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










Publication year range
1.
Int J Clin Oncol ; 28(7): 901-912, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37119370

ABSTRACT

BACKGROUND: Pancreatic cancer has an extremely poor prognosis and is one of the most chemoresistant cancers. Targeting cancer cell transcriptional complexes may enhance chemotherapy effectiveness. RNA-polymerase I (Pol-I)-mediated transcription is an essential initial step for ribosome biogenesis and is related to cancer cell proliferation. RRN3 is a Pol-I-specific transcription initiation factor. In this study, we aimed to elucidate the function and clinical significance of RRN3 in pancreatic cancer. METHODS: We performed immunohistochemical staining to detect RRN3 protein expression in 96 pancreatic cancer tissues and analyzed the relationship between RRN3 protein expression, clinicopathological factors, and cancer patient prognosis. Moreover, we evaluated RRN3 function in vitro and in vivo using proliferation, invasion, and chemosensitivity assays in PANC-1 and SW1990 cell lines, with/without depleting RRN3 expression. RESULTS: RRN3 was mainly expressed in cancer cell nuclei. High levels of RRN3 expression were associated with Ki-67 expression and shorter overall survival. Additionally, proliferation and invasion ability were decreased when RRN3 was silenced with siRNA, compared to non-targeting siRNA-transfected cells. Chemosensitivity analysis showed that inhibition of RRN3 enhanced the sensitivity of pancreatic cancer cell lines to gemcitabine and paclitaxel. RRN3 siRNA-transfected PANC-1 tumors showed significantly reduced tumor volumes and high gemcitabine sensitivity compared to the control in a mouse xenograft model. CONCLUSION: High levels of RRN3 expression are associated with poor prognosis and cancer malignancy, such as proliferation, invasion ability, and chemosensitivity in pancreatic cancer. RRN3 targeting with anticancer drugs may be a promising therapeutic strategy to overcome refractory pancreatic cancer.


Subject(s)
Pancreatic Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Cell Proliferation/genetics , Gemcitabine , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Prognosis , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Pancreatic Neoplasms
2.
Ann Gastroenterol Surg ; 6(4): 505-514, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35847440

ABSTRACT

Background: Connexin is a basic molecule that forms gap junctions and undergoes localization changes to the cytoplasm in association with carcinogenesis. We aimed to investigate and clarify the significance of cytoplasmic Cx26 expression in gastric cancer. Methods: We included 87 patients with intestinal- and mix-type gastric cancer and 111 patients with diffuse type gastric cancer who underwent surgery for gastric cancer between 1999 and 2006. Immunohistochemical staining for Cx26, ß-catenin, and Wnt3a was performed and analyses of the relationship to clinicopathological factors were conducted based on the Lauren classification. In an in vitro study, the gastric cancer cell lines MKN7, MKN74, and MKN45 were used to evaluate the proliferative capacity using the water-soluble tetrazolium salt assay through forced expression of Cx26, and the relationship between Cx26 and ß-catenin was investigated using proximity ligation assay (PLA) and co-immunoprecipitation. Additionally, functional analysis was performed by Cage analysis. Results: In this study, high cytoplasmic Cx26 expression was associated with favorable prognosis in intestinal- and mix-type gastric cancer and could be an independent prognostic factor for overall survival. In terms of the mechanism, in in vitro experiments changes in Cx26 localization to the cytoplasm were shown to suppress the change of localization of ß-catenin to the nucleus by binding to it in the cytoplasm. Conclusions: Cytoplasmic Cx26 was found to be a prognostic factor in intestinal- and mix-type gastric cancer. Regarding the mechanism, in vitro studies revealed that cytoplasmic Cx26 inhibits the translocation of ß-catenin to the nucleus.

3.
Ann Surg Oncol ; 29(11): 7149-7162, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35644860

ABSTRACT

BACKGROUND: Fibroblast growth factor receptor (FGFR)-signaling in lung squamous cell carcinoma (LSCC) is associated with cancer aggressiveness and poor prognosis. Small GTPase RAB11A regulates the recycling of membrane proteins such as FGFR. This study evaluated the potential of RAB11A as a new therapeutic target for LSCC through its regulation of FGFR-signaling. METHODS: Immunohistochemical analysis of 84 LSCC samples was performed to determine the correlation between RAB11A expression, clinicopathologic features, and prognosis. Alterations in FGFR-signaling were assessed in RAB11A-suppressed and RAB11A-overexpressed LSCC cells both in vitro and in vivo. RESULTS: The study identified RAB11A as a strong predictor of poor prognosis in the LSCC cohort. Cell proliferation and invasion were promoted and inhibited respectively in RAB11A-overexpressed and RAB11A -suppressed LSCC cells. In RAB11A-overexpressed and RAB11A-suppressed LSCC cells, FGFR-signaling was respectively up- and downregulated. The viability of the cells treated with nintedanib and lenvatinib was greater in RAB11A-overexpressing cells than in control cells. The in vivo tumor growth and micro-vessel density of RAB11A-overexpressing tumors were significantly higher than in the control cells. CONCLUSION: As a potentially valuable prognostic marker, RAB11A is a promising therapeutic target for LSCC. Evaluation of RAB11A may be useful for identification of LSCC in patients whose cancer is refractory to FGFR inhibitors.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Laryngeal Neoplasms , Lung Neoplasms , Monomeric GTP-Binding Proteins , rab GTP-Binding Proteins , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Laryngeal Neoplasms/pathology , Lung/metabolism , Lung Neoplasms/genetics , Monomeric GTP-Binding Proteins/metabolism , Monomeric GTP-Binding Proteins/therapeutic use , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Fibroblast Growth Factor/therapeutic use
4.
Int J Mol Sci ; 23(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35743281

ABSTRACT

Radiation therapy against cancer cells often causes radiation resistance via accumulation of hypoxia-inducible factor 1 subunit alpha (HIF-1α) under hypoxic conditions and severe side effects. Radiation sensitizers without side effects are required to overcome hypoxia-induced radiation resistance and decrease radiation-related side effects in patients with refractory cancer. We previously developed oxygen nanobubble water (NBO2 water) and demonstrated that it suppresses hypoxia-induced radiation resistance in cancer cell lines within the single-nanometer range. This study aimed to elucidate whether NBO2 water could act as a radiosensitizer via regulation of HIF-1α in a tumor-bearing mouse model. Six-week-old female BALB/c mice subcutaneously injected with tumor cells received control water or NBO2 water for 28 days, after which biochemical examinations and radiation treatment were performed. Hypoxic tumor regions were detected immunohistochemically. We found that NBO2 water sensitized radiation reactivity in the xenografted tumors. Notably, NBO2 water administration downregulated the accumulation of HIF-1α in xenografted tumors and did not affect the vital organs of healthy mice. The combination of radiation and single-nanometer NBO2 water without severe side effects may be a promising therapeutic option to improve radiation sensitivity in cancer patients without tolerance to invasive treatments.


Subject(s)
Oxygen , Radiation-Sensitizing Agents , Animals , Cell Hypoxia , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Mice, Inbred BALB C , Oxygen/pharmacology , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Water/pharmacology
5.
Semin Thorac Cardiovasc Surg ; 34(4): 1170-1177, 2022.
Article in English | MEDLINE | ID: mdl-34688900

ABSTRACT

Aortic stenosis (AS) is a disease characterized by narrowing of the aortic valve (AV) orifice. The purpose of this study was to clarify the significance of bacterial detection and clinicopathological factors, including valve-infiltrating immune cells and disease severity, in relation to AS. After obtaining the written informed consent form from 50 patients with AS, we performed immunohistochemical analysis of lipopolysaccharide (LPS) for gram-negative bacteria and lipoteichoic acid (LTA) for gram-positive bacteria on surgically resected-AVs. Moreover, we evaluated the relationships among the presence of bacteria, immune checkpoint protein PD-L1 expression, and immune cell infiltrations such as CD8-positive T lymphocytes, CD163-positive macrophages, and FOXP3-positive regulatory T cell (Treg) in resected-aortic valves. LPS detection in the resected-aortic valve tissues was significantly associated with stromal PD-L1 expression, valve calcification, and LTA existence in resected samples. We showed that the presence of LPS was significantly related to high PD-L1 expression only in calcified-AVs, not in non-calcified-AVs. Moreover, the high expression of PD-L1 in AS samples without LPS was significantly associated with positive infiltration of CD163-positive macrophages and FOXP3-positive Tregs. Immunohistochemical bacterial detection in resected-aortic valves was associated with PD-L1 accumulation and valve calcification. PD-L1 significantly accumulated only in calcified valves with LPS existence.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Humans , Aortic Valve/surgery , Aortic Valve/pathology , B7-H1 Antigen , Immune Checkpoint Proteins , Lipopolysaccharides/metabolism , Treatment Outcome , Aortic Valve Stenosis/surgery , Aortic Valve Stenosis/pathology , Forkhead Transcription Factors/metabolism , Bacteria
6.
Cancer Rep (Hoboken) ; 4(4): e1364, 2021 08.
Article in English | MEDLINE | ID: mdl-33675293

ABSTRACT

BACKGROUND: Lectin-like oxidized LDL receptor-1 (LOX-1) has been identified as a new marker for functional myeloid-derived suppressor cells (MDSCs) that exhibit an immunosuppressive phenotype in the tumor microenvironment (TME). However, the role of LOX-1+ cells in the TME of colorectal cancer (CRC) remains unknown. AIM: This study aimed to determine the expression and significance of LOX-1 in the TME of clinical CRC specimens. METHODS AND RESULTS: We performed immunohistochemical and genetic analyses of LOX-1, CD8, KRAS, and BRAF in 128 resected CRC specimens and determined the expression of IFN-γ and IL-10 using real-time reverse transcription-polymerase chain reaction. We analyzed the correlation between LOX-1, TME factors, gene alteration, clinicopathological factors, and disease prognosis. The co-expression pattern of LOX-1, hematopoietic markers, and a fibroblast marker was evaluated using multiplex immunofluorescence staining. Low stromal LOX-1 expression and low intratumoral CD8+ cytotoxic T-lymphocyte (CTL) status correlated with poor prognosis. Moreover, stromal LOX-1-low/CD8+ CTL-low status was the most important independent prognostic factor of poor overall survival. Most of the LOX-1+ stromal cells were positive for CD163+ , indicating they were CD163+ M2 macrophages. CONCLUSIONS: The MDSC marker, LOX-1, was mainly expressed by M2 macrophages in CRC tissues. LOX-1+ macrophages and CD8+ CTLs may serve as useful biomarkers for predicting the prognosis of CRC.


Subject(s)
Colorectal Neoplasms/mortality , Scavenger Receptors, Class E/metabolism , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , Chemotherapy, Adjuvant , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Prognosis , Scavenger Receptors, Class E/analysis , T-Lymphocytes, Cytotoxic/metabolism , Young Adult
7.
Ann Surg Oncol ; 28(2): 1217-1227, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32529269

ABSTRACT

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is an important cause of cancer-related death worldwide. CD36, a long-chain fatty acid (FA) receptor, can initiate metastasis in human oral squamous cell carcinoma (SCC), and its expression is associated with poor prognosis in several cancers. The clinical significance of CD36 expression and its function in ESCC remain unknown. METHODS: We examined the clinical significance of CD36 expression in 160 ESCC samples using immunohistochemical staining. Functional analysis was performed to determine the association between CD36 and ESCC characteristics (proliferative ability, invasive ability, and energy source dependency). RESULTS: Thirty (18.8%) ESCC cases showed high CD36 expression, indicating a significant association with progression. CD36 suppression inhibited proliferation and invasiveness in ESCC cells. ESCC cells with CD36 suppression used specific essential amino acids (EAAs) as energy sources. Cell viability depended on FAs under CD36 expression. The viability of ESCC cells with CD36 suppression depended on EAAs but not FAs. CONCLUSIONS: CD36 may be a good biomarker and therapeutic target in ESCC. Our data provide new insights into the basic mechanism of CD36-dependent energy utilization for ESCC survival. CD36 might be a key regulator of the dependency of FAs as energy source in ESCC cells.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Head and Neck Neoplasms , Mouth Neoplasms , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Prognosis , Squamous Cell Carcinoma of Head and Neck
8.
PLoS One ; 15(6): e0234471, 2020.
Article in English | MEDLINE | ID: mdl-32520972

ABSTRACT

Abdominal spacers are useful for maintaining the distance between the target tumors and surrounding tissues, such as the gastrointestinal tract, in patients treated with carbon ion radiotherapy. Surgical intervention to remove the spacers is sometimes necessary because of abdominal infections triggered by long-term spacer placement or intestinal perforation. Therefore, spacers that do not require surgical removal and provide effective drainage against abdominal infections are urgently needed. This study aimed to develop a spacer that could be removed non-surgically and one that provides the therapeutic effect of drainage in patients who receive carbon ion radiotherapy for abdominal tumors. A novel fan-shaped spacer was constructed from a film drain that was folded along the trigger line. Simple withdrawal of the trigger line caused the film drain to fold and the holding lines to become free. We performed laparoscopy-assisted insertion with pneumoperitoneum and blind removal of the spacer fourteen times using a porcine model. Saline in the abdominal cavity was effectively aspirated using the spacer. Our novel fan-shaped spacer could be removed safely without surgery and was able to drain fluid effectively from the abdominal cavity.


Subject(s)
Abdominal Neoplasms/radiotherapy , Catheters , Drainage/methods , Heavy Ion Radiotherapy/methods , Abdomen/physiopathology , Abdominal Neoplasms/surgery , Animals , Drainage/instrumentation , Swine
9.
Ann Surg Oncol ; 27(11): 4360-4368, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32356270

ABSTRACT

BACKGROUND: RAD18 plays an important role in DNA damage repair by inducing monoubiquitinated PCNA (mUB-PCNA) in both cancer and normal tissues. Previous studies have not determined the significance of RAD18 expression in clinical gastric cancer (GC) samples. Thus, this study aimed to clarify the expression and functional significance of RAD18 in GC. METHODS: Overall, 96 resected GC samples were subjected to an immunohistochemical analysis of RAD18. GC cell lines were also subjected to functional RNA interference analyses of RAD18. RESULTS: RAD18 expression was predominantly nuclear and was observed at higher levels in GC tissues than in normal tissues. In GC tissues, strong RAD18 expression was associated with progression of lymph node metastasis (p = 0.0001), lymphatic invasion (p = 0.0255), venous invasion (p < 0.0001), recurrence (p = 0.028), and disease stage (p = 0.0253). Moreover, GC patients with high tumor RAD18 expression had shorter overall survival (p = 0.0061) and recurrence-free survival durations (p = 0.035) than those with low tumor RAD18 expression. RAD18 knockdown inhibited GC proliferation and invasiveness and increased chemosensitivity by suppressing mUB-PCNA. CONCLUSIONS: RAD18 expression may be a useful marker of progression and poor prognosis of GC. Moreover, therapeutic strategies that target RAD18 might be a novel chemosensitizer to eradicate the refractory GC.


Subject(s)
DNA-Binding Proteins , Stomach Neoplasms , Ubiquitin-Protein Ligases , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Disease Progression , Humans , Neoplasm Invasiveness , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
11.
Ann Surg Oncol ; 27(3): 933-942, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31571056

ABSTRACT

BACKGROUND: We investigated whether the expression of transforming growth factor-beta-induced protein (TGFBI) and intratumoral immune cells including CD8- and Forkhead box protein P3 (Foxp3)-positive T cells in clinical lung cancer patients could predict the therapeutic response to nivolumab. METHODS: Thirty-three patients who were treated with nivolumab were enrolled in this study. Immunohistochemical analyses of TGFBI, PD-L1, CD8, Foxp3, and vimentin expression were conducted. Serum concentrations of TGFBI and transforming growth factor-beta1 (TGF-ß1) were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS: Cancer TGFBI was not associated with prognosis and therapeutic response to nivolumab, but cancer stromal TGFBI and intratumoral CD8-positive T cells were associated with them. Therefore, we evaluated cancer stromal TGFBI and intratumoral CD8-positive T cells. The high-TGFBI-expression group had poorer clinical responses than did the low-TGFBI-expression group (p < 0.0001). The number of times nivolumab was administered in the high-CD8-expression group was significantly higher than that in the low-CD8-expression group (p = 0.0046). The high-CD8-expression group had better clinical responses than did the low-CD8-expression group (p = 0.0013). Interestingly, all patients in the high-TGFBI/low-CD8-expression group had progressive disease (PD). In contrast, all patients in the low-TGFBI/high-CD8-expression group had PR + SD (partial response + stable disease) by the Response Evaluation Criteria in Solid Tumors (RECIST 1.1). CONCLUSIONS: The dual evaluation of stromal TGFBI and intratumoral CD8-positive T cells could be a useful predictive marker for nivolumab.


Subject(s)
Adenocarcinoma of Lung/pathology , Antineoplastic Agents, Immunological/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , Stromal Cells/metabolism , Transforming Growth Factor beta1/metabolism , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/metabolism , Aged , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CD8-Positive T-Lymphocytes/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Female , Follow-Up Studies , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Male , Prognosis , Prospective Studies , Survival Rate , Transforming Growth Factor beta1/genetics
12.
Oncotarget ; 10(13): 1306-1319, 2019 Feb 12.
Article in English | MEDLINE | ID: mdl-30863491

ABSTRACT

INTRODUCTION: Thymic epithelial tumors (TETs) comprise several histologies of thymoma and thymic carcinomas (TCs), and TC frequently metastasizes and causes death. We therefore aimed here to identify key molecules closely related to prognosis and their biological roles in high-risk TETs, particularly TCs. RESULTS: RNA sequence analysis demonstrated that hypoxia-related genes were highly expressed in TETs. The expression of the hypoxia-related gene CA9 was noteworthy, particularly in TCs. Immunohistochemical analysis revealed that CA9 was expressed in 81.0% of TCs and 20.7% of all TET samples. CA9 expression was significantly associated with Masaoka stage, WHO classification, and recurrence-free survival after tumor resection (P = 0.005). The down-regulation of CA9 transcription in TC cell lines by small interfering RNAs significantly inhibited CA9 expression, which inhibited proliferation and increased sensitivity to irradiation. CONCLUSIONS: CA9 expression may serve as a significant prognostic marker of TETs and therefore represents a potential target for the development of novel drugs and radiation-sensitizing therapy designed to improve the outcomes of patients with TCs. MATERIALS AND METHODS: We performed comprehensive transcriptome sequencing of 23 TETs and physiologic thymic specimens to identify genes highly and specifically expressed in high-risk TETs, particulary TCs. We performed immunohistochemical analysis of 179 consecutive surgically resected TETs to evaluate the significance of the association of protein expression with clinicopathological features and prognosis. The biological significance of the most promising prognostic marker was further studied using the TC cell lines, Ty-82 and MP57.

13.
Ann Surg Oncol ; 26(6): 1744-1750, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30924018

ABSTRACT

BACKGROUND: Lung combined neuroendocrine carcinomas (NECs) comprise NEC and non-NEC components, such as adenocarcinoma and squamous cell carcinoma. Mutation of epidermal growth factor receptor (EGFR) often is observed in non-NEC but is very rare in sporadic NEC, which almost always has p53 mutation. Therefore, we hypothesized the following research concept: mutation analysis of EGFR and p53 in each component of combined NEC tissues can provide important information on whether such components originate from the same tumor cells or incidentally arise as collision cancers. METHODS: We compared the mutations of EGFR and p53 in laser-microdissected NEC and non-NEC from lungs of eight cases affected by combined NEC. We examined the expression of EGFR and NEC markers in the combined NECs by immunohistochemistry. RESULTS: Five of eight cases of combined NEC had the same mutations of EGFR and/or p53 in both non-NEC and NEC. One case had EGFR mutation in only the non-NEC component, and two cases did not have these mutations. Replacement transformation was observed in borderline areas between non-NEC and NEC. The signal of activated EGFR in non-NEC with the same EGFR mutation was more intense than that in NEC components. CONCLUSIONS: Our study suggests the mechanism behind the carcinogenesis of lung combined NEC, which is partially caused by the transformation from epithelial carcinoma of non-NEC to NEC.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Neuroendocrine/pathology , Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , Mutation , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Aged , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , DNA Mutational Analysis , ErbB Receptors/genetics , Female , Follow-Up Studies , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Prognosis , Survival Rate , Tumor Suppressor Protein p53/genetics
14.
Int J Oncol ; 54(3): 833-844, 2019 03.
Article in English | MEDLINE | ID: mdl-30628666

ABSTRACT

Using whole transcriptome analysis and a lentiviral short hairpin RNA screening library, carboxypeptidase A4 (CPA4) was identified as a novel marker in breast cancer and a therapeutic target in triple­negative breast cancer (TNBC) in the present study. Immunohistochemistry was used to evaluate the presence of CPA4, estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2, Ki67, epidermal growth factor receptor, cytokeratin 5/6, aldehyde dehydrogenase 1, cluster of differentiation (CD)44, CD24, claudins, E­cadherin, vimentin and androgen receptor in 221 cases of breast cancer, including 68 TNBC cases. The effects of CPA4 on the viability and migration ability of TNBC cells were analyzed using RNA interference methods. Increased CPA4 expression, specifically in the cytoplasm of cancer tissue cells, was detected. Furthermore, high CPA4 expression in TNBC cases was associated with low expression of E­cadherin and with the expression of cancer stem cell markers (high CD44/low CD24). Patients with TNBC and high levels of CPA4 expression had a significantly poorer prognosis compared with those with low CPA4 expression. Notably, viability and migration were reduced, but E­cadherin expression was upregulated in CPA4­suppressed TNBC cells. The present data suggested that CPA4 may be a novel inducer for epithelial­mesenchymal transition, which is characterized by the downregulation of E­cadherin and mesenchymal phenotypes. To conclude, CPA4 may be a marker for poor prognosis and a promising therapeutic target in TNBC with aggressive phenotypes.


Subject(s)
Carboxypeptidases A/metabolism , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carboxypeptidases A/genetics , Cell Line, Tumor , Cell Movement , Cell Survival , Cytoplasm/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Middle Aged , Phenotype , Prognosis , RNA, Small Interfering/metabolism , Survival Analysis , Triple Negative Breast Neoplasms/genetics
15.
Oncotarget ; 9(77): 34554-34566, 2018 Oct 02.
Article in English | MEDLINE | ID: mdl-30349649

ABSTRACT

PURPOSE: The identification of genes with synthetic lethality in the context of mutant TP53 is a promising strategy for the treatment of basal-like triple negative breast cancer (TNBC). This study investigated regulators of mutant TP53 (R248Q) in basal-like TNBC and their impact on tumorigenesis. EXPERIMENTAL DESIGN: TNBC cells were analyzed by RNA-seq, and synthetic-lethal shRNA knock-down screening, to identify genes related to the expression of mutant TP53. A tissue microarray of 232 breast cancer samples, that included 66 TNBC cases, was used to assess clinicopathological correlates of tumor protein expression. Functional assays were performed in vitro and in vivo to assess the role of ADORA2B in TNBC. RESULTS: Transcriptome profiling identified ADORA2B as up-regulated in basal-like TNBC cell lines with R248Q-mutated TP53, with shRNA-screening suggesting the potential for a synthetic-lethal interaction between these genes. In clinical samples, ADORA2B was highly expressed in 39.4% (26/66) of TNBC patients. ADORA2B-expression was significantly correlated with ER (P < 0.01), PgR (P = 0.027), EGFR (P < 0.01), and tumor size (P = 0.037), and was an independent prognostic factor for outcome (P = 0.036). In line with this, ADORA2B-transduced TNBC cells showed increased tumorigenesis, and ADORA2B knockdown, along with mutant p53 knockdown, decreased metastasis both in vitro and in vivo. Notably, the cytotoxic cyclic peptide SA-I suppressed ADORA2B expression and tumorigenesis in TNBC cell lines. CONCLUSIONS: ADORA2B expression increases the oncogenic potential of basal-like TNBC and is an independent factor for poor outcome. These data suggest that ADORA2B could serve as a prognostic biomarker and a potential therapeutic target for basal-like TNBC.

16.
J Surg Oncol ; 118(6): 966-974, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30260476

ABSTRACT

BACKGROUND AND OBJECTIVES: Transforming growth factor ß-induced (TGFBI) protein is a secreted extracellular matrix protein with conflicting roles in cancer, acting as a tumour suppressor and a promoter, which appears to be tissue specific. The role of TGFBI in gastric cancer (GC) remains unclear, which we aimed to investigate using the clinical samples as well as an in vitro coculture model of GC. METHODS: The clinical significance of TGFBI was assessed in 208 GC samples using immunohistochemistry. Molecular function of TGFBI in the GC cells was examined by small interfering RNA-mediated TGFBI downregulation in the gastric fibroblasts cocultured with the GC cells. RESULTS: TGFBI expression was localised mainly in the cancer stroma and not in the noncancerous gastric tissue or the GC cells. High TGFBI expression was significantly associated with poor prognosis and cancer progression. Downregulation of TGFBI in the cocultured gastric fibroblasts inhibited the invasion and migration abilities of the GC cells. CONCLUSIONS: High stromal TGFBI expression might be a useful predictive marker for poor prognosis in GC patients. Furthermore, TGFBI in the cancer stromal cells is a promising target for GC treatment.


Subject(s)
Extracellular Matrix Proteins/biosynthesis , Stomach Neoplasms/metabolism , Transforming Growth Factor beta/biosynthesis , Aged , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/physiology , Coculture Techniques , Disease Progression , Extracellular Matrix Proteins/genetics , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured
17.
J Surg Oncol ; 118(1): 101-108, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30098297

ABSTRACT

BACKGROUND AND OBJECTIVES: Esophageal squamous cell carcinoma (ESCC) exhibits good reactivity to chemoradiation therapy (CRT). The dysregulation of F-Box and WD Repeat Domain Containing 7 (FBXW7) is associated with therapeutic resistance in cancer cells. However, the correlation between FBXW7 expression and CRT sensitivity in patients with clinical ESCC has been investigated only in few studies. Therefore, this study aimed to elucidate the significance of FBXW7 expression in pretreatment biopsy specimens from patients with ESCC receiving CRT. METHODS: We investigated the relationship between FBXW7 expression and CRT sensitivity in 30 pretreatment biopsy specimens with histological grades of post-CRT surgically resected tumors. Furthermore, we evaluated the effects of high FBXW7 expression on the sensitivity to cytotoxic agents, including docetaxel and nedaplatin, and radiation in ESCC cells in vitro. RESULTS: High FBXW7 expression before CRT correlated with a good pathological CRT response in patients with advanced ESCC (P < .05). Further, our in vitro data showed that both chemo and radiation sensitivity increased in TE-8 and KYSE140 cells overexpressing FBXW7 compared with mock cells because of the degradation of the anti-apoptotic protein MCL1. CONCLUSIONS: The evaluation of FBXW7 expression before CRT treatment is a potential predictor of good responders among patients with ESCC receiving CRT.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/therapy , F-Box-WD Repeat-Containing Protein 7/biosynthesis , Aged , Biopsy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Nucleus/metabolism , Chemoradiotherapy, Adjuvant , Docetaxel , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/radiotherapy , Esophageal Squamous Cell Carcinoma , Female , Fluorouracil/administration & dosage , Humans , Immunohistochemistry , Male , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoadjuvant Therapy , Organoplatinum Compounds/administration & dosage , Retrospective Studies , Taxoids/administration & dosage
18.
Int J Oncol ; 52(3): 679-686, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29393397

ABSTRACT

Radiation therapy can result in severe side-effects, including the development of radiation resistance. The aim of this study was to validate the use of oxygen nanobubble water to overcome resistance to radiation in cancer cell lines via the suppression of the hypoxia-inducible factor 1-α (HIF­1α) subunit. Oxygen nanobubble water was created using a newly developed method to produce nanobubbles in the single-nanometer range with the ΣPM-5 device. The size and concentration of the oxygen nanobubbles in the water was examined using a cryo-transmission electron microscope. The nanobubble size was ranged from 2 to 3 nm, and the concentration of the nanobubbles was calculated at 2x1018 particles/ml. Cell viability and HIF-1α levels were evaluated in EBC­1 lung cancer and MDA­MB­231 breast cancer cells treated with or without the nanobubble water and radiation under normoxic and hypoxic conditions in vitro. The cancer cells grown in oxygen nanobubble-containing media exhibited a clear suppression of hypoxia-induced HIF­1α expression compared to the cells grown in media made with distilled water. Under hypoxic conditions, the EBC­1 and MDA­MB231 cells displayed resistance to radiation compared to the cells cultured under normoxic cells. The use of oxygen nanobubble medium significantly suppressed the hypoxia-induced resistance to radiation compared to the use of normal medium at 2, 6, 10 and 14 Gy doses. Importantly, the use of nanobubble media did not affect the viability and radiation sensitivity of the cancer cell lines, or the non­cancerous cell line, BEAS­2B, under normoxic conditions. This newly created single-nanometer range oxygen nanobubble water, without any additives, may thus prove to be a promising agent which may be used to overcome the hypoxia-induced resistance of cancer cells to radiation via the suppression of HIF-1α.


Subject(s)
Cell Hypoxia/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/radiotherapy , Oxygen/administration & dosage , Radiation Tolerance/drug effects , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Humans , Oxygen/chemistry , Water/chemistry
19.
Anticancer Res ; 38(2): 939-944, 2018 02.
Article in English | MEDLINE | ID: mdl-29374725

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related deaths worldwide. Stathmin 1 (STMN1) suppression was reported to reduce cellular viability and migration potential. However, no previous studies have addressed whether STMN1 overexpression is associated with malignant potential in PDAC. MATERIALS AND METHODS: To clarify the clinical significance of STMN1 in PDAC, the STMN1 expression in 104 PDAC samples was evaluated by immunohistochemistry. Moreover, we evaluated the proliferative potential and migration ability of pancreatic cancer cell line Suit2 cells highly expressing STMN1. RESULTS: Cytoplasmic STMN1 were higher levels in PDAC than in corresponding non-cancerous tissues. PDAC patients with high STMN1 (n=29) were significantly associated with poor differentiation and distant metastasis compared to those with low STMN1 (n=75). The proliferation rates and migration ability in Suit2-STMN1 were higher than those of Suit2-mock. CONCLUSION: STMN1 evaluation could be a useful progression marker, and STMN1 may be a promising candidate for targeted therapies in PDAC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/secondary , Cell Differentiation , Cell Movement , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/pathology , Stathmin/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Case-Control Studies , Cell Proliferation , Disease Progression , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Middle Aged , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Prognosis , Stathmin/genetics , Tumor Cells, Cultured
20.
J Surg Oncol ; 116(6): 706-715, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28570747

ABSTRACT

BACKGROUND AND OBJECTIVES: The Caspase14 (CASP14) was reported that the low expression of CASP14 in ovarian cancer and colon cancer was associated with cancer progression, on the other hand, that the CASP14 expression in breast cancer was higher than that of non-cancerous tissues. The purpose of this study is to determine the clinical significance of CASP14 in breast cancer. METHODS: We performed immunohistochemistry for CASP14, ER, PgR, HER2, Ki67, EGFR, CK5/6, CD44, CD24, ALDH1, claudins, and androgen receptor in 222 breast cancer patients including 55 TNBC cases, and evaluated the relationship of CASP14, above-mentioned markers, and prognosis. Using public microarray database of breast cancer, the prognostic value of CASP14 was calculated. RESULTS: High CASP14 expression was significantly associated with TNBC subtype (P = 0.015), nuclear grade (P = 0.006), Ki67, EGFR (P < 0.001, P = 0.016), ALDH1, CD44 and CD24 (P < 0.001, P < 0.001, P = 0.001) in 222 breast cancer cases, and the high expression of claudin1 (P = 0.017), and androgen receptor (P = 0.002) in TNBC cases was related to the high CASP14. According to the public database, survival in the high CASP14 breast cancer patients was shorter than low CASP14 patients. CONCLUSIONS: High CASP14 expression is a marker of breast cancer aggressiveness in association with proliferation, TNBC phenotype, and cancer stemness.


Subject(s)
Caspases/biosynthesis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/pathology , Caspases/genetics , Cell Line, Tumor , Female , Humans , Immunohistochemistry , MCF-7 Cells , Middle Aged , Phenotype , Triple Negative Breast Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...