Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Int J Surg Pathol ; 28(7): 711-720, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32362207

ABSTRACT

Background. The expression of terminal deoxynucleotidyl transferase (TdT) in myoepithelial cells (MECs) within the breast was recently incidentally observed in our routine practice. This study aimed to elucidate the expression of TdT in MECs. Methods. TdT immunostaining was performed on 180 mammary, 89 cutaneous, and 94 salivary tissues or lesions. Other myoepithelial markers, including P63, calponin, and SMA as well as double staining for TdT and calponin, were also evaluated in some cases. Selected lesions with basal or myoid differentiation were also included in the investigation. Results. MECs were positive for TdT in mammary lesions that contained MECs (132/135) but negative when they did not contain MECs (45/45). MECs in sweat glands (24/30) and their neoplastic counterparts, including those in hidradenoma papilliferum (2/9), spiradenoma (6/6), and cutaneous mixed tumor (9/9), showed weak to moderate TdT positivity. MECs were variably immunolabeled for TdT in salivary or salivary gland-type tumors with myoepithelial differentiation (pleomorphic adenoma, 24/25; basal cell adenoma, 6/7; adenoid cystic carcinoma, 7/7; Warthin tumor, 0/6; mucoepidermoid carcinoma, 0/8; acinic cell carcinoma, 0/4), but MECs in normal salivary gland barely stained for TdT (30/32). Conclusions. Our findings indicate that TdT may be eligible as an additional auxiliary immunohistochemical marker as P63, but not a surrogate, to identify the MECs in the breast with limited cross-reactivity, particularly in lesions with a prominent proportion of MECs. Positivity for TdT, along with other relevant markers, in a subset of sweat gland lesions and salivary tumors may contribute to their diagnosis.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/diagnosis , DNA Nucleotidylexotransferase/biosynthesis , Salivary Gland Neoplasms/diagnosis , Sweat Gland Neoplasms/diagnosis , Breast/metabolism , DNA Nucleotidylexotransferase/analysis , Female , Humans , Immunohistochemistry , Male , Salivary Glands/metabolism , Sweat Glands/metabolism
2.
Cancer Manag Res ; 11: 6079-6090, 2019.
Article in English | MEDLINE | ID: mdl-31308746

ABSTRACT

BACKGROUND: Taurine-upregulated gene 1 (TUG1) has been documented to be implicated in carcinogenesis and chemoresistance in solid tumors. Here, we explored the biological role and regulatory mechanism of TUG1 in progression and chemoresistance of urothelial carcinoma of the bladder (UCB). METHODS: Nuclear factor-erythroid 2 (NF-E2)-related factor 2 (Nrf2) mRNA and TUG1 expression was determined by quantitative reverse transcription polymerase chain reaction. Western blot was performed to determine the protein levels of Nrf2, p-glycoprotein (p-gp), Ki-67 (Ki67), matrix metalloproteinase (MMP)-2 and MMP-9 and cleaved caspase-3. The effects of either Nrf2 or TUG1 knockdown on the proliferation, invasion, apoptosis and adriamycin (ADM) resistance of UCB cells were evaluated by CCK-8 assay, transwell invasion assay and flow cytometry analysis. Xenograft tumor assay was carried out to confirm the role of Nrf2 and TUG1 in ADM resistance of UCB cells in vivo. RESULTS: Nrf2 and TUG1 were upregulated in UCB tissues and cell lines. A positive correlation between Nrf2 and TUG1 expression was discovered in UCB tissues. Moreover, Nrf2 and TUG1 expression levels were higher in ADM-resistant cells compared with those in parental cells. Furthermore, Nrf2 positively regulated the expression of TUG1 in UCB cells. Knockdown of either Nrf2 or TUG1 led to the inhibition of cell proliferation and invasion and promotion of cell apoptosis, accompanying with down-regulation of Ki67, MMP-2 and MMP-9 and up-regulation of cleaved caspase-3. Knockdown of either Nrf2 or TUG1 enhanced the sensitivity of BIU-87/ADM and T24/ADM cells to ADM, as indicated by decreased expression of p-gp. Besides, knockdown of either Nrf2 or TUG1 inhibited tumor growth in the absence or presence of ADM in vivo. CONCLUSIONS: Nrf2 induces the up-regulation of TUG1 to promote progression and ADM resistance in UCB.

3.
J Cell Biochem ; 120(4): 6789-6797, 2019 04.
Article in English | MEDLINE | ID: mdl-30556250

ABSTRACT

Long noncoding RNA (lncRNA) maternally expressed 3 (MEG3) has been implicated as a tumor suppressor gene in several human cancer types. However, little is known regarding its involvement and potential mechanism in human breast cancer. In this study, we explored the effect of MEG3 on the growth of human breast cancer cell line MDA-MB-231 in vitro and in vivo, and sought to elucidate the potential signaling mechanisms. Ectopic overexpression of MEG3 using a lentiviral vector Lv-MEG3 significantly inhibited breast cancer cell growth in vitro and a cancer xenograft growth in vivo. MEG3 overexpression led to marked increase of apoptosis in breast cancer cells as determined using flow cytometry and fragmented DNA labeling. Moreover, ectopic expression of MEG3 increased the expression of endoplasmic reticulum (ER) stress-related proteins required for unfolded protein response, including glucose-regulated protein 78 (GRP78), inositol-requiring enzyme 1 (IRE1), protein kinase RNA (PKR)-like ER kinase (PERK), and activated transcription factor 6 (ATF6), as well as proapoptotic proteins CCAAT/enhancer binding protein homologous protein (CHOP) and caspase-3. Finally, MEG3 overexpression markedly increased nuclear factor κB (NF-κB) expression, NF-κB translocation to the nucleus, and p53 expression, whereas pharmacological inhibition of NF-κB completely abolished MEG3-induced activation of p53. Together, these results suggest that MEG3 inhibits breast cancer growth and induces breast cancer apoptosis, partially via the activation of the ER stress, NF-κB and p53 pathways, and that NF-κB signaling is required for MEG3-induced p53 activation in breast cancer cells. Our results indicate targeting lncRNA MEG3 may represent a novel strategy for breast cancer therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Endoplasmic Reticulum Stress , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Endoplasmic Reticulum Chaperone BiP , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/genetics , Neoplasm Invasiveness , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Unfolded Protein Response , Xenograft Model Antitumor Assays
4.
Oncotarget ; 8(51): 89142-89148, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29179506

ABSTRACT

The Checkpoint with Forkhead-associated and Ring finger domains (CHFR) is a mitotic checkpoint and tumor-suppressor gene, its loss contributes tumorigenesis of epithelial cancers including colorectal carcinoma (CRC). The diagnostic and prognostic value of CHFR promoter hypermethylation in CRC remains unclear. This study aimed to conduct a meta-analysis and literature review and investigate clinicopathological significance of CHFR promoter hypermethylation in CRC. The following online database were used: PubMed, EMBASE, and Web of Science up to March 2017. Odds Ratios (OR) and Hazard Ratios (HR) with 95% corresponding confidence intervals (CIs) were calculated. A total of seven relevant articles were available for meta-analysis which included 966 patients. The frequency of CHFR promoter hypermethylation significantly increased in CRC compared to normal colorectal mucosa tissue, pooled OR was 8.35, p < 0.00001. CHFR promoter hypermethylation was not significantly correlated to stage, OR was 1.16, p = 0.63. However, CHFR promoter hypermethylation was more frequently observed in CRC with positive lymph nodes metastasis than CRC with negative lymph nodes metastasis, OR was 0.46, p = 0.03. Additionally CHFR promoter hypermethylation was significantly related to poor overall survival in patients with CRC, HR was 0.62, p = 0.008. Based on these results, tumor CHFR promoter hypermethylation is not only a diagnostic biomarker for CRC, but also a prognostic marker. CHFR promoter hypermethylation is significantly associated with worse overall survival in patients with CRC. Our data suggested that CHFR could be a potential drug target for development of demethylation treatment for patients with CRC.

5.
Biomed Pharmacother ; 85: 303-312, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27899257

ABSTRACT

Breast cancer is one of the most common primary malignant tumors of among women, the long-term survival of which has stagnated in the past decades. Juglanin as a natural production mainly extracted from green walnut husks of Juglans mandshurica has been defined as the functional composition among a series of compounds. It showed powerful protective effect in various diseases by inhibiting inflammation and tumor cells growth. However, the effect of juglanin on human breast cancer and the underlying mechanisms remains to be elucidated. We reported here that juglanin could inhibit cell proliferation by leading to G2/M phase arrest. Exposure to juglanin resulted in the activation of cleaved caspase -3, -8, and -9, indicating that juglanin induced apoptosis. Autophagy occurred in juglanin-treated cells as evidenced by formation of autophagosome and up-regulation of LC3B-II. The juglanin-induced cell death was significantly restored by the combination of autophagy and apoptosis. Further, juglanin also induced JNK activation and ROS production. The JNK inhibitor attenuated juglanin-caused apoptosis and autophagy significantly while ROS scavenger could reverse them. In addition, the ROS scavenger also inhibited G2/M phase arrest and phosphorylated JNK. Of note, we found that juglanin had the similar effects on breast cancer cells. Finally, juglanin inhibited tumor growth in the mouse xenograft model in vivo. Together, our results suggested that juglanin led to G2/M phase arrest, induced apoptosis as well as autophagy through the ROS/JNK signaling pathway in human breast cancer cells. Hence, juglanin might be a promising candidate for development of anti-tumor drugs targeting breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Breast Neoplasms/drug therapy , Glycosides/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Kaempferols/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Caspases/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MCF-7 Cells , Male , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Med Sci Monit ; 22: 4516-4522, 2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27876711

ABSTRACT

BACKGROUND Previous studies indicated that calreticulin (CRT) regulated various biological processes. This study was aimed to investigate the function of CRT in Schwann cells (SCs). MATERIAL AND METHODS SCs were separated from sciatic nerves of mice and were transfected with pcDNA3.1-CRT (pc-CRT), small interfering RNA targets CRT (siCRT), or their corresponding negative controls. The expression of CRT was determined by quantitative reverse transcription PCR (qRT-PCR) and Western blot analysis. Then, cell proliferation, migration, and apoptosis were measured by 3-(4, 5-dimethylhiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay, modified 2-chamber migration assay, and flow cytometry, respectively. Finally, the phosphorylation levels of key kinases in the phosphatidylinositol-3-kinase (PI3K)/AKT and the extracellular signal-regulated kinase/ribosomal S6 kinase 2 (ERK/S6) pathways were detected by Western blot analysis. RESULTS Overexpression of CRT remarkably increased viability (P<0.05, P<0.01 or P<0.001) and migration (P<0.001), but inhibited apoptosis (P<0.05). The CRT-knockdown showed the inverse impacts on viability (P<0.05 or P<0.001), migration (P<0.001), and apoptosis (P<0.001). Additionally, the phosphorylation levels of AKT (Thr308 and Ser473), ERK, and S6 were all up-regulated in CRT-overexpressed cells (P<0.001), and were down-regulated in CRT-knockdown cells (P<0.05, P<0.01 or P<0.001). CONCLUSIONS Overexpression of CRT in SCs promoted cell proliferation and migration but suppressed cell apoptosis. The PI3K/AKT and ERK/S6 pathways might be involved in the functional effects of CRT on SCs.


Subject(s)
Calreticulin/biosynthesis , Schwann Cells/metabolism , Schwann Cells/physiology , Animals , Apoptosis/physiology , Calreticulin/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/drug effects , MAP Kinase Signaling System , Mice , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Schwann Cells/cytology , Sciatic Nerve/cytology , Transfection
7.
Int J Clin Exp Pathol ; 8(6): 6031-6, 2015.
Article in English | MEDLINE | ID: mdl-26261480

ABSTRACT

To assess the clinicopathological, immunohistochemical and molecular features of metanephric adenoma (MA). Clinicopathologic data were obtained for 5 cases of MA with follow-up information. Specimens from these patients were stained by HE and immunohistochemistry for the detection of WT1, vimentin, S-100 protein, CK7, P504s, CD10 and renal cell carcinoma marker (RCC). Fluorescence in situ hybridization (FISH) was performed on 4 tumors. The patients included 1 male and 4 females, aged from 30 to 49 (mean=39) years. Tumor diameters ranged from 3 to 5.5 cm. Histologically, the tumors had tubular, papillary, or glomeruloid architectures, and were composed of cells with uniform and round nuclei, inconspicuous nucleoli, and high ratio of nucleus to cytoplasm. Nuclear polymorphism and mitotic figures were not observed. Immunohistochemically, they expressed WT1 (5/5), vimentin (5/5), S-100 (4/5), CK7 (2/5), P504s (2/5), and CD10 (1/5) and not RCC. FISH study was carried out on 4 metanephric adenoma cases, and no abnormalities were observed in chromosomes 3, 7, 17, and P16 gene of chromosomes 9. MA is an uncommon renal tumor. Its diagnosis depends on morphological, immunohistochemical and molecular features.


Subject(s)
Adenoma/chemistry , Biomarkers, Tumor/analysis , Immunohistochemistry , Kidney Neoplasms/chemistry , Adenoma/genetics , Adenoma/pathology , Adenoma/surgery , Adult , Biomarkers, Tumor/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Male , Middle Aged , Nephrectomy , Predictive Value of Tests , Tomography, X-Ray Computed , Tumor Burden
9.
Asian Pac J Cancer Prev ; 15(4): 1511-5, 2014.
Article in English | MEDLINE | ID: mdl-24641359

ABSTRACT

BACKGROUND AND AIMS: To explore the molecular mechanisms of miR-886-5p in breast cancer., we examined roles in inhibiting growth and migration of MCF-7 cells. METHODS: MiR-886-5p mimics and inhibitors were used to express or inhibit MiR-886-5p, respectively, and MTT and clone formation assays were used to determine the survival and proliferation. Hoechst 33342/ PI double staining was applied to detect apoptosis. The expression of caspase-3, caspase-8, caspase-9, MT1-MMP, VEGF-C and VEGF-D was detected by Western blotting, and the levels of MMP2 and MMP9 secreted from MCF-7 cells were assessed by ELISA. MCF-7 cell migration was determined by wound healing and Transwell assays. RESULTS: We found that the growth of MCF-7 cells was inhibited upon decreasing miR-886-5p levels. Inhibiting miR-866-5p also significantly induced apoptosis and decreased the migratory capacity of these cells. The expression of VEGF-C, VEGF-D, MT1-MMP, MMP2, and MMP9 was also found to be decreased as compared to controls. CONCLUSIONS: Our data show that downregulation of miR-886-5p expression in MCF-7 cells could significantly inhibit cell growth and migration. This might imply that inhibiting miR-886-5p could be a therapeutic strategy in breast cancer.


Subject(s)
Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation , MicroRNAs/antagonists & inhibitors , Breast Neoplasms/drug therapy , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Caspase 9/biosynthesis , Cell Line, Tumor , Cell Survival/genetics , Female , Humans , MCF-7 Cells , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , MicroRNAs/genetics , Vascular Endothelial Growth Factor C/biosynthesis , Vascular Endothelial Growth Factor D/biosynthesis
10.
Zhonghua Nan Ke Xue ; 18(2): 164-7, 2012 Feb.
Article in Chinese | MEDLINE | ID: mdl-22568216

ABSTRACT

OBJECTIVE: To study the clinicopathological characteristics of prostatic cystadenoma (PC). METHODS: A sample from surgically removed tissues of a PC patient was examined by conventional pathology and immunohistochemistry. The clinical data and clinicopathological features were analyzed, and the related literature reviewed. RESULTS: The patient was a male aged 55 years, treated by TUVP for dysuria a year before. The tumor was a grey mass, with lots of different sized capsular spaces full of clear white liquid in the cross section. Histologically, the tumor cells were arranged in a sieve-like, microcapsule-shaped or adenoid pattern, lined with cuboidal and columnar epithelial cells, the nuclei located in the base with neither cellular atypia nor mitosis. Concerning the immunophenotype, PSA, PAP and CK7 were positively expressed in the columnar epithelial cells and 34betaE12 in the basal cells, while CK20, P504S, CEA and villin were negatively expressed, with Ki67 + < 2%. CONCLUSION: Prostatic cystadenoma is a rare benign tumor originating in the prostate, with a unique morphological structure, and mostly with the expressions of PSA and PAP.


Subject(s)
Cystadenoma , Prostatic Neoplasms , Cystadenoma/pathology , Humans , Male , Middle Aged , Prostatic Neoplasms/pathology
12.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 39(2): 214-7, 2008 Mar.
Article in Chinese | MEDLINE | ID: mdl-18630686

ABSTRACT

OBJECTIVE: To observe the effects of Ginsenoside Rb1 (GRb1) on neuronal cell apoptosis and the expressions of Bcl-2 and Bax in rats after cerebral ischemia-reperfusion so as to investigate the neuroprotective mechanism of GRb1. METHDOS: The model of cerebral ischemia-reperfusion was established by occluding rat middle cerebral artery for 2 h. The rats were randomly divided into two groups: ischemia-reperfusion group (I/R group) and GRb1 treat group (GRb1 group). GRb1 (40 mg/kg, i.p.) was administered immediately to rats after the onset of reperfusion. Two groups were further subdivided 7 subgroups according to various reperfusion time (3 h, 12 h, 1 d, 2 d, 3 d, 5 d and 10 d, n=4 per time point). HE staining was used to observe histological features. TUNEL and immunohistochemical method were used to analyze the cell apoptosis and expressions of Bcl-2 and Bax, respectively. RESULTS: Compared with I/R group, GRb1 reduced pathological changes, and decreased the number of apoptotic neural cells (P<0.05 on 12 h, 1 d, 2 d and 3 d) and up-regulated the number of Bcl-2 positive cells (P<0.05 on 12 h, 1 d, 3 d, 5 d and 10 d), and meanwhile down-regulated the number of Bax positive cells (P<0.05 on 3 h, 12 h, 1 d, 2 d, 3 d, 5 d and 10 d) in the ipsilateral hemisphere. CONCLUSION: The neuroprotective effect of GRb1 on cerebral ischemia-reperfusion injury is related to inhibit neuronal apoptosis and to up-regulate the expression of Bcl-2 with down-regulating the expression of Bax.


Subject(s)
Apoptosis/drug effects , Ginsenosides/therapeutic use , Neurons/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Reperfusion Injury/prevention & control , bcl-2-Associated X Protein/biosynthesis , Animals , Brain Ischemia/complications , Female , Ginsenosides/pharmacology , Immunohistochemistry , In Situ Nick-End Labeling , Male , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar , Reperfusion Injury/etiology , Reperfusion Injury/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL