Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cancer Sci ; 113(1): 170-181, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34714577

ABSTRACT

The aryl hydrocarbon receptor (AHR) pathway modulates the immune system in response to kynurenine, an endogenous tryptophan metabolite. IDO1 and TDO2 catalyze kynurenine production, which promotes cancer progression by compromising host immunosurveillance. However, it is unclear whether the AHR activation regulates the malignant traits of cancer such as metastatic capability or cancer stemness. Here, we carried out systematic analyses of metabolites in patient-derived colorectal cancer spheroids and identified high levels of kynurenine and TDO2 that were positively associated with liver metastasis. In a mouse colon cancer model, TDO2 expression substantially enhanced liver metastasis, induced AHR-mediated PD-L1 transactivation, and dampened immune responses; these changes were all abolished by PD-L1 knockout. In patient-derived cancer spheroids, TDO2 or AHR activity was required for not only the expression of PD-L1, but also for cancer stem cell (CSC)-related characteristics and Wnt signaling. TDO2 was coexpressed with both PD-L1 and nuclear ß-catenin in colon xenograft tumors, and the coexpression of TDO2 and PD-L1 was observed in clinical colon cancer specimens. Thus, our data indicate that the activation of the TDO2-kynurenine-AHR pathway facilitates liver metastasis of colon cancer via PD-L1-mediated immune evasion and maintenance of stemness.


Subject(s)
B7-H1 Antigen/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Colonic Neoplasms/pathology , Dioxygenases/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Neoplastic Stem Cells/pathology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Line, Tumor , Colonic Neoplasms/metabolism , Humans , Kynurenine , Liver Neoplasms/metabolism , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Tumor Escape , Up-Regulation , Wnt Signaling Pathway
2.
3.
Oncotarget ; 8(35): 57938-57947, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938528

ABSTRACT

MELK play critical roles in human carcinogenesis through activation of cell proliferation, inhibition of apoptosis and maintenance of stemness. Therefore, MELK is a promising therapeutic target for a wide range of cancers. Although p21 is a well-known p53-downstream gene, we found that treatment with a potent MELK inhibitor, OTS167, could induce p21 protein expression in cancer cell lines harboring loss-of-function TP53 mutations. We also confirmed that MELK knockdown by siRNA induced the p21 expression in p53-deficient cancer cell lines and caused the cell cycle arrest at G1 phase. Further analysis indicated that FOXO1 and FOXO3, two known transcriptional regulators of p21, were phosphorylated by MELK and thus be involved in the induction of p21 after MELK inhibition. Collectively, our herein findings suggest that MELK inhibition may be effective for human cancers even if TP53 is mutated.

4.
Neoplasia ; 19(1): 43-53, 2017 01.
Article in English | MEDLINE | ID: mdl-28110670

ABSTRACT

We previously reported that overexpression of an O-type glycosyltransferase, GALNT6 (polypeptide N-acetylgalactosaminyltransferase 6) played critical roles in mammary carcinogenesis. To further investigate the biological function of GALNT6, we screened a substrate protein(s) of GALNT6 using a VVA (Vicia villosa agglutinin) lectin (specific to GalNAc-Ser/Thr) pull-down method followed by mass spectrometry analysis. Here we report GRP78 (glucose-regulated protein 78, also known as HSPA5, heat shock 70 kDa protein 5), which is highly expressed in cancer cells and indicated to play important roles in various cellular processes including ER (endoplasmic reticulum) stress and autophagy, as a novel substrate of GALNT6. We found that GALNT6-induced O-glycosylation is critical for the stability of GRP78, its subcellular localization in ER, and its anti-apoptotic function. Furthermore, we demonstrated that overexpression of GRP78 could be important for Golgi-to-ER relocation of GALNT6. Collectively, our study revealed biological significances of O-glycosylation of GRP78 protein, which might play significant roles in the survival of cancer cells, and thus provided a new insight in cancer cell death and useful information for development of anti-cancer treatment targeting the GALNT6-GRP78 pathway.


Subject(s)
Apoptosis , Heat-Shock Proteins/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Stress, Physiological , Cell Line, Tumor , Cell Survival , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression , Gene Knockout Techniques , Glycosylation , Golgi Apparatus/metabolism , Heat-Shock Proteins/genetics , Humans , Models, Biological , N-Acetylgalactosaminyltransferases/genetics , Protein Binding , Protein Stability , Protein Transport , RNA Interference , Substrate Specificity , Polypeptide N-acetylgalactosaminyltransferase
5.
Medchemcomm ; 8(1): 73-80, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-30108692

ABSTRACT

Cancer stem cells (CSCs) are indicated to play critical roles in drug resistance, recurrence, and metastasis of cancer. Although molecular targeted therapies have contributed to the improvement of cancer treatments by targeting vulnerable pathways indispensable to the proliferation and survival of cancer cells, no relevant therapeutic modalities targeting CSCs have been developed yet. This review focuses on MELK (maternal embryonic leucine zipper kinase), TOPK (T-lymphokine-activated killer cell-originated protein kinase), and TTK (tyrosine threonine kinase), which are over-expressed frequently in human cancers and play indispensable roles in the development and maintenance of cancer stem cells. In addition, we will discuss recently developed small molecules for these protein targets, which have shown remarkable anti-tumor efficacies in several preclinical studies.

6.
Int J Oncol ; 49(3): 868-76, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27572108

ABSTRACT

We identified phosphatidylinositol glycan anchor biosynthesis, class X (PIGX), which plays a critical role in the biosynthetic pathway of glycosylphosphatidylinositol (GPI)-anchor motif, to be upregulated highly and frequently in breast cancer cells. Knockdown of PIGX as well as reticulocalbin 1 (RCN1) and reticulocalbin 2 (RCN2), which we found to interact with PIGX and was indicated to regulate calcium-dependent activities, significantly suppressed the growth of breast cancer cells. We also identified PIGX to be a core protein in an RCN1/PIGX/RCN2 complex. Microarray analysis revealed that the expression of two putative tumor suppressor genes, Zic family member 1 (ZIC1) and EH-domain containing 2 (EHD2), were upregulated commonly in cells in which PIGX, RCN1, or RCN2 was knocked down, suggesting that this RCN1/PIGX/RCN2 complex could negatively regulate the expression of these two genes and thereby contribute to human breast carcinogenesis. Our results imply that PIGX may be a good candidate molecule for development of novel anticancer drugs for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Calcium-Binding Proteins/genetics , Carrier Proteins/genetics , Mannosyltransferases/genetics , Transcription Factors/genetics , Breast Neoplasms/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HeLa Cells , Humans , MCF-7 Cells , Mannosyltransferases/metabolism , Transcription Factors/metabolism , Up-Regulation
7.
Oncotarget ; 7(14): 18171-82, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26918358

ABSTRACT

MELK is upregulated in various types of human cancer and is known to be associated with cancer progression, maintenance of stemness, and poor prognosis. OTS167, a MELK kinase inhibitor, shows potent growth-suppressive effect on human tumors in a xenograft model, but the detailed mode of action has not been fully elucidated. In this study, we demonstrate the molecular mechanism of action of MELK inhibitor OTS167 in a preclinical model. OTS167-treated cells caused morphological transformation, induced the differentiation markers, and reduced stem-cell marker expression. Furthermore, we identified DEPDC1, known as an oncogene, as an additional downstream molecule of the MELK signaling pathway. MELK enhanced DEPDC1 phosphorylation and its stability. The expression of MELK and downstream molecules was decreased in OTS167-treated xenograft tumor tissues, which revealed central necrosis and significant growth suppression. Our data should further shed light on the mechanism of action how OTS167 suppresses tumor growth through the inhibition of the MELK signaling pathway and suggest the possibility of biomarkers for the assessment of clinical efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , GTPase-Activating Proteins/metabolism , Naphthyridines/pharmacology , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , A549 Cells , Animals , Biomarkers, Tumor , COS Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorocebus aethiops , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology , Phosphorylation , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
Oncotarget ; 7(12): 13621-33, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26871945

ABSTRACT

Maternal embryonic leucine zipper kinase (MELK), that plays a critical role in maintenance of cancer stem cells (CSCs), is predominantly expressed in various types of human cancer including small cell lung cancer (SCLC). SCLC usually acquires resistance to anti-cancer drugs and portends dismal prognosis. We have delineated roles of MELK in development/progression of SCLC and examined anti-tumor efficacy of OTS167, a highly potent MELK inhibitor, against SCLC. MELK expression was highly upregulated in both SCLC cell lines and primary tumors. siRNA-mediated MELK knockdown induced significant growth inhibition in SCLC cell lines. Concordantly, treatment with OTS167 exhibited strong cytotoxicity against eleven SCLC cell lines with IC50 of < 10 nM. As similar to siRNA knockdown, OTS167 treatment induced cytokinetic defects with intercellular bridges, and in some cell lines we observed formation of neuronal protrusions accompanied with increase of a neuronal differentiation marker (CD56), indicating that the compound induced differentiation of cancer cells to neuron-like cells. Furthermore, the MELK inhibition decreased its downstream FOXM1 activity and Akt expression in SCLC cells, and led to apoptotic cell death. OTS167 appeared to be more effective to CSCs as measured by the sphere formation assay, thus MELK inhibition might become a promising treatment modality for SCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Forkhead Box Protein M1/metabolism , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Small Cell Lung Carcinoma/pathology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Forkhead Box Protein M1/antagonists & inhibitors , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism , Tumor Cells, Cultured
9.
Clin Cancer Res ; 21(19): 4337-46, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26015512

ABSTRACT

PURPOSE: Paclitaxel is used worldwide in the treatment of breast, lung, ovarian, and other cancers. Sensory peripheral neuropathy is an associated adverse effect that cannot be predicted, prevented, or mitigated. To better understand the contribution of germline genetic variation to paclitaxel-induced peripheral neuropathy, we undertook an integrative approach that combines genome-wide association study (GWAS) data generated from HapMap lymphoblastoid cell lines (LCL) and Asian patients. METHODS: GWAS was performed with paclitaxel-induced cytotoxicity generated in 363 LCLs and with paclitaxel-induced neuropathy from 145 Asian patients. A gene-based approach was used to identify overlapping genes and compare with a European clinical cohort of paclitaxel-induced neuropathy. Neurons derived from human-induced pluripotent stem cells were used for functional validation of candidate genes. RESULTS: SNPs near AIPL1 were significantly associated with paclitaxel-induced cytotoxicity in Asian LCLs (P < 10(-6)). Decreased expression of AIPL1 resulted in decreased sensitivity of neurons to paclitaxel by inducing neurite morphologic changes as measured by increased relative total outgrowth, number of processes and mean process length. Using a gene-based analysis, there were 32 genes that overlapped between Asian LCL cytotoxicity and Asian patient neuropathy (P < 0.05), including BCR. Upon BCR knockdown, there was an increase in neuronal sensitivity to paclitaxel as measured by neurite morphologic characteristics. CONCLUSIONS: We identified genetic variants associated with Asian paclitaxel-induced cytotoxicity and functionally validated the AIPL1 and BCR in a neuronal cell model. Furthermore, the integrative pharmacogenomics approach of LCL/patient GWAS may help prioritize target genes associated with chemotherapeutic-induced peripheral neuropathy.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/etiology , Pharmacogenetics , Adaptor Proteins, Signal Transducing , Antineoplastic Agents, Phytogenic/pharmacology , Carrier Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , DEAD-box RNA Helicases/genetics , Eye Proteins/genetics , Genome-Wide Association Study , Humans , Induced Pluripotent Stem Cells/cytology , Neoplasm Proteins/genetics , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Paclitaxel/pharmacology , Pharmacogenetics/methods , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-bcr/genetics
10.
Breast Cancer Res ; 15(5): R81, 2013.
Article in English | MEDLINE | ID: mdl-24025145

ABSTRACT

INTRODUCTION: Chemotherapy-induced alopecia is one of the most common adverse events caused by conventional cytotoxic chemotherapy, yet there has been very little progress in the prevention or treatment of this side effect. Although this is not a life-threatening event, alopecia is very psychologically difficult for many women to manage. In order to improve the quality of life for these women, it is important to elucidate the molecular mechanisms of chemotherapy-induced alopecia and develop ways to effectively prevent and/or treat it. To identify the genetic risk factors associated with chemotherapy-induced alopecia, we conducted a genome-wide association study (GWAS) using DNA samples from breast cancer patients who were treated with chemotherapy. METHODS: We performed a case-control association study of 303 individuals who developed grade 2 alopecia, and compared them with 880 breast cancer patients who did not show hair loss after being treated with conventional chemotherapy. In addition, we separately analyzed a subset of patients who received specific combination therapies by GWASs and applied the weighted genetic risk scoring (wGRS) system to investigate the cumulative effects of the associated SNPs. RESULTS: We identified an SNP significantly associated with drug-induced grade 2 alopecia (rs3820706 in CACNB4 (calcium channel voltage-dependent subunit beta 4) on 2q23, P = 8.13 × 10(-9), OR = 3.71) and detected several SNPs that showed some suggestive associations by subgroup analyses. We also classified patients into four groups on the basis of wGRS analysis and found that patients who classified in the highest risk group showed 443 times higher risk of antimicrotubule agents-induced alopecia than the lowest risk group. CONCLUSIONS: Our study suggests several associated genes and should shed some light on the molecular mechanism of alopecia in chemotherapy-treated breast cancer patients and hopefully will contribute to development of interventions that will improve the quality of life (QOL) of cancer patients.


Subject(s)
Alopecia/chemically induced , Alopecia/genetics , Antineoplastic Agents/adverse effects , Breast Neoplasms/complications , Genome-Wide Association Study , Adult , Aged , Alleles , Alopecia/diagnosis , Antineoplastic Agents/classification , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/psychology , Calcium Channels/genetics , Case-Control Studies , Female , Genetic Association Studies , Humans , Middle Aged , Paclitaxel/adverse effects , Polymorphism, Single Nucleotide , Quality of Life , Risk , Severity of Illness Index , Tubulin Modulators/adverse effects
12.
Cancer Sci ; 104(8): 1074-82, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23648065

ABSTRACT

Chemotherapeutic agents are notoriously known to have a narrow therapeutic range that often results in life-threatening toxicity. Hence, it is clinically important to identify the patients who are at high risk for severe toxicity to certain chemotherapy through a pharmacogenomics approach. In this study, we carried out multiple genome-wide association studies (GWAS) of 13 122 cancer patients who received different chemotherapy regimens, including cyclophosphamide- and platinum-based (cisplatin and carboplatin), anthracycline-based (doxorubicin and epirubicin), and antimetabolite-based (5-fluorouracil and gemcitabine) treatment, antimicrotubule agents (paclitaxel and docetaxel), and topoisomerase inhibitors (camptothecin and etoposide), as well as combination therapy with paclitaxel and carboplatin, to identify genetic variants that are associated with the risk of severe neutropenia/leucopenia in the Japanese population. In addition, we used a weighted genetic risk scoring system to evaluate the cumulative effects of the suggestive genetic variants identified from GWAS in order to predict the risk levels of individuals who carry multiple risk alleles. Although we failed to identify genetic variants that surpassed the genome-wide significance level (P < 5.0 × 10(-8) ) through GWAS, probably due to insufficient statistical power and complex clinical features, we were able to shortlist some of the suggestive associated loci. The current study is at the relatively preliminary stage, but does highlight the complexity and problematic issues associated with retrospective pharmacogenomics studies. However, we hope that verification of these genetic variants through local and international collaborations could improve the clinical outcome for cancer patients.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Leukopenia/genetics , Neoplasms/blood , Neoplasms/drug therapy , Neutropenia/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Specimen Banks , Female , Genetic Variation/genetics , Genome-Wide Association Study/methods , Humans , Japan , Leukopenia/chemically induced , Male , Middle Aged , Neoplasms/genetics , Neutropenia/chemically induced , Pharmacogenetics/methods , Retrospective Studies , Risk
13.
Med Mol Morphol ; 45(4): 229-33, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23224602

ABSTRACT

The cAMP-dependent protein kinase inhibitor-ß (PKIB) is presumed to be one of the regulatory factors controlling the cAMP-dependent protein kinase A signaling pathway. The aim of this study was to investigate the frequency and patterns of PKIB overexpression in human breast cancer. We also examined the relationship between PKIB and phosphorylated Akt (pAkt) expression in the tumors. Using immunohistochemical techniques, we examined the expression of PKIB, ER, PR, HER2, and pAkt in 148 primary human breast carcinomas. We then analyzed the relationships between PKIB expression and that of pAkt, ER, PR, and HER2, as well as between PKIB expression and various clinicopathological characteristics. We assessed 64 and 27 cases, respectively, as positive for either PKIB or pAkt expression; 20 cases were positive for both PKIB and pAkt. We observed a significant positive correlation between the expression of PKIB and that of pAkt (P = 0.006). We showed by immunohistochemical analyses that PKIB expression was positively correlated with triple-negative breast cancers (P = 0.0004). These findings provide evidence for PKIB overexpression associated with pAkt expression. Furthermore, PKIB expression was strongly correlated with triple-negative breast cancer, suggesting that PKIB expression might contribute to the tumor behavior and development of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Carcinoma/pathology , Female , Humans , Middle Aged , Phosphorylation , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
14.
Oncotarget ; 3(12): 1629-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23283305

ABSTRACT

We previously reported MELK (maternal embryonic leucine zipper kinase) as a novel therapeutic target for breast cancer. MELK was also reported to be highly upregulated in multiple types of human cancer. It was implied to play indispensable roles in cancer cell survival and indicated its involvement in the maintenance of tumor-initiating cells. We conducted a high-throughput screening of a compound library followed by structure-activity relationship studies, and successfully obtained a highly potent MELK inhibitor OTSSP167 with IC50 of 0.41 nM. OTSSP167 inhibited the phosphorylation of PSMA1 (proteasome subunit alpha type 1) and DBNL (drebrin-like), which we identified as novel MELK substrates and are important for stem-cell characteristics and invasiveness. The compound suppressed mammosphere formation of breast cancer cells and exhibited significant tumor growth suppression in xenograft studies using breast, lung, prostate, and pancreas cancer cell lines in mice by both intravenous and oral administration. This MELK inhibitor should be a promising compound possibly to suppress the growth of tumor-initiating cells and be applied for treatment of a wide range of human cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell Proliferation/drug effects , Molecular Targeted Therapy , Naphthyridines/administration & dosage , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Drug Design , Female , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Injections, Intravenous , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Microfilament Proteins/metabolism , Molecular Structure , NIH 3T3 Cells , Naphthyridines/chemistry , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Small Molecule Libraries , Structure-Activity Relationship , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , src Homology Domains
15.
Article in English | MEDLINE | ID: mdl-21986318

ABSTRACT

Keloid represents overgrowth of granulation tissue, which is characterized by collection of atypical fibroblasts with excessive deposition of extracellular matrix components, after skin injury, but its etiology is still largely unknown. We recently performed genome-wide association study (GWAS) of keloid and identified NEDD4 to be one of candidate molecules associated with keloid susceptibility. Here we demonstrate a possible mechanism of NEDD4 involvement in keloid formation through enhancement of the proliferation and invasiveness of fibroblasts as well as upregulation of type 1 collagen expression. Activation of NEDD4 affected subcellular localization and protein stability of p27 which was implied its critical role in contact inhibition. It also induced accumulation of ß-catenin in the cytoplasm and activated the TCF/ß-catenin transcriptional activity. Furthermore, NEDD4 upregulated expressions of fibronectin and type 1 collagen and contributed to the excessive accumulation of extracellular matrix. Our findings provide new insights into mechanism developing keloid and can be applied for development of a novel treatment for keloid.


Subject(s)
Collagen/biosynthesis , Endosomal Sorting Complexes Required for Transport/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Keloid/enzymology , Keloid/pathology , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Communication , Cell Movement , Cell Proliferation , Collagen Type I/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytoplasm/metabolism , Enzyme Activation , Extracellular Matrix/metabolism , Fibronectins/metabolism , Humans , Mice , NIH 3T3 Cells , Nedd4 Ubiquitin Protein Ligases , PTEN Phosphohydrolase/metabolism , Proteolysis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , beta Catenin/metabolism
16.
Neoplasia ; 13(4): 320-6, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21472136

ABSTRACT

A high expression of short and immature O-glycans is one of the prominent features of breast cancer cells, which would be attributed to the upregulated expression of glycosyltransferases. Therefore, a detailed elucidation of glycosyltransferases and their substrate(s) may improve our understandings for their roles in mammary carcinogenesis. Here we report that overexpression of polypeptide N-acetylgalactosaminyltransferase 6 (GALNT6), a glycosyltransferase involved in the initial step of O-glycosylation, has transformational potentials through disruptive acinar morphogenesis and cellular changes similar to epithelial-to-mesenchymal transition in normal mammary epithelial cell, MCF10A. As one of the critical O-glycan substrates, we identified fibronectin that was O-glycosylated in vivo and thereby stabilized by GALNT6. Because knockdown of fibronectin abrogated the disruptive proliferation caused by introduction of GALNT6 into epithelial cells, our findings suggest that GALNT6-fibronectin pathway should be a critical component for breast cancer development and progression.


Subject(s)
Fibronectins/metabolism , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Morphogenesis/genetics , N-Acetylgalactosaminyltransferases/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Cell Line , Disease Progression , Female , Gene Knockdown Techniques , Glycosylation , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Mammary Glands, Human/anatomy & histology , Mammary Glands, Human/pathology , Models, Biological , N-Acetylgalactosaminyltransferases/antagonists & inhibitors , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Protein Stability , Transfection , Polypeptide N-acetylgalactosaminyltransferase
17.
Genes Chromosomes Cancer ; 50(1): 13-24, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20931645

ABSTRACT

To identify novel therapeutic targets for aggressive and therapy-resistant pancreatic cancer, we had previously performed expression profile analysis of pancreatic cancers using microarrays and found dozens of genes trans-activated in pancreatic ductal adenocarcinoma (PDAC) cells. Among them, this study focused on the characterization of a novel gene C12orf48 whose overexpression in PDAC cells was validated by Northern blot and immunohistochemical analysis. Its overexpression was observed in other aggressive and therapy-resistant malignancies as well. Knockdown of C12orf48 by siRNA in PDAC cells significantly suppressed their growth. Importantly, we demonstrated that C12orf48 protein could directly interact with Poly(ADP-ribose) Polymerase-1 (PARP-1), one of the essential proteins in the repair of DNA damage, and positively regulate the poly(ADP-ribosyl)ation activity of PARP-1. Depletion of C12orf48 sensitized PDAC cells to agents causing DNA damage and also enhanced DNA damage-induced G2/M arrest through reduction of PARP-1 enzymatic activities. Hence, our findings implicate C12orf48, termed PARP-1 binding protein (PARPBP), or its interaction with PARP-1 to be a potential molecular target for development of selective therapy for pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/genetics , Carrier Proteins/metabolism , DNA Damage , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Poly(ADP-ribose) Polymerases/metabolism , Blotting, Northern , Blotting, Western , Carrier Proteins/genetics , Cell Cycle , Cell Extracts , Cell Line, Tumor , DNA-Binding Proteins , Gene Expression Profiling , Gene Knockdown Techniques , Histocytochemistry , Humans , Oligonucleotide Array Sequence Analysis , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , Protein Interaction Mapping , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
18.
Cancer Sci ; 102(1): 245-52, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20874843

ABSTRACT

Recent genome-wide association studies reported strong and reproducible associations of multiple genetic variants in a large "gene-desert" region of chromosome 8q24 with susceptibility to prostate cancer (PC). However, the causative or functional variants of these 8q24 loci and their biological mechanisms associated with PC susceptibility remain unclear and should be investigated. Here, focusing on its most centromeric region (so-called Region 2: Chr8: 128.14-128.28 Mb) among the multiple PC loci on 8q24, we performed fine mapping and re-sequencing of this critical region and identified SNPs (single nucleotide polymorphisms) between rs1456315 and rs7463708 (chr8: 128,173,119-128,173,237 bp) to be most significantly associated with PC susceptibility (P = 2.00 × 10(-24) , OR = 1.74, 95% confidence interval = 1.56-1.93). Importantly, we show that this region was transcribed as a ∼13 kb intron-less long non-coding RNA (ncRNA), termed PRNCR1 (prostate cancer non-coding RNA 1), and PRNCR1 expression was upregulated in some of the PC cells as well as precursor lesion prostatic intraepithelial neoplasia. Knockdown of PRNCR1 by siRNA attenuated the viability of PC cells and the transactivation activity of androgen receptor, which indicates that PRNCR1 could be involved in prostate carcinogenesis possibly through androgen receptor activity. These findings could provide a new insight in understanding the pathogenesis of genetic factors for PC susceptibility and prostate carcinogenesis.


Subject(s)
Chromosomes, Human, Pair 8 , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , RNA, Untranslated/genetics , Cell Line, Tumor , Cell Survival , Chromosome Mapping , Cloning, Molecular , Genome-Wide Association Study , Haplotypes , Humans , Male , Polymorphism, Single Nucleotide , Prostatic Neoplasms/pathology , Receptors, Androgen/physiology
19.
Nat Genet ; 42(9): 768-71, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20711176

ABSTRACT

Keloid is a dermal fibroproliferative growth that results from dysfunction of the wound healing processes. Through a multistage genome-wide association study using 824 individuals with keloid (cases) and 3,205 unaffected controls in the Japanese population, we identified significant associations of keloid with four SNP loci in three chromosomal regions: 1q41, 3q22.3-23 and 15q21.3. The most significant association with keloid was observed at rs873549 (combined P = 5.89 x 10(-23), odds ratio (OR) = 1.77) on chromosome 1. Associations on chromosome 3 were observed at two separate linkage disequilibrium (LD) blocks: rs1511412 in the LD block including FOXL2 with P = 2.31 x 10(-13) (OR = 1.87) and rs940187 in another LD block with P = 1.80 x 10(-13) (OR = 1.98). Association of rs8032158 located in NEDD4 on chromosome 15 yielded P = 5.96 x 10(-13) (OR = 1.51). Our findings provide new insights into the pathophysiology of keloid formation.


Subject(s)
Asian People/genetics , Genetic Loci , Genetic Predisposition to Disease , Keloid/genetics , Case-Control Studies , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 3 , Gene Frequency , Genetics, Population , Genome-Wide Association Study , Humans , Linkage Disequilibrium , Polymorphism, Single Nucleotide
20.
Cancer Sci ; 101(8): 1897-904, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20560974

ABSTRACT

Prostate cancer often relapses during androgen-depletion therapy, even under the castration condition in which circulating androgens are drastically reduced. High expressions of androgen receptor (AR) and genes involved in androgen metabolism indicate a continued role for AR in castration-resistant prostate cancers (CRPCs). There is increasing evidence that some amounts of 5alpha-dihydrotestosterone (DHT) and other androgens are present sufficiently to activate AR within CRPC tissues, and enzymes involved in the androgen and steroid metabolism, such as 5alpha-steroid reductases, are activated in CRPCs. In this report, we screened eight natural 5alphaDH-steroids to search for novel products of 5alpha-steroid reductases, and identified 11-deoxycorticosterone (DOC) as a novel substrate for 5alpha-steroid reductases in CRPCs. 11-Deoxycorticosterone (DOC) and 5alpha-dihydro-deoxycorticosterone (5alphaDH-DOC) could promote prostate cancer cell proliferation through AR activation, and type 1 5alpha-steroid reductase (SRD5A1) could convert from DOC to 5alphaDH-DOC. Sensitive liquid chromatography-tandem mass spectrometric analysis detected 5alphaDH-DOC in some clinical CRPC tissues. These findings implicated that under an extremely low level of DHT, 5alphaDH-DOC and other products of 5alpha-steroid reductases within CRPC tissues might activate the AR pathway for prostate cancer cell proliferation and survival under castration.


Subject(s)
Pregnanediones/pharmacology , Prostatic Neoplasms/pathology , Receptors, Androgen/drug effects , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Desoxycorticosterone/pharmacology , Humans , Male , Orchiectomy , Pregnanediones/analysis , Pregnanediones/metabolism , Prostatic Neoplasms/drug therapy , Receptors, Androgen/physiology , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...