Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 15: 972308, 2022.
Article in English | MEDLINE | ID: mdl-36483569

ABSTRACT

Introduction: Transferrin receptor protein 1 (TFRC), an ananda molecule associated with ferroptosis, has been identified as affecting a wide spectrum of pathological processes in various cancers, but the prognostic value correlates with the tumor microenvironment of TFRC in lower-grade glioma (LGG) is still unclear. Materials and methods: Clinical pathological information and gene expression data of patients with LGG come from The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), GTEx, Oncomine, UCSC Xena, and GEO databases. We then used various bioinformatics methods and mathematical models to analyze those data, aiming to investigate the clinical significance of TFRC in LGG and illustrate its association with tumor immunity. In addition, the molecular function and mechanisms of TFRC were revealed by gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA). Immunohistochemical experiments and single-cell analysis have been performed. Results: TFRC expression was highly expressed in many tumors and showed a poor prognosis. Including gliomas, it was significantly associated with several poor clinical prognostic variables, tumor immune microenvironment, tumor mutational burden (TMB), m6a modification, and ferroptosis in LGG. TFRC as a key factor was further used to build a prediction nomogram. The C-index, calibration curve, and decision curve analysis showed the nomogram was clinically useful and calibration was accurate. At the same time, we also demonstrated that promoter hypomethylation of DNA upstream of TFRC could lead to high TFRC expression and poor overall survival. There is a significant correlation between TFRC and CD8 + T cell, macrophage cell infiltration, and several immune checkpoints, such as PD-L1(cd274), CTLA4, and PD1, suggesting a novel direction for future clinical application. Functional and molecular mechanism analysis showed an association of TFRC expression with immune-related pathways through GSEA, GO, and KEGG analysis. Finally, immunohistochemical experiments and single-cell analysis confirmed the expression of TFRC in glioma. Conclusion: TFRC may be a potential prognostic biomarker and an immunotherapeutic target for glioma.

2.
Med Sci Monit ; 24: 8899-8909, 2018 Dec 09.
Article in English | MEDLINE | ID: mdl-30531691

ABSTRACT

BACKGROUND Lin28 is a gene involved in many biological processes, including development, glucose metabolism, and tumorigenesis. Let-7 miRNA is a tumor-suppressor gene that is frequently inactivated in cancer cells. The role of c-Myc (a target gene of let-7) and the Lin28-let-7-c-Myc pathway in the growth and malignancy of thyroid cancer is unclear. The purpose of the present study was to evaluate the expression of Lin28A, let-7a, and c-Myc in human papillary thyroid carcinoma (PTC) and to investigate their potential mechanisms in the progression of PTC. MATERIAL AND METHODS Lin28A and c-Myc expression were assessed in PTC tissues and PTC cell lines using immunohistochemistry, Western blotting, and real-time PCR. CCK-8 and Transwell assays were performed to evaluate PTC cell proliferation, migration, and invasion in cells in which the expression of Lin28A was downregulated by RNA interference or in which let-7a was overexpressed after transfection with let-7a mimics. RESULTS The expression of Lin28A and c-Myc was upregulated in PTC tissues and cell lines, whereas the expression of let-7a was downregulated in PTC cell lines. Clinically, Lin28A was linked to a higher tumor/node/metastasis stage and the presence of lymph node metastases. Moreover, knockdown of Lin28A activated let-7a processing and inhibited the expression of the downstream gene c-Myc, suppressing cell proliferation, migration, and invasion. Similar results were obtained after let-7a overexpression. CONCLUSIONS The Lin28A/let-7a/c-Myc pathway is involved in cancer growth and malignant behavior in PTC and is a potential target for therapeutic intervention in this disease.


Subject(s)
MicroRNAs/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/metabolism , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/genetics , Middle Aged , RNA Interference , RNA-Binding Proteins/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
3.
Cell Physiol Biochem ; 40(3-4): 538-548, 2016.
Article in English | MEDLINE | ID: mdl-27889764

ABSTRACT

BACKGROUND/AIMS: Hyperuricemia is part of the metabolic-syndrome cluster of abdominal obesity, impaired glucose tolerance, insulin resistance, dyslipidemia, and hypertension. Monocytes/macrophages are critical in the development of metabolic syndrome, including gout, obesity and atherosclerosis. However, how high uric acid (HUA) exposure affects monocyte/macrophage function remains unclear. In this study, we investigated the molecular mechanism of HUA exposure in monocytes/macrophages and its impact on oxidized low-density lipoprotein (oxLDL)-induced foam-cell formation in a human monocytic cell line, THP-1. METHODS: We primed THP-1 cells with phorbol-12-myristate-13-acetate (PMA) for differentiation, then exposed cells to HUA and detected the production of reactive oxygen species (ROS) and analyzed the level of phospho-AMPKα. THP-1 cells were pre-incubated with Compound C, an AMPK inhibitor, or N-acetyl-L-cysteine (NAC), a ROS scavenger, or HUA before PMA, to assess CD68 expression and phospho-AMPKα level. PMA-primed THP-1 cells were pre-treated with oxLDL before Compound C and HUA treatment. Western blot analysis was used to examine the levels of phospho-AMPKα, CD68, ABCG1, ABCA1, cyclooxygenase-2 (COX-2) and NF-κB (p65). Flow cytometry was used to assess ROS production and CD68 expression in live cells. Oil-red O staining was used to observe oxLDL uptake in cells. RESULTS: HUA treatment increased ROS production in PMA-primed THP-1 cells; NAC blocked HUA-induced oxidative stress. HUA treatment time-dependently increased phospho-AMPKα level in PMA-primed THP-1 cells. The HUA-induced oxidative stress increased phospho-AMPKα levels, which was blocked by NAC. HUA treatment impaired CD68 expression during cell differentiation by activating the AMPK pathway, which was reversed by Compound C treatment. Finally, HUA treatment inhibited oxLDL uptake in the formation of foam cells in THP-1 cells, which was blocked by Compound C treatment. HUA treatment significantly increased the expression of ABCG1 and reversed the oxLDL-reduced ABCG1 expression but did not affect the expression of ABCA1, NF-κB (p65) or COX-2. CONCLUSIONS: HUA exposure activated the ROS-AMPK pathway, impaired CD68 expression, and inhibited oxLDL-induced foam-cell formation in a human monocytic cell line, THP-1.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Foam Cells/cytology , Lipoproteins, LDL/pharmacology , Monocytes/cytology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Uric Acid/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Acetylcysteine/pharmacology , Cell Differentiation/drug effects , Cell Line , Cyclooxygenase 2/metabolism , Foam Cells/drug effects , Foam Cells/metabolism , Humans , Models, Biological , Oxidative Stress/drug effects , Phosphorylation/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transcription Factor RelA/metabolism
4.
Cancer Sci ; 107(12): 1806-1817, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27636742

ABSTRACT

Metformin is an oral biguanide commonly used for treating type II diabetes and has recently been reported to possess antiproliferative properties that can be exploited for the prevention and treatment of a variety of cancers. The mechanisms underlying this effect have not been fully elucidated. Our study shows a marked loss of AMP-activated protein kinase (AMPK) phosphorylation and nuclear human Forkhead box O1 (FOXO1) protein in estrogen-dependent endometrial cancer (EC) tumors compared to normal control endometrium. Metformin treatment suppressed EC cell growth in a time-dependent manner in vitro; this effect was cancelled by cotreatment with an AMPK inhibitor, compound C. Metformin decreased FOXO1 phosphorylation and increased FOXO1 nuclear localization in Ishikawa and HEC-1B cells, with non-significant increase in FOXO1 mRNA expression. Moreover, compound C blocked the metformin-induced changes of FOXO1 and its phosphorylation protein, suggesting that metformin upregulated FOXO1 activity by AMPK activation. Similar results were obtained after treatment with insulin. In addition, transfection with siRNA for FOXO1 cancelled metformin-inhibited cell growth, indicating that FOXO1 mediated metformin to inhibit EC cell proliferation. A xenograft mouse model further revealed that metformin suppressed HEC-1B tumor growth, accompanied by downregulated ki-67 and upregulated AMPK phosphorylation and nuclear FOXO1 protein. Taken together, these data provide a novel mechanism of antineoplastic effect for metformin through the regulation of FOXO1, and suggest that the AMPK-FOXO1 pathway may be a therapeutic target to the development of new antineoplastic drugs.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Estrogens/metabolism , Forkhead Box Protein O1/metabolism , Metformin/pharmacology , Signal Transduction/drug effects , Adult , Aged , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Enzyme Activation , Female , Humans , Mice , Middle Aged , Phosphorylation , Xenograft Model Antitumor Assays
5.
BMC Med Imaging ; 15: 23, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26179753

ABSTRACT

BACKGROUND: Primary hyperparathyroidism is an endocrinopathic condition characterized by hypersecretion of parathyroid hormone. Excess parathyroid hormone results in an altered state of osseous metabolism involving bone resorption and tissue change known as osteitis fibrosa cystica, which is the end stage of primary hyperparathyroidism. Osteitis fibrosa cystica is associated with the development of brown tumors, which are rare because hyperparathyroidism is now usually diagnosed and treated before symptoms develop. Brown tumors are rarely the first symptom of hyperparathyroidism and can occasionally be mistaken for malignancy. CASE PRESENTATION: We herein report three cases of primary hyperparathyroidism with an unusual presentation of brown tumors. All three patients were Asian. In the first case, a 42-year-old man was admitted with a mass mimicking a malignant bone neoplasm in the right mandible as the first manifestation of primary hyperparathyroidism. The second case involved a 25-year-old man admitted with a fracture of his right femur. The third case involved a 43-year-old man with multiple brown tumors in both lower limbs. All three patients underwent successful parathyroidectomy for parathyroid adenomas; one case was complicated by a papillary thyroid carcinoma. CONCLUSION: Complete evaluation of the medical history and biochemical and radiographic findings is necessary to achieve a correct diagnosis and avoid unnecessary bone resections in patients with primary hyperparathyroidism.


Subject(s)
Diagnostic Imaging/methods , Hyperparathyroidism, Primary/diagnosis , Hyperparathyroidism, Primary/etiology , Parathyroid Neoplasms/complications , Parathyroid Neoplasms/diagnosis , Adult , Diagnosis, Differential , Humans , Hyperparathyroidism, Primary/surgery , Male , Parathyroid Neoplasms/surgery , Treatment Outcome
6.
PLoS One ; 9(4): e93256, 2014.
Article in English | MEDLINE | ID: mdl-24709998

ABSTRACT

AMP-activated protein kinase (AMPK) is a central metabolic sensor and plays an important role in regulating glucose, lipid and cholesterol metabolism. Therefore, AMPK is a key therapeutic target in diabetes. Recent pilot studies have suggested that diabetes drugs may reduce the risk of cancer by affecting the AMPK pathway. However, the association between AMPK and the proliferation of hepatocellular carcinoma (HCC) is unknown. In this study, we investigated the relationship between AMPK activity and the proliferation of HCC in cell lines, nude mice and human clinic samples. We first investigated the relationship between AMPK activity and cell proliferation in two HCC cell lines, PLC/PRF/5 and HepG2, by two AMPK activators, 5-aminoimidazole-4-carboxamide-1-h-D-ribofuranoside (AICAR) and metformain. AICAR and metformin treatment significantly inhibited the proliferation of HCC cells and induced cell cycle arrest at G1-S checkpoint. We then observed that metformin abrogated the growth of HCC xenografts in nude mice. The clinical pathology of AMPK activity in HCC, including cell proliferation, differential grade, tumor size and microvessel density, was studied by using 30 clinical tissue samples. In HCC tissue samples, phosphorylated AMPK was expressed mainly in cytoplasm. AMPK activity decreased significantly in HCC in comparison with paracancerous liver tissues (P<0.05). AMPK activity was negatively correlated with the level of Ki-67 (a marker of cell proliferation), differential degradation and tumor size (P<0.05), but not with microvessel density, hemorrhage or necrosis in HCC. Our findings suggest that AMPK activity inhibits the proliferation of HCC and AMPK might be an effective target for prevention and treatment of HCC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/enzymology , Cell Proliferation , Liver Neoplasms/enzymology , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Carcinoma, Hepatocellular/mortality , Hep G2 Cells , Heterografts , Humans , Hypoglycemic Agents/pharmacology , Ki-67 Antigen/metabolism , Liver Neoplasms/pathology , Metformin/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/mortality , Ribonucleotides/pharmacology
7.
Oncol Rep ; 30(5): 2449-57, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24008375

ABSTRACT

Metformin is used as a first-line therapy for type 2 diabetes, with reports of its usefulness for the prevention and control of several types of cancers. This study investigated the effects of metformin on hepatocellular carcinoma (HCC). The human HCC cell lines HepG2 and PLC/PRF/5 were cultured and treated with metformin or 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), an activator of adenosine monophosphate (AMP)-activated protein kinase. Changes in cell viability and cell cycle distribution were evaluated by MTT and flow cytometry, respectively. Apoptosis was assessed by fluorescent-dye staining. An HCC model was established in 6- to 8-week-old BALB/c-nu mice by subcutaneous injection of PLC/PRF/5 cells. After 1 week, mice were treated intragastrically with metformin or vehicle. Tumor xenograft tissues were examined using immunohistochemistry for evaluation of the the expression of cyclin D1, p21CIP and p27KIP. HCC cells and tissues from the in vitro and in vivo experiments, respectively, were subjected to protein extraction and western blotting. We found that metformin treatment reduced HCC cell viability in a dose-dependent manner similar to AICAR treatment. In addition, metformin treatment induced HCC cell cycle arrest at G1/G0 phase and apoptosis. Intragastric treatment of the mouse PLC/PRF/5 cell xenograft model with metformin showed that metformin not only blocked tumor progression, but also reduced tumor morbidity. Treatment with metformin upregulated the expression of p21CIP and p27KIP, but downregulated cyclin D1 levels, both in vitro and in vivo. Metformin treatment also upregulated the expression of phosphorylated AMPK protein in xenograft tissues. These findings indicate that metformin warrants further evaluation as a novel therapeutic and preventive strategy against HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Liver Neoplasms/drug therapy , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Cyclin D1/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Metformin/pharmacology , Mice , Ribonucleotides/pharmacology , Xenograft Model Antitumor Assays
8.
Cancer Invest ; 29(4): 286-92, 2011 May.
Article in English | MEDLINE | ID: mdl-21469977

ABSTRACT

A higher expression of S-100 in tissue of thyroid papillary carcinoma (TPC) vs. thyroid follicular adenoma (TFA) (p < .001) was observed as well as a higher expression of CD83 in the peri-cancerous tissues vs. TFA (p < .001), oppositely, CD83 was negative in the cancerous net. TPC showed greater decreases in levels of CD80 and CD86 than did the TFA. These findings suggest that impaired immune function, absence of CD83-positive mature and activated dendritic cells in cancer nodules may have a role in the pathogenesis of TPC. The low expression of CD80 and CD86 in TPC may help them evade the immune system.


Subject(s)
Adenoma/immunology , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Dendritic Cells/immunology , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , S100 Proteins/metabolism , Adult , Aged , Carcinoma , Carcinoma, Papillary , Case-Control Studies , Cells, Cultured , Female , Humans , Immunohistochemistry , Middle Aged , Thyroid Cancer, Papillary , Thyroid Neoplasms/immunology , Time Factors , Tumor Escape , CD83 Antigen
9.
Cell Mol Immunol ; 1(5): 378-82, 2004 Oct.
Article in English | MEDLINE | ID: mdl-16285898

ABSTRACT

To investigate the expression and distribution of S-100 protein and CD 83 in the thyroid tissues of autoimmune thyroid diseases (ATDs), and to study the role of the dendritic cells in the pathogenesis of ATDs, immunohistochemical staining was used on pathological tissues of 20 patients with Hashimoto's thyroiditis (HT) and 20 patients with Graves' disease (GD) to check the expression and distribution of S-100 protein and CD 83. Compared with control group (20 cases of thyroid follicular adenoma, TFA), the higher expressions of S-100 in HT (139.38+/-5.92 vs 59.47+/-11.69) and GD (119.42+/-14.48 vs 59.47+/-11.69) were observed respectively (p<0.001). The increased positive expressions of CD 83 which is known as a marker of mature and activated DCs in HT (22.58+/-13.96 vs 5.19+/-8.08) and GD (29.92 +/-14.43 vs 5.19+/-8.08) were also found respectively (p<0.001). Serum TPO antibody (TPO-Ab, 67.3+/-11.6%) and Tg antibody (Tg-Ab, 59.8+/-10.1%) in HT were higher than those in GD (28.4+/-5.7%, 23.1+/-4.9%) and TFA (6.1+/-3.4%, 7.2 +/-4.6%) (p<0.01). Serum TR-Ab in GD (16.3+/-5.6 U/L) was higher than those in HT (4.8+/- 2.3 U/L) and TFA (2.5+/-1.2 U/L) (p<0.01). Our findings suggest that the high expression of DCs' markers may be related to the pathogenesis of HT and GD. The upregulation of both the number and the matured functions of DCs, may lead to present more antigens and to produce more auto-antibodies (such as Tg-Ab and TPO-Ab in HT, TR-Ab in GD), which may be involved in pathogenesis of the autoimmune thyroid diseases.


Subject(s)
Antigens, CD/biosynthesis , Gene Expression Regulation , Graves Disease/metabolism , Hashimoto Disease/metabolism , Immunoglobulins/biosynthesis , Membrane Glycoproteins/biosynthesis , S100 Proteins/biosynthesis , Thyroid Gland/metabolism , Adult , Aged , Antigens, CD/genetics , Autoantibodies/blood , Biomarkers/metabolism , Dendritic Cells/pathology , Female , Graves Disease/blood , Graves Disease/pathology , Hashimoto Disease/blood , Hashimoto Disease/genetics , Humans , Immunoglobulins/genetics , Membrane Glycoproteins/genetics , Middle Aged , S100 Proteins/genetics , Thyroid Gland/pathology , CD83 Antigen
10.
Zhongguo Fei Ai Za Zhi ; 7(4): 354-6, 2004 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-21241560

ABSTRACT

BACKGROUND: To explore a rapid and accurate method for examining cancer cell from the sputum in patients with lung cancer. METHODS: One hundred and twenty patients with lung cancer diagnosed by operation and pathologically confirmed were enrolled in this study. Sputum sediment section and sputum smear examinations were performed and compared for diagnosis of lung cancer. RESULTS: (1) The positive rate of lung cancer cell was 71.67% (86/120) by sputum sediment section examination, however, only 31.67% (38/120) by sputum smear examination ( P < 0.001). The diagnostic rate of combination of two methods for lung cancer was 90.83% (109/120), which was significantly higher than that of single sputum sediment section examination ( P < 0.001). (2) With routine HE staining, 55 patients (55/86,63.95%) could be histologically identified by sputum sediment section examination, but only 6 patients (6/38,15.79%) by sputum smear examination ( P < 0.001). (3) In 31 patients unidentified with routine HE staining, 29 were further histologically confirmed by sputum sediment section examination with immunohistochemistry. CONCLUSIONS: Compared to sputum smear examination, sputum sediment section examination can make use of more sputum materials, show a higher sensitivity for cancer cell, and accurately identify the histological classification of tumor. It is supposed to be a good examination for lung cancer and deserved to be extended in clinical application.

SELECTION OF CITATIONS
SEARCH DETAIL