Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
iScience ; 25(12): 105501, 2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36404918

ABSTRACT

Loss of epithelial integrity is associated with colorectal cancer (CRC) aggressiveness. Protein kinase C (PKC) is frequently implicated in human cancers, but the role of PKCγ in CRC remains poorly understood. Here, we show that PKCγ, a conventional PKC, is expressed in normal colonic epithelium, but this is lower in dedifferentiated CRC. PKCγ expression was downregulated by SNAI1 overexpression, and low PKCγ expression was associated with poor prognosis in patients with CRC. Transient or stable knockdown of PKCγ reduced E-cadherin expression in CRC cells. PKCγ knockdown enhanced proliferation, anchorage-independent cell growth, resistance to anti-cancer drugs, and in vivo tumor growth of DLD-1 cells. We have also identified phosphorylation substrates for PKCγ. Among them, ARHGEF18, a RhoA activator that stabilizes cell-cell junctions, was phosphorylated and stabilized by PKCγ. Thus, these results suggest that the downregulation of PKCγ decreases the epithelial property of CRC cells and enhances its malignant phenotypes.

2.
J Cell Mol Med ; 26(22): 5680-5689, 2022 11.
Article in English | MEDLINE | ID: mdl-36282887

ABSTRACT

Zic family member 5 (ZIC5) is a transcription factor that promotes the survival of several cancer cell types. As ZIC5 is expressed at minimal levels in normal human adult tissues, it is a potential therapeutic target. In this study, we screened a chemical library containing 3398 compounds that includes pre-existing drugs and compounds with known effects to identify ZIC5 inhibitors. In the first screening, 18 hit compounds decreased GFP intensity in melanoma A375 cells overexpressing GFP-tagged ZIC5. In the second screening, five compounds that attenuated ZIC5 protein levels in A375 cells were identified. Among them, LL-Z1640-2 and patulin selectively induced apoptosis in melanoma cells expressing ZIC5, while only inducing very low levels of apoptosis in normal human melanocytes, which have no detectable ZIC5 expression. LL-Z1640-2 and patulin also induced apoptosis in BRAF inhibitor-resistant melanoma, pancreatic cancer, cholangiocarcinoma and colorectal cancer cells. LL-Z1640-2- and patulin-mediated suppression of melanoma proliferation were rescued by ZIC5 overexpression. These results suggest that LL-Z1640-2 and patulin are promising compounds that decrease ZIC5 expression to induce apoptosis in cancer cells.


Subject(s)
Melanoma , Patulin , Adult , Humans , DNA-Binding Proteins/genetics , Patulin/pharmacology , Apoptosis , Melanoma/genetics , Family , Transcription Factors/genetics
3.
Biochem Biophys Rep ; 31: 101289, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35669984

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) and cholangiocarcinoma (CCA) are malignant tumors with poor prognosis because of the limited effectiveness of traditional chemotherapy and few effective molecular therapeutic agents. Here, we determined the essential roles of Zic family member 5 (ZIC5) in the survival of PDAC and CCA cells. The results showed that ZIC5 is strongly expressed in PDAC and CCA tissues, while ZIC5 expression is barely observed in most normal human adult tissues. Furthermore, ZIC5 expression is related to poor prognosis of patients with PDAC. ZIC5 knockdown via small interfering RNA decreased the phosphorylation of signal transducer and activator of transcription 3 (STAT3), a protein that is associated with PDAC and CCA aggressiveness. However, ZIC5 knockdown induced cell death regardless of STAT3 activation, which is promoted by interleukin (IL) -6, a factor associated with inflammation. Furthermore, knockdown of ZIC5 in PDAC and CCA cells additively or synergistically induced apoptosis with the anti-cancer drug gemcitabine. Thus, ZIC5 constitutes a potential therapeutic target for the treatment of PDAC and CCA.

4.
Nat Commun ; 12(1): 5930, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34635648

ABSTRACT

Malignancy is associated with changes in cell mechanics that contribute to extensive cell deformation required for metastatic dissemination. We hypothesized that the cell-intrinsic physical factors that maintain epithelial cell mechanics could function as tumor suppressors. Here we show, using optical tweezers, genetic interference, mechanical perturbations, and in vivo studies, that epithelial cells maintain higher plasma membrane (PM) tension than their metastatic counterparts and that high PM tension potently inhibits cancer cell migration and invasion by counteracting membrane curvature sensing/generating BAR family proteins. This tensional homeostasis is achieved by membrane-to-cortex attachment (MCA) regulated by ERM proteins, whose disruption spontaneously transforms epithelial cells into a mesenchymal migratory phenotype powered by BAR proteins. Consistently, the forced expression of epithelial-mesenchymal transition (EMT)-inducing transcription factors results in decreased PM tension. In metastatic cells, increasing PM tension by manipulating MCA is sufficient to suppress both mesenchymal and amoeboid 3D migration, tumor invasion, and metastasis by compromising membrane-mediated mechanosignaling by BAR proteins, thereby uncovering a previously undescribed mechanical tumor suppressor mechanism.


Subject(s)
Cell Membrane/chemistry , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Homeostasis/genetics , Mechanotransduction, Cellular/genetics , Biomechanical Phenomena , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Neoplasm Invasiveness , Optical Tweezers , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Surface Tension , Transcription Factors/genetics , Transcription Factors/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
5.
Biochem Biophys Res Commun ; 527(4): 1050-1056, 2020 07 05.
Article in English | MEDLINE | ID: mdl-32439160

ABSTRACT

Phospholipids are distributed asymmetrically in the plasma membrane (PM) of mammalian cells. Phosphatidylinositol (PI) and its phosphorylated forms are primarily located in the inner leaflet of the PM. Among them, phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is a well-known substrate for phospholipase C (PLC) or phosphoinositide-3 kinase, and is also a regulator for the actin cytoskeleton or ion channels. Although functions of PI(4,5)P2 in the inner leaflet are well characterized, those in the outer leaflet are poorly understood. Here, PI(4,5)P2 was detected in the cell surface of non-permeabilized cells by anti-PI(4,5)P2 antibodies and the pleckstrin-homology (PH) domain of PLCδ1 that specifically binds PI(4,5)P2. Cell surface PI(4,5)P2 signal was universally detected in various cell lines and freshly isolated mouse bone marrow cells and showed a punctate pattern in a cholesterol, sphingomyelin, and actin polymerization-dependent manner. Furthermore, blocking cell surface PI(4,5)P2 by the addition of anti-PI(4,5)P2 antibody or the PH domain of PLCδ1 inhibited cell attachment, spreading, and migration. Taken together, these results indicate a unique localization of PI(4,5)P2 in the outer leaflet that may have a crucial role in cell attachment, spreading, and migration.


Subject(s)
Cell Adhesion , Cell Membrane/metabolism , Cell Movement , Phosphatidylinositol 4,5-Diphosphate/metabolism , Actins/metabolism , Cell Line , Cholesterol/metabolism , Humans , Phosphatidylinositol 4,5-Diphosphate/analysis , Pleckstrin Homology Domains , Sphingomyelins/metabolism , Type C Phospholipases/analysis , Type C Phospholipases/metabolism
6.
FASEB J ; 33(3): 3851-3869, 2019 03.
Article in English | MEDLINE | ID: mdl-30508500

ABSTRACT

The BRAF inhibitor PLX4032 is effective in treating BRAF-mutated melanoma; however, because drug resistance develops in most cases, it is critical to develop a new strategy for inhibiting drug-resistant melanoma growth. The melanoma-associated membrane glycoprotein CD63 is involved in cell proliferation and metastasis. Here, we found that cell surface CD63 suppresses the proliferation of human melanoma cells and PLX4032-resistant cells. Endogenous CD63 protein levels were negatively correlated with PLX4032 resistance of human melanoma cell lines. CD63 overexpression in these cells, in which endogenous CD63 levels are low, suppressed cell proliferation under PLX4032 treatment. The cell surface levels and average molecular mass of CD63 were increased with PLX4032 treatment because of the up-regulated polylactosamine modification caused by induced ß1,3- N-acetylglucosaminyltransferase 2 expression, which is involved in polylactosamine synthesis. Forced cell surface localization of CD63 led to reduced melanoma cell proliferation without PLX4032 treatment. CD63 overexpression in PLX4032-resistant cells, in which CD63 levels were lower and cell surface polylactosamine levels were higher than those in parental cells, effectively suppressed proliferation. Our study shows the potential of CD63 to sensitize melanoma cells to PLX4032 and to reduce the proliferation of PLX4032-resistant cells.-Kudo, K., Yoneda, A., Sakiyama, D., Kojima, K., Miyaji, T., Yamazaki, M., Yaita, S., Hyodo, T., Satow, R., Fukami, K. Cell surface CD63 increased by up-regulated polylactosamine modification sensitizes human melanoma cells to the BRAF inhibitor PLX4032.


Subject(s)
Amino Sugars/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Melanoma/metabolism , Polysaccharides/metabolism , Protein Processing, Post-Translational , Tetraspanin 30/metabolism , Vemurafenib/pharmacology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/drug effects , Humans , Tetraspanin 30/genetics
7.
Cancer Sci ; 108(12): 2405-2412, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29024195

ABSTRACT

Identification of specific drug targets is very important for cancer therapy. We recently identified zinc finger protein of the cerebellum 5 (ZIC5) as a factor that promotes melanoma aggressiveness by platelet-derived growth factor D (PDGFD) expression. However, its roles in other cancer types remain largely unknown. Here we determined the roles of ZIC5 in prostate cancer (PCa) and colorectal cancer (CRC) cells. Results showed that ZIC5 was highly expressed in CRC and dedifferentiated PCa tissues, whereas little expression was observed in relevant normal tissues. Knockdown of ZIC5 decreased proliferation of several PCa and CRC cell lines with induction of cell death. ZIC5 knockdown significantly suppressed PDGFD expression transcriptionally, and PDGFD suppression also decreased proliferation of PCa and CRC cell lines. In addition, suppression of ZIC5 or PDGFD expression decreased levels of phosphorylated focal adhesion kinase (FAK) and signal transducer and activator of transcription 3 (STAT3) which are associated with PCa and CRC aggressiveness. Furthermore, knockdown of ZIC5 or PDGFD enhanced death of PCa and CRC cells induced by the anti-cancer drugs docetaxel or oxaliplatin, respectively. These results suggest that ZIC5 and PDGFD promote survival of PCa and CRC cells by enhancing FAK and STAT3 activity, and that the roles of ZIC5 are consistent across several cancer types.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transcription Factors/metabolism , Adult , Aged , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , DNA-Binding Proteins , Female , Humans , Male , Middle Aged
8.
Biochem Biophys Res Commun ; 488(4): 578-583, 2017 07 08.
Article in English | MEDLINE | ID: mdl-28528980

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of cancer-related death worldwide. Kirsten rat sarcoma viral oncogene homolog (KRAS) is frequently mutated in CRC, and KRAS mutations promote cell motility, growth, and survival. We previously revealed that the expression of phospholipase C (PLC) δ1, one of the most basal PLCs, is down-regulated in colon adenocarcinoma, and that the KRAS signaling pathway suppresses PLCδ1 expression. Although recent studies revealed that KRAS mutations activate autophagy in cancer cells, a relation between PLCδ1 and autophagy remains unclear. Here, we found that PLCδ1 overexpression suppresses the formation of autophagosomes, which are key structures of autophagy, whereas endogenous PLCδ1 knockdown increases autophagosome formation in CRC cells. We also showed that PLCδ1 overexpression promotes cell death under nutrient deprivation. Furthermore, PLCδ1 overexpression suppresses the autophagy induced by the anti-cancer drug oxaliplatin and promotes cell death under oxaliplatin treatment. These data suggest that PLCδ1 negatively regulates autophagy, and PLCδ1 suppression contributes to the tolerance of CRC cells harboring KRAS mutations to nutrient deprivation and anti-cancer drug treatment.


Subject(s)
Autophagy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Phospholipase C delta/metabolism , Colorectal Neoplasms/metabolism , HCT116 Cells , Humans , Tumor Cells, Cultured
9.
Cancer Res ; 77(2): 366-377, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27671679

ABSTRACT

Insights into mechanisms of drug resistance could extend the efficacy of cancer therapy. To probe mechanisms in melanoma, we performed siRNA screening of genes that mediate the development of neural crest cells, from which melanocytes are derived. Here, we report the identification of ZIC5 as a mediator of melanoma drug resistance. ZIC5 is a transcriptional suppressor of E-cadherin expressed highly in human melanoma. ZIC5 enhanced melanoma cell proliferation, survival, drug resistance, in vivo growth and metastasis. Microarray analysis revealed that ZIC5 downstream signaling included PDGFD and FAK activation, which contributes to drug resistance by enhancing STAT3 activation. Silencing of ZIC5 or PDGFD enhanced the apoptotic effects of BRAF inhibition and blocked survival of melanoma cells resistant to BRAF inhibitors. Furthermore, inhibition of FAK or STAT3 suppressed expression of ZIC5, which was positively regulated by PDGFD, FAK, and STAT3 in a positive feedback loop. Taken together, our results identify ZIC5 and PDGFD as candidate therapeutic targets to overcome drug resistance in melanoma. Cancer Res; 77(2); 366-77. ©2016 AACR.


Subject(s)
Drug Resistance, Neoplasm/physiology , Focal Adhesion Kinase 1/metabolism , Lymphokines/metabolism , Melanoma/pathology , Platelet-Derived Growth Factor/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Blotting, Western , Cell Line, Tumor , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic/physiology , Humans , Melanoma/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
10.
Proc Natl Acad Sci U S A ; 111(37): 13505-10, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25197077

ABSTRACT

Colorectal cancer (CRC) is one of the most common causes of cancer-related deaths worldwide, and Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations in CRC predict the ineffectiveness of EGF receptor-targeted therapy. Previous transcriptional microarray analysis suggests the association between phospholipase Cδ1 (PLCδ1) expression and KRAS mutation status in CRC. However, both the roles and the regulatory mechanisms of PLCδ1 in CRC are not known. Here, we found that the expression of PLCδ1, one of the most basal PLCs, is down-regulated in CRC specimens compared with normal colon epithelium by immunohistochemistry. Furthermore, we examined the roles of PLCδ1 in CRC cell lines that harbor an activating KRAS mutation. Ectopic expression of PLCδ1 in CRC cells induced the expression of E-cadherin, whereas knockdown of PLCδ1 repressed the expression of E-cadherin. Moreover, the overexpression of PLCδ1 suppressed the expression of several mesenchymal genes and reduced cell motility, invasiveness, and in vivo tumorigenicity of SW620 CRC cells. We also showed that PLCδ1 expression is repressed by the KRAS/mitogen-activated protein kinase kinase (MEK) pathway. Furthermore, PLCδ1 suppressed the phosphorylation of extracellular signal-regulated kinase (ERK)1/2 through E-cadherin induction in CRC cells, suggesting the presence of a negative regulatory loop between KRAS/MEK/ERK signaling and PLCδ1. These data indicate that PLCδ1 has tumor-suppressive functions in CRC through E-cadherin induction and KRAS/MEK/ERK signal attenuation.


Subject(s)
Cadherins/metabolism , Carcinogenesis/pathology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Phospholipase C delta/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antigens, CD , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Invasiveness , Phenotype , Phosphorylation , Signal Transduction
11.
Oncology ; 86(2): 94-103, 2014.
Article in English | MEDLINE | ID: mdl-24457375

ABSTRACT

OBJECTIVE: Osteosarcoma (OS) is the most frequent primary malignant bone tumor in children and young adults. Although the introduction of combined neoadjuvant chemotherapy has significantly prolonged survival, the outcome for OS patients showing a poor response to chemotherapy is still unfavorable. In order to develop new therapeutic approaches, elucidation of the entire molecular pathway regulating OS cell proliferation would be desirable. METHODS: MicroRNA (miRNA) are highly conserved noncoding RNA that play important roles in the development and progression of various other cancers. Using miRNA microarrays capable of detecting a known number of 933 miRNA, 108 miRNA were found to be commonly expressed in 24 samples of OS tissue and subjected to a cell proliferation assay. RESULTS: We found that inhibition of 5 let-7 family miRNA (hsa-let-7a, b, f, g and i) significantly suppressed the proliferation of OS cells. Using a quantitative shotgun proteomics approach, we also found that the let-7 family miRNA regulated the expression of vimentin and serpin H1 proteins. CONCLUSIONS: Our present results indicate the involvement of let-7 family miRNA in regulation of the cell proliferation as well as epithelial-mesenchymal transition of OS. Thus, let-7 family miRNA may potentially provide novel targets for the development of therapeutic strategies against OS.


Subject(s)
Bone Neoplasms/metabolism , MicroRNAs/metabolism , Osteosarcoma/metabolism , Adolescent , Adult , Amino Acid Sequence , Bone Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Child , Female , Gene Expression Regulation, Neoplastic , HSP47 Heat-Shock Proteins/chemistry , HSP47 Heat-Shock Proteins/genetics , HSP47 Heat-Shock Proteins/metabolism , Humans , Male , MicroRNAs/genetics , Molecular Sequence Data , Osteosarcoma/genetics , RNA Interference , Transcriptome , Vimentin/chemistry , Vimentin/genetics , Vimentin/metabolism , Young Adult
12.
Biochem Pharmacol ; 86(10): 1419-29, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24035834

ABSTRACT

Tumor dissemination and invasive behavior are associated with a majority of cancer-related mortality cases. Loss of E-cadherin, which is caused by several tumor-promoting factors, is associated with metastasis and poor prognosis in many neoplasms. In this study, we aimed to identify small molecule compounds that restore the expression of E-cadherin, because these molecules are most likely to suppress tumor malignancy by restoring E-cadherin function and/or by inhibiting signals that suppress E-cadherin expression. Here, we developed a fluorescence screen system based on E-cadherin expression. A pilot drug library screen revealed that methotrexate (MTX) strongly induces E-cadherin expression in a colorectal cancer cell line, SW620. From the screen for 9600 compounds, we identified 9 hit compounds, which restored the expression of E-cadherin in SW620 and/or a melanoma cell line, SK-MEL-28. We confirmed that MTX and the other identified compounds transcriptionally promote E-cadherin expression. Among these, 2 compounds suppressed migration/invasion capacity in colorectal cancer cells and 3 in melanoma cells. A compound reduced SW620 migration and invasion with subtle effects on cell viability in SW620, SK-MEL-28, and a non-tumor cell line, HaCaT, with decrease in AKT and ERK1/2 protein levels. One of the other compounds reduced SK-MEL-28 cell migration and invasion and affected the viability only of SW620 and SK-MEL-28 cells but not HaCaT cells. These results suggest that these compounds would be attractive lead molecules as anti-metastasis agents.


Subject(s)
Antineoplastic Agents/chemistry , Cadherins/biosynthesis , Neoplasm Invasiveness/pathology , Small Molecule Libraries/chemistry , Adenocarcinoma , Antineoplastic Agents/pharmacology , Cell Line , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Colorectal Neoplasms , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Melanoma , Methotrexate/chemistry , Methotrexate/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Small Molecule Libraries/pharmacology
13.
Gastroenterology ; 142(3): 572-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22155184

ABSTRACT

BACKGROUND & AIMS: Loss of promyelocytic leukemia protein (PML) nuclear body (NB) formation has been reported in colorectal and other solid tumors. However, genetic alteration of PML is rarely observed in these tumors; the exact mechanisms that mediate loss of PML function are not known. METHODS: We previously used a comprehensive shotgun mass spectrometry approach to identify PML as 1 of 70 proteins that coimmunoprecipitate with anti-T-cell factor 4 in DLD-1 and HCT116 colorectal cancer cell lines; we investigated the effects of altered ß-catenin expression on PML function in these cells. RESULTS: ß-catenin specifically interacted with the product of PML transcript variant IV (PML-IV) through the armadillo repeat domain of ß-catenin. Overexpression of ß-catenin in colorectal cancer cells disrupted the subcellular compartmentalization of PML-IV, whereas knockdown of ß-catenin restored formation of PML-NB. Modification of PML by the small ubiquitin-related modifier (SUMO) is required for proper assembly of PML-NB. ß-catenin inhibited Ran-binding protein 2-mediated SUMOylation of PML-IV. CONCLUSIONS: ß-catenin interacts with PML isoform IV and disrupts PML-IV function and PML-NB formation by inhibiting Ran-binding protein 2-mediated SUMO modification of PML-IV. These findings indicate the involvement of a posttranslational mechanism in disruption of PML-NB organization in cancer cells and provide more information about the oncogenic functions of ß-catenin.


Subject(s)
Colorectal Neoplasms/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Lysine , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Binding , Protein Isoforms , RNA Interference , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Transcription Factor 4 , Transcription Factors/genetics , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics
14.
Diabetes ; 60(7): 1926-37, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21617180

ABSTRACT

OBJECTIVE: Regulation of obesity development is an important issue to prevent metabolic syndromes. Gene-disrupted mice of phospholipase Cδ1 (PLCδ1), a key enzyme of phosphoinositide turnover, seemed to show leanness. Here we examined whether and how PLCδ1 is involved in obesity. RESEARCH DESIGN AND METHODS: Weight gain, insulin sensitivity, and metabolic rate in PLCδ1(-/-) mice were compared with PLCδ1(+/-) littermate mice on a high-fat diet. Thermogenic and adipogenetic potentials of PLCδ1(-/-) immortalized brown adipocytes and adipogenesis of PLCδ1-knockdown (KD) 3T3L1 cells, or PLCδ1(-/-) white adipose tissue (WAT) stromal-vascular fraction (SVF) cells, were also investigated. RESULTS: PLCδ1(-/-) mice showed marked decreases in weight gain and mass of epididymal WAT and preserved insulin sensitivity compared with PLCδ1(+/-) mice on a high-fat diet. In addition, PLCδ1(-/-) mice have a higher metabolic rate such as higher oxygen consumption and heat production. When control immortalized brown adipocytes were treated with thermogenic inducers, expression of PLCδ1 was decreased and thermogenic gene uncoupling protein 1 (UCP1) was upregulated to a greater extent in PLCδ1(-/-) immortalized brown adipocytes. In contrast, ectopic expression of PLCδ1 in PLCδ1(-/-) brown adipocytes induced a decrease in UCP expression, indicating that PLCδ1 negatively regulates thermogenesis. Importantly, accumulation of lipid droplets was severely decreased when PLCδ1-KD 3T3L1 cells, or PLCδ1(-/-) WAT SVF cells, were differentiated, whereas differentiation of PLCδ1(-/-) brown preadipocytes was promoted. CONCLUSIONS: PLCδ1 has essential roles in thermogenesis and adipogenesis and thereby contributes to the development of obesity.


Subject(s)
Adipogenesis/physiology , Obesity/prevention & control , Phospholipase C delta/genetics , Thermogenesis/physiology , 3T3-L1 Cells , Adipocytes, Brown/physiology , Adipose Tissue, White/growth & development , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dietary Fats/administration & dosage , Gene Expression Profiling , Insulin , Ion Channels/genetics , Mice , Mice, Nude , Mitochondrial Proteins/genetics , NFATC Transcription Factors/physiology , Phospholipase C delta/deficiency , Protein Kinase C/physiology , Protein Kinase C-epsilon/physiology , Uncoupling Protein 1
15.
Jpn J Clin Oncol ; 41(7): 847-53, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21576114

ABSTRACT

OBJECTIVE: Emerging molecular targeting therapeutics have been incorporated into the management of advanced renal cell carcinoma; however, their efficacy remains limited. The aim of this study was to catalog potential therapeutic target molecules for renal cell carcinoma. METHODS: We first selected genes up-regulated in clear cell renal cell carcinoma relative to surrounding normal kidney tissues in 10 patients (Study Cohort) using high-density exon arrays that detect all potential transcripts predicted in the human genome. The selected genes were subjected to independent validation in another set of 10 patients (Validation Cohort) using real-time reverse transcriptase polymerase chain reaction and functional screening using small interfering RNA in six clear cell renal cell carcinoma cell lines. RESULTS: We identified 164 genes whose expression was significantly elevated in clear cell renal cell carcinoma (P< 0.0001 [Student's t-test] and at least a 3-fold change in transcription signal). We finally extracted 33 genes required for maintaining cell proliferation in at least two clear cell renal cell carcinoma cell lines. The 33 genes included 13 genes known to be associated with the development/progression of renal cell carcinoma, including CAIX and FLT-1, confirming the robustness of the current strategy. CONCLUSIONS: Through a combination of genome-wide expression and functional assays, we identified a set of genes with high potential as targets for drug development. This method is rapid and comprehensive and could be applied to the discovery of diagnostic biomarkers and therapeutic targets for cancers other than clear cell renal cell carcinoma.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Biomarkers, Tumor/genetics , Kidney Neoplasms/genetics , Molecular Targeted Therapy , Transcription, Genetic , Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Cell Line, Tumor , Cell Proliferation , Exons , Female , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Genome-Wide Association Study , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , RNA, Small Interfering/analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
16.
J Biol Chem ; 285(34): 26289-94, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20566648

ABSTRACT

Wnt signaling pathways play important roles in various stages of developmental events and several aspects of adult homeostasis. Aberrant activation of Wnt signaling has also been associated with several types of cancer. We have recently identified Traf2- and Nck-interacting kinase (TNIK) as a novel activator of Wnt signaling through a comprehensive proteomic approach in human colorectal cancer cell lines. TNIK is an activating kinase for T-cell factor-4 (TCF4) and essential for the beta-catenin-TCF4 transactivation and colorectal cancer growth. Here, we report the essential role of TNIK in Wnt signaling during Xenopus development. We found that Xenopus TNIK (XTNIK) was expressed maternally and that the functional knockdown of XTNIK by catalytically inactive XTNIK (K54R) or antisense morpholino oligonucleotides resulted in significant malformations with a complete loss of head and axis structures. XTNIK enhanced beta-catenin-induced axis duplication and the expression of beta-catenin-TCF target genes, whereas knockdown of XTNIK inhibited it. XTNIK was recruited to the promoter region of beta-catenin-TCF target genes in a beta-catenin-dependent manner. These results demonstrate that XTNIK is an essential factor for the transcriptional activity of the beta-catenin-TCF complex and dorsal axis determination in Xenopus embryos.


Subject(s)
Body Patterning , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , Xenopus Proteins/physiology , Abnormalities, Multiple/etiology , Animals , Embryo, Nonmammalian , Germinal Center Kinases , Transcription, Genetic , Xenopus/embryology , beta Catenin
17.
Cancer Res ; 70(12): 5024-33, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20530691

ABSTRACT

T-cell factor-4 (TCF4) is a transcription factor essential for maintaining the undifferentiated status and self-renewal of intestinal epithelial cells. It has therefore been considered that constitutive activation of TCF4 by aberrant Wnt signaling is a major force driving colorectal carcinogenesis. We previously identified Traf2- and Nck-interacting kinase (TNIK) as one of the proteins that interact with TCF4 in colorectal cancer cells, but its functional significance has not been elucidated. Here, we report that TNIK is an activating kinase for TCF4 and essential for colorectal cancer growth. TNIK, but not its catalytically inactive mutant, phosphorylated the conserved serine 154 residue of TCF4. Small interfering RNA targeting TNIK inhibited the proliferation of colorectal cancer cells and the growth of tumors produced by injecting colorectal cancer cells s.c. into immunodeficient mice. The growth inhibition was abolished by restoring the catalytic domain of TNIK, thus confirming that its enzyme activity is essential for the maintenance of colorectal cancer growth. Several ATP-competing kinase inhibitors have been applied to cancer treatment and have shown significant activity. Our findings suggest TNIK as a feasible target for pharmacologic intervention to ablate aberrant Wnt signaling in colorectal cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 2/metabolism , Wnt Proteins/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Cell Proliferation , Chromatography, Liquid , Colony-Forming Units Assay , Colorectal Neoplasms/genetics , Female , Germinal Center Kinases , Humans , Immunoblotting , Immunoenzyme Techniques , Immunoprecipitation , Luciferases/metabolism , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TNF Receptor-Associated Factor 2/genetics , Tumor Cells, Cultured , Two-Hybrid System Techniques , Wnt Proteins/genetics , beta Catenin/genetics , beta Catenin/metabolism
18.
Clin Cancer Res ; 16(9): 2518-28, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20388846

ABSTRACT

PURPOSE: The outcome of patients with advanced hepatocellular carcinoma (HCC) has remained unsatisfactory. Patients with HCC suffer from chronic hepatitis or liver cirrhosis, and their reserve liver function is often limited. EXPERIMENTAL DESIGN: To develop new therapeutic agents that act specifically on HCC but interfere only minimally with residual liver function, we searched for genes that were upregulated in 20 cases of HCC [namely, discovery sets 1 (n = 10) and 2 (n = 10)] in comparison with corresponding nontumorous liver and a panel representing normal organs using high-density microarrays capable of detecting all exons in the human genome. RESULTS: Eleven transcripts whose expression was significantly increased in HCC were subjected to siRNA-based secondary screening of genes required for HCC cell proliferation as well as quantitative reverse transcription-PCR analysis [validation sets 1 (n = 20) and 2 (n = 44)] and immunohistochemistry (n = 19). We finally extracted four genes, AKR1B10, HCAP-G, RRM2, and TPX2, as candidate therapeutic targets for HCC. siRNA-mediated knockdown of these candidate genes inhibited the proliferation of HCC cells and the growth of HCC xenografts transplanted into immunodeficient mice. CONCLUSIONS: The four genes we identified were highly expressed in HCC, and HCC cells are highly dependent on these genes for proliferation. Although many important genes must have been overlooked, the selected genes were biologically relevant. The combination of genome-wide expression and functional screening described here is a rapid and comprehensive approach that could be applied in the identification of therapeutic targets in any type of human malignancy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Genome, Human/genetics , Genome-Wide Association Study/methods , Liver Neoplasms/genetics , Aged , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease/genetics , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
19.
Mol Cancer Ther ; 9(3): 535-44, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20159990

ABSTRACT

Pulmonary metastasis is the most significant prognostic determinant for osteosarcoma, but methods for its prediction and treatment have not been established. Using oligonucleotide microarrays, we compared the global gene expression of biopsy samples between seven osteosarcoma patients who developed pulmonary metastasis within 4 years after neoadjuvant chemotherapy and curative resection, and 12 patients who did not relapse. We identified argininosuccinate synthetase (ASS) as a gene differentially expressed with the highest statistical significance (Welch's t test, P = 2.2 x 10(-5)). Immunohistochemical analysis of an independent cohort of 62 osteosarcoma cases confirmed that reduced expression of ASS protein was significantly correlated with the development of pulmonary metastasis after surgery (log-rank test, P < 0.05). Cox regression analysis revealed that ASS was the sole significant predictive factor (P = 0.039; hazard ratio, 0.319; 95% confidence interval, 0.108-0.945). ASS is one of the enzymes required for the production of a nonessential amino acid, arginine. We showed that osteosarcoma cells lacking ASS expression were auxotrophic for arginine and underwent G(0)-G(1) arrest in arginine-free medium, suggesting that an arginine deprivation therapy could be effective in patients with osteosarcoma. Recently, phase I and II clinical trials in patients with melanoma and hepatocellular carcinoma have shown the safety and efficacy of plasma arginine depletion by stabilized arginine deiminase. Our data indicate that in patients with osteosarcoma, reduced expression of ASS is not only a novel predictive biomarker for the development of metastasis, but also a potential target for pharmacologic intervention.


Subject(s)
Argininosuccinate Synthase/genetics , Bone Neoplasms/diagnosis , Lung Neoplasms/diagnosis , Lung Neoplasms/secondary , Osteosarcoma/diagnosis , Adolescent , Adult , Argininosuccinate Synthase/metabolism , Argininosuccinate Synthase/physiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Child , Down-Regulation/physiology , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Oligonucleotide Array Sequence Analysis , Osteosarcoma/genetics , Osteosarcoma/mortality , Osteosarcoma/pathology , Prognosis , Tumor Cells, Cultured , Young Adult
20.
Cancer Sci ; 100(12): 2268-74, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19725836

ABSTRACT

Osteosarcoma (OS) is the most frequent primary malignant bone tumor of children and young adults. Although the introduction of combined neoadjuvant chemotherapy has markedly improved survival, the outcome of OS patients with distant metastasis and/or poor response to chemotherapy is still unsatisfactory. Therefore there is a need to develop new therapeutic agents that suppress OS cell proliferation with higher efficacy. The protein kinases are a family of genes that play critical roles in various signaling pathways. Some cancer cells show addiction to constitutive activation of certain signaling pathways for proliferation and survival. To identify new drug targets for OS, we screened a panel of small interfering RNAs (siRNAs) that target 691 genes encoding human protein kinases and related proteins. We found that different constructs of siRNA specifically targeting polo-like 1 kinase (PLK1) significantly caused mitotic cell cycle arrest and subsequent apoptotic cell death in a variety of OS cell lines. siRNA targeting PLK1 also suppressed the growth of OS xenografts established in immunodeficient mice. Recently, phase I clinical trials of PLK1 chemical inhibitors have been reported. Our results indicate that PLK1 is a promising molecular target for pharmacologic intervention in OS.


Subject(s)
Bone Neoplasms/therapy , Cell Cycle Proteins/antagonists & inhibitors , Osteosarcoma/therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , RNA, Small Interfering/therapeutic use , Animals , Bone Neoplasms/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Osteosarcoma/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...