Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Genes Cells ; 29(1): 73-85, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38016691

ABSTRACT

Bladder cancer is a urothelial cancer and effective therapeutic strategies for its advanced stages are limited. Here, we report that CD271, a neurotrophin receptor, promotes the proliferation and migration of bladder cancer cells. CD271 knockdown decreased proliferation in both adherent and spheroid cultures, and vice versa when CD271 was overexpressed in bladder cancer cell lines. CD271 depletion impaired tumorigenicity in vivo. Migration activity was reduced by CD271 knockdown and TAT-Pep5, a known CD271-Rho GDI-binding inhibitor. Apoptosis was induced by CD271 knockdown. Comprehensive gene expression analysis revealed alterations in E2F- and Myc-related pathways upon CD271 expression. In clinical cases, patients with high CD271 expression showed significantly shortened overall survival. In surgically resected specimens, pERK, a known player in proliferation signaling, colocalizes with CD271. These data indicate that CD271 is involved in bladder cancer malignancy by promoting cell proliferation and migration, resulting in poor prognosis.


Subject(s)
Receptors, Nerve Growth Factor , Urinary Bladder Neoplasms , Humans , Adapalene , Receptors, Nerve Growth Factor/genetics , Cell Proliferation , Signal Transduction , Urinary Bladder Neoplasms/genetics , Cell Movement , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
2.
Biochem Biophys Res Commun ; 655: 59-67, 2023 05 07.
Article in English | MEDLINE | ID: mdl-36933308

ABSTRACT

Gastric cancer is the fifth most common malignancy worldwide. However, targeted therapy for advanced gastric cancer is still limited. Here, we report BEX2 (Brain expressed X-linked 2) as a poor prognostic factor in two gastric cancer cohorts. BEX2 expression was increased in spheroid cells, and its knockdown decreased aldefluor activity and cisplatin resistance. BEX2 was found to upregulate CHRNB2 (Cholinergic Receptor Nicotinic Beta 2 Subunit) expression, a cancer stemness-related gene, in a transcriptional manner, and the knockdown of which also decreases aldefluor activity. Collectively, these data are suggestive of the role of BEX2 in the malignant process of gastric cancer, and as a promising therapeutic target.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Prognosis , Cell Line, Tumor , Oncogenes , Nerve Tissue Proteins/metabolism
3.
Cancer Sci ; 113(5): 1613-1624, 2022 May.
Article in English | MEDLINE | ID: mdl-35247012

ABSTRACT

To examine effects of PP6Ā gene (Ppp6c) deficiency on pancreatic tumor development, we developed pancreas-specific, tamoxifen-inducible Cre-mediated KP (KRAS(G12D) plus Trp53-deficient) mice (cKP mice) and crossed them with Ppp6cflox / flox mice. cKP mice with the homozygous Ppp6c deletion developed pancreatic tumors, became emaciated and required euthanasia within 150Ā days of mutation induction, phenotypes that were not seen in heterozygous or wild-type (WT) mice. At 30Ā days, a comparative analysis of genes commonly altered in homozygous versus WT Ppp6c cKP mice revealed enhanced activation of Erk and NFκB pathways in homozygotes. By 80Ā days, the number and size of tumors and number of precancerous lesions had significantly increased in the pancreas of Ppp6c homozygous relative to heterozygous or WT cKP mice. Ppp6c-/- tumors were pathologically diagnosed as pancreatic ductal adenocarcinoma (PDAC) undergoing the epithelial-mesenchymal transition (EMT), and cancer cells had invaded surrounding tissues in three out of six cases. Transcriptome and metabolome analyses indicated an enhanced cancer-specific glycolytic metabolism in Ppp6c-deficient cKP mice and the increased expression of inflammatory cytokines. Individual Ppp6c-/- cKP mice showed weight loss, decreased skeletal muscle and adipose tissue, and increased circulating tumor necrosis factor (TNF)-α and IL-6Ā levels, suggestive of systemic inflammation. Overall, Ppp6c deficiency in the presence of K-ras mutations and Trp53Ā gene deficiency promoted pancreatic tumorigenesis with generalized cachexia and early death. This study provided the first evidence that Ppp6c suppresses mouse pancreatic carcinogenesis and supports the use of Ppp6c-deficient cKP mice as a model for developing treatments for cachexia associated with pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Phosphoprotein Phosphatases/metabolism , Animals , Cachexia/genetics , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/pathology , Humans , Mice , Mutation , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms
4.
Cancer Sci ; 113(8): 2878-2887, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35343032

ABSTRACT

Various proteins are highly expressed in cancer (e.g., epidermal growth factor receptor); however, the majority are also expressed in normal cells, although they may differ in expression intensity. Recently, we reported that CD271 (nerve growth factor receptor), a glycosylated protein, increases malignant behavior of cancer, particularly stemlike phenotypes in squamous cell carcinoma (SCC). CD271 is expressed in SCC and in normal epithelial basal cells. Glycosylation alterations generally occur in cancer cells; therefore, we attempted to establish a cancer-specific anti-glycosylated CD271 antibody. We purified recombinant glycosylated CD271 protein, immunized mice with the protein, and screened hybridomas using an ELISA assay with cancer cell lines. We established a clone G4B1 against CD271 which is glycosylated with O-glycan and sialic acid. The G4B1 antibody reacted with the CD271 protein expressed in esophageal cancer, but not in normal esophageal basal cells. This specificity was confirmed in hypopharyngeal and cervical cancers. G4B1 antibody recognized the fetal esophageal epithelium and Barrett's esophagus, which possess stem cell-like characteristics. In conclusion, G4B1 antibody could be useful for precise identification of dysplasia and cancer cells in SCC.


Subject(s)
Barrett Esophagus , Carcinoma, Squamous Cell , Esophageal Neoplasms , Adapalene , Animals , Antibodies, Monoclonal/metabolism , Barrett Esophagus/pathology , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Glycosylation , Immunohistochemistry , Mice , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
5.
Cancer Sci ; 112(6): 2233-2244, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33743547

ABSTRACT

According to TCGA database, mutations in PPP6C (encoding phosphatase PP6) are found in c. 10% of tumors from melanoma patients, in which they coexist with BRAF and NRAS mutations. To assess PP6 function in melanoma carcinogenesis, we generated mice in which we could specifically induce BRAF(V600E) expression and delete Ppp6c in melanocytes. In these mice, melanoma susceptibility following UVB irradiation exhibited the following pattern: Ppp6c semi-deficient (heterozygous) > Ppp6c wild-typeĀ >Ā Ppp6c-deficient (homozygous) tumor types. Next-generation sequencing of Ppp6c heterozygous and wild-type melanoma tumors revealed that all harbored Trp53 mutations. However, Ppp6c heterozygous tumors showed a higher Signature 1 (mitotic/mitotic clock) mutation index compared with Ppp6c wild-type tumors, suggesting increased cell division. Analysis of cell lines derived from either Ppp6c heterozygous or wild-type melanoma tissues showed that both formed tumors in nude mice, but Ppp6c heterozygous tumors grew faster compared with those from the wild-type line. Ppp6c knockdown via siRNA in the Ppp6c heterozygous line promoted the accumulation of genomic damage and enhanced apoptosis relative to siRNA controls. We conclude that in the presence of BRAF(V600E) expression and UV-induced Trp53 mutation, Ppp6c haploinsufficiency promotes tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Melanoma/genetics , Phosphoprotein Phosphatases/genetics , Proto-Oncogene Proteins B-raf/genetics , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Exome/genetics , Exome/radiation effects , Genotype , Haploinsufficiency , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanocytes/radiation effects , Melanoma/pathology , Mice , Mice, Nude , Mice, Transgenic , Mutation/radiation effects , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
6.
Int J Mol Sci ; 22(16)2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34445227

ABSTRACT

Osimertinib is the latest generation epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor used for patients with EGFR-mutated non-small cell lung cancer (NSCLC). We aimed to explore the novel mechanisms of osimertinib by particularly focusing on EGFR-independent effects, which have not been well characterized. We explored the EGFR-independent effects of osimertinib on cell proliferation using NSCLC cell lines, an antibody array analysis, and the association between the action of osimertinib and the ephrin receptor B4 (EphB4). We also studied the clinicopathological significance of EphB4 in 84 lung adenocarcinoma patients. Osimertinib exerted significant inhibitory effects on cell growth and cell cycle progression by promoting the phosphorylation of p53 and p21 and decreasing cyclin D1 expression independently of EGFR. EphB4 was significantly suppressed by osimertinib and promoted cell growth and sensitivity to osimertinib. The EphB4 status in carcinoma cells was positively correlated with tumor size, T factor, and Ki-67 labeling index in all patients and was associated with poor relapse-free survival in EGFR mutation-positive patients. EphB4 is associated with the EGFR-independent suppressive effects of osimertinib on cell cycle and with a poor clinical outcome. Osimertinib can exert significant growth inhibitory effects in EGFR-mutated NSCLC patients with a high EphB4 status.


Subject(s)
Acrylamides/pharmacology , Adenocarcinoma of Lung , Aniline Compounds/pharmacology , Carcinoma, Non-Small-Cell Lung , Cell Cycle/drug effects , Lung Neoplasms , Neoplasm Proteins/metabolism , Receptor, EphB4/metabolism , A549 Cells , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/mortality , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cell Cycle/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Mutation , Neoplasm Proteins/genetics , Receptor, EphB4/genetics
7.
Br J Cancer ; 123(1): 61-71, 2020 07.
Article in English | MEDLINE | ID: mdl-32336752

ABSTRACT

BACKGROUND: Understanding the status of intratumoural immune microenvironment is necessary to ensure the efficacy of immune-checkpoint (IC) blockade therapy. Cortisol plays pivotal roles in glucocorticoid interactions in the immune system. We examined the correlation between intratumourally synthesised cortisol through 11Ɵ hydroxysteroid dehydrogenase (HSD) 1 and the immune microenvironment in non-small-cell lung carcinoma (NSCLC). METHODS: We correlated 11ƟHSD1 immunoreactivity in 125 cases of NSCLC with the amount of intratumoural immune cells present, and 11ƟHSD1 immunoreactivity with the efficacy of IC blockade therapy in 18 specimens of NSCLC patients. In vitro studies were performed to validate the immunohistochemical examination. RESULTS: 11ƟHSD1 immunoreactivity showed a significant inverse correlation with the number of tumour-infiltrating lymphocytes and CD3- or CD8-positive T cells. 11ƟHSD1 immunoreactivity tended to be inversely correlated with the clinical efficacy of the IC blockade therapy. In vitro studies revealed that 11ƟHSD1 promoted the intratumoural synthesis of cortisol. This resulted in a decrease in cytokines and in the inhibition of monocyte migration. CONCLUSIONS: Our study is the first report clarifying the inhibitory effects of intratumourally synthesised cortisol through 11ƟHSD1 on immune cell migration. We propose that the response to IC blockade therapy in NSCLC may be predicted by 11ƟHSD1.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Immune Checkpoint Inhibitors/administration & dosage , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Female , Humans , Immune Checkpoint Inhibitors/immunology , Male , Middle Aged , Tumor Microenvironment/drug effects
8.
Biochem Biophys Res Commun ; 522(3): 690-696, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31787238

ABSTRACT

Lung adenocarcinoma (LUAC) is a unique lung cancer subtype that is responsive to several therapeutic agents. The KRAS gene is the second most frequently mutated gene in LUAC and the majority of KRAS mutations are one of three classical activating mutations (G12, G13, and Q61). Recently, other types of "minor" KRAS mutation have been identified among LUAC patients and some may have similar transforming activities to those of the classical KRAS mutations. Here we describe minor KRAS mutations in LUAC patients, some of which (A66T, A66V, and G75E) may have tumor-forming activity in mouse embryonic fibroblasts in an allograft model. RNA-Seq analysis revealed that mouse embryonic fibroblasts overexpressing these three minor KRAS mutations have distinct expression profiles compared with overexpression of the wild type but similar expression profiles compared with overexpression of the classical KRAS mutants. Our results indicate that some of the minor KRAS mutations cause varying tumor formation activity and are important targets for developing anti-RAS agents as chemotherapeutic agents.


Subject(s)
Adenocarcinoma of Lung/genetics , Lung Neoplasms/genetics , Point Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma of Lung/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lung Neoplasms/pathology , Mice , Models, Molecular , Transcriptome
9.
Pancreatology ; 20(3): 493-500, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32007357

ABSTRACT

BACKGROUND: Accumulating evidence indicates that CD109, a glycosylphosphatidylinositol-anchored glycoprotein, is highly expressed in human epithelial carcinomas of multiple organs including the pancreas, but its functional role in carcinoma development has not yet been fully clarified. The aim of this study was to investigate the role of CD109 in the malignancy of pancreatic ductal adenocarcinoma (PDAC). METHODS: PDAC specimens of 145 cases were immunostained for CD109, and correlations between CD109 expression and clinicopathological conditions were analyzed. CD109 expression in PANC-1Ā cells, a PDAC-derived cell line, was decreased by siRNA or shRNA and its effect on the malignancy of PANC-1Ā cells was examined. RESULTS: Suppression of CD109 expression in PANC-1Ā cells resulted in reduction of inĀ vitro cell motility and tumorigenicity in xenografts. Based on these results, we investigated the relationship between CD109 expression and metastasis of PDAC using tumor tissue specimens. Among 106 recurrent cases of 145 PDAC, there was a tendency for CD109-positive cases to be accompanied by distant metastasis. CONCLUSIONS: CD109 plays a critical role in the promotion of tumorigenic ability and cellular motility relating to metastasis of PDAC cells.


Subject(s)
Antigens, CD/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement/genetics , Female , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Middle Aged , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , RNA, Small Interfering/pharmacology , Wound Healing , Xenograft Model Antitumor Assays
10.
Lab Invest ; 99(9): 1349-1362, 2019 09.
Article in English | MEDLINE | ID: mdl-31019292

ABSTRACT

Squamous cell carcinoma is a major type of cancer in the lung. While several therapeutic target molecules for lung adenocarcinoma have been identified, little is known about lung squamous cell carcinoma (LSCC). We recently reported that CD271 (p75 neurotrophin receptor) serves as a marker for tumor initiation and is a key regulator of cell proliferation in hypopharyngeal squamous cell carcinoma. In this study, we found that CD271 was also expressed in squamous cell carcinoma, but not in adenocarcinoma, of several tissues, including the lung, and the expression of CD271 was associated with a poor prognosis in LSCC. To examine CD271's role in LSCC, we established xenograft cell lines from LSCC patients. Within the sorted live LSCC cell population, the CD271high cells were primarily cycling through the G2/M phase, while the CD271low cells were mostly in the G0 phase. CD271 knockdown in the LSCC cells completely suppressed their proliferation and tumor-formation capability, and increased their cell-cycle arrest in the G0 phase. In the CD271-knockdown cells, ERK-phosphorylation was decreased, while no change was observed in the IκBα-phosphorylation, p65-phosphorylation, or Akt-phosphorylation. Treatment with the MEK inhibitor U0126 decreased the LSCC cells' proliferation capability. Microarray analysis revealed that CD271 knockdown attenuated the RAS-related pathways. The knockdown of TrkB, which forms a heterodimer with CD271 and accelerates its downstream signaling, partially inhibited the LSCC cell proliferation. These results indicated that LSCC exclusively depends on CD271 for cell proliferation, in part through ERK-signaling activation, and CD271 is a promising target for LSCC therapy.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Proliferation/genetics , Lung Neoplasms/metabolism , Nerve Tissue Proteins , Receptors, Nerve Growth Factor , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/mortality , Cell Line, Tumor , Cell Movement/genetics , Disease-Free Survival , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/mortality , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prognosis , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
11.
Cancer Sci ; 109(7): 2178-2187, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29758119

ABSTRACT

Here, we address the function of protein phosphatase 6 (PP6) loss on K-ras-initiated tumorigenesis in keratinocytes. To do so, we developed tamoxifen-inducible double mutant (K-rasG12D -expressing and Ppp6c-deficient) mice in which K-rasG12D expression is driven by the cytokeratin 14 (K14) promoter. Doubly-mutant mice showed early onset tumor formation in lips, nipples, external genitalia, anus and palms, and had to be killed by 3Ā weeks after induction by tamoxifen, while comparably-treated K-rasG12D -expressing mice did not. H&E-staining of lip tumors before euthanasia revealed that all were papillomas, some containing focal squamous cell carcinomas. Immunohistochemical analysis of lips of doubly-mutant vs K-rasG12D mice revealed that cell proliferation and cell size increased approximately 2-fold relative to K-rasG12D -expressing mutants, and epidermal thickness of lip tissue greatly increased relative to that seen in K-rasG12D -only mice. Moreover, AKT phosphorylation increased in K-rasG12D -expressing/Ppp6c-deficient cells, as did phosphorylation of the downstream effectors 4EBP1, S6 and GSK3, suggesting that protein synthesis and survival signals are enhanced in lip tissues of doubly-mutant mice. Finally, increased numbers of K14-positive cells were present in the suprabasal layer of doubly-mutant mice, indicating abnormal keratinocyte differentiation, and ƎĀ³H2AX-positive cells accumulated, indicating perturbed DNA repair. Taken together, Ppp6c deficiency enhances K-rasG12D -dependent tumor promotion.


Subject(s)
Carcinogenesis/genetics , Keratinocytes/enzymology , Phosphoprotein Phosphatases/metabolism , Skin Neoplasms/enzymology , Animals , Mice , Mice, Mutant Strains , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms/genetics
12.
Biochem Biophys Res Commun ; 505(2): 466-470, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30268496

ABSTRACT

S100A10 is one of the members of the S100 protein family and is a key plasminogen receptor. Its upregulation has been reported in many types of tumors. In lung cancer, an association between upregulation of S100A10 and poor prognoses has been reported only in adenocarcinoma. We pursued the possibility of significance in lung squamous cell carcinoma (SCC). We first examined S100A10 protein expression by immunohistochemistry in 120 primary resected lung SCCs; 33 (27.5%) tumors showed strong membranous-immunopositivity particularly at the invasive front, i.e., the cancer-cell surface in contact with the stroma. Expression levels were significantly associated with higher pathological TNM stage (PĆ¢Ā€ĀÆ=Ć¢Ā€ĀÆ0.0119), tumor size (PĆ¢Ā€ĀÆ=Ć¢Ā€ĀÆ0.0003), lymphatic invasion (PĆ¢Ā€ĀÆ=Ć¢Ā€ĀÆ0.0005), lymph node metastasis (PĆ¢Ā€ĀÆ=Ć¢Ā€ĀÆ0.0006), and poorer prognosis (PĆ¢Ā€ĀÆ=Ć¢Ā€ĀÆ0.0064). Our present results suggest that high S100A10 expression of the lung SCC cells, particularly adjacent to stroma, plays an important role in tumor progression, probably caused by lymphatic invasion and nodal metastasis.


Subject(s)
Annexin A2/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , S100 Proteins/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Prognosis , Up-Regulation
13.
Cancer Sci ; 108(3): 435-443, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28075522

ABSTRACT

Carbonic anhydrase IX (CA9) is a membrane-associated carbonic anhydrase that regulates cellular pH, is upregulated in various solid tumors, and is considered to be a therapeutic target. Here, we describe the essential role of CA9 in the tumorigenicity of cells derived from human adult T-cell leukemia/lymphoma (ATL). We previously established the highly tumorigenic ST1-N6 subline from the ATL-derived ST1 cell line by serial xenotransplantation in NOG mice. In the present study, we first show that CA9 expression is strongly enhanced in ST1-N6 cells. We then sorted ST1 cells by high or low CA9 expression and established ST1-CA9high and ST1-CA9low sublines. ST1-CA9high cells, like ST1-N6 cells, were more strongly tumorigenic than ST1-CA9low or parental ST1 cells when injected into NOG mice. Knockdown of CA9 with shRNAs suppressed the ability of ST1-CA9high cells to initiate tumors, and the tumorigenicity of ST1 cells was significantly enhanced by introducing wild-type CA9 or a CA9 mutant with deletion of an intracytoplasmic domain. However, a CA9 with point mutations in the catalytic site did not increase the tumorigenicity of ST1 cells. Furthermore, we detected a small population of CA9+ CD25+ cells in lymph nodes of ATL patients. These findings suggest that CA9, and particularly its carbonic anhydrase activity, promotes the tumorigenicity of ATL-derived cells and may be involved in malignant development of lymphoma-type ATL.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase IX/metabolism , Cell Transformation, Neoplastic/pathology , Leukemia-Lymphoma, Adult T-Cell/pathology , Animals , Catalytic Domain/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Humans , Lymph Nodes/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering/genetics , Transplantation, Heterologous
14.
Cancer Sci ; 108(5): 931-940, 2017 May.
Article in English | MEDLINE | ID: mdl-28235245

ABSTRACT

Recent studies have indicated that increased expression of the M2 isoform of pyruvate kinase (PKM2) is involved in glycolysis and tumor development. However, little is known about the role of PKM2 in gastric cancer (GC). Therefore, we examined the expression and function of PKM2 in human GC. We evaluated PKM1 and PKM2 expression by quantitative RT-PCR in gastric tissues from 10 patients who underwent gastric endoscopic submucosal dissection, 80 patients who underwent gastrectomy, and seven healthy volunteers, and analyzed the correlation with clinicopathological variables. To assess the function of PKM2, we generated PKM2-knockdown GC cells, and investigated the phenotypic changes. Furthermore, we examined the induction of PKM2 expression by cytotoxin-associated gene A (CagA), a pathogenic factor of Helicobacter pylori, using CagA-inducible GC cells. We found that PKM2 was predominantly expressed not only in GC lesions but also in the normal gastric regions of GC patients and in the gastric mucosa of healthy volunteers. The PKM2 expression was significantly higher in carcinoma compared to non-cancerous tissue and was associated with venous invasion. Knockdown of PKM2 in GC cells caused significant decreases in cellular proliferation, migration, anchorage-independent growth, and sphere formation inĀ vitro, and in tumor growth and liver metastasis inĀ vivo. The serine concentration-dependent cell proliferation was also inhibited by PKM2 silencing. Furthermore, we found that PKM2 expression was upregulated by CagA by way of the Erk pathway. These results suggested that enhanced PKM2 expression plays a pivotal role in the carcinogenesis and development of GC in part by regulating cancer-specific metabolism.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Isoforms/genetics , Stomach Neoplasms/genetics , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Aged , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Glycolysis/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Signaling System/genetics , Male , Protein Isoforms/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Up-Regulation/genetics , Thyroid Hormone-Binding Proteins
15.
J Oral Pathol Med ; 45(8): 565-72, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26711722

ABSTRACT

OBJECTIVE: Recent evidence suggests that human papillomavirus (HPV)-related head and neck squamous cell carcinoma (HNSCC) is a separate HNSCC subgroup with distinct epidemiology, histopathological characteristics, therapeutic response to chemotherapy and radiation, and clinical outcome. This study aimed to investigate the role of HPV infection in oral squamous cell carcinoma (OSCC) and the correlation between HPV infection, tumor suppressor protein p16 expression, and clinicopathological features in Japanese patients. METHODS: In total, 174 OSCC specimens were examined for p16 levels by immunohistochemistry, and p16-positive OSCCs were analyzed for HPV DNA by in situ hybridization (ISH) and HPV genotypes by real-time PCR. The results were evaluated for the association with clinicopathological characteristics of OSCC patients. RESULTS: Twenty-four OSCC samples were found positive for p16 expression; all of them were well-differentiated tumors. P16 immunoreactivity was significantly associated with the invasion depth and tended to correlate with sex, site in the oral cavity, stromal reaction, TNM stage, and survival. HPV DNA was detected in 13 of 24 (54%) p16-positive OSCC by real-time PCR; HPV 16, 18, and other high-risk genotypes were the most prevalent. However, ISH failed to detect HPV DNA in p16-positive OSCCs. CONCLUSION: P16 immunoreactivity and HPV genotyping by real-time PCR may be useful markers of HPV infection in OSCC. However, although HPV-related OSCC showed good outcomes, HPV infection may have a minor role in oral oncogenesis in Japanese patients.


Subject(s)
Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Mouth Neoplasms/pathology , Mouth Neoplasms/virology , Papillomaviridae/isolation & purification , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Adult , Aged , Aged, 80 and over , Cohort Studies , DNA, Viral/isolation & purification , Female , Humans , Japan , Male , Middle Aged , Papillomaviridae/genetics , Prevalence , Squamous Cell Carcinoma of Head and Neck
16.
Int J Cancer ; 137(7): 1560-73, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25810027

ABSTRACT

The plasma membrane-associated sialidase NEU3 is a key enzyme for ganglioside degradation. We previously demonstrated remarkable up-regulation of NEU3 in various human cancers, with augmented malignant properties. Here, we provide evidence of a close link between NEU3 expression and Wnt/Ɵ-catenin signaling in colon cancer cells by analyzing tumorigenic potential and cancer stem-like characteristics. NEU3 silencing in HT-29 and HCT116 colon cancer cells resulted in significant decrease in clonogenicity on soft agar and in vivo tumor growth, along with down-regulation of stemness and Wnt-related genes. Analyses further revealed that NEU3 enhanced phosphorylation of the Wnt receptor LRP6 and consequently Ɵ-catenin activation by accelerating complex formation with LRP6 and recruitment of GSK3Ɵ and Axin, whereas its silencing exerted the opposite effects. NEU3 activity-null mutants failed to demonstrate the activation, indicating the requirement of ganglioside modulation by the sialidase for the effects. Under sphere-forming conditions, when stemness genes are up-regulated, endogenous NEU3 expression was found to be significantly increased, whereas NEU3 silencing suppressed sphere-formation and in vivo tumor incidence in NOD-SCID mice. Increased ability of clonogenicity on soft agar and sphere formation by Wnt stimulation was abrogated by NEU3 silencing. Furthermore, NEU3 was found to regulate phosphorylation of ERK and Akt via EGF receptor and Ras cascades, thought to be additionally required for tumor progression. The results indicate an essential contribution of NEU3 to tumorigenic potential through maintenance of stem-like characteristics of colon cancer cells by regulating Wnt signaling at the receptor level, in addition to tumor progression via Ras/MAPK signaling.


Subject(s)
Colonic Neoplasms/metabolism , Gangliosides/metabolism , Neuraminidase/metabolism , Animals , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , HCT116 Cells , HEK293 Cells , HT29 Cells , Heterografts , Humans , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Proteins/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , TCF Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
17.
Cancer Sci ; 106(11): 1544-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26470851

ABSTRACT

Regional lymph node metastasis in head and neck squamous cell carcinoma (HNSCC) is a crucial event for its progression, associated with a high rate of mortality. Sialidase, a key enzyme for the regulation of cellular sialic acids through catalyzing the initial step of degradation of glycoproteins and glycolipids, has been implicated in cancer progression. To facilitate the development of novel treatments for HNSCC, we have investigated whether sialidase is involved in the progression of this cancer. We found plasma membrane-associated sialidase (NEU3) to be significantly upregulated in tumor compared to non-tumor tissues; particularly, an increase in its mRNA levels was significantly associated with lymph node metastasis. To understand the mechanisms, we analyzed the NEU3-mediated effects on the malignant phenotype using squamous carcinoma HSC-2 and SAS cells. NEU3 promoted cell motility and invasion, accompanied by the increased expression of MMP-9, whereas NEU3 silencing or the activity-null mutant did not. NEU3 enhanced phosphorylation of epidermal growth factor receptor (EGFR), and an EGFR inhibitor, AG1478, abrogated the NEU3-induced MMP9 augmentation. These findings identify NEU3 as a participant in HNSCC progression through the regulation of EGFR signaling and thus as a potential target for inhibiting EGFR-mediated tumor progression.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Neuraminidase/biosynthesis , Adult , Aged , Aged, 80 and over , Chromatography, Thin Layer , Disease Progression , Female , Humans , Immunoblotting , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Neuraminidase/analysis , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Squamous Cell Carcinoma of Head and Neck , Transfection , Up-Regulation
18.
Cancer Sci ; 106(4): 383-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25652216

ABSTRACT

Aberrant sialylation in glycoproteins and glycolipids is a characteristic feature of malignancy. Human sialidases, which catalyze the removal of sialic acid residues from glycoconjugates, have been implicated in cancer progression. They have been detected in a wide variety of human cells and tissues, but few studies have focused on their existence in human serum. Among the four types identified to date, we previously demonstrated that plasma membrane-associated ganglioside sialidase (NEU3) is markedly upregulated in various human cancers, including examples in the colon and prostate. Here, using a sensitive assay method, we found a significant increase of sialidase activity in the serum of patients with prostate cancer compared with that in healthy subjects having low activity, if any. Activity was apparent with gangliosides as substrates, but only to a very limited extent with 4-methylumbelliferyl sialic acid, a good synthetic substrate for sialidases other than human NEU3. The serum sialidase was also almost entirely immunoprecipitated with anti-NEU3 antibody, but not with antibodies for other sialidases. Interestingly, sera additionally contained inhibitory activity against the sialidase and also against recombinant human NEU3. The sialidase and inhibitor activities could be separated by exosome isolation and by hydrophobic column chromatography. The serum sialidase was assessed by a sandwich ELISA method using two anti-NEU3 antibodies. The results provide strong evidence that the serum sialidase is, in fact, NEU3, and this subtype may, therefore, be a potential utility for novel diagnosis of human cancers.


Subject(s)
Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/blood , N-Acetylneuraminic Acid/metabolism , Neuraminidase/antagonists & inhibitors , Neuraminidase/blood , Prostatic Neoplasms/blood , Biomarkers, Tumor/biosynthesis , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Gangliosides/metabolism , Humans , Male , Neuraminidase/biosynthesis , Neuraminidase/immunology , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
19.
J Pathol ; 234(2): 277-88, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24687913

ABSTRACT

The development of therapeutic resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs, ie erlotinib or gefitinib) has been the major clinical problem when treating lung adenocarcinoma patients with these agents. However, its mechanisms have not necessarily been well studied to this date. Autophagy has been recently considered to play pivotal roles in escaping from the effects of anti-neoplastic agents. Therefore, in this study, we examined its roles in the development of resistance to EGFR-TKIs in lung adenocarcinoma. We first established erlotinib-resistant cell lines (PC9/ER) from parental PC9 cells by exposing the cells to erlotinib. In PC9/ER, autophagy-related LC3A expression came to be up-regulated and constitutive activation of LC3A-mediated autophagy became more pronounced through the process of acquiring therapeutic resistance. In addition, inhibition of LC3A or autophagy restores sensitivity to EGFR-TKIs in PC9/ER. LC3A was also activated at the transcriptional level in de novo resistant cells via demethylation of the MAP1LC3A gene. We then evaluated the status of LC3A in 169 lung adenocarcinoma patients using immunohistochemistry. LC3A immunoreactivity was only detected in carcinoma cells (89/169 patients), not in non-tumoural cells. In addition, LC3A immunoreactivity was significantly correlated with progression-free survival (p = 0.0039) and overall survival (p = 0.0040) of 35 patients treated with EGFR-TKIs. The results of our present study demonstrated that LC3A-mediated autophagy in carcinoma cells was involved in the development of resistance to EGFR-TKIs, and that LC3A could serve as a promising therapeutic target for overcoming resistance to EGFR-TKIs and a novel predictor of response to EGFR-TKIs in lung adenocarcinoma patients.


Subject(s)
Adenocarcinoma/genetics , Antineoplastic Agents/pharmacology , Autophagy/genetics , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Lung Neoplasms/genetics , Microtubule-Associated Proteins/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Erlotinib Hydrochloride , Gefitinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology
20.
Nihon Hinyokika Gakkai Zasshi ; 106(3): 199-205, 2015 Jul.
Article in Japanese | MEDLINE | ID: mdl-26419079

ABSTRACT

The sexual dysfunction and infertility after treatment of bilateral germ cell tumors (GCT) becomes the serious problem. Therefore andrological aspects as well as cancer curability should be considered in planning of bilateral GCT treatment. Here we report 3 cases of metachronous bilateral GCT treated with different regimens, and discuss from the viewpoint of preservation of sexual function. Case presentations: (1) A 38-year-old man underwent right-sided orchitectomy for a right testicular tumor at the age of 26 years. Pathological diagnosis was seminoma and clinical stage was T1N0M0S2. 12 years later, contralateral testicular tumor developed. Left-sided orchitectomy was performed. Pathological diagnosis was seminoma and clinical stage was T1N0M0S2. He has been followed up for 4 years after the second operation without any evidence of tumor recurrence. Endocrinological examination show low testosterone level, and high LH and FSH levels. Erection and ejaculation are impossible but he does not request androgen replacement therapy. (2) A 21-year-old man underwent right-sided orchitectomy for a right testicular seminoma at the age of 20 years (T1 N0M0S0). 1 year later, contralateral seminoma (T1N0M0S0) developed and left-sided organ-preserving operation was performed. Histologic specimens showed seminoma and intratubular malignant germ cells (ITMGC) in surrounding seminiferous tubules. 2 cycles of BEP was added after the operation. He has been followed up for 5 years without any evidence of tumor recurrence. Endocrinological examination shows normal levels of testosterone and LH, but FSH is slightly high. Erection and ejaculation are possible. (3) A 36-year-old man underwent right-sided orchitectomy for a right testicular embryonal carcinoma at the age of 30 years. Clinical T1N0M0S1 was confirmed. 6 years later, he noticed the induration at his left testis. The result of fine needle aspiration cytology was embryonal carcinoma. At first, organ-preserving operation after chemotherapy was planned. However, he refused the operation considering the possibility of erectile dysfunction and infertility. As a result, he received only chemotherapy (3 cycles of BEP), and has been free of the disease for 11 years after chemotherapy. The level of testosterone, LH, and FSH are all normal. Erection and ejaculation are possible.


Subject(s)
Ejaculation , Fertility , Penile Erection , Testicular Neoplasms/physiopathology , Adult , Humans , Male , Neoplasm Staging , Orchiectomy , Testicular Neoplasms/pathology , Testicular Neoplasms/surgery , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL