Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Hepatocell Carcinoma ; 10: 1587-1593, 2023.
Article in English | MEDLINE | ID: mdl-37791067

ABSTRACT

Background: Advanced-stage hepatocellular carcinoma (HCC), especially huge HCC or portal vein tumour thrombus (PVTT), is difficult to treat, and the prognosis is poor. The advantages of hepatic artery infusion chemotherapy (HAIC) combined with targeted therapy and immunotherapy for this complex disease are gradually becoming apparent. However, HAIC still has some inevitable disadvantages, such as arterial perfusion therapy requiring a long time, which results in many patients having difficulty completing the procedure. Modified HAIC (mHAIC)-based oxaliplatin and S-1 is a new treatment option for huge HCC or PVTT that can reduce complications and improve patient compliance. We report two cases of huge HCC or PVTT that were successfully treated with mHAIC combined with lenvatinib and camrelizumab. The clinical presentations, treatment strategies, and outcomes of these cases are presented. Case Presentation: Case 1: A 52-year-old female was found to have a huge HCC with a size of 14×11 cm. She was treated with one cycle of mHAIC combined with transcatheter arterial chemoembolization (TACE), lenvatinib and camrelizumab and 3 cycles of mHAIC in combination with lenvatinib and camrelizumab. The patient's follow-up maintenance therapy with lenvatinib and camrelizumab has been evaluated for efficacy in achieving complete response (CR). Case 2: A 57-year-old man was diagnosed with advanced HCC in combination with PVTT. He achieved partial remission (PR) after four cycles of mHAIC combined with lenvatinib and camrelizumab. This was followed by treatment with lenvatinib and camrelizumab with an efficacy assessment for CR, and progression-free survival (PFS) was 7 months. Conclusion: For advanced HCC with a large mass or PVTT, mHAIC combined with lenvatinib and camrelizumab is a safe and effective treatment with good patient compliance.

2.
Front Oncol ; 13: 1164368, 2023.
Article in English | MEDLINE | ID: mdl-37124509

ABSTRACT

Second-line treatment for metastatic or locally advanced urothelial cancer (UC) is limited. Immunotherapy is approved as a second-line treatment for metastatic UC. Its use as a first-line agent is limited to patients who are ineligible for cisplatin-based treatments. The fibroblast growth factor receptor (FGFR) inhibitor, erdafitinib, can be applied as a third-line approach after the failure of these prior treatments in eligible patients. Therefore, it is especially important to combine limited drugs for second-line treatment of advanced or metastatic UC. Anlotinib is a multiple tyrosine kinase inhibitor agent with both anti-angiogenic and FGFR inhibitory effects. For two patients with advanced and metastatic UC, we combined anlotinib and tislelizumab therapy even though there is no indication of its use. We describe two patients with programmed death ligand-1 (PD-L1)-negative advanced bladder cancer, one with FGFR3 mutation and another with FGFR3 wild type. Both patients had progressed after first-line chemotherapy with gemcitabine and cisplatin. We selected anlotinib in combination with tislelizumab, a programmed death-1 (PD-1) immune checkpoint inhibitor, for second-line treatment. Responses were evaluated as partial remission in both cases, who achieved up to 12 months of progression-free survival with no significant adverse events. Two patients with PD-L1-negative UC underwent second-line therapy using tislelizumab in combination with anlotinib, and the efficacy was better than that of tislelizumab alone. These results suggest that anlotinib may act synergistically with tislelizumab in the treatment of UC.

3.
Front Oncol ; 13: 1015976, 2023.
Article in English | MEDLINE | ID: mdl-36937414

ABSTRACT

Background: Irinotecan-loaded drug-eluting beads transarterial chemoembolization (DEBIRI-TACE) is a safe and effective therapeutic option for unresectable colorectal liver metastases (CRLM). The evaluation of treatment response after DEBIRI-TACE is very important for assessing the patient's condition. At present, the Response Evaluation Criteria in Solid Tumors (RECIST) with the tumor size obtained by CT and/or MRI and PET Response Criteria in Solid Tumors (PERCIST) based on fluorodeoxyglucose-positron emission tomography/computed tomography (FDG PET/CT) are used for evaluating the response to therapy of solid tumors; however, their value in the assessment of treatment response after DEBIRI-TACE remains unclear. Case presentation: A 52-year-old male with unresectable simultaneous CRLM was treated in the Affiliated Hospital of Yanbian University with DEBIRI-TACE combined with systemic chemotherapy and targeted therapy. Carcinoembryonic antigen levels decreased by 82.50% after 27 days of treatment. At 6 weeks post-surgery, FDG-PET/CT showed that the maximum standardized uptake value (SUVmax) of intrahepatic lesions was reduced to 62.14%. Abdominal MRI revealed that the sum of target lesion diameters was less than 30% that at baseline. PERCIST indicated partial metabolic response, whereas RECIST suggested stable disease. Conclusion: FDG PET/CT-based PERCIST may be accurate in determining treatment response and evaluating patient prognosis after DEBIRI-TACE in unresectable CRLM.

4.
Histol Histopathol ; 38(4): 467-474, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36259602

ABSTRACT

OBJECTIVE: To explore the correlation between the expression level of Desmoglein 2 (DSG2) and the epithelial-mesenchymal transition (EMT) progression in gallbladder cancer (GBC). METHOD: 106 GBC tissue specimens and corresponding clinical information were collected to make a tissue microarray. Immunohistochemical method was used to test the expression level of DSG2 in GBC tissues. DSG2 was knocked down in the GBC cell line GBC-SD to detect the change of its invasion and metastasis ability. Then RT-qPCR and Western Blot were applied on the DSG2-knocked down GBC-SD cells to detect the expression level change of genes associated with EMT. RESULT: The high expression rate of DSG2 was significantly correlated with the N, M and TNM staging of patients (P<0.05). Survival analysis identified that GBC patients with high DSG2 expression level had significantly better survival (P<0.05). To further investigate the potential mechanism of DSG2 on regulating GBC tumor progression, we used knockdown DSG2 on GBC-SD cell lines. The results showed that GBC-SD cell lines with DSG2 knockdown showed a promotion of cell invasion and metastatic ability. The mRNA levels of EMT-related genes E-Cadherin, Snail, Twist, ZEB1, and ß-catenin, which is a key protein in the Wnt signaling pathway, were also significantly altered. Besides, protein levels of E-cadherin and Snail showed consistent results. CONCLUSION: The downregulation of DSG2 in gallbladder cancer is hypothesized to be associated with the invasion and metastasis progression of gallbladder cancer cells by regulating EMT-related pathways. Its expression level can be a novel biomarker for gallbladder cancer, providing new perspectives for diagnosis and treatment strategies.


Subject(s)
Desmoglein 2 , Epithelial-Mesenchymal Transition , Gallbladder Neoplasms , Humans , Cell Line, Tumor , Cell Movement , Cell Proliferation , Desmoglein 2/genetics , Desmoglein 2/metabolism , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Prognosis
5.
Technol Cancer Res Treat ; 19: 1533033820967455, 2020.
Article in English | MEDLINE | ID: mdl-33267707

ABSTRACT

USP15 is a member of ubiquitin-specific proteases (USPs, the largest subfamily of deubiquitinases) and functions as a stabilize factor of target proteins in reversible ubiquitiantion progression. Dysregulated expression of USP15 has been observed in various cancers. However the expression profile and regulatory mechanism of USP15 in hepatocellular carcinoma (HCC) remains largely elusive. To exam the USP15 expression changes in the progression of HCC, we performed IHC analysis to test USP15 expression in a series of cancer-prone diseases including 2 normal liver tissues, 6 liver cirrhosis, 16 primary liver lesions and 15 metastases of hepatocellular carcinoma. The expression of USP15 was upregulated in various liver diseases in compared with normal tissue significantly (p < 0.05). Although no significant different of USP15 expression were discovered between cirrhotic tissue and primary tissue, its expression in HCC metastatic tissue was upregulated. Subsequently, we test the USP15 expression profile in a cohort of 66 HCC patients. USP15 expression was positively correlated with the recurrence of HCC significantly (p = 0.004). HCC patients with high USP15 expression had shorter disease free survival time in compare with those with low USP15 expression (56.9% VS 26.7%, P = 0.012). Subsequently, Cox multivariate analyses of clinical factors associated with disease free survival were performed and USP15 expression (p = 0.008) together with tumor size (p = 0.034) were proved to be independent predict factors in HCC. Then, we silenced USP15 expression in HCC cells and the results showed that downregulated USP15 expression resulting proliferation inhibition and apoptosis induction. In conclusion, our results suppose USP15 to be a potential target in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Ubiquitin-Specific Proteases/genetics , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Liver/pathology , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Prognosis , Ubiquitin/genetics
6.
BMC Cancer ; 19(1): 898, 2019 Sep 09.
Article in English | MEDLINE | ID: mdl-31500591

ABSTRACT

BACKGROUND: Although the leucine zipper-EF-hand-containing transmembrane protein 1 (LETM1) is one of the mitochondrial inner membrane proteins that is involved in cancer prognosis in various tumors, LETM1 as a biomarker for prognostic evaluation of non-small cell lung carcinoma (NSCLC) has not been well studied. METHODS: To address this issue, we used 75 cases NSCLC, 20 cases adjacent normal lung tissues and NSCLC cell lines. We performed immunohistochemistry staining and western blot analysis as well as immunofluorescence imaging. RESULTS: Our studies show that expression of LETM1 is significantly correlated with the lymph node metastasis (p = 0.003) and the clinical stage (p = 0.005) of NSCLC. The Kaplan-Meier survival analysis revealed that NSCLC patients with positive expression of LETM1 exhibits a shorter overall survival (OS) rate (p = 0.005). The univariate and multivariate Cox regression analysis indicated that LETM1 is a independent poor prognostic marker of NSCLC. In addition, the LETM1 expression is correlated with cancer stemness-related gene LGR5 (p < 0.001) and HIF1α expression (p < 0.001), but not with others. Moreover, LETM1 expression was associated with the expression of cyclin D1 (p = 0.003), p27 (p = 0.001), pPI3K(p85) (p = 0.025), and pAkt-Thr308 (p = 0.004). Further, our studies show in LETM1-positive NSCLC tissues the microvessel density was significantly higher than in the negative ones (p = 0.024). CONCLUSION: These results indicate that LETM1 is a potential prognostic biomarker of NSCLC.


Subject(s)
Calcium-Binding Proteins/analysis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/analysis , Aged , Biomarkers, Tumor/analysis , Calcium-Binding Proteins/genetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Male , Membrane Proteins/genetics , Middle Aged , Prognosis , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics
7.
Mol Med Rep ; 19(5): 3367-3375, 2019 May.
Article in English | MEDLINE | ID: mdl-30896806

ABSTRACT

Leucine zipper/EF­hand­containing transmembrane protein 1 (LETM1) has been identified as the gene responsible for Wolf­Hirschhorn syndrome (WHS), which is characterized by intellectual disability, epilepsy, growth delay and craniofacial dysgenesis. LETM1 is a mitochondrial inner membrane protein that encodes a homolog of the yeast protein Mdm38, which is involved in mitochondrial morphology. In the present review, the importance of LETM1 in WHS and its role within the mitochondrion was explored. LETM1 governs the mitochondrion ion channel and is involved in mitochondrial respiration. Recent studies have reported that LETM1 acts as a mitochondrial Ca2+/H+ antiporter. LETM1 has also been identified as a K+/H+ exchanger, and serves a role in Mg2+ homeostasis. The function of LETM1 in mitochondria regulation is regulated by its binding partners, carboxyl­terminal modulator protein and mitochondrial ribosomal protein L36. Therefore, we describe the remarkable role of LETM1 in mitochondrial network physiology and its function in mitochondrion­mediated cell death. In the context of these findings, we suggest that the participation of LETM1 in tumorigenesis through the alteration of cancer metabolism should be investigated. This review provides a comprehensive description of LETM1 function, which is required for mitochondrial homeostasis and cellular viability.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Survival/genetics , Homeostasis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Animals , Calcium-Binding Proteins/chemistry , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Energy Metabolism , Humans , Membrane Proteins/chemistry , Structure-Activity Relationship
8.
Int J Surg ; 58: 37-45, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30121307

ABSTRACT

BACKGROUND: Recently, long non-coding RNA activated by transforming growth factor beta (TGF-ß) (lncRNA ATB) was shown to be useful in cancer prognosis, however, its prognostic value in human cancer has been inconsistent. Our study aimed to explore the prognostic role of lncRNA ATB expression in cancer prognosis. METHODS: PubMed, Embase, and Cochrane Library databases were thoroughly searched to retrieve studies focusing on the prognostic role of lncRNA ATB expression in cancer, and meta-analysis was performed. RESULTS: A total of 15 studies were included into this meta-analysis. High lncRNA ATB expression was significantly related to shorter overall survival (OS) (HR = 2.44, 95%CI = 1.98-3.01, P < 0.01), recurrence-free survival (RFS) (HR = 1.85, 95%CI = 1.42-2.40, P < 0.01), disease-free survival (DFS) (HR = 3.61, 95%CI = 2.45-5.33, P < 0.01), and progression-free survival (PFS) (HR = 2.97, 95%CI = 2.12-4.16, P < 0.01) when compared with low lncRNA ATB expression in cancer. Moreover, Patients with high lncRNA ATB expression tended to have worse tumor differentiation (P < 0.01), more advanced clinical stage (P < 0.01), deeper tumor invasion (P < 0.01), earlier distant metastases (P = 0.02), lymph node metastases (P = 0.04), and vascular invasion (P < 0.01) when compared with those with low lncRNA ATB expression. CONCLUSIONS: High lncRNA ATB expression was significantly associated with worse prognosis in cancer. LncRNA ATB expression could be used as a prognostic biomarker for human cancer.


Subject(s)
Neoplasms/mortality , RNA, Long Noncoding/physiology , Transforming Growth Factor beta/physiology , Female , Humans , Lymphatic Metastasis , Neoplasms/genetics , Neoplasms/pathology , Prognosis , RNA, Long Noncoding/analysis
9.
Hum Pathol ; 81: 148-156, 2018 11.
Article in English | MEDLINE | ID: mdl-30031102

ABSTRACT

Leucine zipper-EF-hand containing transmembrane protein 1 (LETM1) is closely related to the occurrence and development of malignant tumors. This study discusses the expression of LETM1 in esophageal squamous cell carcinoma (ESCC) and its association with cancer stem-like cells (CSC). We used immunohistochemistry in 166 ESCC tissue samples, as well as Western blot and immunofluorescent methods in ESCC cell lines, to study the role of LETM1 and its association with CSC in ESCC. The expression of LETM1 was significantly higher in ESCC, and it was closely related to the primary tumor stage and clinical stage. LETM1 expression was significantly associated with lower overall survival and disease-free survival. In addition, the protein expression of LETM1 and CSC markers was higher in TE11 and ECG10 than in other ESCC cell lines. Moreover, the expression of LETM1 positively correlated with LSD1, CD44, and OCT4. Immunofluorescence revealed that LETM1 costained with CD44 and OCT4 in ECG10. The expression of LETM1 was associated with not only HIF-1α but also higher microvessel density and tumor-associated macrophage infiltration. Furthermore, LETM1 significantly correlated with cyclinD1 and pAkt. High expression of LETM1 indicates poor prognosis and may be a potential CSC marker in ESCC. Moreover, LETM1 may be a novel therapeutic target for the treatment of ESCC.


Subject(s)
Calcium-Binding Proteins/metabolism , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Membrane Proteins/metabolism , Neoplastic Stem Cells/metabolism , Aged , Cell Line, Tumor , Cyclin D1/metabolism , Disease-Free Survival , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/surgery , Female , Histone Demethylases/metabolism , Humans , Hyaluronan Receptors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Middle Aged , Neoplasm Staging , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic , Octamer Transcription Factor-3/metabolism , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Time Factors , Tumor Microenvironment
10.
Hum Pathol ; 80: 152-162, 2018 10.
Article in English | MEDLINE | ID: mdl-29885401

ABSTRACT

B7H4 is overexpressed in human cancers and often correlates with poor clinical outcome. There is a lack of data on the role of B7H4 as a cancer stem cell (CSC) regulator in esophageal squamous cell carcinoma (ESCC) and its expression levels compared to other stemness genes in ESCC. In this study, we have assessed the expression of B7H4 and cancer stemness proteins in 156 paraffin-embedded ESCC tissue samples using immunohistochemistry as well as in ESCC cell lines using Western blotting and immunofluorescence imaging. The correlation of B7H4 expression with clinicopathological parameters, cell cycle regulating genes, and PI3K/Akt/NF-κB signaling genes was investigated. The expression of B7H4 in ESCC tissue was correlated with the primary tumor (pT) stage, stromal activity, and the expression of CD68 and HIF-1α. However, B7H4 expression was negatively associated with CD8+ T cell infiltration in ESCC tissues. Moreover, B7H4 was found to be strongly linked to prognostic factors leading to poor clinical outcome. B7H4-expressing cancer cells also expressed known cancer stemness proteins (Sox9, LSD1, Oct4, and LGR5). Moreover, B7H4, Sox9, LSD1, Oct4, and LGR5 were highly expressed in more poorly differentiated ESCC cell lines. Notably, B7H4 expression was positively associated with the expression of cell cycle regulators such as cyclin D1, p27, and PI3K/Akt/NFκB signaling proteins. B7H4 could be a novel cancer stem cell marker for the prognostic evaluation of ESCC patients as well as a potential therapeutic target against ESCC.


Subject(s)
Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Proliferation/genetics , Esophageal Neoplasms/genetics , Female , Humans , Immunohistochemistry/methods , Male , Neoplastic Stem Cells/pathology , Phosphatidylinositol 3-Kinases/genetics , Prognosis
11.
Anticancer Res ; 37(10): 5415-5423, 2017 10.
Article in English | MEDLINE | ID: mdl-28982851

ABSTRACT

Unopposed phosphoinositide 3-kinase (PI3K) activity and 3-phosphoinositide production in Jurkat cells, due to a mutation in the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor-suppressor protein, results in deregulation of PH domain-containing proteins including the serine/threonine kinase PKB. In Jurkat cells, PKB is constitutively active and phosphorylated at the activation-loop residue (Thr308). 3-Phosphoinositide-dependent protein kinase-1 (PDK1), an enzyme that also contains a PH domain, catalyses Thr308 phosphorylation of PKB in addition to other kinase families such as PKC isoforms. It is unknown, however, whether the loss of PTEN in Jurkat cells also results in unregulated PDK1 activity and whether such loss has an impact on activation-loop phosphorylation of other PDK1 substrates e.g. PKC. In this study, we addressed whether loss of PTEN in Jurkat cells affects PDK1 catalytic activity and intracellular localization. We demonstrated that reducing the level of 3-phosphoinositides in Jurkat cells with pharmacological inhibitors of PI3K or expression of PTEN does not affect PDK1 activity or its intracellular localization. We conclude, therefore, that although Jurkat cells lack PTEN expression, only a subset of pathways downstream of PDK1 are perturbed as a consequence of PTEN loss.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Leukemia, T-Cell/enzymology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Catalysis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Time Factors , Transfection
12.
Exp Mol Pathol ; 98(2): 254-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25617527

ABSTRACT

Triple negative breast cancer (TNBC) is a heterogeneous disease with higher rates of relapse and decreased overall survival in metastatic tumors. Due to its poor prognosis, it is necessary to identify effective biomarkers that are associated with tumor growth and metastasis. The leucine zipper/EF hand-containing transmembrane-1 (LETM1) protein, which is a mitochondrial inner membrane protein, can reduce mitochondrial biogenesis and ATP production. The expression levels of LETM1 were significantly increased in numerous human malignancies. However, the clinicopathological characteristics and prognostic value of LETM1 overexpression in TNBC remains unclear. LETM1 protein was detected in 107 TNBC, 42 ductal carcinoma in situ (DCIS) and 65 adjacent non-tumor breast tissues using immunohistochemical (IHC) staining. Immunofluorescence (IF) staining was also performed to detect the localization of LETM1 protein in MCF-7 BC cells. The correlations between LETM1 overexpression and clinicopathological features of TNBC were evaluated using Chi-squared test and Fisher's exact tests. The survival rate was calculated using the Kaplan-Meier method. LETM1 protein showed cytoplasmic staining patterns in TNBC. The strongly positive rate of LETM1 in TNBC was 69.2% (74/107), which was significantly higher than in both DCIS 35.7% (15/42) and adjacent non-tumor tissues 12.3% (8/65). High-level expression of LETM1 was positively correlated with late clinical stage, poor differentiation, lymph node metastasis, disease-free survival (DFS) and 10-year overall survival (OS) rates in TNBC. Further analysis showed that high LETM1 expression along with clinical stage emerged as significant independent risk factors in patients with TNBC. In conclusion, LETM1 protein overexpression is associated with TNBC progression, and may be a potential biomarker for poor prognostic evaluation of TNBC.


Subject(s)
Biomarkers, Tumor/biosynthesis , Calcium-Binding Proteins/biosynthesis , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Membrane Proteins/biosynthesis , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Disease-Free Survival , EF Hand Motifs , Female , Humans , Leucine Zippers , Lymphatic Metastasis , MCF-7 Cells , Middle Aged , Mitochondrial Membranes/pathology , Mitochondrial Proteins/biosynthesis , Triple Negative Breast Neoplasms/mortality
13.
Int J Clin Exp Pathol ; 8(10): 12893-900, 2015.
Article in English | MEDLINE | ID: mdl-26722481

ABSTRACT

BACKGROUND: Leucine zipper/EF hand-containing transmembrane-1 (LETM1) is a mitochondrial inner membrane protein that was first identified in Wolf-Hirschhorn syndrome. However, high-level expression of LETM1 has been correlated with multiple human malignancies, suggesting roles in carcinogenesis and tumor progression. This study is aimed to explore the clinicopathological characteristics and prognostic value of LETM1 overexpression in breast cancer. METHODS: Immunohistochemical (IHC) staining, and immunofluorescence (IF) were performed to examine LETM1 expression in breast cancer cell line/tissues compared with adjacent normal tissues. Statistical analysis was applied to evaluate the correlation between LETM1 overexpression and the clinicopathological features of breast cancer. Survival rates were calculated using the Kaplan-Meier method, and the relationship between prognostic factors and patient survival was analyzed using the Cox proportional hazard models. RESULTS: LETM1 protein showed cytoplasmic staining pattern in breast cancer. The strongly positive rate of LETM1 protein was 61.6% (98/159) in breast cancer, which was significantly higher than in DCIS (29.7%, 11/37), hyperplasia (16.7%, 3/18) and adjacent normal breast tissues (15.9%, 7/44). High-level expression of LETM1 protein was correlated with lymph node metastasis, poor differentiation, late clinical stage, disease-free survival (DFS) and overall survival (OS) rates in breast cancer. Moreover, multivariate analysis suggested that LETM1 emerged as a significant independent prognostic factor along with clinical stage of patients with breast cancer. CONCLUSIONS: LETM1 plays an important role in the progression of breast cancer. High level expression of LETM1 is an independent poor prognostic factor of breast cancer.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Calcium-Binding Proteins/biosynthesis , Carcinoma, Ductal, Breast/pathology , Membrane Proteins/biosynthesis , Adult , Aged , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Calcium-Binding Proteins/analysis , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/mortality , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Disease Progression , Disease-Free Survival , Female , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Membrane Proteins/analysis , Middle Aged , Prognosis , Proportional Hazards Models , Up-Regulation
14.
Biomed Res Int ; 2014: 850316, 2014.
Article in English | MEDLINE | ID: mdl-24689060

ABSTRACT

Leucine zipper-EF-hand containing transmembrane protein 1 (LETM1) is a mitochondrial inner membrane protein and plays an important role in mitochondrial ATP production and biogenesis. High expression levels of LETM1 have been correlated with numerous human malignancies. This study explored the clinicopathological significance of LETM1 expression as a prognostic determinant in head and neck squamous cell carcinoma (HNSCC). HNSCC samples from 176 patients were selected for immunohistochemical staining of LETM1 protein. Correlations between LETM1 overexpression and clinicopathological features of HNSCC were evaluated by Chi-squared tests and Fisher's exact tests, and relationships between prognostic factors and patient survival were analyzed using Cox proportional hazards models. Our results demonstrated that the strongly positive rate of LETM1 protein was 65.3% in HNSCC, which was significantly higher than in either adjacent nontumor tissue (25.0%) or normal squamous epithelia (6.7%). LETM1 overexpression correlated with poor differentiation, presence of lymph node metastasis, advanced stage, absence of chemoradiotherapy, and 5-year disease-free survival and overall survival rates in HNSCC. Further analysis showed that high LETM1 expression, advanced stage, and nonchemoradiotherapy were significant independent risk factors for mortality in HNSCC. In conclusion, LETM1 plays an important role in the progression of HNSCC and is an independent poor prognostic factor for HNSCC.


Subject(s)
Calcium-Binding Proteins/metabolism , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Membrane Proteins/metabolism , Aged , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Epithelium/metabolism , Epithelium/pathology , Female , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , Squamous Cell Carcinoma of Head and Neck
15.
Int J Mol Med ; 33(5): 1364-70, 2014 May.
Article in English | MEDLINE | ID: mdl-24626642

ABSTRACT

Antipsychotic drugs are regularly used for the treatment of many types of psychiatric disorders. The administration of second-generation antipsychotics is often associated with weight gain and the development of diabetes mellitus; however, the molecular mechanisms underlying the effects of these drugs remain poorly understood. Leptin and insulin play key roles in the regulation of energy balance and glucose homeostasis, and resistance to the actions of these hormones can occur with obesity and inflammation, resulting in the pathogenesis of obesity and type 2 diabetes. In this study, the effects of risperidone on the insulin-induced protein kinase B (PKB) phosphorylation and leptin-stimulated signal transducer and activator of transcription 3 (STAT3) phosphorylation were investigated in the human SH-SY5Y neuroblastoma cell line. The treatment of these cells with risperidone induced the activation of extracellular signal-related kinase (ERK) by cellular cyclic adenosine 3-monophosphate (cAMP)-dependent protein kinase (also known as protein kinase A; PKA) and the mechanisms involved include the induction of suppressor of cytokine signaling 3 (SOCS3) and suppressor of cytokine signaling 6 (SOCS6) expression. The risperidone-induced ERK activation induced an upregulation of SOCS3 and SOCS6 mRNA expression levels. Taken together, these results suggest that risperidone modulates SOCS3 and SOCS6 expression through adenylate cyclase-mediated ERK activation, which, in turn, leads to an inhibition of insulin-induced PKB phosphorylation and leptin-stimulated STAT3 phosphorylation. Eventually, these effects result in excessive body weight gain due to the inhibition of both the leptin and insulin signaling pathways.


Subject(s)
Insulin/pharmacology , Leptin/pharmacology , Neuroblastoma/metabolism , Risperidone/pharmacology , Cell Line, Tumor , Humans , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
16.
Cancer Cell Int ; 14(1): 10, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24472145

ABSTRACT

INTRODUCTION: Although advanced gastric cancer has many limitations and response rate is marginal in chemotherapy. Overexpression of human epidermal growth factor receptor 2(HER-2/neu) gene and its protein are associated with increased cell division and a high rate of tumor growth and have been reported in several malignancies. Especially, approximately 30% of breast cancer patients have overexpression of HER-2/neu protein and the overexpression metastasize faster, induces resistance of the chemotherapy and down-regulate function of estrogen receptor. Recombinant humanized anti-HER2 antibody (Herceptin) inhibits proliferation of HER-2/neu overexpressing tumor cells and the use of that in combination in metastatic breast cancer have increased cytotoxicity of chemotherapeutic agents. METHODS: We evaluated the expression of HER-2/neu protein in gastric cell lines by FACS and then comparing the cytotoxicity in chemotherapeutics (doxorubicin, cisplatin, paclitaxel, 5-FU) alone and in combination with Herceptin according to the expression of HER-2/neu protein by MTT assay. RESULTS: 1. NCI-N87 (88%) gastric cancer cell line and SK-BR-3 (89%) breast cancer cell line with strong positivity of HER-2/neu expression. YBC-2 (55%) and YBC-3 (48%) gastric cancer cell line with intermediated, weak positivity respectively. Negative control U-87 MG (6%) brain cancer cell line were showed low expression of HER-2/neu. 2. Cell growth was dose-dependently inhibited in HER-2/neu positive, control cell line SK-BR-3 by Herceptin treatment but not observed in HER-2/neu negative control cell line U-87 MG. Effective growth inhibition was not observed in gastric cancer cell lines with single treatment of Herceptin, all cell lines observed the dose-dependent growth inhibition to chemotherapeutic agents (doxorubicin, cisplatin, paclitaxel and 5-FU). 3. Combination of Herceptin with doxorubicin observed synergistic effects in all cancer cell lines except YBC-3, combination of Herceptin with cisplatin observed NCI-N87 and SK-BR-3 and combination of Herceptin with paclitaxel observed synergistic effects in YBC-2. Combination of Herceptin with 5-FU observed antagonistic effects in all cancer cell lines. CONCLUSIONS: According to HER-2/neu expression level, effect of anti-cancer agents was observed differently in combination of Herceptin with chemotherapeutic agents. This suggests that HER-2/neu expression level can be applied standard of combination drug selection in combination of Herceptin With chemotherapeutic agents in gastric cancer.

17.
Asian Pac J Cancer Prev ; 14(6): 3897-901, 2013.
Article in English | MEDLINE | ID: mdl-23886204

ABSTRACT

BACKGROUND: Optimal treatment for prostate cancer remains a challenge worldwide. Recently, T cell immunoglobulin mucin-3 (TIM-3) has been implicated in tumor biology but its contribution prostate cancer remains unclear. The aim of this study was to investigate the role of TIM-3 as a prognostic marker in patients with prostate cancer. METHODS: TIM-3 protein expression was determined by immunohistochemistry and Western blotting in 137 prostate cancer tumor samples and paired adjacent benign tissue. We also performed cell proliferation assays using 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyl- 2H tetrazolium bromide (MTT) and cell invasion assays. The effects of small interfering RNA (siRNA)-mediated knockdown of TIM-3 (TIM-3 siRNA) in two human prostate cancer cell lines were also evaluated. RESULTS: TIM-3 expression was higher in prostate cancer tissue than in the adjacent benign tissue (P<0.001). High TIM-3 expression was an independent predictor of both recurrence-free survival and progression-free survival. TIM-3 protein was expressed in both prostate cancer cell lines and knockdown suppressed their proliferation and invasion capacity. CONCLUSIONS: TIM-3 expression is associated with a poor prognosis in prostate cancer. Taken together, our results indicate that TIM-3 is a potential prognostic marker in prostate cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/mortality , Lung Neoplasms/mortality , Membrane Proteins/metabolism , Prostatic Neoplasms/mortality , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Proliferation , Follow-Up Studies , Hepatitis A Virus Cellular Receptor 2 , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Membrane Proteins/genetics , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured
18.
Cell Signal ; 25(1): 74-84, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22975685

ABSTRACT

PHD finger protein 20 (PHF20) is a transcription factor, which was originally identified in glioma patients. PHF20 appears to be a novel antigen in glioma, and has also termed glioma-expressed antigen 2. PHF20 is thought to contribute to the development of cancers, including glioblastoma, lung cancer, colon cancer and ovarian cancer. However, little is known about the function of PHF20 in various cancers. Here we report that PHF20 contains two consensus sites for protein kinase B (PKB) phosphorylation (RxRxxS/T). PKB can directly phosphorylate PHF20 on Ser291 in vitro and in vivo. It has been shown that PKB participates in the tumor suppressor p53 regulated gene expression program and has a direct effect on p21 regulation after DNA damage. UV-induced DNA damage results in accumulation of p53 and PKB activation. Interestingly, PKB-mediated PHF20 phosphorylation led to an inhibition of p53 induction following UV treatment, leading to the reduction of p21 transcriptional activity. Using anti PHF20 and anti pPKB (S473) antibodies, these events were mapped in various human cancer tissues. Taken together, these data suggest that PHF20 is a novel substrate for PKB and its phosphorylation by PKB plays an important role in tumorigenesis via regulating of p53 mediated signaling.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , DNA Damage/radiation effects , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Amino Acid Sequence , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins , Down-Regulation , HCT116 Cells , HEK293 Cells , Humans , Insulin/pharmacology , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/drug effects , Serine/metabolism , Transcription Factors , Tumor Suppressor Protein p53/antagonists & inhibitors
19.
Cancer Genet ; 205(4): 147-55, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22559975

ABSTRACT

The identification of molecular markers for diagnosis, treatment, and prognosis is a significant issue in the management of patients with gastric cancer. We compared the expression profiles of 23 gastric cancers and 22 normal gastric tissues using cDNA microarrays. We divided the samples into two sets, 11 pairs as a training set and 12 unpaired gastric cancer and 11 unpaired normal gastric tissues as a test set. We selected significant genes in the training set and validated the significance of the genes in the test set. We obtained 238 classifier genes that showed a maximum cross-validation probability and clear hierarchical clustering pattern in the training set, and showed excellent class prediction probability in the independent test set. The classifier genes consisted of known genes related to the biological features of cancer and 28% unknown genes. We obtained genome-wide molecular signatures of gastric cancer, which provides preliminary exploration data for the pathophysiology of gastric cancer.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Genetic Markers , Stomach Neoplasms/genetics , Aged , Aged, 80 and over , Base Sequence , Cell Line, Tumor , DNA, Complementary , Female , Gene Expression Regulation, Neoplastic , Genetic Variation , Genome-Wide Association Study , Humans , Lymphatic Metastasis/genetics , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA , Stomach/pathology , Stomach Neoplasms/diagnosis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
20.
Asian Pac J Cancer Prev ; 12(12): 3257-63, 2011.
Article in English | MEDLINE | ID: mdl-22471463

ABSTRACT

Metastasis is the major feature of malignant tumors that causes 90% of cancer deaths. Our laboratory has already established liver metastatic clones with YCC-16, isolated from the blood of a gastric cancer patient and expanded in vitro culture using a repeated orthotopic implantation method, and had reported biologic behaviour of the parental YCC-16, the orthotopic primary S1L0, and S1L1, S2L2 and S3L3 liver metastatic clones. Here, using these cell lines, we screened from chromosomal abnormalities using karyotype analysis and micro-CGH matching. There were 31 genes screened using PCA method which were functionally related to cell adhesion. Also, there were 23 genes selected which were related to the liver specific metastasis but excluded genes related to adhesion. There were 4 genes which demonstrated reduced or increased expression stepwise with passage. In conclusion, our results should contribute to exploring the mechanisms of liver metastasis by gastric cancer.


Subject(s)
Biomarkers, Tumor/genetics , Chromosome Aberrations , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Animals , Cell Adhesion , Female , Gene Expression Profiling , Genetic Testing , Humans , Karyotyping , Mice , Mice, Inbred BALB C , Mice, Nude , Oligonucleotide Array Sequence Analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...