Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
Mol Cancer Ther ; 12(11): 2569-80, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24013097

ABSTRACT

miRNAs are associated with various types of cancer due to their ability to affect expression of genes that modulate tumorigenesis. In this study, we explored the role of miR-141 in pancreatic cancer. The analysis of clinical characteristics showed that miR-141 was significantly downregulated in tissues and cell lines of pancreatic cancer. Moreover, the decreased miR-141 level was significantly associated with tumor size and TNM stage, as well as lymph node and distant metastasis. Meanwhile, both Kaplan-Meier and multivariate survival analysis showed decreased miR-141 were associated with overall survival. Overexpression of miR-141 in pancreatic cancer cells inhibited cell proliferation, clonogenicity, and invasion; induced G1 arrest and apoptosis; and enhanced chemosensitivity. To understand how miR-141 mediates the phenotype of pancreatic cancer cells, a bioinformatics tool was used to identify MAP4K4 as a potential target of miR-141. The Dual-Luciferase reporter gene assay showed that miR-141 binds directly to the 3'-untranslated region (3'UTR) of MAP4K4 to inhibit MAP4K4 expression. Western blot and quantitative real-time PCR (qRT-PCR) analyses revealed that MAP4K4 expression was inversely correlated with miR-141 expression both in pancreatic cancer samples and cell lines. Knockdown of MAP4K4 inhibited cell proliferation, clonogenicity, and invasion, induced G1 arrest and apoptosis, and enhanced chemosensitivity. In a nude mouse xenograft model, both overexpression of miR-141 and knockdown of MAP4K4 significantly repressed pancreatic cancer cell growth. Therefore, we conclude that miR-141 targets MAP4K4, acts as a tumor suppressor in pancreatic cancer cells, and may serve as a novel therapeutic agent for miRNA-based pancreatic cancer therapy.


Subject(s)
Cell Proliferation/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , 3' Untranslated Regions , Adult , Aged , Animals , Apoptosis/drug effects , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/pharmacology , Middle Aged , Molecular Targeted Therapy , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Xenograft Model Antitumor Assays
6.
PLoS One ; 8(9): e73803, 2013.
Article in English | MEDLINE | ID: mdl-24040078

ABSTRACT

Accumulating evidence indicates that microRNAs (miRNAs) are aberrantly expressed in human cancer and contribute to the tumorigenesis, but their roles in pancreatic cancer are still largely unknown. In this study, our data showed that miR-130b was significantly downregulated in 52 pairs of pancreatic cancer tissues and five cell lines. Furthermore, the deregulated miR-130b was correlated with worse prognosis, increased tumor size, late TNM stage, lymphatic invasion and distant metastasis. Multivariate analysis showed that miR-130b expression was a significant and independent prognostic predictor for pancreatic cancer patients. Functional studies indicated that the overexpression of miR-130b dramatically suppressed the proliferation of pancreatic cancer cells both in vitro and in vivo, which could be attributed to the induction of apoptosis and cell cycle arrest at S phase. Meanwhile, an overexpressed miR-130b remarkably inhibited the invasive ability of pancreatic cancer cells. Moreover, the dual luciferase assay revealed that STAT3 was directly targeted by miR-130b, which was further confirmed by the inverse expression of miR-130b and STAT3 in pancreatic cancer samples. Our findings suggested that miR-130b might have a considerable potential in prognosis identification and application of therapy for pancreatic cancer.


Subject(s)
Biomarkers, Tumor/genetics , Cell Proliferation , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , STAT3 Transcription Factor/genetics , 3' Untranslated Regions/genetics , Animals , Apoptosis/genetics , Blotting, Western , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Transplantation, Heterologous
7.
Pancreatology ; 13(2): 140-6, 2013.
Article in English | MEDLINE | ID: mdl-23561972

ABSTRACT

BACKGROUND: Nicotinamide (NAM), the precursor for the synthesis of NAD(+) and also an inhibitor of SIRT1, has been discovered to inhibit some types of cancer. However, little is known about the effects of NAM on pancreatic cancer cells. Since previous research showed that SIRT1 and K-Ras/Akt signaling acted as a promoter in tumorigenesis of pancreatic cancer, our present research set out to explore whether NAM inhibits proliferation and facilitates chemosensitivity in pancreatic cancer cells as well as the potential mechanisms involving SIRT1 and K-Ras/Akt pathway. METHODS: Cell viability was assessed by MTT assay, and apoptosis and cell cycle were measured by flow cytometry. Cell invasive ability was evaluated by matrigel invasion assays. The activity of SIRT1 was measured by the Fluor de Lys deacetylation assay. Expression levels of SIRT1, K-Ras, Phosphated Akt (P-Akt, Ser-473) and Akt were measured using western blot. In vivo tumor growth was performed in pancreatic cancer cells xenografts. RESULTS: NAM inhibited the proliferation of pancreatic cancer cells in a dose-dependent manner, and significantly induced apoptosis and cell cycle arrest in G2/M phase. Moreover, NAM obviously restrained cell invasive ability and increased the chemosensitivity. NAM significantly inhibited the activity of SIRT1 and decreased expression of SIRT1, K-Ras and P-Akt. Further, NAM prohibited proliferation and enhanced GEM antitumor activity in vivo. CONCLUSIONS: Our results implied that NAM might be a potential therapeutic agent for human pancreatic cancer treatment through downregulating SIRT1, K-Ras and P-Akt expression.


Subject(s)
Genes, ras/physiology , Niacinamide/pharmacology , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Neoplasms, Experimental , Niacinamide/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics
8.
World J Gastroenterol ; 19(13): 2044-52, 2013 Apr 07.
Article in English | MEDLINE | ID: mdl-23599623

ABSTRACT

AIM: To compare effects of different resuscitation fluid on microcirculation, inflammation, intestinal barrier and clinical results in severe acute pancreatitis (SAP). METHODS: One hundred and twenty patients with SAP were enrolled at the Pancreatic Disease Institute between January 2007 and March 2010. The patients were randomly treated with normal saline (NS group), combination of normal saline and hydroxyethyl starch (HES) (SH group), combination of normal saline, hydroxyethyl starch and glutamine (SHG group) in resuscitation. The ratio of normal saline to HES in the SH and SHG groups was 3:1. The glutamine (20% glutamine dipeptide, 100 mL/d) was supplemented into the resuscitation liquid in the SHG group. Complications and outcomes including respiratory and abdominal infection, sepsis, abdominal hemorrhage, intra-abdominal hypertension, abdominal compartment syndrome (ACS), renal failure, acute respiratory distress syndrome (ARDS), multiple organ dysfunction syndrome (MODS), operation intervention, length of intensive care unit stay, length of hospital stay, and mortality at 60 d were compared. Moreover, blood oxygen saturation (SpO2), gastric intramucosal pH value (pHi), intra-abdominal pressure (IAP), inflammation cytokines, urine lactulose/mannitol (L/M) ratio, and serum endotoxin were investigated to evaluate the inflammatory reaction and gut barrier. RESULTS: Compared to the NS group, patients in the SH and SHG groups accessed the endpoint more quickly (3.9 ± 0.23 d and 4.1 ± 0.21 d vs 5.8 ± 0.25 d, P < 0.05) with less fluid volume (67.26 ± 28.53 mL/kg/d, 61.79 ± 27.61 mL/kg per day vs 85.23 ± 21.27 mL/kg per day, P < 0.05). Compared to the NS group, incidence of renal dysfunction, ARDS, MODS and ACS in the SH and SHG groups was obviously lower. Furthermore, incidence of respiratory and abdominal infection was significantly decreased in the SH and SHG groups, while no significant difference in sepsis was seen. Moreover, less operation time was needed in the SH and SHG group than the NS group, but the difference was not significant. The mortality did not differ significantly among these groups. Blood SpO2 and gastric mucosal pHi in the SH and SHG groups increased more quickly than in the NS group, while IAP was significantly decreased in the SH and SHG group. Moreover, the serum tumor necrosis factor-α, interleukin-8 and C-reactive protein levels in the SH and SHG groups were obviously lower than in the NS group at each time point. Furthermore, urine L/M ratio and serum endotoxin were significantly lower in the SH group and further decreased in the SHG group. CONCLUSION: Results indicated that combination of normal saline, HES and glutamine are more efficient in resuscitation of SAP by relieving inflammation and sustaining the intestinal barrier.


Subject(s)
Fluid Therapy , Pancreatitis/therapy , Resuscitation/methods , Acute Disease , Adult , Capillaries , Cytokines/blood , Endotoxins/metabolism , Female , Glutamine/therapeutic use , Humans , Hydrogen-Ion Concentration , Hydroxyethyl Starch Derivatives/therapeutic use , Inflammation , Intestinal Mucosa/metabolism , Intestines/drug effects , Lactulose/urine , Male , Mannitol/urine , Microcirculation , Middle Aged , Sodium Chloride/therapeutic use , Treatment Outcome
9.
Mol Cancer Ther ; 12(1): 83-93, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23171948

ABSTRACT

miRNAs are small noncoding RNAs that participate in a variety of biologic processes, and dysregulation of miRNA is always associated with cancer development and progression. Aberrant expression of miR-148b has been found in some types of cancer, but its expression and potential biologic role in pancreatic cancer are still largely unknown. In this study, our data showed that miR-148b was significantly downregulated in 48 pairs of human pancreatic cancer tissues and five cell lines. Furthermore, the deregulated miR-148b was correlated with increased tumor size, late tumor-node-metastasis stage, lymphatic invasion, distant metastasis, and worse prognosis in pancreatic cancer. Functional studies indicated overexpression of miR-148b dramatically suppressed the growth of cancer cells, attributable to induction of apoptosis and cell-cycle arrest at S-phase. Meanwhile, miR-148b remarkably inhibited invasion and enhanced chemosensitivity of pancreatic cancer cells. Moreover, ectopic expression of miR-148b was able to inhibit tumorigenicity in nude mice. Further studies revealed that AMPKα1 might be the direct target gene of miR-148b, and overexpressed AMPKα1 inversely correlated with miR-148b in pancreatic cancer. Silencing of AMPKα1 with RNA interference inhibited the growth of pancreatic cancer cells in vitro and in vivo and also induced apoptosis, cell-cycle arrest, and inhibited invasion of cancer cells, which is consistent with the effects of miR-148b overexpression. In conclusion, miR-148b can inhibit cell proliferation, invasion, and enhance chemosensitivity of pancreatic cancer by targeting AMPKα1. Our present results implicate the potential effects of miR-148b on prognosis and treatment of pancreatic cancer.


Subject(s)
AMP-Activated Protein Kinases/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Pancreatic Neoplasms/enzymology , RNA Interference , AMP-Activated Protein Kinases/metabolism , Adult , Aged , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Cell Cycle Checkpoints , Cell Proliferation , Female , Fluorouracil/pharmacology , Gene Expression , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Genes, Tumor Suppressor , Humans , Inhibitory Concentration 50 , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...