Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Anal Biochem ; 654: 114794, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35777456

ABSTRACT

Gastric cancer seriously affects the health of modern people. The immune microenvironment of gastric cancer tissue is key to gastric cancer progression. We downloaded training and validation sets data from The Cancer Genome Atlas and Gene Expression Omnibus. Single-sample gene set enrichment analysis was used to sort patients into high, middle, and low immunity groups, of which immune infiltration in the high immunity group was substantially higher than of other two groups. Genes in high and low immunity groups expressed prominent differences. Further, the enrichment of differentially expressed genes was found mainly in immune-related pathways. Subsequently, an immune-related prognostic model was established, composed of ten prognosis-related genes identified by univariate risk regression, least absolute shrinkage and selection operator Cox, and multivariate risk regression. Survival analysis and receiver operating characteristic curves suggested good diagnostic efficacy of this model, and feature genes were linked to the degree of immune infiltration. An independent test suggested that the risk score could independently determine patient outcomes. We combined all clinical information and risk scores to establish a nomogram that could predict patient's prognosis. A prognostic model composed of 10 prognosis-related genes was generated with good diagnostic efficacy in predicting prognoses of gastric cancer patients.


Subject(s)
Stomach Neoplasms , Biomarkers, Tumor/analysis , Humans , Nomograms , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Tumor Microenvironment/genetics
2.
Theranostics ; 9(16): 4795-4810, 2019.
Article in English | MEDLINE | ID: mdl-31367258

ABSTRACT

Rationale: Emerging evidences have highlighted the critical roles of lncRNAs in human cancer development. The work sought to assess the biological role and potential underlying mechanisms of lncRNA-CF129 (CF129) which is significantly reduced in pancreatic cancer (PC). Methods: CF129 expression and its association with multiple clinicopathologic characteristics in PC specimens were analyzed. The role of CF129 both in vitro and in vivo was assessed, with RNA pull-down and immunoprecipitation assays being performed to detect the interaction between CF129 and p53 and E3 ligase MKRN1. Chromatin immunoprecipitation and luciferase assays were utilized to identify the interaction between p53 and FOXC2 promoter, HIF-1α/HDAC1 complex and CF129 promoter, FOXC2 and HIF-1α promoter, respectively. Results: CF129 levels were markedly lower in PC compared with paired non-tumor adjacent tissues. Low CF129 expression predicted short overall survival in PC patients. CF129 inhibited invasion and metastasis of PC cells in a FOXC2-dependent manner. In addition, CF129 regulates FOXC2 transcription through association with mutant p53. CF129 directly binds to p53 and E3 ligase MKRN1, and such an interaction leading to p53 protein ubiquitination and degradation. Furthermore, CF129 is a hypoxia-responsive lncRNA, which is transcriptionally downregulated by binding between HIF-1α/HDAC1 complex and CF129 promoter. Finally, it is revealed that HIF-1α is reciprocally regulated by FOXC2 in transcriptional level. Clinically, CF129 downregulation coordinates overexpression of FOXC2. Conclusions: Our study suggests that CF129 inhibits pancreatic cell proliferation and invasion by suppression of FOXC2 transcription, which depends on MKRN1-mediated ubiquitin-dependent p53 degradation. The HIF-1α/CF129/ p53/FOXC2 axis may function as a potential biomarker and therapeutic target.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pancreatic Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA, Long Noncoding/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Tumor Suppressor Protein p53/genetics
3.
Theranostics ; 9(18): 5298-5314, 2019.
Article in English | MEDLINE | ID: mdl-31410216

ABSTRACT

Rationale: Hypoxia has been proved to contribute to aggressive phenotype of cancers, while functional and regulatory mechanism of long noncoding RNA (lncRNA) in the contribution of hypoxia on pancreatic cancer (PC) tumorigenesis is incompletely understood. The aim of this study was to uncover the regulatory and functional roles for hypoxia-induced lncRNA-MTA2TR (MTA2 transcriptional regulator RNA, AF083120.1) in the regulation of PC tumorigenesis. Methods: A lncRNA microarray confirmed MTA2TR expression in tissues of PC patients. The effects of MTA2TR on proliferation and metastasis of PC cells and xenograft models were determined, and the key mechanisms by which MTA2TR promotes PC were further dissected. Furthermore, the expression and regulation of MTA2TR under hypoxic conditions in PC cells were assessed. We also assessed the correlation between MTA2TR expression and PC patient clinical outcomes. Results: We found that metastasis associated protein 2 (MTA2) transcriptional regulator lncRNA (MTA2TR) was overexpressed in PC patient tissues relative to paired noncancerous tissues. Furthermore, we found that depletion of MTA2TR significantly inhibited PC cell proliferation and invasion both in vitro and in vivo. We further demonstrated that MTA2TR transcriptionally upregulates MTA2 expression by recruiting activating transcription factor 3 (ATF3) to the promoter area of MTA2. Consequentially, MTA2 can stabilize the HIF-1α protein via deacetylation, which further activates HIF-1α transcriptional activity. Interestingly, our results revealed that MTA2TR is transcriptionally regulated by HIF-1α under hypoxic conditions. Our clinical samples further indicated that the overexpression of MTA2TR was correlated with MTA2 upregulation, as well as with reduced overall survival (OS) in PC patients. Conclusions: These results suggest that feedback between MTA2TR and HIF-1α may play a key role in regulating PC tumorigenesis, thus potentially highlighting novel avenues PC treatment.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pancreatic Neoplasms/genetics , RNA, Long Noncoding/metabolism , Acetylation , Activating Transcription Factor 3/metabolism , Animals , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Prognosis , Promoter Regions, Genetic , Protein Stability , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic
4.
Cancer Res ; 79(7): 1398-1412, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30563888

ABSTRACT

Cancer cells are known to undergo metabolic reprogramming, such as glycolysis and glutamine addiction, to sustain rapid proliferation and metastasis. It remains undefined whether long noncoding RNAs (lncRNA) coordinate the metabolic switch in pancreatic cancer. Here we identify a nuclear-enriched antisense lncRNA of glutaminase (GLS-AS) as a critical regulator involved in pancreatic cancer metabolism. GLS-AS was downregulated in pancreatic cancer tissues compared with noncancerous peritumor tissues. Depletion of GLS-AS promoted proliferation and invasion of pancreatic cancer cells both in vitro and in xenograft tumors of nude mice. GLS-AS inhibited GLS expression at the posttranscriptional level via formation of double stranded RNA with GLS pre-mRNA through ADAR/Dicer-dependent RNA interference. GLS-AS expression was transcriptionally downregulated by nutrient stress-induced Myc. Conversely, GLS-AS decreased Myc expression by impairing the GLS-mediated stability of Myc protein. These results imply a reciprocal feedback loop wherein Myc and GLS-AS regulate GLS overexpression during nutrient stress. Ectopic overexpression of GLS-AS inhibited proliferation and invasion of pancreatic cancer cells by repressing the Myc/GLS pathway. Moreover, expression of GLS-AS and GLS was inversely correlated in clinical samples of pancreatic cancer, while low expression of GLS-AS was associated with poor clinical outcomes. Collectively, our study implicates a novel lncRNA-mediated Myc/GLS pathway, which may serve as a metabolic target for pancreatic cancer therapy, and advances our understanding of the coupling role of lncRNA in nutrition stress and tumorigenesis.Significance: These findings show that lncRNA GLS-AS mediates a feedback loop of Myc and GLS, providing a potential therapeutic target for metabolic reprogramming in pancreatic cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/7/1398/F1.large.jpg.See related commentary by Mafra and Dias, p. 1302.


Subject(s)
Pancreatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Animals , Glutaminase , Mice , Mice, Nude , Nutrients
5.
Oncogene ; 37(44): 5811-5828, 2018 11.
Article in English | MEDLINE | ID: mdl-29970904

ABSTRACT

The contribution of long noncoding RNAs (lncRNAs) to pancreatic cancer progression and the regulatory mechanisms of their expression are attractive areas. In the present study, the overexpression of lncRNA-BX111887 (BX111) in pancreatic cancer tissues was detected by microarray and further validated in a cohort of pancreatic cancer tissues. We further demonstrated that knockdown or overexpression of BX111 dramatically repressed or enhanced proliferation and invasion of pancreatic cancer cells. Mechanically, BX111 activated transcription of ZEB1, a key regulator for epithelia-mesenchymal transition (EMT), via recruiting transcriptional factor Y-box protein (YB1) to its promoter region. Moreover, we revealed that BX111 transcription was induced by hypoxia-inducible factor (HIF-1α) in response to hypoxia. In addition, BX111 contributed to the hypoxia-induced EMT of pancreatic cells by regulating expression of ZEB1 and its downstream proteins E-cadherin and MMP2. Coincidence with in vitro results, BX111 depletion effectively inhibited growth and metastasis of xenograft tumor in vivo. The clinical samples of pancreatic cancer further confirmed a positive association between BX111 and ZEB1. Moreover, high BX111 expression was correlated with late TNM stage, lymphatic invasion and distant metastasis, as well as short overall survival time in patients. Taken together, our findings implicate a hypoxia-induced lncRNA contributes to metastasis and progression of pancreatic cancer, and suggest BX111 might be applied as a potential biomarker and therapeutic target for pancreatic cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , RNA, Long Noncoding/physiology , Zinc Finger E-box-Binding Homeobox 1/genetics , Animals , Cell Hypoxia , Cell Line, Tumor , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/physiopathology , Pancreatic Neoplasms/secondary , RNA, Long Noncoding/metabolism , Transcription, Genetic , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism
6.
J Pathol ; 245(3): 349-360, 2018 07.
Article in English | MEDLINE | ID: mdl-29708271

ABSTRACT

Metastasis-associated protein 2 (MTA2) is overexpressed in certain malignancies, and plays important roles in tumour metastasis and progression. The present study highlights the function of MTA2 in pancreatic carcinoma through its role as a deacetylator of hypoxia-inducible factor-1α (HIF-1α) and a cotranscriptional factor for E-cadherin expression. We found that overexpression of MTA2 promoted, and knockdown of MTA2 inhibited, the invasion and proliferation of pancreatic carcinoma cells both in vitro and in xenograft models in vivo. We also found that MTA2 is transcriptionally upregulated by HIF-1α through a hypoxia response element (HRE) of the MTA2 promoter in response to hypoxia. Reciprocally, MTA2 deacetylates HIF-1α and enhances its stability through interacting with histone deacetylase 1 (HDAC1). Consequently, HIF-1α recruits MTA2 and HDAC1 to the HRE of the E-cadherin promoter, by which E-cadherin transcription is repressed. In agreement with these experimental results, MTA2 is positively associated with HIF-1α, but inversely correlated with E-cadherin, in pancreatic carcinoma samples. Moreover, data from The Cancer Genome Atlas on 172 pancreatic carcinomas indicate an association between high expression of MTA2 and short overall survival. Taken together, our study identifies MTA2 as a critical hub and potential therapeutic target to inhibit the progression and metastasis of pancreatic carcinoma. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Adenocarcinoma/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Histone Deacetylases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pancreatic Neoplasms/metabolism , Repressor Proteins/metabolism , Acetylation , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Animals , Antigens, CD/genetics , Cadherins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/metabolism , Histone Deacetylases/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice, Inbred BALB C , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Processing, Post-Translational , Protein Stability , Repressor Proteins/genetics , Response Elements , Signal Transduction , Transcription, Genetic , Tumor Burden
7.
Oncogene ; 37(13): 1743-1758, 2018 03.
Article in English | MEDLINE | ID: mdl-29343850

ABSTRACT

Migration and invasion inhibitory protein (MIIP) is recently identified as an inhibitor in tumor development. However, the regulatory mechanism and biological contributions of MIIP in pancreatic cancer (PC) have been not elucidated. In this study, we demonstrated a negative feedback of MIIP and hypoxia-induced factor-1α (HIF-1α), which was mediated by a hypoxia-induced microRNA. Compared with paracarcinoma tissues, MIIP was downregulated in PC tissues. Overexpression of MIIP significantly impeded the proliferation and invasion of PC cells both in vitro and in mouse xenograft models. We further verified MIIP was downregulated under hypoxia in a HIF-1α-mediated manner. Interestingly, although MIIP promoter containing two putative hypoxia response elements (HREs), the chromatin immunoprecipitation (ChIP) and luciferase reporter assays did not support an active interaction between HIF-1α and MIIP promoter. Meanwhile, microRNA array revealed a hypoxia-induced microRNA, miR-646, impaired stability of MIIP mRNA and consequently inhibited its expression by targeting the coding sequence (CDS). Coincidently, knockdown of miR-646 significantly repressed proliferation and invasion ability of PC cells both in vitro and in vivo by upregulating MIIP expression. Besides, ChIP and luciferase reporter assays further validated that HIF-1α activated transcription of miR-646 in hypoxia condition. Therefore, these results suggested HIF-1α indirectly regulated MIIP expression in post-transcriptional level through upregulating miR-646 transcription. Conversely, our results further revealed that MIIP suppressed deacetylase ability of histone deacetylase 6 (HDAC6) to promote the acetylation and degradation of HIF-1α, by which impairing HIF-1α accumulation. What is more, a specific relationship between downregulated MIIP and upregulated miR-646 expression was validated in PC samples. Moreover, the dysregulated miR-646 and MIIP expression was correlated with advanced tumor stage, lymphatic invasion, metastasis and shorter overall survival in PC patients. Together, our results highlight that the reciprocal loop of HIF-1α/miR-646/MIIP might be implemented as an applicable target for pancreatic cancer therapy.


Subject(s)
Carcinogenesis/genetics , Carrier Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MicroRNAs/physiology , Pancreatic Neoplasms/genetics , Adult , Aged , Animals , Cell Proliferation/genetics , Cells, Cultured , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Tumor Hypoxia/genetics
8.
Cell Death Dis ; 9(2): 133, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29374159

ABSTRACT

Cancer cells predominantly produce energy at a high rate of glycolysis even in aerobic environment. It is termed as Warburg effect and is necessary for the tumorigenesis. Studies showed pyruvate kinase M2 (PKM2), a key regulator of the Warburg effect, is overexpressed and involved in numerous cancers. However, the expression and function of PKM2 in pancreatic cancer (PC) remain undefined. Our results showed that PKM2 is overexpressed in the PC tissue compared to the peritumoral tissue. Unexpected, the downregulation of PKM2 did not affect the proliferation, invasion, and chemoresistance of PC cells. Since pancreatic cancer is a hypovascular tumor with comparably insufficient energy supply, we further investigate the relationship between PKM2 and hypoglucose. Interestingly, we further discovered that decreased expression of PKM2 was detected in PC samples with lower microvessel density as well as in PC cells treated with hypoglucose condition (0.5 mM). Furthermore, the downregulation of PKM2 facilitated, while the upregulation of PKM2 inhibited, PC cells survival during hypoglucose. We further revealed that the repressed PKM2 induced autophagy, high NADPH/NADP ratio, and biomacromolecule production, but reduced ROS accumulation. Moreover, AMPKα1 knockdown repressed the autophagy and survival of PC cells during hypoglucose, which were promoted by PKM2 knockdown. Collectively, our study indicates that decreased PKM2 diverts glucose metabolism to biomacromolecule accumulation and antioxidants generation during glucose deprivation. This metabolism alteration elevates AMPKα1-dependent autophagy, which facilitates PC cell survival during glucose deprivation. Therefore, functions of PKM2 are complicated and cannot be defined as oversimplified promoter or inhibitor in PC.


Subject(s)
Carrier Proteins/metabolism , Glucose/pharmacology , Membrane Proteins/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Thyroid Hormones/metabolism , Adenylate Kinase/metabolism , Autophagy/drug effects , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lactic Acid/biosynthesis , Membrane Proteins/genetics , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Thyroid Hormones/genetics , Tumor Microenvironment/drug effects , Up-Regulation/drug effects , Thyroid Hormone-Binding Proteins
9.
Cell Death Dis ; 8(5): e2806, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28518134

ABSTRACT

Extracellular acid can have important effects on cancer cells. Acid-sensing ion channels (ASICs), which emerged as key receptors for extracellular acidic pH, are differently expressed during various diseases and have been implicated in underlying pathogenesis. This study reports that ASIC1 and ASIC3 are mainly expressed on membrane of pancreatic cancer cells and upregulated in pancreatic cancer tissues. ASIC1 and ASIC3 are responsible for an acidity-induced inward current, which is required for elevation of intracellular Ca2+ concentration ([Ca2+]i). Inhibition of ASIC1 and ASIC3 with siRNA or pharmacological inhibitor significantly decreased [Ca2+]i and its downstream RhoA during acidity and, thus, suppressed acidity-induced epithelial-mesenchymal transition (EMT) of pancreatic cancer cells. Meanwhile, downregulating [Ca2+]i with calcium chelating agent BAPTA-AM or knockdown of RhoA with siRNA also significantly repressed acidity-induced EMT of pancreatic cancer cells. Significantly, although without obvious effect on proliferation, knockdown of ASIC1 and ASIC3 in pancreatic cancer cells significantly suppresses liver and lung metastasis in xenograft model. In addition, ASIC1 and ASIC3 are positively correlated with expression of mesenchymal marker vimentin, but inversely correlated with epithelial marker E-cadherin in pancreatic cancer cells. In conclusion, this study indicates that ASICs are master regulator of acidity-induced EMT. In addition, the data demonstrate a functional link between ASICs and [Ca2+]i/RhoA pathway, which contributes to the acidity-induced EMT.


Subject(s)
Acid Sensing Ion Channels/metabolism , Acids/pharmacology , Calcium/metabolism , Epithelial-Mesenchymal Transition/drug effects , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects , rhoA GTP-Binding Protein/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Female , Gene Knockdown Techniques , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Up-Regulation/drug effects
10.
Mol Cancer Ther ; 15(9): 2209-19, 2016 09.
Article in English | MEDLINE | ID: mdl-27353169

ABSTRACT

Hypoxic microenvironments contribute to the tumorigenesis of numerous cancers by regulating the expression of a subset of miRNAs called "hypoxiamiRs." However, the function and mechanism of these deregulated miRNAs in hypoxic microenvironments within pancreatic cancers remain undefined. This study demonstrates that miR-548an is significantly downregulated in pancreatic cancer tissues and correlates with increased tumor size, advanced TNM stage, distant metastasis, and poor prognosis. Moreover, the overexpression of miR-548an significantly inhibited the proliferation and invasion of pancreatic cancer cells in vitro and in vivo We further revealed that hypoxia-induced factor-1α (HIF-1α) induces the downregulation of miR-548an in pancreatic cancer cells during hypoxia. Our co-IP and ChIP assays revealed that HIF-1α and histone deacetylase 1 (HDAC1) form a complex and bind to the hypoxia response elements (HRE) on the miR-548an promoter. In addition, inhibition of HDAC1 with trichostatin A antagonizes the suppression of miR-548 by hypoxia. Our dual luciferase assay validated that miR-548an directly binds to the 3' untranslated region of vimentin mRNA. The downregulation of vimentin suppresses the proliferation and invasion of pancreatic cancer cells in vitro and in vivo In addition, vimentin was inversely correlated with miR-548an expression in pancreatic cancer samples. In conclusion, our findings suggest that the HIF-1α-HDAC1 complex transcriptionally inhibits miR-548an expression during hypoxia, resulting in the upregulation of vimentin that facilitates the pancreatic tumorigenesis. Mol Cancer Ther; 15(9); 2209-19. ©2016 AACR.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/genetics , Vimentin/genetics , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease Models, Animal , Down-Regulation , Gene Knockdown Techniques , Humans , Male , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , RNA Interference , Transcription, Genetic , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(5): 6000-14, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26755660

ABSTRACT

Recent studies indicate that long non-coding RNAs (lncRNAs) play crucial roles in numerous cancers, while their function in pancreatic cancer is rarely elucidated. The present study identifies a functional lncRNA and its potential role in tumorigenesis of pancreatic cancer. Microarray co-assay for lncRNAs and mRNAs demonstrates that lncRNA-NUTF2P3-001 is remarkably overexpressed in pancreatic cancer and chronic pancreatitis tissues, which positively correlates with KRAS mRNA expression. After downregulating lncRNA-NUTF2P3-001, the proliferation and invasion of pancreatic cancer cell are significantly inhibited both in vitro and vivo, accompanying with decreased KRAS expression. The dual-luciferase reporter assay further validates that lncRNA-NUTF2P3-001 and 3'UTR of KRAS mRNA competitively bind with miR-3923. Furthermore, miR-3923 overexpression simulates the inhibiting effects of lncRNA-NUTF2P3-001-siRNA on pancreatic cancer cell, which is rescued by miR-3923 inhibitor. Specifically, the present study further reveals that lncRNA-NUTF2P3-001 is upregulated in pancreatic cancer cells under hypoxia and CoCl2 treatment, which is attributed to the binding of hypoxia-inducible factor-1α (HIF-1α) to hypoxia response elements (HREs) in the upstream of KRAS promoter. Data from pancreatic cancer patients show a positive correlation between lncRNA-NUTF2P3-001 and KRAS, which is associated with advanced tumor stage and worse prognosis. Hence, our data provide a new lncRNA-mediated regulatory mechanism for the tumor oncogene KRAS and implicate that lncRNA-NUTF2P3-001 and miR-3923 can be applied as novel predictors and therapeutic targets for pancreatic cancer.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Long Noncoding/biosynthesis , Animals , Carcinogenesis/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Long Noncoding/genetics
12.
Oncotarget ; 6(37): 39661-75, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26498682

ABSTRACT

Recent evidences suggest that the acidic microenvironment might facilitate epithelial mesenchymal transition (EMT) of tumor cells, while the effects of acidity on EMT of pancreatic cancer (PC) remain undefined. The present study demonstrated that acidity suppressed miR-652 expression, which further promoted EMT process by absenting inhibition on the transcriptional factor ZEB1 expression. At first, we found that acidity remarkably enhanced invasion ability of PC cells accompanying with increased mesenchymal and decreased epithelial markers. Meanwhile, miRNAs-microarray showed that miR-652, the potential regulator of ZEB1, was distinctly decreased in acidity-treated PC cells. Furthermore, restoration of miR-652 reversed acidity-induced EMT by inhibiting ZEB1 expression, while miR-652 inhibitor induced EMT in normal PC cells through promoting ZEB1 expression. Nevertheless, knockdown of ZEB1 significantly suppressed acidity-induced EMT in PC cells, but ZEB1 overexpression rescued the EMT which was inhibited by miR-652 overexpression. The in vivo results showed that the tumor growth and liver metastasis were remarkably retarded by both miR-652 overexpression and ZEB1 knockdown. The clinical samples further revealed that miR-652 was decreased in PC tissues and antagonistically correlated with ZEB1 expression, associating with late tumor stage, lymphatic invasion and metastasis. In conclusion, our study indicated a novel acidity/miR-652/ZEB1/EMT axis in the tumorigenesis of PC.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Transcription Factors/genetics , Tumor Microenvironment/genetics , Acids/chemistry , Adult , Aged , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Humans , Hydrogen-Ion Concentration , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA Interference , RNAi Therapeutics/methods , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Xenograft Model Antitumor Assays/methods , Zinc Finger E-box-Binding Homeobox 1
13.
Cancer Lett ; 355(2): 184-91, 2014 Dec 28.
Article in English | MEDLINE | ID: mdl-25172416

ABSTRACT

Epithelial-mesenchymal transition (EMT) is supposed to be an inflammation induced response which may take a central role in tumorigenesis. Since recent evidence indicates that microRNAs may be involved in EMT, the present study set out to reveal the miRNA which might regulate the EMT in CP (chronic pancreatitis) and PC (pancreatic cancer) and its potential mechanism. Firstly, we provided evidence that both CP and PC tissues demonstrated active EMT profile. Consistently, miR-217 was obviously down-regulated in CP, PC and TGF-ß1 treated PC cells, while negatively correlated to its direct target SIRT1. Moreover, either ectopic expression of miR-217 or inhibition of SIRT1 remarkably induced mesenchymal to epithelial transition (MET) in TGF-ß1 treated PC cells. On the contrary, miR-217 inhibitor promoted EMT in PC cells but not in SIRT-knockdown PC cells. Clinical information from a cohort of 54 PC patients demonstrated that down-regulated miR-217 was positively correlated with late tumor stage, lymphatic invasion, vascular infiltration and distant metastasis. These results suggest that the overexpressed TGF-ß1 in inflammation triggers the deregulation of the miR-217-SIRT1 pathway and then promotes the EMT process, which might be involved in the tumorigenesis of PC. Additionally, miR-217 may function as a novel target and predictor for PC prevention and therapy.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatitis, Chronic/genetics , Sirtuin 1/genetics , Cell Movement/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/metabolism , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/pathology , Signal Transduction , Sirtuin 1/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...