Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 22836, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819572

ABSTRACT

The present study aimed to map the location and frequency of fracture lines on the coronal articular and sagittal planes in multifragmentary patellar fractures. 66 multifragmentary patellar fractures were digitally reconstructed using the 3D CT mapping technique. The coronal articular surface and midsagittal fracture maps were produced by superimposing each case over a single template. Each fracture line was classified based on the initial displacement and orientation. We evaluated the frequency and direction of the fracture line, coronal split fragment area, and satellite and inferior pole fragment presence. Coronal articular surface fracture mapping identified primary horizontal fracture lines between the middle and inferior one-third of the articular surface in 63 patients (95.4%). Secondary horizontal fracture lines running on the inferior border of the articular facet were confirmed (83.3%). Secondary vertical fracture lines creating satellite fragments were mostly located on the periphery of the bilateral facet. Midsagittal fracture mapping of primary and secondary horizontal fracture lines with the main coronal fracture line revealed a predominantly X-shaped fracture map. The consequent coronal split fragment and inferior pole fracture were combined in most cases. In conclusion, the multifragmentary patellar fracture has a distinct pattern which makes coronal split, inferior pole, or satellite fragments.

2.
Science ; 374(6567): eabb3420, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34709885

ABSTRACT

Immune cells identify and destroy damaged cells to prevent them from causing cancer or other pathologies by mechanisms that remain poorly understood. Here, we report that the cell-cycle inhibitor p21 places cells under immunosurveillance to establish a biological timer mechanism that controls cell fate. p21 activates retinoblastoma protein (Rb)­dependent transcription at select gene promoters to generate a complex bioactive secretome, termed p21-activated secretory phenotype (PASP). The PASP includes the chemokine CXCL14, which promptly attracts macrophages. These macrophages disengage if cells normalize p21 within 4 days, but if p21 induction persists, they polarize toward an M1 phenotype and lymphocytes mount a cytotoxic T cell response to eliminate target cells, including preneoplastic cells. Thus, p21 concurrently induces proliferative arrest and immunosurveillance of cells under duress.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Immunologic Surveillance , Animals , Cell Cycle Checkpoints , Cell Line , Chemokines, CXC/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Genes, ras , Hepatocytes/immunology , Hepatocytes/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/metabolism , Retinoblastoma Protein/metabolism , Stress, Physiological , T-Lymphocytes, Cytotoxic/immunology , Transcription, Genetic
3.
Sci Total Environ ; 749: 142375, 2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33370884

ABSTRACT

Composted livestock manures, in both solid and liquid form, are used as fertilizers in cropland. However, excess solid and liquid manures in agricultural watersheds are considered as nonpoint pollution sources because of their high nutrient and heavy metal contents of, as well as their antibiotic contents, especially veterinary antibiotics (VAs). In this study, 21 VAs under nine classes (i.e., cephems, ionophores, lincosamides, penicillins, pleuromutilins, quinolones, streptogramins, sulfonamides, and tetracyclines) found in agricultural watersheds were simultaneously analyzed via UHPLC-q-orbitrap high-resolution mass spectrometry using an on-line solid-phase extraction system. The residues of VAs in the surface water of two intensive livestock rearing watersheds (Cheongmi and Gwangcheon streams) in Korea were successfully quantified, and the values were found to range from 1.84 ± 0.42 ng L-1 to 835.6 ± 31.9 ng L-1. Time lags of 2-3 months were observed between the periods of liquid manure application and the periods with the maximum concentrations of VAs. In both watersheds, samples from points close to areas with extensive application of liquid manure exhibited high concentrations of most of the 21 VAs. Between the watersheds, the one with heavier application of liquid manure showed higher concentrations of the target VAs. To the best of our knowledge, this study represents the first attempt at evaluating the correlation between liquid manure application and environmental occurrence of VAs in surface water. The findings reveal that liquid manure application plays an important role in introducing VAs into aquatic environments.


Subject(s)
Livestock , Manure , Agriculture , Animals , Anti-Bacterial Agents , Chromatography, High Pressure Liquid , Manure/analysis , Republic of Korea
4.
Cancer Res ; 80(19): 4158-4171, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32816906

ABSTRACT

The key functional molecules involved in inflammatory bowel disease (IBD) and IBD-induced colorectal tumorigenesis remain unclear. In this study, we found that the apoptosis repressor with caspase recruitment domain (ARC) protein plays critical roles in IBD. ARC-deficient mice exhibited substantially higher susceptibility to dextran sulfate sodium (DSS)-induced IBD compared with wild-type mice. The inflammatory burden induced in ARC-deficient conditions was inversely correlated with CCL5 and CXCL5 levels in immune cells, especially CD4-positive T cells. Pathologically, ARC expression in immune cells was significantly decreased in clinical biopsy specimens from patients with IBD compared with normal subjects. In addition, ARC levels inversely correlated with CCL5 and CXCL5 levels in human biopsy specimens. ARC interacted with TNF receptor associated factor (TRAF) 6, regulating ubiquitination of TRAF6, which was associated with NF-κB signaling. Importantly, we identified a novel ubiquitination site at lysine 461, which was critical in the function of ARC in IBD. ARC played a critical role in IBD and IBD-associated colon cancer in a bone marrow transplantation model and azoxymethane/DSS-induced colitis cancer mouse models. Overall, these findings reveal that ARC is critically involved in the maintenance of intestinal homeostasis and protection against IBD through its ubiquitination of TRAF6 and subsequent modulation of NF-κB activation in T cells. SIGNIFICANCE: This study uncovers a crucial role of ARC in the immune system and IBD, giving rise to a novel strategy for IBD and IBD-associated colon cancer therapy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Colorectal Neoplasms/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Muscle Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Azoxymethane/toxicity , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , Chemokine CCL5/metabolism , Chemokine CXCL5/metabolism , Colitis/chemically induced , Colorectal Neoplasms/chemically induced , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/chemistry , Muscle Proteins/genetics , Ubiquitination
5.
Oncogene ; 37(26): 3501-3513, 2018 06.
Article in English | MEDLINE | ID: mdl-29563609

ABSTRACT

Inflammation is a complex biological host reaction to tissue damage, infection and trauma. Extensive study of the inflammatory response has led to the identification of several protein kinases that are essential for signaling and could be potential therapeutic targets. The RSK family of kinases has multiple cellular functions. In our study, we found that RSK2 is a mediator for inflammation signaling and interacts with TRAF6. In vitro kinase assay results indicated that RSK2 strongly phosphorylates TRAF6 at serines 46, 47 and 48. Ectopic overexpression of TRAF6 or knocking down RSK2 expression confirmed that RSK2 is a positive regulator of TRAF6 K63 ubiquitination. TRAF6 is also required for RSK2 ubiquitination. TRAF6 bridges the TNF receptor superfamily and intracellular signaling for the induction of proinflammatory cytokines. We developed a colon inflammation model using RSK2 wild type (WT) and knockout (KO) mice. As expected, F4/80 and CD3 infiltration were significantly upregulated in WT mice compared to RSK2 KO mice. Furthermore, inflammation signaling, including Ikkα/ß, p38 and JNKs, was dramatically upregulated in WT mice. Colon tissue immunoprecipitation results further confirmed that TRAF6 K63 ubiquitination was lower in RSK2 KO mice. Overall, these results indicate that phosphorylation of TRAF6 (S46, 47, 48) by RSK2 is required for TRAF6 K63 ubiquitination and inflammation signaling.


Subject(s)
Colitis/pathology , Colon/pathology , Ribosomal Protein S6 Kinases, 90-kDa/genetics , TNF Receptor-Associated Factor 6/metabolism , Animals , Antigens, Differentiation/metabolism , CD3 Complex/metabolism , Cell Line, Tumor , Colon/immunology , Female , HEK293 Cells , Humans , Inflammation/pathology , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , RAW 264.7 Cells , Ubiquitination/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Mol Cancer Ther ; 17(5): 1101-1113, 2018 05.
Article in English | MEDLINE | ID: mdl-29483219

ABSTRACT

The biological functions of the p53-related protein kinase (PRPK) remain unclear. We have previously demonstrated that PRPK is phosphorylated by the T-LAK cell-originated protein kinase (TOPK) and that phosphorylated PRPK (p-PRPK) promotes colon cancer metastasis. Here, we analyzed colon adenocarcinomas from 87 patients and found that higher expression levels of p-PRPK were associated with later stages of metastatic dissemination (stage III and IV) as compared with earlier stages (stages I and II). Indeed, levels of p-PRPK were higher in metastatic versus malignant human colon adenocarcinomas. Knocking down PRPK expression attenuated colorectal liver and lung metastasis of colon cancer cells in vivo An in vitro kinase assay indicated that active PRPK does not phosphorylate p53 directly. We found that PRPK phosphorylates survivin, a regulator of colon cancer metastasis. PRPK phosphorylates survivin at Thr34, which is important for survivin stability. Taken together, our data strongly suggest that the PRPK signaling pathway promotes colon cancer metastasis by modulating survivin stability, and that PRPK could be a new prognostic marker for the survival of colon cancer patients. In addition, we identified an FDA-approved bacteriostatic antibiotic, fusidic acid sodium salt (fusidic acid or FA) as an inhibitor of PRPK, and show that FA combined with 5-fluorouracil (5-FU) inhibited PRPK activity and colon cancer metastasis to the lung in mice. We contend that the combination of FA with 5-FU could be an alternative therapeutic strategy to traditional chemotherapy for colon cancer patients with poor prognosis. Mol Cancer Ther; 17(5); 1101-13. ©2018 AACR.


Subject(s)
Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Fluorouracil/administration & dosage , Fusidic Acid/administration & dosage , HCT116 Cells , HT29 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Survivin/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Stem Cell Reports ; 9(6): 2050-2064, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29153991

ABSTRACT

The POU transcription factor OCT4 is critical for maintaining the undifferentiated state of embryonic stem cells (ESCs) and generating induced pluripotent stem cells (iPSCs), but its precise mechanisms of action remain poorly understood. Here, we investigated the role of OCT4 phosphorylation in the biological functions of ESCs. We observed that c-Jun N-terminal kinases (JNKs) directly interacted with and phosphorylated OCT4 at serine 347, which inhibited the transcriptional activity of OCT4. Moreover, phosphorylation of OCT4 induced binding of FBXW8, which reduced OCT4 protein stability and enhanced its proteasomal degradation. We also found that the mutant OCT4 (S347A) might delay the differentiation process of mouse ESCs and enhance the efficiency of generating iPSCs. These results demonstrated that OCT4 phosphorylation on serine 347 by JNKs plays an important role in its stability, transcriptional activities, and self-renewal of mouse ESCs.


Subject(s)
Cell Differentiation/genetics , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/metabolism , Animals , Humans , MAP Kinase Kinase 4/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Phosphorylation , Pluripotent Stem Cells/cytology , Protein Stability , Proteolysis , Serine/metabolism
8.
Carcinogenesis ; 38(11): 1136-1146, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29029040

ABSTRACT

Herbacetin is a flavonol compound that is found in plants such as flaxseed and ramose scouring rush herb, it possesses a strong antioxidant capacity, and exerts anticancer effects on colon and breast cancer. However, the effect of herbacetin on skin cancer has not been investigated. Herein, we identified herbacetin as a dual V-akt murine thymoma viral oncogene homolog (AKT) and ornithine decarboxylase (ODC) inhibitor, and illustrated its anticancer effects in vitro and in vivo against cutaneous squamous cell carcinoma (SCC) and melanoma cell growth. To identify the direct target(s) of herbacetin, we screened several skin cancer-related protein kinases, and results indicated that herbacetin strongly suppresses both AKT and ODC activity. Results of cell-based assays showed that herbacetin binds to both AKT and ODC, inhibits TPA-induced neoplastic transformation of JB6 mouse epidermal cells, and suppresses anchorage-independent growth of cutaneous SCC and melanoma cells. The inhibitory activity of herbacetin was associated with markedly reduced NF-κB and AP1 reporter activity. Interestingly, herbacetin effectively attenuated TPA-induced skin cancer development and also exhibited therapeutic effects against solar-UV-induced skin cancer and melanoma growth in vivo. Our findings indicate that herbacetin is a potent AKT and ODC inhibitor that should be useful for preventing skin cancers.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Flavonoids/pharmacology , Melanoma/drug therapy , Ornithine Decarboxylase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/drug therapy , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Humans , Melanoma/metabolism , Mice , NF-kappa B , Ornithine Decarboxylase Inhibitors/pharmacology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
9.
EBioMedicine ; 25: 22-31, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29033371

ABSTRACT

Colorectal cancer is associated with aberrant activation of the Wnt pathway. ß-Catenin plays essential roles in the Wnt pathway by interacting with T-cell factor 4 (TCF4) to transcribe oncogenes. We synthesized a small molecule, referred to as HI-B1, and evaluated signaling changes and biological consequences induced by the compound. HI-B1 inhibited ß-catenin/TCF4 luciferase activity and preferentially caused apoptosis of cancer cells in which the survival is dependent on ß-catenin. The formation of the ß-catenin/TCF4 complex was disrupted by HI-B1 due to the direct interaction of HI-B1 with ß-catenin. Colon cancer patient-derived xenograft (PDX) studies showed that a tumor with higher levels of ß-catenin expression was more sensitive to HI-B1 treatment, compared to a tumor with lower expression levels of ß-catenin. The different sensitivities of PDX tumors to HI-B1 were dependent on the ß-catenin expression level and potentially could be further exploited for biomarker development and therapeutic applications against colon cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Small Molecule Libraries/administration & dosage , Transcription Factor 4/genetics , beta Catenin/genetics , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Multiprotein Complexes/drug effects , Multiprotein Complexes/genetics , Small Molecule Libraries/chemical synthesis , Transcription Factor 4/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays , beta Catenin/antagonists & inhibitors
10.
Carcinogenesis ; 38(12): 1228-1240, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29040381

ABSTRACT

The Wilms' tumor 1 (WT1) gene is believed to act as a canonical tumor suppressor. However, it has also been reported to function as an oncogene. Germline WT1 deletion is associated with Wilms' tumor, and exogenous WT1 cDNA introduction into cells induces the transcriptional suppression of its oncogenic target genes. In contrast, high WT1 expression is associated with poor prognosis in patients with various cancers. Why WT1 acts as a tumor suppressor under certain conditions but as an oncogene under other conditions is unknown. Here, we report that CUG initiation site for WT1 protein synthesis (CUG)-translated WT1 (cugWT1), an N-terminally extended form of canonical AUG initiation site for WT1 protein synthesis (AUG)-translated WT1 (augWT1), was overexpressed in most cancer cell lines and cancer tissues and functioned as an oncogene, whereas the classical augWT1 acted as a tumor suppressor as reported previously and inhibited the function of cugWT1. Translation of cugWT1 is initiated from a CUG codon upstream and in-frame with the coding region of augWT1. cugWT1 induced cell transformation and increased the gene expression of c-myc, bcl-2 and egfr, whereas overexpression of augWT1 repressed colony formation of cancer cells and inhibited the expression of the same target genes by recruiting histone deacetylase 1 (HDAC1). In addition, we found that protein kinase B (AKT)-phosphorylated cugWT1 on Ser62 and protected cugWT1 from proteasomal degradation induced by the F-box/WD repeat-containing protein 8 (FBXW8). These results provide an important breakthrough in the field of cancer biology and contribute significantly to the resolution of the chameleon function of WT1.


Subject(s)
Genes, Wilms Tumor , Oncogenes/genetics , Protein Biosynthesis/genetics , Transcription Initiation Site , WT1 Proteins/genetics , Animals , Cell Line, Tumor , Female , Heterografts , Humans , Male , Mice , Mice, Nude
11.
EBioMedicine ; 18: 73-82, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28412249

ABSTRACT

Approximately 90% of all cancer deaths arise from the metastatic dissemination of primary tumors. Metastasis is the most lethal attribute of colorectal cancer. New data regarding the molecules contributing to the metastatic phenotype, the pathways they control and the genes they regulate are very important for understanding the processes of metastasis prognosis and prevention in the clinic. The purpose of this study was to investigate the role of T-LAK cell-originated protein kinase (TOPK) in the promotion of colorectal cancer metastasis. TOPK is highly expressed in human metastatic colorectal cancer tissue compared with malignant adenocarcinoma. We identified p53-related protein kinase (PRPK) as a new substrate of TOPK. TOPK binds with and phosphorylates PRPK at Ser250 in vitro and ex vivo. This site plays a critical role in the function of PRPK. Cell lines stably expressing mutant PRPK (S250A), knockdown TOPK, knockdown PRPK or knockdown of both TOPK and PRPK significantly inhibited liver metastasis of human HCT116 colon cancer cells in a xenograft mouse model. Therefore, we conclude that TOPK directly promotes metastasis of colorectal cancer by modulating PRPK. Thus, these findings may assist in the prediction of prognosis or development of new therapeutic strategies against colon cancer.


Subject(s)
Colorectal Neoplasms/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/metabolism , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
12.
Oncotarget ; 8(19): 32027-32042, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28410190

ABSTRACT

Previously, we reported that high-fat-diet (HFD)-induced obesity stimulates melanoma progression in the B16F10 allograft model. In this study, we examined whether oleuropein (OL), the most abundant phenolic compound in olives, inhibits HFD-induced melanoma progression. Four-week-old male C57BL/6N mice were fed a HFD-diet with or without OL. After 16 weeks of feeding, B16F10-luc cells were subcutaneously injected and the primary tumor was resected 3 weeks later. OL suppressed HFD-induced solid tumor growth. In the tumor tissues, OL reduced HFD-induced expression of angiogenesis (CD31, VE-cadherin, VEGF-A, and VEGFR2), lymphangiogenesis (LYVE-1, VEGF-C, VEGF-D, and VEGFR3), and hypoxia (HIF-1α and GLUT-1) markers as well as HFD-induced increases in lipid vacuoles and M2 macrophages (MΦs). All animals were euthanized 2.5 weeks after tumor resection. OL suppressed HFD-induced increases in lymph node (LN) metastasis; expression of VEGF-A, VEGF-C, and VEGF-D in the LN; and M2-MΦs and the size of adipocytes in adipose tissues surrounding LNs. Co-culture results revealed that the crosstalk between B16F10s, M2-MΦs, and differentiated 3T3-L1 cells under hypoxic conditions increased the secretion of VEGF-A and -D, which stimulated tube formation and migration of endothelial cells (HUVECs) and lymphatic endothelial cells (LEC), respectively. Additionally, OL directly inhibited the differentiation of 3T3-L1 preadipocytes and tube formation by HUVECs and LECs. The overall results indicated that dietary OL inhibits lipid and M2-MΦ accumulation in HFD-fed mice, which contributes to decreases in VEGF secretion, thereby leading to inhibition of angiogenesis and lymphangiogenesis.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Dietary Supplements , Iridoids/administration & dosage , Lymphangiogenesis/drug effects , Melanoma, Experimental/pathology , Neovascularization, Pathologic , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Allografts , Animals , Apoptosis , Cell Proliferation/drug effects , Diet, High-Fat , Hypoxia/metabolism , Iridoid Glucosides , Lipid Metabolism , Lymphatic Metastasis , Macrophages/drug effects , Macrophages/metabolism , Melanoma, Experimental/drug therapy , Mice , Neovascularization, Pathologic/drug therapy , Tumor Burden , Vascular Endothelial Growth Factors/metabolism
13.
Oncotarget ; 7(23): 35001-14, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27167001

ABSTRACT

Various carcinogens induce EGFR/RAS/MAPK signaling, which is critical in the development of lung cancer. In particular, constitutive activation of extracellular signal-regulated kinase 2 (ERK2) is observed in many lung cancer patients, and therefore developing compounds capable of targeting ERK2 in lung carcinogenesis could be beneficial. We examined the therapeutic effect of catechol in lung cancer treatment. Catechol suppressed anchorage-independent growth of murine KP2 and human H460 lung cancer cell lines in a dose-dependent manner. Catechol inhibited ERK2 kinase activity in vitro, and its direct binding to the ERK2 active site was confirmed by X-ray crystallography. Phosphorylation of c-Myc, a substrate of ERK2, was decreased in catechol-treated lung cancer cells and resulted in reduced protein stability and subsequent down-regulation of total c-Myc. Treatment with catechol induced G1 phase arrest in lung cancer cells and decreased protein expression related to G1-S progression. In addition, we showed that catechol inhibited the growth of both allograft and xenograft lung cancer tumors in vivo. In summary, catechol exerted inhibitory effects on the ERK2/c-Myc signaling axis to reduce lung cancer tumor growth in vitro and in vivo, including a preclinical patient-derived xenograft (PDX) model. These findings suggest that catechol, a natural small molecule, possesses potential as a novel therapeutic agent against lung carcinogenesis in future clinical approaches.


Subject(s)
Antineoplastic Agents/pharmacology , Catechols/pharmacology , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase 1/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mice, SCID , Mitogen-Activated Protein Kinase 1/drug effects , Proto-Oncogene Proteins c-myc/drug effects , Xenograft Model Antitumor Assays
14.
Int J Mol Sci ; 17(2): 264, 2016 02 22.
Article in English | MEDLINE | ID: mdl-26907265

ABSTRACT

Benzyl isothiocyanate (BITC) is a hydrolysis product of glucotropaeolin, a compound found in cruciferous vegetables, and has been shown to have anti-tumor properties. In the present study, we investigated whether BITC inhibits the development of prostate cancer in the transgenic adenocarcinoma mouse prostate (TRAMP) mice. Five-week old, male TRAMP mice and their nontransgenic littermates were gavage-fed with 0, 5, or 10 mg/kg of BITC every day for 19 weeks. The weight of the genitourinary tract increased markedly in TRAMP mice and this increase was suppressed significantly by BITC feeding. H and E staining of the dorsolateral lobes of the prostate demonstrated that well-differentiated carcinoma (WDC) was a predominant feature in the TRAMP mice. The number of lobes with WDC was reduced by BITC feeding while that of lobes with prostatic intraepithelial neoplasia was increased. BITC feeding reduced the number of cells expressing Ki67 (a proliferation marker), cyclin A, cyclin D1, and cyclin-dependent kinase (CDK)2 in the prostatic tissue. In vitro cell culture results revealed that BITC decreased DNA synthesis, as well as CDK2 and CDK4 activity in TRAMP-C2 mouse prostate cancer cells. These results indicate that inhibition of cell cycle progression contributes to the inhibition of prostate cancer development in TRAMP mice treated with BITC.


Subject(s)
Antineoplastic Agents/administration & dosage , G1 Phase Cell Cycle Checkpoints/drug effects , Isothiocyanates/administration & dosage , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isothiocyanates/pharmacology , Male , Mice , Mice, Transgenic , Organ Size/drug effects , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
15.
Cancer Res ; 76(5): 1146-1157, 2016 03 01.
Article in English | MEDLINE | ID: mdl-26676750

ABSTRACT

Ornithine decarboxylase (ODC) is a rate-limiting enzyme in the first step of polyamine biosynthesis that is associated with cell growth and tumor formation. Existing catalytic inhibitors of ODC have lacked efficacy in clinical testing or displayed unacceptable toxicity. In this study, we report the identification of an effective and nontoxic allosteric inhibitor of ODC. Using computer docking simulation and an in vitro ODC enzyme assay, we identified herbacetin, a natural compound found in flax and other plants, as a novel ODC inhibitor. Mechanistic investigations defined aspartate 44 in ODC as critical for binding. Herbacetin exhibited potent anticancer activity in colon cancer cell lines expressing high levels of ODC. Intraperitoneal or oral administration of herbacetin effectively suppressed HCT116 xenograft tumor growth and also reduced the number and size of polyps in a mouse model of APC-driven colon cancer (ApcMin/+). Unlike the well-established ODC inhibitor DFMO, herbacetin treatment was not associated with hearing loss. Taken together, our findings defined the natural product herbacetin as an allosteric inhibitor of ODC with chemopreventive and antitumor activity in preclinical models of colon cancer, prompting its further investigation in clinical trials.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavonoids/pharmacology , Ornithine Decarboxylase Inhibitors/pharmacology , Allosteric Regulation , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anticarcinogenic Agents/pharmacology , Eflornithine/pharmacology , Female , Flavonoids/toxicity , HCT116 Cells , Hearing Loss/chemically induced , Humans , Male , Mice , Mice, Inbred C57BL , Ornithine Decarboxylase/chemistry , Ornithine Decarboxylase/metabolism
16.
Carcinogenesis ; 36(11): 1372-80, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26271098

ABSTRACT

Solar ultraviolet irradiation is an environmental carcinogen that causes skin cancer. Caspase-7 is reportedly expressed at reduced levels in many cancers. The present study was designed to examine the role of caspase-7 in solar-simulated light (SSL)-induced skin cancer and to elucidate its underlying molecular mechanisms. Our study revealed that mice with genetic deficiency of caspase-7 are highly susceptible to SSL-induced skin carcinogenesis. Epidermal hyperplasia, tumor volume and the average number of tumors were significantly increased in caspase-7 knockout (KO) mice compared with SKH1 wild-type mice irradiated with SSL. The expression of cell proliferation markers, such as survivin and Ki-67, was elevated in SSL-irradiated skin of caspase-7 KO mice compared with those observed in SSL-exposed wild-type SKH1 mouse skin. Moreover, SSL-induced apoptosis was abolished in skin from caspase-7 KO mice. Two-dimensional gel electrophoresis, followed by matrix-assisted laser desorption/ionization-time-of-flight analysis of skin tissue lysates from SSL-irradiated SKH1 wild-type and caspase-7 KO mice revealed an aberrant induction of keratin-17 in caspase-7 KO mice. Immunohistochemical analysis of skin tumors also showed an increase of keratin-17 expression in caspase-7 KO mice compared with SKH1 wild-type mice. The expression of keratin-17 was also elevated in SSL-irradiated caspase-7 KO keratinocytes as well as in human basal cell carcinomas. The in vitro caspase activity assay showed keratin-17 as a substrate of caspase-7, but not caspase-3. Overall, our study demonstrates that genetic loss of caspase-7 promotes SSL-induced skin carcinogenesis by blocking caspase-7-mediated cleavage of keratin-17.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Caspase 7/genetics , Keratins/physiology , Radiation Injuries, Experimental/enzymology , Skin Neoplasms/enzymology , Sunlight/adverse effects , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Caspase 7/metabolism , Cells, Cultured , Epidermis/enzymology , Epidermis/pathology , Epidermis/radiation effects , Female , Gene Knockout Techniques , Keratinocytes/enzymology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proteolysis , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Burden
17.
Mol Carcinog ; 54(4): 322-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24974831

ABSTRACT

Lung cancer is a leading cause of death worldwide and MET amplification is a major therapeutic limitation in acquired-resistance lung cancer. We hypothesized that butein, a phytochemical, can overcome gefitinib-induced resistance by targeting both EGFR and MET in non-small cell lung cancer (NSCLC). To investigate the ability of butein to target EGFR and MET, we used in silico docking, a library of natural compounds and kinase assays. The effects of butein on growth, induction of apoptosis and expression of EGFR/MET signaling targets were examined in HCC827 (gefitinib-sensitive) and HCC827GR (gefitinib-resistant) NSCLC cells. Results were confirmed in vivo by a HCC827 or HCC827GR cell xenograft mouse model, each treated with vehicle, butein or gefitinib. Butein inhibited phosphorylation and kinase activity of EGFR and MET as well as soft agar colony formation and decreased viability of HCC827 and HCC827GR cells. Butein increased apoptosis-related protein expression in these cells. Results were confirmed by co-treatment with inhibitors of EGFR/MET or double knock-down. Finally, xenograft study results showed that butein strongly suppressed HCC827 and HCC827GR tumor growth. Immunohistochemical data suggest that butein inhibited Ki-67 expression. These results indicate that butein has potent anticancer activity and targets both EGFR and MET in acquired-resistance NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Chalcones/pharmacology , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinazolines/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/metabolism , Gefitinib , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism
18.
Mol Carcinog ; 54(12): 1596-604, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25328014

ABSTRACT

9-cis-UAB30 (UAB30) and Targretin are well-known retinoid X receptor (RXR) agonists. They were highly effective in decreasing the incidence of methylnitrosourea (MNU)-induced mammary cancers. However, whether the anti-mammary cancer effects of UAB30 or Targretin originate from the activation of RXR is unclear. In the present study, we hypothesized that UAB30 and Targretin not only affect RXR, but likely influence one or more off-target proteins. Virtual screening results suggest that Src is a potential target for UAB30 and Targretin that regulates extracellular matrix (ECM) molecules and cell motility and invasiveness. In vitro kinase assay data revealed that UAB30 or Targretin interacted with Src and attenuated its kinase activity. We found that UAB30 or Targretin substantially inhibited invasiveness and migration of MCF-7 and SK-BR-3 human breast cancer cells. We examined the effects of UAB30 and Targretin on the expression of matrix metalloproteinases (MMP)-9, which are known to play an essential role in tumor invasion. We show that activity and expression of MMP-9 were decreased by UAB30 or Targretin. Western blot data showed that UAB30 or Targretin decreased AKT and its substrate molecule p70(s6k), which are downstream of Src in MCF-7 and SK-BR-3 cells. Moreover, knocking down the expression of Src effectively reduced the sensitivity of SK-BR-3 cells to the inhibitory effects of UAB30 and Targretin on invasiveness. Taken together, our results demonstrate that UAB30 and Targretin each inhibit invasion and migration by targeting Src in human breast cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Fatty Acids, Unsaturated/pharmacology , Naphthalenes/pharmacology , Oncogene Protein pp60(v-src)/genetics , Retinoid X Receptors/agonists , Tetrahydronaphthalenes/pharmacology , Bexarotene , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Female , Humans , MCF-7 Cells , Matrix Metalloproteinase 9/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/prevention & control , Proto-Oncogene Proteins c-akt/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics
19.
J Biol Chem ; 289(52): 35839-48, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25368326

ABSTRACT

Non-small-cell lung cancer (NSCLC) is associated with diverse genetic alterations including mutation of epidermal growth factor receptor (EGFR). Isoliquiritigenin (ILQ), a chalcone derivative, possesses anticancer activities. In the present study, we investigated the effects of ILQ on the growth of tyrosine kinase inhibitor (TKI)-sensitive and -resistant NSCLC cells and elucidated its underlying mechanisms. Treatment with ILQ inhibited growth and induced apoptosis in both TKI-sensitive and -resistant NSCLC cells. ILQ-induced apoptosis was associated with the cleavage of caspase-3 and poly-(ADP-ribose)-polymerase, increased expression of Bim, and reduced expression of Bcl-2. In vitro kinase assay results revealed that ILQ inhibited the catalytic activity of both wild type and double mutant (L858R/T790M) EGFR. Treatment with ILQ inhibited the anchorage-independent growth of NIH3T3 cells stably transfected with either wild type or double-mutant EGFR with or without EGF stimulation. ILQ also reduced the phosphorylation of Akt and ERK1/2 in both TKI-sensitive and -resistant NSCLC cells, and attenuated the kinase activity of Akt1 and ERK2 in vitro. ILQ directly interacted with both wild type and double-mutant EGFR in an ATP-competitive manner. A docking model study showed that ILQ formed two hydrogen bonds (Glu-762 and Met-793) with wild type EGFR and three hydrogen bonds (Lys-745, Met-793, and Asp-855) with mutant EGFR. ILQ attenuated the xenograft tumor growth of H1975 cells, which was associated with decreased expression of Ki-67 and diminished phosphorylation of Akt and ERK1/2. Taken together, ILQ suppresses NSCLC cell growth by directly targeting wild type or mutant EGFR.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Mutation, Missense , NIH 3T3 Cells , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
Cancer Prev Res (Phila) ; 7(10): 1056-66, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25104643

ABSTRACT

Caffeic acid (3,4-dihydroxycinnamic acid) is a well-known phenolic phytochemical present in coffee and reportedly has anticancer activities. However, the underlying molecular mechanisms and targeted proteins involved in the suppression of carcinogenesis by caffeic acid are not fully understood. In this study, we report that caffeic acid significantly inhibits colony formation of human skin cancer cells and EGF-induced neoplastic transformation of HaCaT cells dose-dependently. Caffeic acid topically applied to dorsal mouse skin significantly suppressed tumor incidence and volume in a solar UV (SUV)-induced skin carcinogenesis mouse model. A substantial reduction of phosphorylation in mitogen-activated protein kinase signaling was observed in mice treated with caffeic acid either before or after SUV exposure. Caffeic acid directly interacted with ERK1/2 and inhibited ERK1/2 activities in vitro. Importantly, we resolved the cocrystal structure of ERK2 complexed with caffeic acid. Caffeic acid interacted directly with ERK2 at amino acid residues Q105, D106, and M108. Moreover, A431 cells expressing knockdown of ERK2 lost sensitivity to caffeic acid in a skin cancer xenograft mouse model. Taken together, our results suggest that caffeic acid exerts chemopreventive activity against SUV-induced skin carcinogenesis by targeting ERK1 and 2.


Subject(s)
Caffeic Acids/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Skin Neoplasms/metabolism , Animals , Antineoplastic Agents/chemistry , Binding, Competitive , Carcinogenesis , Crystallization , Female , Humans , Keratinocytes/drug effects , Keratinocytes/radiation effects , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Radiation-Induced/prevention & control , Phosphorylation , Recombinant Proteins/chemistry , Signal Transduction , Skin/drug effects , Skin/radiation effects , Skin Neoplasms/prevention & control , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL