Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Biochem Biophys Res Commun ; 662: 66-75, 2023 06 25.
Article in English | MEDLINE | ID: mdl-37099812

ABSTRACT

Thyroid hormone (T3)-induced autophagy and its biological significance have been extensively investigated in recent years. However, limited studies to date have focused on the important role of lysosomes in autophagy. In this study, we explored the effects of T3 on lysosomal protein expression and trafficking in detail. Our findings showed that T3 activates rapid lysosomal turnover and expression of numerous lysosomal genes, including TFEB, LAMP2, ARSB, GBA, PSAP, ATP6V0B, ATP6V0D1, ATP6V1E1, CTSB, CTSH, CTSL, and CTSS, in a thyroid hormone receptor-dependent manner. In a murine model, LAMP2 protein was specifically induced in mice with hyperthyroidism. T3-promoted microtubule assembly was significantly disrupted by vinblastine, resulting in accumulation of the lipid droplet marker PLIN2. In the presence of the lysosomal autophagy inhibitors bafilomycin A1, chloroquine and ammonium chloride, we observed substantial accumulation of LAMP2 but not LAMP1 protein. T3 further enhanced the protein levels of ectopically expressed LAMP1 and LAMP2. Upon knockdown of LAMP2, cavities of lysosomes and lipid droplets accumulated in the presence of T3, although the changes in LAMP1 and PLIN2 expression were less pronounced. More specifically, the protective effect of T3 against ER stress-induced death was abolished by knockdown of LAMP2. Our collective results indicate that T3 not only promotes lysosomal gene expression but also LAMP protein stability and microtubule assembly, leading to enhancement of lysosomal activity in digesting any additional autophagosomal burden.


Subject(s)
Lysosomes , Thyroid Hormones , Animals , Mice , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/metabolism , Thyroid Hormones/metabolism , Autophagy/physiology
2.
Cancer Sci ; 113(1): 205-220, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34773335

ABSTRACT

Lung adenocarcinoma (ADC) is the predominant histological type of lung cancer, and radiotherapy is one of the current therapeutic strategies for lung cancer treatment. Unfortunately, biological complexity and cancer heterogeneity contribute to radioresistance development. Karyopherin α2 (KPNA2) is a member of the importin α family that mediates the nucleocytoplasmic transport of cargo proteins. KPNA2 overexpression is observed across cancer tissues of diverse origins. However, the role of KPNA2 in lung cancer radioresistance is unclear. Herein, we demonstrated that high expression of KPNA2 is positively correlated with radioresistance and cancer stem cell (CSC) properties in lung ADC cells. Radioresistant cells exhibited nuclear accumulation of KPNA2 and its cargos (OCT4 and c-MYC). Additionally, KPNA2 knockdown regulated CSC-related gene expression in radioresistant cells. Next-generation sequencing and bioinformatic analysis revealed that STAT1 activation and nuclear phospholipid scramblase 1 (PLSCR1) are involved in KPNA2-mediated radioresistance. Endogenous PLSCR1 interacting with KPNA2 and PLSCR1 knockdown suppressed the radioresistance induced by KPNA2 expression. Both STAT1 and PLSCR1 were found to be positively correlated with dysregulated KPNA2 in radioresistant cells and ADC tissues. We further demonstrated a potential positive feedback loop between PLSCR1 and STAT1 in radioresistant cells, and this PLSCR1-STAT1 loop modulates CSC characteristics. In addition, AKT1 knockdown attenuated the nuclear accumulation of KPNA2 in radioresistant lung cancer cells. Our results collectively support a mechanistic understanding of a novel role for KPNA2 in promoting radioresistance in lung ADC cells.


Subject(s)
Adenocarcinoma of Lung/metabolism , Cell Nucleus/metabolism , Lung Neoplasms/metabolism , Phospholipid Transfer Proteins/metabolism , Radiation Tolerance , STAT1 Transcription Factor/metabolism , alpha Karyopherins/metabolism , Adenocarcinoma of Lung/genetics , Cell Line, Tumor , Feedback, Physiological , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/radiation effects , Gene Knockout Techniques , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Phospholipid Transfer Proteins/genetics , STAT1 Transcription Factor/genetics , Up-Regulation , alpha Karyopherins/genetics
3.
Pharmacogenomics J ; 20(1): 57-68, 2020 02.
Article in English | MEDLINE | ID: mdl-31611591

ABSTRACT

Although targeted agents are recommended as the first-line treatments for advanced hepatocellular carcinoma (aHCC), systemic chemotherapy or hepatic arterial infusion chemotherapy (HAIC) are still being used in Asian countries. Beside economic considerations, it was found that targeted drugs could not significantly prolong overall survival in aHCC patients with distant metastasis. In addition, chemotherapy could achieve complete response in a small proportion of patients. Here, we aimed to investigate whether combination of three previously identified single nucleotide polymorphism (SNP) predictors (GALNT14-rs9679162, WWOX-rs13338697, and rs6025211) could guide our choice between systemic chemotherapy, HAIC, and targeted agents in aHCC patients. A cohort of 237 real-world aHCC patients (171 receiving systemic chemotherapy followed by various anticancer treatments including sorafenib; 66 receiving HAIC) were included for outcome analysis. By combining the three SNP markers with or without addition of two clinical criteria (tumor diameter <8 cm, neutrophils <80%), small groups of patients were found to harbor high complete response rates to systemic chemotherapy (35.3% if the 3-SNP signature alone matched; 60.0% if clinical criteria also matched). Subsequent sorafenib treatment for chemotherapy non-responders was associated with longer overall survival (P < 0.001). In HAIC-treated patients, GALNT14-rs9679162 genotype "GG" was associated with longer overall survival (P = 0.019, median survival > 10.5 months). In conclusion, pre-test for the 3-SNP signature in aHCC patients could identify potential systemic chemotherapy or HAIC responders. Chemotherapy non-responders still benefited from subsequent sorafenib treatment. Accordingly, we propose a roadmap for aHCC patients when chemotherapy or HAIC is to be used.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Infusions, Intra-Arterial/methods , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , N-Acetylgalactosaminyltransferases/genetics , Adult , Aged , Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/mortality , Female , Genotype , Humans , Liver Neoplasms/mortality , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Survival Rate/trends , Treatment Outcome
4.
J Med Virol ; 92(1): 124-127, 2020 01.
Article in English | MEDLINE | ID: mdl-31469172

ABSTRACT

We examined the seroprevalence change of anti-hepatitis D virus (HDV) antibodies in Taiwan from 2006 to 2019. A total of 1147 patients who had chronic hepatitis B virus (HBV) infection were assessed. Of them, 51 (4.4%) were positive for anti-HDV antibodies. Comparison between anti-HDV-positive and negative groups was performed to examine clinical and virological factors related to anti-HDV positivity. It was found that the median HBV-DNA concentration was 1.6 × 105 IU/mL (range, <20-4.5 × 1010 IU/mL) and <20 IU/mL (range, <20-2.0 × 109 IU/mL) for patients with negative and positive anti-HDV antibodies, respectively (P < .001). In addition, a progressive year-to-year decrease of anti-HDV seroprevalence was unveiled. For patients who had HBV-DNA >15 000 IU/mL, the year-to-year (calculated every 2 years) seropositive rates of anti-HDV were 10.0%, 7.9%, 0.7%, 0.3%, 0%, 0%, and 0% (P < .001). For patients who had HBV-DNA <15 000 IU/mL, the year-to-year seropositive rates were 18.6%, 12.8%, 7.8%, 5.0%, 7.3%, 8.0%, and 3.7% (P < .001). In conclusion, seropositive of anti-HDV was inversely associated with HBV-DNA levels. A progressive decrease of anti-HDV seroprevalence was found with no anti-HDV-positive cases detected in high HBV-DNA patient group after 2014.


Subject(s)
DNA, Viral/blood , Hepatitis Antibodies/blood , Hepatitis B virus/genetics , Hepatitis B, Chronic/epidemiology , Hepatitis D/epidemiology , Adult , Aged , Coinfection/epidemiology , Coinfection/immunology , Coinfection/virology , Female , Hepatitis B, Chronic/immunology , Hepatitis D/immunology , Humans , Male , Middle Aged , Retrospective Studies , Seroepidemiologic Studies , Taiwan/epidemiology
6.
Int J Mol Sci ; 21(9)2020 04 30.
Article in English | MEDLINE | ID: mdl-32366022

ABSTRACT

The authors wish to make the following corrections to this paper [...].

7.
Int J Mol Sci ; 21(5)2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32182776

ABSTRACT

Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related deaths worldwide. For patients who are resistant to monotherapy, multimodal therapy is a basic oncologic principle that incorporates surgery, radiotherapy (RT), and chemotherapy providing survival benefits for patients with most types of cancer. Although liver has low tolerance for radiation, high-precision RT for local HCC minimizes the likelihood of radiation-induced liver disease (RILD) in noncancerous liver tissue. RT have several therapeutic benefits, including the down-staging of tumors to make them resectable and repression of metastasis. The DNA damage response (DDR) is a cellular response to irradiation (IR), including DNA repair of injured cells and induction of programmed cell death, thereby resulting in maintenance of cell homeostasis. Molecules that block the activity of proteins in DDR pathways have been found to enhance radiotherapeutic effects. These molecules include antibodies, kinase inhibitors, siRNAs and miRNAs. MicroRNAs (miRNAs) are short non-coding regulatory RNAs binding to the 3'-untranslated regions (3'-UTR) of the messenger RNAs (mRNAs) of target genes, regulating their translation and expression of proteins. Thus, miRNAs and their target genes constitute complicated interactive networks, which interact with other molecules during carcinogenesis. Due to their promising roles in carcinogenesis, miRNAs were shown to be the potential factors that mediated radiosensitivity and optimized outcomes of the combination of systemic therapy and radiotherapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Radiation Tolerance/genetics , 3' Untranslated Regions/genetics , Animals , DNA Repair/genetics , Humans , Signal Transduction/genetics
8.
Hepatology ; 67(1): 188-203, 2018 01.
Article in English | MEDLINE | ID: mdl-28802060

ABSTRACT

Cancer cells display altered glucose metabolism characterized by a preference for aerobic glycolysis. The aerobic glycolytic phenotype of hepatocellular carcinoma (HCC) is often correlated with tumor progression and poorer clinical outcomes. However, the issue of whether glycolytic metabolism influences metastasis in HCC remains unclear. In the current study, we showed that knockdown of taurine up-regulated gene 1 (TUG1) induces marked inhibition of cell migration, invasion, and glycolysis through suppression of microRNA (miR)-455-3p. MiR-455-3p, which is transcriptionally repressed by p21, directly targets the 3' untranslated region of adenosine monophosphate-activated protein kinase subunit beta 2 (AMPKß2). The TUG1/miR-455-3p/AMPKß2 axis regulates cell growth, metastasis, and glycolysis through regulation of hexokinase 2 (HK2). TUG1 is clearly associated with HK2 overexpression and unfavorable prognosis in HCC patients. CONCLUSION: Our data collectively highlight that novel regulatory associations among TUG1, miR-455-3p, AMPKß2, and HK2 are an important determinant of glycolytic metabolism and metastasis in HCC cells and support the potential utility of targeting TUG1/HK2 as a therapeutic strategy for HCC. (Hepatology 2018;67:188-203).


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/genetics , Glycolysis/genetics , Liver Neoplasms/genetics , RNA, Long Noncoding/genetics , Biopsy, Needle , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Male , MicroRNAs/genetics , Neoplasm Metastasis/genetics , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Signal Transduction/drug effects , Taurine/pharmacology , Up-Regulation
9.
FASEB J ; 32(5): 2601-2614, 2018 05.
Article in English | MEDLINE | ID: mdl-29401583

ABSTRACT

Argininosuccinate synthetase 1 (ASS1) is a rate-limited enzyme in arginine biosynthesis. The oncogenic potential of ASS1 in terms of prognosis and cancer metastasis in arginine prototrophic gastric cancer (GC) remains unclear at present. We identify differentially expressed proteins in microdissected GC tumor cells relative to adjacent nontumor epithelia by isobaric mass tag for relative and absolute quantitation proteomics analysis. GC cells with stable expression or depletion of ASS1 were further analyzed to identify downstream molecules. We investigated their effects on chemoresistance and cell invasion in the presence or absence of arginine. ASS1 was highly expressed in GC and positively correlated with GC aggressiveness and poor outcome. Depletion of ASS1 led to inhibition of tumor growth and decreased cell invasion via induction of autophagy-lysosome machinery, resulting in degradation of active ß-catenin, Snail, and Twist. Ectopic expression of ASS1 in GC cells reversed these effects and protected cancer cells from chemotherapy drug-induced apoptosis via activation of the AKT-mammalian target of rapamycin signaling pathway. ASS1 contributes to GC progression by enhancing aggressive potential resulting from active ß-catenin, Snail, and Twist accumulation. Our results propose that ASS1 might contribute to GC metastasis and support its utility as a prognostic predictor of GC.-Tsai, C.-Y., Chi, H.-C., Chi, L.-M., Yang, H.-Y., Tsai, M.-M., Lee, K.-F., Huang, H.-W., Chou, L.-F., Cheng, A.-J., Yang, C.-W., Wang, C.-S., Lin, K.-H. Argininosuccinate synthetase 1 contributes to gastric cancer invasion and progression by modulating autophagy.


Subject(s)
Argininosuccinate Synthase/biosynthesis , Autophagy , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Signal Transduction , Stomach Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Argininosuccinate Synthase/genetics , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
10.
J Biomed Sci ; 26(1): 24, 2019 Mar 08.
Article in English | MEDLINE | ID: mdl-30849993

ABSTRACT

The liver is controlled by several metabolic hormones, including thyroid hormone, and characteristically displays high lysosomal activity as well as metabolic stress-triggered autophagy, which is stringently regulated by the levels of hormones and metabolites. Hepatic autophagy provides energy through catabolism of glucose, amino acids and free fatty acids for starved cells, facilitating the generation of new macromolecules and maintenance of the quantity and quality of cellular organelles, such as mitochondria. Dysregulation of autophagy and defective mitochondrial homeostasis contribute to hepatocyte injury and liver-related diseases, such as non-alcoholic fatty liver disease (NAFLD) and liver cancer.Thyroid hormones (TH) mediate several critical physiological processes including organ development, cell differentiation, metabolism and cell growth and maintenance. Accumulating evidence has revealed dysregulation of cellular TH activity as the underlying cause of several liver-related diseases, including alcoholic or non-alcoholic fatty liver disease and liver cancer. Data from epidemiologic, animal and clinical studies collectively support preventive functions of THs in liver-related diseases, highlighting the therapeutic potential of TH analogs. Elucidation of the molecular mechanisms and downstream targets of TH should thus facilitate the development of therapeutic strategies for a number of major public health issues.Here, we have reviewed recent studies focusing on the involvement of THs in hepatic homeostasis through induction of autophagy and their implications in liver-related diseases. Additionally, the potential underlying molecular pathways and therapeutic applications of THs in NAFLD and HCC are discussed.


Subject(s)
Autophagy , Homeostasis , Liver Neoplasms/physiopathology , Liver/physiology , Non-alcoholic Fatty Liver Disease/physiopathology , Thyroid Hormones/physiology , Animals , Humans , Liver/physiopathology , Mice , Rats
11.
Int J Mol Sci ; 20(20)2019 Oct 09.
Article in English | MEDLINE | ID: mdl-31600974

ABSTRACT

Several physiological processes, including cellular growth, embryonic development, differentiation, metabolism and proliferation, are modulated by genomic and nongenomic actions of thyroid hormones (TH). Several intracellular and extracellular candidate proteins are regulated by THs. 3,3,5-Triiodo-L-thyronine (T3) can interact with nuclear thyroid hormone receptors (TR) to modulate transcriptional activities via thyroid hormone response elements (TRE) in the regulatory regions of target genes or bind receptor molecules showing no structural homology to TRs, such as the cell surface receptor site on integrin αvß3. Additionally, L-thyroxine (T4) binding to integrin αvß3 is reported to induce gene expression through initiating non-genomic actions, further influencing angiogenesis and cell proliferation. Notably, thyroid hormones not only regulate the physiological processes of normal cells but also stimulate cancer cell proliferation via dysregulation of molecular and signaling pathways. Clinical hypothyroidism is associated with delayed cancer growth. Conversely, hyperthyroidism is correlated with cancer prevalence in various tumor types, including breast, thyroid, lung, brain, liver and colorectal cancer. In specific types of cancer, both nuclear thyroid hormone receptor isoforms and those on the extracellular domain of integrin αvß3 are high risk factors and considered potential therapeutic targets. In addition, thyroid hormone analogs showing substantial thyromimetic activity, including triiodothyroacetic acid (Triac), an acetic acid metabolite of T3, and tetraiodothyroacetic acid (Tetrac), a derivative of T4, have been shown to reduce risk of cancer progression, enhance therapeutic effects and suppress cancer recurrence. Here, we have reviewed recent studies focusing on the roles of THs and TRs in five cancer types and further discussed the potential therapeutic applications and underlying molecular mechanisms of THs.


Subject(s)
Apoptosis , Neoplasms/etiology , Neoplasms/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Gene Expression Regulation , Humans , Neoplasms/pathology , Protein Binding , Receptors, Thyroid Hormone/metabolism
12.
Int J Mol Sci ; 20(20)2019 Oct 21.
Article in English | MEDLINE | ID: mdl-31640265

ABSTRACT

Oxidative stress occurs as a result of imbalance between the generation of reactive oxygen species (ROS) and antioxidant genes in cells, causing damage to lipids, proteins, and DNA. Accumulating damage of cellular components can trigger various diseases, including metabolic syndrome and cancer. Over the past few years, the physiological significance of microRNAs (miRNA) in cancer has been a focus of comprehensive research. In view of the extensive level of miRNA interference in biological processes, the roles of miRNAs in oxidative stress and their relevance in physiological processes have recently become a subject of interest. In-depth research is underway to specifically address the direct or indirect relationships of oxidative stress-induced miRNAs in liver cancer and the potential involvement of the thyroid hormone in these processes. While studies on thyroid hormone in liver cancer are abundantly documented, no conclusive information on the potential relationships among thyroid hormone, specific miRNAs, and oxidative stress in liver cancer is available. In this review, we discuss the effects of thyroid hormone on oxidative stress-related miRNAs that potentially have a positive or negative impact on liver cancer. Additionally, supporting evidence from clinical and animal experiments is provided.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Thyroid Hormones/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
13.
Int J Mol Sci ; 20(22)2019 Nov 13.
Article in English | MEDLINE | ID: mdl-31766266

ABSTRACT

Gastric cancer (GC) is the second most widespread cause of cancer-related mortality worldwide. The discovery of novel biomarkers of oncoproteins can facilitate the development of therapeutic strategies for GC treatment. In this study, we identified novel biomarkers by integrating isobaric tags for relative and absolute quantitation (iTRAQ), a human plasma proteome database, and public Oncomine datasets to search for aberrantly expressed oncogene-associated proteins in GC tissues and plasma. One of the most significantly upregulated biomarkers, DEK, was selected and its expression validated. Our immunohistochemistry (IHC) (n = 92) and quantitative real-time polymerase chain reaction (qRT-PCR) (n = 72) analyses disclosed a marked increase in DEK expression in tumor tissue, compared with paired nontumor mucosa. Importantly, significantly higher preoperative plasma DEK levels were detected in GC patients than in healthy controls via enzyme-linked immunosorbent assay (ELISA). In clinicopathological analysis, higher expression of DEK in both tissue and plasma was significantly associated with advanced stage and poorer survival outcomes of GC patients. Data from receiver operating characteristic (ROC) curve analysis disclosed a better diagnostic accuracy of plasma DEK than carcinoembryonic antigen (CEA), carbohydrate antigen 19.9 (CA 19.9), and C-reactive protein (CRP), highlighting its potential as an effective plasma biomarker for GC. Plasma DEK is also more sensitive in tumor detection than the other three biomarkers. Knockdown of DEK resulted in inhibition of GC cell migration via a mechanism involving modulation of matrix metalloproteinase MMP-2/MMP-9 level and vice versa. Our results collectively support plasma DEK as a useful biomarker for making diagnosis and prognosis of GC patients.


Subject(s)
Chromosomal Proteins, Non-Histone/analysis , Oncogene Proteins/analysis , Poly-ADP-Ribose Binding Proteins/analysis , Stomach Neoplasms/pathology , Aged , Biomarkers, Tumor/analysis , Cell Line, Tumor , Cell Movement , Chromosomal Proteins, Non-Histone/blood , Female , Humans , Male , Middle Aged , Neoplasm Metastasis/diagnosis , Neoplasm Metastasis/pathology , Oncogene Proteins/blood , Poly-ADP-Ribose Binding Proteins/blood , Prognosis , Stomach Neoplasms/blood , Stomach Neoplasms/diagnosis , Survival Analysis
14.
Mol Carcinog ; 57(9): 1087-1101, 2018 09.
Article in English | MEDLINE | ID: mdl-29672920

ABSTRACT

Heat shock protein 60 (HSP60) overexpresses in various types of cancer, but its expression levels and functions in hepatocellular carcinoma (HCC) are still in dispute. We aim to clarify this issue and examine whether HSP60 could be a therapeutic target for HCC. We found drastically enhanced cell apoptosis and suppressed cell proliferation in two HCC cell lines with HSP60-silencing, and also indicated survivin was involved in this regulatory process in vitro and in vivo. However, HSP60-silencing in normal human hepatocytes only resulted in a minimal reduction of cell proliferation but without effects on cell apoptosis. We also showed HSP60 interacted with cytosolic but not mitochondrial survivin by immunoprecipitation assay. A rigorous method was used to standardize quantification from immunoblot assay to obtain more precise expression levels of HSP60 and survivin. The expression of HSP60 and survivin positively correlated in both cancerous and non-cancerous liver tissues (P < 0.001) after analyzing 145 surgically removed HCC tissues. A total of 56.6% of HCC patients overexpressed HSP60 in cancerous tissues, and 40.0% under-expressed HSP60. Higher expression of HSP60 and survivin in non-cancerous tissues both correlated with shorter overall survival (P = 0.029 and P < 0.001, respectively). Finally, we evaluated the therapeutic potential of HSP60 using extraneous delivery of jetPEI/shHSP60 complexes. The treatment results showed significant reduction of tumor weight by 44.3% (P < 0.05), accompanied by under-expression of survivin. These studies suggested that HSP60 not only served as a prognostic marker but also served as a novel therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Chaperonin 60/genetics , Liver Neoplasms/therapy , RNA, Small Interfering/therapeutic use , RNAi Therapeutics , Survivin/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Chaperonin 60/analysis , Cytoplasm/genetics , Cytoplasm/pathology , Gene Expression Regulation, Neoplastic , Injections, Subcutaneous , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNAi Therapeutics/methods , Survivin/analysis
15.
Dig Dis Sci ; 63(9): 2301-2308, 2018 09.
Article in English | MEDLINE | ID: mdl-29736829

ABSTRACT

BACKGROUND: Gastric cancer has a poor outcome and identifying useful biomarkers from peripheral blood or tissue could allow its early detection, or potentially precancerous changes, thus improving the curative rates. MicroRNAs (miRNAs) have been shown to offer great potential in cancer diagnosis and prediction. AIM: Here, we investigated the role of plasma miRNAs in the natural course of gastric cancer, from intestinal metaplasia to early cancer. The findings were used to understand whether patients at a high risk of malignancy could be given appropriate interventions in the early disease process, such as using endoscopic submucosal dissection to treat gastric dysplasia or early gastric cancer. METHODS: Participants were divided into healthy control, intestinal metaplasia (IM), and dysplasia/early cancer (pT1a/b) groups. Microarray was used to select potential markers in tissue. RESULTS: Quantitative real-time polymerase chain reaction data showed circulating miRNA-22-3p had significantly different expression in patients with precancerous lesions or gastric adenocarcinoma. The areas under the curve of incomplete IM versus healthy control, low-grade/high-grade dysplasia, early gastric cancer, and GED were 0.8080, 0.8040, 0.8494, and 0.8095, respectively (all P values < 0.05). CONCLUSIONS: Circulating miRNA-22-3p could be a potential biomarker for gastric precancerous dysplasia and early cancer detection.


Subject(s)
Adenocarcinoma/blood , Biomarkers, Tumor/blood , Circulating MicroRNA/blood , MicroRNAs/blood , Precancerous Conditions/blood , Stomach Neoplasms/blood , Adenocarcinoma/diagnosis , Aged , Cohort Studies , Disease Progression , Female , Humans , Male , Metaplasia/blood , Metaplasia/diagnosis , Middle Aged , Precancerous Conditions/diagnosis , Predictive Value of Tests , Stomach Neoplasms/diagnosis
16.
Int J Mol Sci ; 19(12)2018 Nov 24.
Article in English | MEDLINE | ID: mdl-30477236

ABSTRACT

The tumor microenvironment is an important concept that defines cancer development not only through tumor cells themselves but also the surrounding cellular and non-cellular components, including stromal cells, blood vessels, infiltrating inflammatory cells, cancer stem cells (CSC), cytokines, and growth factors, which act in concert to promote tumor cell survival and metastasis. Hepatocellular carcinoma (HCC) is one of the most common and aggressive human malignancies worldwide. Poor prognosis is largely attributable to the high rate of tumor metastasis, highlighting the importance of identifying patients at risk in advance and developing novel therapeutic targets to facilitate effective intervention. Long non-coding RNAs (lncRNA) are a class of non-protein coding transcripts longer than 200 nucleotides frequently dysregulated in various cancer types, which have multiple functions in widespread biological processes, including proliferation, apoptosis, metastasis, and metabolism. lncRNAs are involved in regulation of the tumor microenvironment and reciprocal signaling between cancer cells. Targeting of components of the tumor microenvironment or cancer cells has become a considerable focus of therapeutic research and establishing the effects of different lncRNAs on this network should aid in the development of effective treatment strategies. The current review provides a summary of the essential properties and functional roles of known lncRNAs associated with the tumor microenvironment in HCC.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Neoplasm Proteins/genetics , RNA, Long Noncoding/genetics , Tumor Microenvironment/drug effects , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cytokines/genetics , Cytokines/metabolism , Epigenesis, Genetic , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , Signal Transduction , Tumor Microenvironment/genetics
17.
Int J Mol Sci ; 19(2)2018 Feb 12.
Article in English | MEDLINE | ID: mdl-29439529

ABSTRACT

Radiotherapy is a well-established regimen for nearly half the cancer patients worldwide. However, not all cancer patients respond to irradiation treatment, and radioresistance is highly associated with poor prognosis and risk of recurrence. Elucidation of the biological characteristics of radioresistance and development of effective prognostic markers to guide clinical decision making clearly remain an urgent medical requirement. In tumorigenic and radioresistant cancer cell populations, phenotypic switch is observed during the course of irradiation treatment, which is associated with both stable genetic and epigenetic changes. While the importance of epigenetic changes is widely accepted, the irradiation-triggered specific epigenetic alterations at the molecular level are incompletely defined. The present review provides a summary of current studies on the molecular functions of DNA and RNA m6A methylation, the key epigenetic mechanisms involved in regulating the expression of genetic information, in resistance to irradiation and cancer progression. We additionally discuss the effects of DNA methylation and RNA N6-methyladenosine (m6A) of specific genes in cancer progression, recurrence, and radioresistance. As epigenetic alterations could be reversed by drug treatment or inhibition of specific genes, they are also considered potential targets for anticancer therapy and/or radiotherapy sensitizers. The mechanisms of irradiation-induced alterations in DNA and RNA m6A methylation, and ways in which this understanding can be applied clinically, including utilization of methylation patterns as prognostic markers for cancer radiotherapy and their manipulation for anticancer therapy or use as radiotherapy sensitizers, have been further discussed.


Subject(s)
DNA Methylation , Neoplasms/genetics , RNA Processing, Post-Transcriptional , Radiation Tolerance/genetics , Animals , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/pathology , Neoplasms/radiotherapy
18.
Int J Mol Sci ; 19(5)2018 May 14.
Article in English | MEDLINE | ID: mdl-29758012

ABSTRACT

Anaplastic lymphoma kinase (ALK)-negative anaplastic large-cell lymphoma (ALCL) is a rare type of highly malignant, non-Hodgkin lymphoma. Currently, only a few gene rearrangements have been linked to ALK-negative ALCL progression. However, the specific molecular mechanisms underlying the growth of ALK-negative ALCL tumors remain unclear. Here, we investigated aberrantly expressed, long non-coding RNAs (lncRNAs) in ALK-negative ALCL and assessed their potential biological function. MIR503HG (miR-503 host gene) was highly expressed in ALK-negative cell lines and was significantly upregulated in tumors in mice formed from ALK-negative ALCL cell lines. Depletion of MIR503HG suppressed tumor cell proliferation in vivo and in vitro; conversely, its overexpression enhanced tumor cell growth. MIR503HG-induced proliferation was mediated by the induction of microRNA-503 (miR-503) and suppression of Smurf2, resulting in stabilization of the tumor growth factor-ß receptor (TGFBR) and enhanced tumor cell growth. Collectively, these findings support a potential role for MIR503HG in cancer cell proliferation through the miR-503/Smurf2/TGFBR axis and indicate that MIR503HG is a potential marker in ALK-negative ALCL.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, Large-Cell, Anaplastic/genetics , RNA, Long Noncoding/genetics , Receptor Protein-Tyrosine Kinases/deficiency , Anaplastic Lymphoma Kinase , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Heterografts , Humans , Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Mice , MicroRNAs/genetics , RNA Interference , Receptors, Transforming Growth Factor beta , Ubiquitin-Protein Ligases/genetics
19.
Int J Cancer ; 140(7): 1581-1596, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27925179

ABSTRACT

NEK2 (NIMA-related expressed kinase 2) is a serine/threonine centrosomal kinase that acts as a critical regulator of centrosome structure and function. Aberrant NEK2 activities lead to failure in regulating centrosome duplication. NEK2 overexpression promotes tumorigenesis and is associated with poor prognosis in several cancers. Increased NEK2 expression during the late pathological stage has been detected in the Oncomine liver dataset and hepatocellular carcinoma (HCC) specimens. Elevated NEK2 protein is associated with poor overall survival in patients with HCC. However, the precise roles and mechanisms of NEK2 in liver cancer progression remain largely unknown. An earlier functional study revealed that NEK2 mediates drug resistance (cisplatin or lipo-doxorubicin) via expression of an ABCC10 transporter. Active angiogenesis and metastasis underlie the rapid recurrence and poor survival of HCC. Results from the current study showed that NEK2 mediates tumor growth, metastasis and angiogenesis in vivo. NEK2-mediated drug resistance was blocked by a specific PI3K or AKT inhibitor. Moreover, NEK2 mediated liver cancer cell migration via pAKT/NF-κB signaling and matrix metalloproteinase (MMP) activation. Angiogenesis was induced via the same signaling pathway and IL-8 stimulation. Our findings collectively indicate that NEK2 modulates hepatoma cell functions, including growth, drug resistance, metastasis and angiogenesis via downstream genes activation.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , NIMA-Related Kinases/physiology , Aged , Animals , Apoptosis , Carcinogenesis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Centrosome/metabolism , Cisplatin/chemistry , Disease Progression , Doxorubicin/chemistry , Drug Resistance, Neoplasm , Female , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic , Prognosis , RNA, Small Interfering/metabolism , Treatment Outcome
20.
Int J Mol Sci ; 18(9)2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28872613

ABSTRACT

Radiotherapy is a well-established therapeutic regimen applied to treat at least half of all cancer patients worldwide. Radioresistance of cancers or failure to treat certain tumor types with radiation is associated with enhanced local invasion, metastasis and poor prognosis. Elucidation of the biological characteristics underlying radioresistance is therefore critical to ensure the development of effective strategies to resolve this issue, which remains an urgent medical problem. Cancer stem cells (CSCs) comprise a small population of tumor cells that constitute the origin of most cancer cell types. CSCs are virtually resistant to radiotherapy, and consequently contribute to recurrence and disease progression. Metastasis is an increasing problem in resistance to cancer radiotherapy and closely associated with the morbidity and mortality rates of several cancer types. Accumulating evidence has demonstrated that radiation induces epithelial-mesenchymal transition (EMT) accompanied by increased cancer recurrence, metastasis and CSC generation. CSCs are believed to serve as the basis of metastasis. Previous studies indicate that CSCs contribute to the generation of metastasis, either in a direct or indirect manner. Moreover, the heterogeneity of CSCs may be responsible for organ specificity and considerable complexity of metastases. Long noncoding RNAs (lncRNAs) are a class of noncoding molecules over 200 nucleotides in length involved in the initiation and progression of several cancer types. Recently, lncRNAs have attracted considerable attention as novel critical regulators of cancer progression and metastasis. In the current review, we have discussed lncRNA-mediated regulation of CSCs following radiotherapy, their association with tumor metastasis and significance in radioresistance of cancer.


Subject(s)
Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/genetics , Animals , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Humans , Radiotherapy , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL