Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Chromatogr A ; 1727: 464990, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38744188

ABSTRACT

An approach for the controllable separation and concentration of nucleic acid using a circular nonuniform electric field was proposed and developed. Using six different lengths of DNA molecules as standard samples, the distribution of the gradient electric field was increased from the outer circular electrode to the inner rod-shaped electrode, contributing to the migration of DNA molecules at a velocity gradient towards the region with the strongest inner electric field. The DNA molecules were arranged in a distribution of concentric circles that aligned with the distribution of concentric equipotential lines. The concentration of DNA multiplied with the alternation of radius. As a result, this platform allowed simultaneous DNA separation, achieving a resolution range of 1.17-3.03 through an extended electrophoresis time, resulting in enhanced concentration factors of 1.08-6.27. Moreover, the manipulation of the relative height of the inner and outer electrodes enabled precise control over the distribution and the deflection degree of electric field lines, leading to accurate control over DNA deflection.


Subject(s)
DNA , DNA/isolation & purification , DNA/analysis , DNA/chemistry , Electrodes , Electricity , Electrophoresis, Capillary/methods
2.
Life Sci ; 327: 121698, 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37080351

ABSTRACT

Cholangiocarcinoma (CCA) with a high malignancy is usually diagnosed as advanced and is prone to metastasis and leads to a poor prognosis. It is reported that cordycepin has anti-tumor effect. However, the molecular targets and mechanisms of cordycepin in inhibiting CCA metastasis remains unclear. In order to evaluate the therapeutic effect of cordycepin on CCA metastasis, experiments were conducted in vivo and in vitro. The results showed that cordycepin inhibited the migration and EMT progression of HuCCT1 and QBC939 cells. Cordycepin has a strong hypolipidemic effects, therefore, we examined its effect on lipid metabolism in CCA. Cordycepin inhibits SREBP1 mediated fatty acid synthesis through the AKT/mTOR signaling pathway. Meanwhile, cordycepin can reduce ERO1A expression in HuCCT1 and QBC939 cells. ERO1A plays a role in malignant tumors. ERO1A promotes migration and lipid metabolism of CCA cells through AKT/mTOR signaling pathway. In addition, cordycepin significantly inhibited the tumor metastasis and the serum levels of TG and T-CHO in mice. Taken together, we demonstrate that cordycepin mediated ERO1A/mTOR/SREBP1 axis inhibits lipid metabolism and metastasis in CCA cells in vitro and in vivo. These data suggest that cordycepin can be used as a novel drug for the clinical treatment of CCA and to improve the prognosis.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Animals , Mice , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cholangiocarcinoma/metabolism , Lipid Metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Humans , Epithelial-Mesenchymal Transition
3.
J Cell Biochem ; 124(1): 3-16, 2023 01.
Article in English | MEDLINE | ID: mdl-36334309

ABSTRACT

The hallmark feature of metabolic reprogramming is now considered to be widespread in many malignancies, including colorectal cancer (CRC). Of the gastrointestinal tumors, CRC is one of the most common with a high metastasis rate and long insidious period. The incidence and mortality of CRC has increased in recent years. Metabolic reprogramming also has a significant role in the development and progression of CRC, especially lipid metabolic reprogramming. Many studies have reported that lipid metabolism reprogramming is similar to the Warburg effect with typical features affecting tumor biology including proliferation, migration, local invasion, apoptosis, and other biological behaviors of cancer cells. Therefore, studying the role of lipid metabolism in the occurrence and development of CRC will increase our understanding of its pathogenesis, invasion, metastasis, and other processes and provide new directions for the treatment of CRC. In this paper, we mainly describe the molecular mechanism of lipid metabolism reprogramming and its important role in the occurrence and development of CRC. In addition, to provide reference for subsequent research and clinical diagnosis and treatment we also review the treatments of CRC that target lipid metabolism.


Subject(s)
Colorectal Neoplasms , Lipid Metabolism , Humans , Colorectal Neoplasms/metabolism , Cell Proliferation , Cell Movement , Apoptosis
4.
Chin Med ; 17(1): 69, 2022 Jun 13.
Article in English | MEDLINE | ID: mdl-35698073

ABSTRACT

BACKGROUND: Cholangiocarcinoma (CCA) is a rare biliary adenocarcinoma related to poor clinical prognosis. Crowberry is an herbal medicine used to control inflammatory diseases and reestablish antioxidant enzyme activity. Although crowberry shows significant therapeutic efficacy in various tumors and diseases, its anticancer effects and specific molecular mechanisms in CCA are poorly understood. AIM OF THE STUDY: This study was conducted to characterize crowberry effects on CCA cells behavior. MATERIALS AND METHODS: The chemical profiles of crowberry extract was qualitatively analyzed by high-performance liquid chromatography (HPLC) and HPLC-tandem mass spectrometry. MTT, colony formation and EdU assays were performed to measure cell proliferation. The effect of crowberry treatment on CCA cell migration was assessed by wound healing and migration assays. Moreover, Hoechst staining assay and flow cytometry were performed to assess the cell apoptosis rate. Western blotting was used to assess the protein expression levels of key factors associated with apoptosis, the Akt signaling pathway, and the epithelial-mesenchymal transition. A xenograft model was established and immunohistochemical and H&E staining was performed to assess crowberry antitumor effects in vivo. RESULTS: Crowberry clearly inhibited CCA cells proliferation and migration in a dose-dependent manner and induced apoptosis in vitro. Crowberry inactivated the PI3K/Akt signaling pathway by regulating DEK in vitro and significantly inhibited tumor growth by downregulating the DEK expression in xenograft models. CONCLUSION: Crowberry inhibits CCA cells proliferation and migration through a molecular mechanism that includes inhibition of DEK and Akt signaling pathway inhibition in vitro and in vivo.

5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(6): 513-521, 2022 Jun.
Article in Chinese | MEDLINE | ID: mdl-35732609

ABSTRACT

Objective To explore the inhibitory effect of cordycepin on the proliferation and migration of gastric cancer cells and its molecular mechanism. Methods MGC-803 cells were treated with 0, 25, 50, 100 µmol/L of cordycepin and HGC-27 cells with 0, 5, 25, 50 µmol/L of cordycepin for 48 hours. The proliferation ability of MGC-803 and HGC-27 cells was detected by MTT assay and EdU assay; the colony formation ability of cells was detected by colony formation assay; both wound healing assay and cell migration assay were applied to detect the cell migration ability of MGC-803 and HGC-27 cells; the chromatin agglutination was detected by Hoechst 33342 staining; the apoptosis of gastric cancer cells was detected by annexin V-FITC/PI double labeling combined with flow cytometry; Western blot was used to measure the protein expression levels of lipid metabolism-related proteins including sterol regulatory element binding transcription factor 1 (SREBF1), fatty acid synthase (FASN), and acetyl coA carboxylase 1 (ACC1), epithelial-mesenchymal transition (EMT)-related proteins including E-cadherin, vimentin, Snail, Slug, matrix metalloproteinase 2 (MMP2), MMP9, AMPK, and phosphorylated AMPK (p-AMPK), MAPK signaling pathway-related proteins including JNK, phosphorylated JNK (p-JNK), p38 MAPK, and p-p38 MAPK, and apoptosis-related proteins including cleaved caspase-9 (c-caspase-9), c-caspase-3, and cleaved poly (ADP-ribose) polymerase (c-PARP). Results Cordycepin significantly inhibited the proliferation, colony formation, and migration of gastric cancer cells. After cordycepin treatment, the karyopycnosis, karyorrhexis, and apoptosis rate of cancer cells increased, and the expressions of apoptosis-related proteins c-caspase-3, c-caspase-9, and c-PARP increased. The expression of E-cadherin increased, while the expressions of vimentin, Snail, Slug, SREBF1, FASN, ACC1, MMP2, MMP9 significantly decreased; the phosphorylation levels of AMPK, JNK and P38 proteins significantly increased. Conclusion Cordycepin inhibits the proliferation and migration of gastric cancer cells by suppressing the lipid metabolism and EMT process via activating AMPK and MAPK signaling pathway.


Subject(s)
Deoxyadenosines , Stomach Neoplasms , AMP-Activated Protein Kinases/metabolism , Apoptosis , Cadherins/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation , Deoxyadenosines/pharmacology , Humans , Lipid Metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Vimentin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Front Pharmacol ; 12: 727275, 2021.
Article in English | MEDLINE | ID: mdl-34539408

ABSTRACT

Melanoma is aggressive and can metastasize in the early stage of tumor. It has been proved that dihydroartemisinin (DHA) positively affects the treatment of tumors and has no apparent toxic and side effects. Our previous research has shown that DHA can suppress the formation of melanoma. However, it remains poorly established how DHA impacts the invasion and metastasis of melanoma. In this study, B16F10 and A375 cell lines and metastatic tumor models will be used to investigate the effects of DHA. The present results demonstrated that DHA inhibited the proliferative capacity in A375 and B16F10 cells. As expected, the migration capacity of A375 and B16F10 cells was also reduced after DHA administration. DHA alleviated the severity and histopathological changes of melanoma in mice. DHA induced expansion of CD8+CTL in the tumor microenvironment. By contrast, DHA inhibited Treg cells infiltration into the tumor microenvironment. DHA enhanced apoptosis of melanoma by regulating FasL expression and Granzyme B secretion in CD8+CTLs. Moreover, DHA impacts STAT3-induced EMT and MMPS in tumor tissue. Furthermore, Metabolomics analysis indicated that PGD2 and EPA significantly increased after DHA administration. In conclusion, DHA inhibited the proliferation, migration and metastasis of melanoma in vitro and in vivo. These results have important implications for the potential use of DHA in the treatment of melanoma in humans.

7.
Am J Chin Med ; 49(2): 525-541, 2021.
Article in English | MEDLINE | ID: mdl-33641654

ABSTRACT

Gastric cancer is a common malignancy worldwide and is associated with high morbidity and mortality rates. However, very little is known about the underlying mechanism in human gastric cancer cells. Baicalein (BAI), a widely used Chinese herbal medicine, has shown anticancer effects on many types of human cancer cell lines. Here, we investigated the molecular mechanisms underlying BAI action on gastric cancer cell proliferation and migration. The results showed that BAI can expressively inhibit cell proliferation, colony-forming ability and migration ability in a dose-dependent manner, while in the meantime inducing cell apoptosis. Additionally, we found that BAI can suppress FAK and the phosphorylation of PI3K, AKT and mTOR in a dose-dependent manner. Furthermore, BAI significantly inhibited tumor growth in a xenograft model. Also, BAI can inhibit the proliferation and migration of gastric cancer cells and the expression of the pathway by downregulating the expression of FAK. In short, we demonstrated that BAI inhibited gastric cancer cell proliferation and migration through FAK interaction via downregulation in AKT/mTOR signaling, which signifies that BAI may be a latent therapeutic factor for the treatment of gastric cancer patients and that FAK might be a hopeful therapy target for the disease.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Flavanones/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Flavanones/chemistry , Humans , Mice , Mice, Inbred BALB C
8.
J Dermatol Sci ; 99(3): 193-202, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32859456

ABSTRACT

BACKGROUND: It has been shown that dihydroartemisinin (DHA) is effective in the treatment of malaria. Recently studies have demonstrated that DHA also regulates tumor cell growth, angiogenesis, T cell differentiation and generation. However, how DHA affects melanoma development remains poorly defined. OBJECTIVES: To investigate the effects of DHA on the proliferation and migration of melanoma in vivo and in vitro, and to explore its possible mechanism. METHODS: B16F10 cells and melanoma-bearing BALB/c mice were used to investigate the effects of DHA on melanoma. RESULTS: DHA had inhibitory effect on melanoma proliferation in a time-and dose-dependent manner. Treatment of DHA attenuated melanoma severity and histopathological changes in BALB/c mice. DHA also inhibited melanoma invasion, migration, and community formation in a dose-dependent manner. Flow cytometry revealed a significant increase in IFN-γ+CD8+ T cells in the DHA groups. In tumor microenvironment and spleen, DHA induced expansion of CD8+CTL, while, CD4+CD25+Foxp3+ regulatory T (Treg) cells and IL-10+CD4+CD25+ T cells were normalized by DHA treatment. DHA diminished expression of IL-10 and IL-6, and increased the expression of IFN-γ in the tumor and spleen. Moreover, DHA administration significantly promoted the mitochondrial apoptosis of melanoma by regulating the STAT3 pathway. CONCLUSION: DHA induces mitochondrial apoptosis and alters cytokines expression by inhibiting the phosphorylation of STAT3. DHA improves anti-tumor immunity in mice through controlling CD8+CTL function by counteracting IL-10-dependent Treg cells suppression, which promises to be an alternative drug for melanoma.


Subject(s)
Artemisinins/pharmacology , Interleukin-10/metabolism , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Tumor Escape/drug effects , Administration, Oral , Animals , Apoptosis/drug effects , Apoptosis/immunology , Artemisinins/therapeutic use , Cell Line, Tumor/transplantation , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Interleukin-10/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mitochondria/drug effects , Mitochondria/pathology , Phosphorylation/drug effects , Phosphorylation/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Escape/genetics
9.
J Pharmacol Exp Ther ; 373(2): 279-289, 2020 05.
Article in English | MEDLINE | ID: mdl-32102917

ABSTRACT

Cholangiocarcinoma (CCA) is a malignant tumor that arises from the epithelial cells of the bile duct and is notorious for its poor prognosis. The clinical outcome remains disappointing, and thus more effective therapeutic options are urgently required. Cordycepin, a traditional Chinese medicine, provides multiple pharmacological strategies in antitumors, but its mechanisms have not been fully elucidated. In this study, we reported that cordycepin inhibited the viability and proliferation capacity of CCA cells in a time- and dose-dependent manner determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and colony formation assay. Flow cytometry and Hoechst dye showed that cordycepin induced cancer cell apoptosis via extracellular signal-regulated kinase (ERK) 1/2 deactivation. Moreover, cordycepin significantly reduced the angiogenetic capabilities of CCA in vitro as examined by tube formation assay. We also discovered that cordycepin inhibited DEK expression by using Western blot assay. DEK serves as an oncogenic protein that is overexpressed in various gastrointestinal tumors. DEK silencing inhibited CCA cell viability and angiogenesis but not apoptosis induction determined by Western blot and flow cytometry. Furthermore, cordycepin significantly inhibited tumor growth and angiogenic capacities in a xenograft model by downregulating the expression of DEK, phosphorylated ERK1/2 CD31 and von Willebrand factor (vWF). Taken together, we demonstrated that cordycepin inhibited CCA cell proliferation and angiogenesis with a DEK interaction via downregulation in ERK signaling. These data indicate that cordycepin may serve as a novel agent for CCA clinical treatment and prognosis improvement. SIGNIFICANCE STATEMENT: Cordycepin provides multiple strategies in antitumors, but its mechanisms are not fully elucidated, especially on cholangiocarcinoma (CCA). We reported that cordycepin inhibited the viability of CCA cells, induced apoptosis via extracellular signal-regulated kinase 1/2 deactivation and DEK inhibition, and reduced the angiogenetic capabilities of CCA both in vivo and in vitro.


Subject(s)
Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Deoxyadenosines/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/drug effects , Neovascularization, Pathologic/prevention & control , Oncogene Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Animals , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/pathology , Humans , Male , Mice , Xenograft Model Antitumor Assays
10.
Onco Targets Ther ; 12: 6565-6576, 2019.
Article in English | MEDLINE | ID: mdl-31496746

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a major leading cause of cancer mortality worldwide. Polyadenylate (poly(A))-binding protein (PABP)-interacting protein 1 (Paip1) is a key regulator in the initiation of translation; however, its role in GC remains to be investigated. PURPOSE: The purpose of this study is to determine Paip1 expression levels and investigate its underlying molecular mechanism in GC. PATIENTS AND METHODS: In the present study, a total of 90 GC samples and 90 adjacent noncancerous tissues were used to examine the expression of Paip1. In order to gain a deep insight into the molecular mechanism of Paip1 in GC, the Paip1 siRNA sequences were transfected into GC cell lines (MGC-803 and SGC-7901), respectively. Meanwhile, Paip1 plasmid was used to mediate overexpression of Paip1. Cell proliferation were examined via colony formation assay, EdU assay and flow cytometry assay. Cell metastasis were discovered via wound healing assay and Transwell assays. In addition, key EMT makers were detected by Western blotting assay. RESULTS: In this study, Paip1 expression was observed to be upregulated in GC and was associated with shorter overall survival. Knockdown of Paip1 inhibited cell proliferation, migration and caused cell cycle arrest in GC cells, whereas its overexpression reversed these effects. Another mechanistic study showed that Paip1 overexpression promoted EMT progression and regulated its targets expression. CONCLUSION: High expression of Paip1 plays a significant role in the progression of GC and may be a potential biomarker of poor prognosis as well as a therapeutic target.

11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 35(7): 613-618, 2019 Jul.
Article in Chinese | MEDLINE | ID: mdl-31537246

ABSTRACT

Objective To investigate the effect of baicalein (BAI) on autophagy in gastric cancer cell line MGC-803. Methods MGC-803 cells were treated with 0, 5, 15, 25, 50 µmol/L BAI for 24, 48, 72 hours. The proliferation activity of MGC-803 cells was detected by MTT assay. Acridine orange (AO) staining combined with immunofluorescence cytochemical staining was performed to observe the expression of microtubule-associated protein 1 light chain 3 (LC3) and P62 to determine autophagy in MGC-803 cells. The protein levels of LC3, P62, phosphatidylinositol 3-kinase (PI3K), phosphorylated PI3K (p-PI3K), AKT, and p-AKT were detected by Western blot analysis. Results Compared with the control group, BAI significantly inhibited the proliferation of MGC-803 cells in a time- and dose-dependent manner. BAI-treated MGC-803 cells showed a significant increase in acid lysosomes and increased LC3 expression. BAI treatment significantly decreased phosphorylation of PI3K and AKT proteins, increased the ratio of LC3-II/LC3-I and up-regulated the expression of P62 protein. Conclusion Baicalein could inhibit PI3K/AKT signaling pathway and induce autophagy in MGC-803 cells.


Subject(s)
Autophagy , Flavanones/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Cell Line, Tumor , Cell Proliferation , Humans , Microtubule-Associated Proteins/metabolism , RNA-Binding Proteins/metabolism
12.
Cancer Manag Res ; 11: 6727-6739, 2019.
Article in English | MEDLINE | ID: mdl-31410063

ABSTRACT

ABSTRACT: Endoplasmic reticulum oxidoreductase 1-α (ERO1A) is a kind of hypoxia-induced endoplasmic reticulum oxidase that regulates translation and folding of oxidized proteins. This study aimed to explore the clinicopathological significance of ERO1A and the effect on the biological behavior of cholangiocarcinoma (CCA) cells. METHODS: Immunohistochemical staining was used to detect the expression of ERO1A, carcinoembryonic antigen (CEA), and carbohydrate antigen 19-9 (CA19-9) in cholangiocarcinoma. Immunofluorescence staining was performed to detect the subcellular localization of ERO1A in CCA cells. The expression of ERO1A in CAA cells after depletion or overexpression was verified by Western blot assay. Then, the effect of ERO1A on proliferation in CCA cells was verified by MTT assay and colony formation assay. Wound healing assays and migration assays were performed to detect the effect of ERO1A on cell migration ability. Finally, we explored the role of ERO1A in EMT and Akt/mTOR signaling pathway. RESULTS: In this study, our data demonstrated that ERO1A, CEA, and CA19-9 were expressed in cholangiocarcinoma tissues, and the positive rates were 95%, 95%, and 55%, respectively. The high expression of ERO1A is associated with clinical stage and pathological stage of CCA. In vitro data indicate that deletion of ERO1A can inhibit the proliferation and migration of CCA cells and vice versa. In addition, ERO1A has been shown to be closely related to EMT and Akt/mTOR pathways. CONCLUSION: In summary, we found that high expression of ERO1A is associated with poor prognosis in patients, and ERO1A can promote the proliferation and migration of CCA cells. In conclusion, ERO1A can be used as an independent biomarker for predicting the prognosis of CCA.

13.
Cancer Res Treat ; 51(4): 1653-1665, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31010277

ABSTRACT

PURPOSE: This study was aimed to investigate the role of poly(A)-binding protein-interacting protein 1 (Paip1) in cervical carcinogenesis. MATERIALS AND METHODS: The expression of Paip1 in normal cervical epithelial tissues and cervical cancer (CC) tissues were detected by immunohistochemistry. In vivo and in vitro assays were performed to validate effect of Paip1 on CC progression. RESULTS: Paip1 was found to be up-regulated in CC, which was linked with shorter survival. Knockdown of Paip1 inhibited cell growth, induced apoptosis and cell cycle arrest in CC cells, whereas its overexpression reversed these effects. The in vivo tumor model confirmed the pro-tumor role of Paip1 in CC growth. CONCLUSION: Altogether, the investigation demonstrated the clinical significance of Paip1 expression, which prompted that the up-regulated of Paip1 can presumably be a potential prognostic and progression marker for CC.


Subject(s)
Biomarkers, Tumor/metabolism , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Up-Regulation , Uterine Cervical Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Mice , Neoplasm Transplantation , Prognosis , Survival Analysis , Uterine Cervical Neoplasms/metabolism
14.
Int J Clin Exp Pathol ; 7(8): 5016-23, 2014.
Article in English | MEDLINE | ID: mdl-25197373

ABSTRACT

Oncoprotein DEK plays an important role in cancer tumorigenesis. To explore the clinical implication of DEK expression on prognostic evaluation in small cell lung cancer (SCLC), 130 cases of SCLC with strict follow-up were selected for immunohistochemical (IHC) staining of DEK protein. The correlation between DEK expression and clinicopathological features of SCLC was evaluated using the Chi-square and Fisher's exact tests, survival rates were calculated using the Kaplan-Meier method and univariate and multivariate analyses were performed using the Cox proportional hazards regression model. IHC analysis demonstrated that DEK protein staining was strongly positive and significantly higher (44.62%) in SCLC compared with either adjacent non-tumor or normal lung tissues (P < 0.001 for both). DEK expression correlated with large tumor size (P = 0.025) and late pathologic stage (P = 0.005). Moreover, it correlated with low disease-free (P = 0.004) and 5-year (P = 0.005) survival rates. In the late-stage group, disease-free and 5-year survival rates of patients with high level DEK expression were significantly lower than those with low level DEK expression (P = 0.006 and P = 0.001, respectively). Furthermore, Cox analysis revealed that DEK expression emerged as a significant independent hazard factor for the overall survival rate of patients with SCLC (HR: 1.594, 95% CI: 1.087-2.336, P = 0.017). In conclusion, DEK plays an important role in the progression of SCLC. DEK may potentially be used as an independent biomarker for the prognostic evaluation of SCLC.


Subject(s)
Chromosomal Proteins, Non-Histone/biosynthesis , Lung Neoplasms/pathology , Oncogene Proteins/biosynthesis , Small Cell Lung Carcinoma/pathology , Adult , Aged , Biomarkers, Tumor/analysis , Disease Progression , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Poly-ADP-Ribose Binding Proteins , Prognosis , Proportional Hazards Models , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/mortality
15.
BMC Cancer ; 14: 414, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24912939

ABSTRACT

BACKGROUND: NQO1 (NAD(P)H: quinone oxidoreductase-1), located on chromosome 16q22, functions primarily to protect normal cells from oxidant stress and electrophilic attack. Recent studies have revealed that NQO1 is expressed at a high level in most human solid tumors including those of the colon, breast, pancreas, ovaries and thyroid, and it has also been detected following the induction of cell cycle progression and proliferation of melanoma cells. In this study, we aimed to investigate the clinicopathological significance of upregulated NQO1 protein expression in squamous cell carcinomas (SCCs) of the uterine cervix. METHODS: The localization of the NQO1 protein was determined in the SiHa cervical squamous cancer cell line using immunofluorescence (IF) staining, and immunohistochemical (IHC) staining performed on paraffin-embedded cervical SCC specimens from 177 patients. For comparison, 94 cervical intraepithelial neoplasia (CIN) and 25 normal cervical epithelia samples were also included. QRT-PCR was performed on RNA from fresh tissues to detect NQO1 mRNA expression levels, and HPV infection status was genotyped using oligonucleotide microarray. Disease-free survival (DFS) and 5-year overall survival (OS) rates for all cervical SCC patients were calculated using the Kaplan-Meier method, and univariate and multivariate analysis was performed using the Cox proportional hazards regression model. RESULTS: The NQO1 protein showed a mainly cytoplasmic staining pattern in cervical cancer cells, and only three cases of cervical SCC showed a nuclear staining pattern. The strongly positive rate of NQO1 protein expression was significantly higher in cervical SCCs and CINs than in normal cervical epithelia. High-level NQO1 expression was closely associated with poor differentiation, late-stage, lymph node metastasis and high-risk for HPV infection. Additionally, high-level NQO1 expression was associated with lower DFS and 5-year OS rates, particularly for patients with early-stage cervical SCCs. Furthermore, Cox analysis revealed that NQO1 expression emerged as a significant independent hazard factor for DFS rate in patients with cervical SCC. CONCLUSIONS: NQO1 overexpression might be an independent biomarker for prognostic evaluation of cervical SCCs.


Subject(s)
Carcinoma, Squamous Cell/genetics , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Uterine Cervical Neoplasms/genetics , Adult , Aged , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Middle Aged , NAD(P)H Dehydrogenase (Quinone)/genetics , Papillomaviridae/pathogenicity , Prognosis , Proportional Hazards Models , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
16.
Hepatology ; 56(1): 157-64, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22290731

ABSTRACT

UNLABELLED: IgG4 reactions consisting of marked infiltration by immunoglobulin G4 (IgG4)-positive plasma cells in affected organs is found in cancer patients as well as patients with IgG4-related diseases. Notably, extrahepatic cholangiocarcinomas accompanying marked IgG4 reactions clinicopathologically mimic IgG4-related sclerosing cholangitis. The regulatory cytokine interleukin (IL)-10 is thought to induce the differentiation of IgG4-positive cells. In this study, to clarify the mechanism of the IgG4 reaction in extrahepatic cholangiocarcinoma, we investigated nonprofessional antigen-presenting cells (APCs) generating IL-10-producing regulatory T cells (anergy T cells) and Foxp3-positive regulatory cells producing IL-10. Immunohistochemistry targeting IgG4, HLA-DR, CD80, CD86, and Foxp3 was performed using 54 cholangiocarcinoma specimens from 24 patients with gallbladder cancer, 22 patients with common bile duct cancer, and eight patients with cancer of the Papilla of Vater. Moreover, a molecular analysis of Foxp3 and IL-10 was performed using a cultured human cholangiocarcinoma cell line. Consequently, 43% of the cholangiocarcinomas were found to be abundant in IgG4. Those expressing HLA-DR but lacking costimulatory molecules (CD80 and CD86) and those expressing Foxp3 detected by an antibody recognizing the N terminus accounted for 54% and 39% of cases, respectively. Moreover, the number of IgG4-positive cells was larger in these cases than in other groups. In cultured cells, the presence of a splicing variant of Foxp3 messenger RNA and the expression of IL-10 were demonstrated. CONCLUSION: Extrahepatic cholangiocarcinoma is often accompanied by significant infiltration of IgG4-positive cells. Cholangiocarcinoma cells could play the role of nonprofessional APCs and Foxp3-positive regulatory cells, inducing IgG4 reactions via the production of IL-10 indirectly and directly, respectively.


Subject(s)
Bile Duct Neoplasms/immunology , Cholangiocarcinoma/immunology , Gallbladder Neoplasms/immunology , Immunoglobulin G/metabolism , Aged , Ampulla of Vater/pathology , Bile Duct Neoplasms/pathology , Bile Ducts, Extrahepatic/pathology , Biomarkers/metabolism , Cholangiocarcinoma/pathology , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Gallbladder Neoplasms/pathology , Humans , Immunohistochemistry , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Middle Aged , Molecular Biology , Plasma Cells/immunology , Plasma Cells/metabolism , Polymerase Chain Reaction/methods , Registries , Sensitivity and Specificity , Tissue Embedding
17.
World J Hepatol ; 1(1): 35-42, 2009 Oct 31.
Article in English | MEDLINE | ID: mdl-21160963

ABSTRACT

Flat-type "biliary intraepithelial neoplasia (BilIN)" and papillary-type "intraductal papillary neoplasm of the bile duct (IPN-B)" are proposed as precursors of invasive, perihilar intrahepatic cholangiocarcinoma (ICC). Three carcinogenetic pathways are proposed: BilIN progressing to tubular adenocarcinoma, and IPN-B progressing to tubular adenocarcinoma or to colloid carcinoma. Carcinogenesis via BilIN was characterized by mucin core protein 2-/cytokeratin 20-(MUC2-/CK20-) with MUC1 expression, while carcinogenesis via IPN-B leading to tubular adenocarcinoma was associated with MUC1 expression or that to colloid carcinoma with MUC1-negativity. In both the BilIN and IPNB series, the expression of p21, p53, and cyclin D1 was upregulated with histological progression. Interestingly, p53 expression was upregulated at the invasive stage of BilIN, but was low in noninvasive BilIN, while p53 expression was upregulated in IPN-B1 and reached a plateau in IPN-B2 and invasive ICC. Expression of p16(INK4a), which was frequent in BilIN1, was decreased in BilIN-2/3 and invasive carcinoma. EZH2 expression showed a stepwise increase from BilIN to invasive carcinoma. Membranous expression of ß-catenin and E-cadherin was more markedly decreased in ICC with BilIN than in ICC with IPNB. Interestingly, disruption of the membranous distribution of ß-catenin and E-cadherin seems to result in the invasion and metastasis of carcinoma cells of BilIN and IPN-B expressing MMP-7 and MT1-MMP. Increased expression of cyclin D1 and c-myc was more frequent in the IPNB lineage than BilIN lineage, possibly related to the Wnt signaling pathway associated with the nuclear accumulation of ß-catenin. In conclusion, BilIN and IPN-B progress to invasive ICC through characteristic multistep processes.

SELECTION OF CITATIONS
SEARCH DETAIL
...