidant and anti-obesity effects. However, the wound healing function of P. aviculare extract has not been assessed. In this study, we identified a novel property of P. aviculare extract as a Wnt/ß-catenin pathway activator based on a screen of 350 plant extracts using HEK293-TOP cells retaining the Wnt/ß-catenin signaling reporter gene. P. aviculare extract accelerated the migration of HaCaT keratinocytes without showing significant cytotoxicity. Moreover, P. aviculare extract efficiently re-epithelized wounds generated on mice. Additionally, ingredients of P. aviculare extract, such as quercitrin hydrate, caffeic acid, and rutin, also accelerated the motility of HaCaT keratinocytes with the activation of Wnt/ß-catenin signaling. Therefore, based on our findings, P. aviculare extract and its active ingredients could be potential therapeutic agents for wound healing. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Caffeic Acids/chemistry , Plant Extracts/chemistry , Polygonum/chemistry , Quercetin/analogs & derivatives , Wound Healing/drug effects , beta Catenin/metabolism , Animals , Humans , Male , Mice , Plant Extracts/pharmacology , Quercetin/chemistry , Transfection , Wnt Signaling Pathway
15.
Exp Mol Med ; 47: e152, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25792220

ABSTRACT

The Wnt/ß-catenin pathway has a role in osteoblast differentiation and bone formation. We screened 100 plant extracts and identified an extract from Euodia sutchuenensis Dode (ESD) leaf and young branch as an effective activator of the Wnt/ß-catenin pathway. ESD extract increased ß-catenin levels and ß-catenin nuclear accumulation in murine primary osteoblasts. The ESD extract also increased mRNA levels of osteoblast markers, including RUNX2, BMP2 and COL1A1, and enhanced alkaline phosphatase (ALP) activity in murine primary osteoblasts. Both ESD extract-induced ß-catenin increment and ALP activation were abolished by ß-catenin knockdown, confirming that the Wnt/ß-catenin pathway functions in osteoblast differentiation. ESD extract enhanced terminal osteoblast differentiation as shown by staining with Alizarin Red S and significantly increased murine calvarial bone thickness. This study shows that ESD extract stimulates osteoblast differentiation via the Wnt/ß-catenin pathway and enhances murine calvarial bone formation ex vivo.


Subject(s)
Cell Differentiation/drug effects , Evodia/chemistry , Osteoblasts/drug effects , Osteoblasts/metabolism , Plant Extracts/pharmacology , Wnt Signaling Pathway/drug effects , Animals , HEK293 Cells , Humans , Mice , Osteoblasts/cytology , Osteogenesis/drug effects , Plant Extracts/chemistry , Skull/anatomy & histology , Skull/drug effects , Skull/metabolism , beta Catenin/genetics , beta Catenin/metabolism
16.
World J Gastroenterol ; 20(29): 9862-71, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25110417

ABSTRACT

Administration of monoclonal antibodies (mAbs) against epidermal growth factor receptor (EGFR) such as cetuximab and panitumumab in combination with conventional chemotherapy substantially prolongs survival of patients with metastatic colorectal cancer (mCRC). However, the efficacy of these mAbs is limited due to genetic variation among patients, in particular K-ras mutations. The discovery of K-ras mutation as a predictor of non-responsiveness to EGFR mAb therapy has caused a major change in the treatment of mCRC. Drugs that inhibit transformation caused by oncogenic alterations of Ras and its downstream components such as BRAF, MEK and AKT seem to be promising cancer therapeutics as single agents or when given with EGFR inhibitors. Although multiple therapeutic strategies to overcome EGFR mAb-resistance are under investigation, our understanding of their mode of action is limited. Rational drug development based on stringent preclinical data, biomarker validation, and proper selection of patients is of paramount importance in the treatment of mCRC. In this review, we will discuss diverse approaches to overcome the problem of resistance to existing anti-EGFR therapies and potential future directions for cancer therapies related to the mutational status of genes associated with EGFR-Ras-ERK and PI3K signalings.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Animals , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , ErbB Receptors/immunology , ErbB Receptors/metabolism , Humans , Molecular Targeted Therapy , Neoplasm Metastasis , Signal Transduction/drug effects , Treatment Outcome
17.
Bone ; 65: 60-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24815917

ABSTRACT

Obesity is a growing issue of the modern world, and its negative impact on bones in obese male patients has been recently reported. The Wnt/ß-catenin pathway has an established role in the regulation of body fat content and bone density. We investigated the effects of indirubin-3'-oxime (I3O), the GSK3ß inhibitor that activates Wnt/ß-catenin signaling, on trabecular bone in high-fat diet (HFD)-induced obese male mice. I3O reverses the downregulating effect of fatty acid (FA) on Wnt/ß-catenin signaling and enhances the osteogenic commitment of the bone marrow-derived stromal cell line ST2. FA induces the adipogenic differentiation of bone marrow stromal cells in vitro. In a male mouse model of HFD-induced obesity, trabecular bone loss was observed in the femora, with a gross increase in abdominal fat; however, the HFD effects were rescued with the activation of Wnt/ß-catenin signaling by I3O treatment. I3O administration also reversed the increase in the number of HFD-induced adipocytes in the femur bone marrow in trabecular bone. Overall, our results indicate that I3O could be a potential therapeutic agent for obese male patients through downregulation of abdominal fat and net increment in trabecular bone density.


Subject(s)
Diet, High-Fat , Indoles/pharmacology , Obesity/metabolism , Osteogenesis/drug effects , Oximes/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Adipogenesis/drug effects , Animals , Cell Line , Gene Expression , Male , Mice
18.
PLoS One ; 9(1): e85546, 2014.
Article in English | MEDLINE | ID: mdl-24465596

ABSTRACT

The Wnt/ß-catenin pathway is a potential target for development of anabolic agents to treat osteoporosis because of its role in osteoblast differentiation and bone formation. However, there is no clinically available anti-osteoporosis drug that targets this Wnt/ß-catenin pathway. In this study, we screened a library of aqueous extracts of 350 plants and identified Hovenia dulcis Thunb (HDT) extract as a Wnt/ß-catenin pathway activator. HDT extract induced osteogenic differentiation of calvarial osteoblasts without cytotoxicity. In addition, HDT extract increased femoral bone mass without inducing significant weight changes in normal mice. In addition, thickness and area of femoral cortical bone were also significantly increased by the HDT extract. Methyl vanillate (MV), one of the ingredients in HDT, also activated the Wnt/ß-catenin pathway and induced osteoblast differentiation in vitro. MV rescued trabecular or cortical femoral bone loss in the ovariectomized mice without inducing any significant weight changes or abnormality in liver tissue when administrated orally. Thus, natural HDT extract and its ingredient MV are potential anabolic agents for treating osteoporosis.


Subject(s)
Bone Density/drug effects , Rhamnaceae/chemistry , Vanillic Acid/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/prevention & control , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Female , HEK293 Cells , Humans , Immunoblotting , Mice , Mice, Inbred ICR , Microscopy, Confocal , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Ovariectomy/adverse effects , Plant Extracts/chemistry , Plant Extracts/pharmacology , RNA Interference , Skull/cytology , Vanillic Acid/analogs & derivatives , Wnt Signaling Pathway/genetics , beta Catenin/genetics
19.
J Bone Miner Res ; 29(5): 1196-205, 2014.
Article in English | MEDLINE | ID: mdl-24243753

ABSTRACT

Osteoporosis is a major global health issue in elderly people. Because Wnt/ß-catenin signaling plays a key role in bone homeostasis, we screened activators of this pathway through cell-based screening, and investigated indirubin-3'-oxime (I3O), one of the positive compounds known to inhibit GSK3ß, as a potential anti-osteoporotic agent. Here, we show that I3O activated Wnt/ß-catenin signaling via inhibition of the interaction of GSK3ß with ß-catenin, and induced osteoblast differentiation in vitro and increased calvarial bone thickness ex vivo. Intraperitoneal injection of I3O increased bone mass and improved microarchitecture in normal mice and reversed bone loss in an ovariectomized mouse model of age-related osteoporosis. I3O also increased thickness and area of cortical bone, indicating improved bone strength. Enhanced bone mass and strength correlated with activated Wnt/ß-catenin signaling, as shown by histological analyses of both trabecular and cortical bones. I3O also restored mass and density of bone in hindlimb-unloaded mice compared with control, suspended mice, demonstrating bone-restoration effects of I3O in non-aged-related osteoporosis as well. Overall, I3O, a pharmacologically active small molecule, could be a potential therapeutic agent for the treatment and prevention of osteoporosis.


Subject(s)
Hindlimb Suspension , Indoles/pharmacology , Osteoporosis/drug therapy , Osteoporosis/metabolism , Ovariectomy , Oximes/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Bone Density/drug effects , Female , HEK293 Cells , Humans , Mice , Osteoporosis/etiology , Osteoporosis/pathology
20.
Immunol Res ; 56(1): 181-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23564227

ABSTRACT

House dust mites (HDMs) are a common cause of allergic asthma. The group 2 allergen from Dermatophagoides farinae, Der f 2, is one of the major HDM allergens. Elevated Der f 2 immunoglobulin E (IgE) levels are observed in most of the allergic patients. Interleukin-13 (IL-13), a gene associated with asthma pathology, was induced by Der f 2 in BEAS-2B human airway epithelial cells; however, the signaling pathways associated with Der f 2 are not fully understood. In this study, we identified a role of the phosphatidylinositol-3-kinase (PI3K)/Akt pathway, a well-known potential target for anti-asthma drugs, in the IL-13 induction by Der f 2. First, Der f 2 activated the PI3K/Akt pathway, which subsequently activated the nuclear factor-kappa B (NF-κB) pathway and induced IL-13 expression in BEAS-2B cells. Treatment with the PI3K inhibitor LY294002 abolished Der f 2-induced activation of Akt and NF-κB and the expression of IL-13. Furthermore, Der f 2-induced activation of the PI3K/Akt and NF-κB pathways, expression of IL-13, and the blockade of these effects with a PI3K inhibitor were confirmed in the lungs of mice that were intranasally exposed to Der f 2. Taken together, these results indicate that the PI3K/Akt pathway regulates Der f 2-induced IL-13 expression via activation of the NF-κB pathway.


Subject(s)
Antigens, Dermatophagoides/immunology , Arthropod Proteins/immunology , Asthma/immunology , Interleukin-13/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Respiratory Mucosa/immunology , Animals , Cell Line , Gene Expression Regulation/immunology , Humans , Immunoglobulin E/blood , Interleukin-13/genetics , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , Pyroglyphidae/immunology , Signal Transduction/immunology , Transcriptional Activation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL