Antitumor activity of Coptis chinensis rhizome extract against PANC-1 cells

  • Abstract
  • Similar Papers
Abstract
Translate article icon Translate Article Star icon
Take notes icon Take Notes

The rhizome of Coptis chinensis is known for its antibacterial, anti-inflammatory, and anticancer activities. This study aimed to investigate the anti-pancreatic cancer activity of C. chinensis rhizome and elucidate the molecular mechanism involved in such activity. The C. chinensis rhizome extract (CRE) significantly inhibited the cell viability of PANC-1 pancreatic cancer cells and arrested the cell cycle at the G0/G1 phase. Western-blot analysis revealed that CRE downregulated the protein level of Forkhead box M1 (FoxM1), an oncogenic transcription factor, and its downstream target proteins such as cyclin D1, c-Myc, and survivin. Berberine was identified as the main component in CRE, effectively reducing cell viability and FoxM1 expression. These findings indicate that CRE and berberine can exert anticancer activity by downregulating FoxM1 expression in pancreatic cancer cells and that berberine may partially contribute to the anti-pancreatic cancer properties of CRE. This study highlights that C. chinensis rhizome extract and its main ingredient, berberine, might have therapeutic potential against pancreatic cancer.

Similar Papers
  • Research Article
  • 10.1158/1538-7445.am2017-4405
Abstract 4405: A new energy restriction mimetic decreases survival and FOXM1 expression in pancreatic cancer cells
  • Jul 1, 2017
  • Cancer Research
  • Laura Scolaro + 1 more

Pancreatic cancer is a deadly disease with only 6% of the patients living longer than 5 years. Even if the disease is diagnosed when it is localized, the 5-year survival rate is approximately 19%. A characteristic of cancer cell metabolism is a shift from oxidative phosphorylation to aerobic glycolysis known as the Warburg effect. It is well known that calorie restriction (CR) is a powerful intervention that suppresses tumorigenesis in animal models by targeting glycolysis and oncogenic signaling pathways. Recent studies have shown that the metabolic responses characteristic of CR can be obtained with the use of energy restriction mimetics (ERM). The purpose of this study is to evaluate a new ERM, OSU-CG5 in altering growth of pancreatic cancer cells and nutrient-sensitive signaling pathways. Tumors including those of the pancreas have uncontrolled proliferation necessitating increased fuel demands by increasing glucose availability and altering metabolic signaling pathways. Since ERM have been reported to target glucose transport, we evaluated whether OSU-CG5 alters glucose transport (Glut) proteins and glucose uptake. Glut1, but not Glut2, 3, 4, and 6, were overexpressed in Panc-1 cells with a 6-fold higher expression compared to HPNE cells. Glut proteins were not altered in MiaPaca cells. OSU-CG5 did not alter Glut1 protein expression in either cell line. Silencing Glut1 also had no effect on the cytoxicity of OSU-CG5. OSU-CG5 did not alter glucose uptake when measured in different concentrations of glucose in the media. Cell survival, as measured with Pico green, was decreased after OSU-CG5 treatment with an IC 50 of 3uM when cultured in physiological concentration of 5 and 10mM of glucose. FOXM1 is unregulated in pancreatic cancer and a promoter of the Warburg effect. We found that OSU-CG5 significantly decreased expression of FOXM1 in Panc1 and MiaPaca cells. Our findings indicate that OSU-CG5 does not inhibit the growth of Panc1 and MiaPaca pancreatic cancer cells by decreasing glucose uptake but may target the FOXM1 signaling pathways. Citation Format: Laura Scolaro, Susan Lanza-Jacoby. A new energy restriction mimetic decreases survival and FOXM1 expression in pancreatic cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4405. doi:10.1158/1538-7445.AM2017-4405

  • Research Article
  • Cite Count Icon 6
  • 10.3748/wjg.v30.i21.2793
Thymoquinone affects hypoxia-inducible factor-1α expression in pancreatic cancer cells via HSP90 and PI3K/AKT/mTOR pathways.
  • Jun 7, 2024
  • World journal of gastroenterology
  • Zhan-Xue Zhao + 2 more

Pancreatic cancer (PC) is associated with some of the worst prognoses of all major cancers. Thymoquinone (TQ) has a long history in traditional medical practice and is known for its anti-cancer, anti-inflammatory, anti-fibrosis and antioxidant pharmacological activities. Recent studies on hypoxia-inducible factor-1α (HIF-1α) and PC have shown that HIF-1α affects the occurrence and development of PC in many aspects. In addition, TQ could inhibit the development of renal cancer by decreasing the expression of HIF-1α. Therefore, we speculate whether TQ affects HIF-1α expression in PC cells and explore the mechanism. To elucidate the effect of TQ in PC cells and the regulatory mechanism of HIF-1α expression. Cell counting kit-8 assay, Transwell assay and flow cytometry were performed to detect the effects of TQ on the proliferative activity, migration and invasion ability and apoptosis of PANC-1 cells and normal pancreatic duct epithelial (hTERT-HPNE) cells. Quantitative real-time polymerase chain reaction and western blot assay were performed to detect the expression of HIF-1α mRNA and protein in PC cells. The effects of TQ on the HIF-1α protein initial expression pathway and ubiquitination degradation in PANC-1 cells were examined by western blot assay and co-immunoprecipitation. TQ significantly inhibited proliferative activity, migration, and invasion ability and promoted apoptosis of PANC-1 cells; however, no significant effects on hTERT-HPNE cells were observed. TQ significantly reduced the mRNA and protein expression levels of HIF-1α in PANC-1, AsPC-1, and BxPC-3 cells. TQ significantly inhibited the expression of the HIF-1α initial expression pathway (PI3K/AKT/mTOR) related proteins, and promoted the ubiquitination degradation of the HIF-1α protein in PANC-1 cells. TQ had no effect on the hydroxylation and von Hippel Lindau protein mediated ubiquitination degradation of the HIF-1α protein but affected the stability of the HIF-1α protein by inhibiting the interaction between HIF-1α and HSP90, thus promoting its ubiquitination degradation. The regulatory mechanism of TQ on HIF-1α protein expression in PC cells was mainly to promote the ubiquitination degradation of the HIF-1α protein by inhibiting the interaction between HIF-1α and HSP90; Secondly, TQ reduced the initial expression of HIF-1α protein by inhibiting the PI3K/AKT/mTOR pathway.

  • Research Article
  • Cite Count Icon 32
  • 10.1620/tjem.215.149
Selective Inhibition of Cyclooxygenase-2 Suppresses the Growth of Pancreatic Cancer Cells in Vitro and in Vivo
  • Jan 1, 2008
  • The Tohoku Journal of Experimental Medicine
  • Xuan-Fu Xu + 6 more

Cyclooxygenase-2 (COX-2), a prostaglandin synthetase, is involved in development of certain tumors. We therefore analyzed COX-2 expression in pancreatic cancer tissues (53 samples) and Panc-1 human pancreatic cancer cells by immunohistochemistry, RT-PCR and western-blotting analyses. Also, immunohistochemistry of proliferating cell nuclear antigen (PCNA) was performed. We found expression of COX-2 was dramatically upregulated in 36 of 53 cases (67.9%) and the expression of COX-2 was associated with the diameter (> 3 cm) of the tumors (p < 0.05), but not with the age, gender, tumor location, differentiation, lymph-node metastases and TNM stage. The positivity rate of PCNA expression in the pancreatic cancer cells of the COX-2 positive group (32.88 +/- 13.26%) was significantly higher than that in the COX-2 negative group (24.56 +/- 11.51%) (p < 0.05). Then we investigated the effect of selective inhibitors of COX-2 (NS398 and celecoxib) on proliferation of Panc-1 cells by 3-(4,5 dimethyl-2-thiazolyl)-2.5-diphenyl-2H-tetrazolium bromide (MTT) assay. Either NS398 or celecoxib suppressed proliferation of Panc-1 cells dose-dependently in vitro. Furthermore, Panc-1 cells were implanted into nude mice, and celecoxib was administrated orally with feed. The volume of the tumor xenografted into nude mice was decreased by 51.6% in the celecoxib group (p < 0.01). In conclusion, the increased expression of COX-2 may be responsible for rapid proliferation of pancreatic cancer, and specific inhibition of COX-2 suppresses proliferation of Panc-1 cells in vitro and in nude mice. The selective inhibitor of COX-2 may be an effectual agent for pancreatic cancer chemoprevention.

  • Research Article
  • Cite Count Icon 35
  • 10.1016/j.abb.2020.108469
LINC00261 inhibits progression of pancreatic cancer by down-regulating miR-23a-3p
  • Jun 24, 2020
  • Archives of Biochemistry and Biophysics
  • Xin Wang + 6 more

LINC00261 inhibits progression of pancreatic cancer by down-regulating miR-23a-3p

  • Research Article
  • Cite Count Icon 99
  • 10.4161/cbt.6.7.4328
IL-6 stimulates Th2 type cytokine secretion and upregulates VEGF and NRP-1 expression in pancreatic cancer cells
  • Jul 11, 2007
  • Cancer Biology & Therapy
  • Louis W Feurino + 9 more

BACKGROUND. Although upregulation of interleukin-6 (IL-6) is associated with many solid tumors, its role in pancreatic cancer has not been well elucidated. In this study, we examined the expression of IL-6 in pancreatic cancer cells, and determined the effect of exogenous IL-6 on cytokine secretion, gene expression, and signaling in human pancreatic cancer cells. METHODS. The mRNA levels of IL-6, VEGF165, neuropilin-1 (NRP-1), and neuropilin-2 (NRP-2) were determined by real-time RT PCR. Phosphorylation of ERK2 in pancreatic cancer cells was determined by using Bio-Plex phosphoprotein assay. The expression of IL-6 and other cytokines in human pancreatic cancer cell lines was determined with Bio-Plex cytokine assay. RESULTS. Pancreatic cancer cell lines expressed higher levels of IL-6 than normal human pancreatic ductal epithelium (HPDE) cells. Exogenous IL-6 increased the secretion of multiple Th2 type of cytokines in Panc-1, MIA PaCa-2, and BxPC-3 cells. IL-6 also upregulated the expression of VEGF165, and NRP-1, and both IL-6 and VEGF165 were inducible by hypoxia. In addition, IL-6 activated ERK2 signaling pathways in pancreatic cancer cells. CONCLUSIONS. IL-6 may be involved in promoting human pancreatic cancer development by furnishing Th2 type of cytokine environment and upregulating cell proliferation and angiogenesis related genes. Targeting IL-6 might be an effective treatment for pancreatic cancer.

  • Research Article
  • Cite Count Icon 33
  • 10.26355/eurrev_201812_16628
MiR-210 knockdown promotes the development of pancreatic cancer via upregulating E2F3 expression.
  • Dec 1, 2018
  • European review for medical and pharmacological sciences
  • Sun Fb + 5 more

The aim of this study was to explore the role of microRNA-210 (miR-210) and E2F3 in the development of pancreatic cancer and to investigate the possible underlying mechanism. The expression level of miR-210 in pancreatic cancer tissues, para-cancerous tissues, and normal pancreatic tissues was detected by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). The correlation between miR-210 expression and pathological indicators of pancreatic cancer was analyzed. Meanwhile, the expression of miR-210 in pancreatic cancer cells and normal pancreatic ductal epithelial cells was detected by qRT-PCR. After transfection with miR-210 mimics and inhibitor, the viability and cell cycle of pancreatic cancer cells were detected by cell counting kit-8 (CCK-8) assay and flow cytometry, respectively. The binding condition of miR-210 and E2F3 was verified by Dual-Luciferase reporter gene assay. MiR-210 was lowly expressed in pancreatic cancer tissues than that of para-cancerous tissues. The expression of miR-210 was negatively correlated with TNM stage and tumor size of pancreatic cancer. In vitro experiments showed that the miR-210 was downregulated in pancreatic cancer cells than that of normal pancreatic ductal epithelial cells. Meanwhile, overexpression of miR-210 arrested cell cycle decreased cell viability and downregulated E2F3 expression in pancreatic cancer cells. Dual-Luciferase reporter gene assay indicated that E2F3 bound to mi-210. Further experiments confirmed that E2F3 was negatively regulated by miR-210. MiR-210 knockdown promotes cell proliferation by upregulating E2F3 expression, thereby promoting the progression of pancreatic cancer.

  • Research Article
  • Cite Count Icon 1
  • 10.1158/1538-7445.panca16-a41
Abstract A41: GPRC5A acts as a potent oncogene in pancreatic cancer
  • Dec 14, 2016
  • Cancer Research
  • Honglei Zhou + 7 more

Background: GPRC5A is an orphan G-protein coupled receptor acting as an oncogene in some cancers and as a tumor suppressor in other. Its role in pancreatic cancer has not been studied systematically. ELAV-like protein 1, also known as “HuR,” is an RNA binding protein that is encoded by the ELAVL1 gene. HuR post-transcriptionally regulates multiple genes, plays key roles in human malignancies, and is known to mediate the efficacy of gemcitabine. Our preliminary data indicated that, in pancreatic cells, GPRC5A transcription responded to gemcitabine treatment and that its mRNA likely interacted with HuR. Methods: We immunohistochemically analyzed GPRC5A protein expression in primary and metastatic pancreatic cancer samples. We examined the effects of GPRC5A on pancreatic cells’ ability to proliferate, migrate, and invade, and how gemcitabine modulates this ability. We also studied the interaction between GPRC5A and HuR and examined how it is affected by gemcitabine. Lastly, we used microarray expression analyses to identify the genes and pathways linked to GPRC5A in pancreatic cancer cells. Results: We analyzed public mRNA expression data from 675 human cancer cell lines and 10,609 samples from The Cancer Genome Atlas (TCGA): GPRC5A mRNA abundance in pancreatic cancer was the highest (cell lines) or second highest (TCGA) among all tissues and cancer types. We analyzed an independent set of 252 pancreatic normal and cancer samples: we found GPRC5A mRNA to be up-regulated in primary tumor samples compared to normal pancreas, and even further up-regulated in metastatic pancreatic cancer. We immunostained 208 cores (103 primary and metastatic pancreatic cancer samples): we found GPRC5A protein to be barely expressed in normal pancreatic ductal cells and highly expressed in both primary and metastatic pancreatic ductal cancer cells. We carried out in vitro studies of multiple pancreatic cancer cell lines: we found that increasing GPRC5A protein levels promoted pancreatic cancer cell growth and migration whereas siRNA-mediated knockdown of GPRC5A impaired proliferation and invasion. We found that gemcitabine treatment enhanced GPRC5A expression in pancreatic cancer cells. Conversely, siRNA-mediated knockdown of GPRC5A sensitized the cells to gemcitabine. We showed that HuR/ELAVL1 up-regulated GPRC5A protein in the first 18 hours following gemcitabine treatment by stabilizing GPRC5A’s mRNA through binding to at least on site in GPRC5A’s 3´UTR. Microarray analysis showed that GPRC5A overexpression impacts several signaling networks. Conclusions: Our results indicate that GPRC5A acts as an oncogene in pancreatic cancer. Unexpectedly, gemcitabine was found to increase GPRC5A's mRNA and protein levels. We showed that this increase is mediated by HuR, a known enabler of gemcitabine efficacy, through a direct interaction between HuR and GPRC5A’s mRNA. It appears that the sensitivity of pancreatic cancer cells to gemcitabine can be augmented through down-regulation of GPRC5A. Citation Format: Honglei Zhou, Aristeidis Telonis, Yi Jing, Masaya Jimbo, Fernando Blanco, Eric Londin, Jonathan Brody, Isidore Rigoutsos.{Authors}. GPRC5A acts as a potent oncogene in pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2016 May 12-15; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(24 Suppl):Abstract nr A41.

  • Research Article
  • Cite Count Icon 9
  • 10.1007/s13277-014-2315-0
Secreted protein acidic and rich in cysteine inhibits the growth of human pancreatic cancer cells with G1 arrest induction.
  • Jul 16, 2014
  • Tumor Biology
  • Zhengfa Mao + 7 more

Aberrant secreted protein acidic and rich in cysteine (SPARC) expression has been reported to play an important role in the tumor development. However, the pattern and the role of SPARC in pancreatic cancer remain largely unknown. Therefore, we further deciphered the role of SPARC played in pancreatic cancer. We first evaluated the SPARC expression in human pancreatic cancer tissues and pancreatic cancer cells. Then we forced expression and silenced SPARC expression in pancreatic cancer cell lines MIA PaCa2 and PANC-1, respectively, using lentivirus vectors. We characterized the stable cells in vitro. In this study, we found that SPARC expression was weak in cancer cells in specimens which negatively correlated with the expression level of phosphorylated pRB and poorer outcome. Moreover, our results demonstrated that SPARC negatively regulated pancreatic cell growth in vitro. Furthermore, we disclosed that the activation of p53 and p27(Kip1) may involve in the effect of SPARC on pancreatic cancer cells. SPARC is downregulated in pancreatic cancer cells and retards the growth of pancreatic cancer cell. Taken together, these results indicate SPARC may be a potential target for pancreatic cancer therapy.

  • Research Article
  • 10.7150/ijms.98252
Cadherin 23 is a prognostic marker of pancreatic cancer and promotes cell viability in floating culture conditions
  • Jan 1, 2025
  • International Journal of Medical Sciences
  • Chaohui Zhen + 9 more

Purpose: Pancreatic cancer has the worst prognosis of all common cancers worldwide. Cadherin plays important roles in cancer cell invasion and metastasis. This study investigated the role and mechanism of Cadherin 23 (CDH23) action in the viability of pancreatic cancer cells.Methods: We examined CDH23 expression in 70 surgical pancreatic cancer samples and examined relationships among the level of CDH23 expression, clinicopathological characteristics, and the prognosis of the pancreatic cancer patients. Furthermore, we silenced CDH23 expression in pancreatic cancer cell lines (Panc-1, SUIT-2, MIA PaCa-2, CFPAC-1, and Capan-2) and assessed the viability of these cells. CDH23 expression in pancreatic cancer patients and cell lines was examined using immunohistochemistry and western blotting.Results: High levels of CDH23 in pancreatic cancer patients led to shorter overall survival and correlated with local recurrence and distance metastasis. The viability of pancreatic cancer cells in floating culture conditions decreased sharply when CDH23 was silenced. The viability and migration of pancreatic cancer cells in monolayer culture conditions did not change when CDH23 was silenced. The level of phosphorylated AKT was significantly decreased in the CDH23 knockdown cells in floating culture conditions.Conclusion: High levels of CDH23 expression are correlated with a poor prognosis in pancreatic cancer and may serve as a novel prognostic marker.

  • Research Article
  • Cite Count Icon 57
  • 10.1007/s10620-015-4001-5
IL-6/STAT3 Plays a Regulatory Role in the Interaction Between Pancreatic Stellate Cells and Cancer Cells.
  • Jan 6, 2016
  • Digestive Diseases and Sciences
  • Shin Hamada + 4 more

Pancreatic stellate cells (PSCs) play a pivotal role in pancreatic fibrosis, a characteristic feature of pancreatic cancer. Although it is still controversial, previous studies have suggested that PSCs promote the progression of pancreatic cancer by regulating the cell functions of cancer cells. PSCs produce large amounts of IL-6, which promotes the accumulation of myeloid-derived suppressor cells via a signal transducers and activator of transcription 3 (STAT3)-dependent mechanism. But the role of IL-6/STAT3 pathway in the interaction between PSCs and pancreatic cancer cells remains largely unknown. To clarify the role of IL-6/STAT3 in the interaction between PSCs and cancer cells. Human pancreatic cancer cells (Panc-1 and SUIT-2 cells) were treated with conditioned medium of immortalized human PSCs (PSC-CM). The effects of PSC-CM and IL-6 neutralization on the mRNA expression profiles were examined using Agilent's microarray. Activation of STAT3 was assessed by Western blotting using an anti-phospho-specific antibody. Cellular migration was examined by a two-chamber assay. The expression of markers related to epithelial-mesenchymal transition (EMT) was assessed by real-time reverse transcription PCR. PSC-CM induced the activation of STAT3 in pancreatic cancer cells. Neutralization of IL-6 suppressed the PSC-CM-induced upregulation of genes including complement factor B, lipocalin, and chemokine (C-C motif) ligand 20. Inhibition of IL-6/STAT3 pathway by anti-IL-6 antibody or a STAT3 inhibitor (NSC74859) inhibited the PSC-CM-induced migration and the expression of EMT-related markers (Snail and cadherin-2) in pancreatic cancer cells. IL-6/STAT3 pathway regulates the PSC-induced EMT and alterations in gene expression in pancreatic cancer cells.

  • Research Article
  • Cite Count Icon 18
  • 10.1007/s13277-012-0434-z
CCN1 promotes tumorigenicity through Rac1/Akt/NF-κB signaling pathway in pancreatic cancer
  • Jul 3, 2012
  • Tumor Biology
  • Xuqing Wang + 8 more

Aberrant CCN1 expression has been reported to play an important role in the tumor development. However, the pattern and the role of CCN1 in pancreatic cancer remain largely unknown. Therefore, we further deciphered the role CCN1 played in pancreatic cancer. We first evaluated the CCN1 expression in human pancreatic cancer tissues and pancreatic cancer cells. Then we forced expression and silenced CCN1 expression in pancreatic cancer cell lines MIA PaCa2 and PANC-1 respectively, using lentivirus vectors. We characterized the stable cells in vitro and in vivo using a nude mouse xenograft model. In this study, we found that CCN1 expression was significantly higher in cancer specimens which positively correlated with the expression level of phosphorylated Akt and p65. and poorer outcome. Moreover, our results demonstrated that CCN1 positively regulated pancreatic cell growth in vitro and in vivo and helped cancer cells resist to tumor necrosis factor alpha-induced apoptosis. Furthermore, we disclosed that activation of CCN1/ras-related c3 botulinum toxin substrate 1 (Rac1)/V-akt murine thymoma viral oncogene homolog (Akt)/nuclear factor-kappa B pathway inhibited apoptosis in pancreatic cancer cells. CCN1 is upregulated in pancreatic cancer and promotes the survival of pancreatic cancer cells. Taken together, these results indicate that CCN1 may be a potential target for pancreatic cancer therapy.

  • Research Article
  • Cite Count Icon 1
  • 10.1158/1538-7445.am2015-5028
Abstract 5028: CXCL10 within the tumor microenvironment induces gemcitabine resistance in pancreatic cancer cells
  • Aug 1, 2015
  • Cancer Research
  • Daniel Delitto + 15 more

Background: The systemic treatment of pancreatic cancer (PC) is hindered by the rapid development of chemoresistance to current cytotoxic therapies. Mechanisms governing the development of chemoresistance remain poorly characterized, particularly with respect to contributions from the tumor microenvironment. Thus, the goal of this study was to identify novel mechanisms acting within the tumor microenvironment which lead to PC chemoresistance. Methods: Intratumoral soluble mediator concentrations from resected PC specimens (n = 26) as well as supernatants from co-cultures of primary tumor-associated pancreatic stellate cells (PSCs) and PC cells (n = 12) were evaluated using a panel of 41 growth factors, chemokines and cytokines. The effect of CXCL10, a highly expressed soluble mediator during co-culture, on viability, proliferation, and apoptosis of PC cells was evaluated with and without gemcitabine treatment. In addition, the contribution of CXCL10 on migration patterns of peripheral blood mononuclear cells (PBMCs) was assessed. Results: Co-culture of tumor-associated PSCs with PC cells revealed increased CXCL10 levels compared to either cell type cultured alone. In addition, high intratumoral CXCL10 concentrations correlated with reduced overall survival (HR 6.9; P = .006). While CXCL10 treatment had a small effect on the viability of PC cells, it led to significantly increased PC cell viability in the presence of gemcitabine. Further, gemcitabine treatment induced the expression of the CXCL10 receptor, CXCR3, and this induction of CXCR3 was associated with the absence of apoptotic markers in PC cells. Finally, constitutive expression of CXCL10 by PC cells preferentially led to the migration of regulatory immune cell subsets. Conclusion: Paracrine CXCL10 signaling between stromal, PC and immune cells may be responsible not only for chemoresistance to gemcitabine, but also the recruitment and potential polarization of regulatory immune cell subsets in the pancreatic cancer microenvironment. Citation Format: Daniel Delitto, Chelsey Perez, Brian S. Black, Heather L. Sorenson, Andrea E. Knowlton, Song Han, Dongyu Zhang, George A. Sarosi, Lyle L. Moldawer, Kevin E. Behrns, Chen Liu, Thomas J. George, Ryan M. Thomas, Jose G. Trevino, Shannon M. Wallet, Steven J. Hughes. CXCL10 within the tumor microenvironment induces gemcitabine resistance in pancreatic cancer cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5028. doi:10.1158/1538-7445.AM2015-5028

  • Research Article
  • Cite Count Icon 33
  • 10.2217/epi-2016-0175
MiR-377 reverses cancerous phenotypes of pancreatic cells via suppressing DNMT1 and demethylating tumor suppressor genes.
  • Jul 31, 2017
  • Epigenomics
  • Masoumeh Azizi + 6 more

The aim was to investigate the effect of miR-377 on DNMT1 expression and cancer phenotype in pancreatic cancer cells. Real-time PCR, luciferase assay, MTT and Annexin-PI staining were used. Decreased miR-377 and increased DNMT1 (verified as a target for mir-377) levels in pancreatic cancer tissues and cell lines in comparison with normal tissues was confirmed to be influenced by promoter methylation. Also hypermethylation of BNIP3, SPARC, TFPI2 and PENK promoters was observed in tumor samples but not in normal tissues which negatively correlated with their expression. Restoration of miR-377 resulted in a reduction of the expression of DNMT1 and reactivation of BNIP3 and SPARC genes via promoter demethylation. Furthermore, enhanced expression of miR-377 could significantly inhibit cell proliferation and induce apoptosis. Our findings showed that miR-377 through targeting DNMT1 could reduce DNA methylation of some tumor suppressor genes and restore their expression in pancreatic cancer cells.

  • Research Article
  • Cite Count Icon 30
  • 10.1016/j.amjsurg.2005.07.013
Effect of cyclophilin A on gene expression in human pancreatic cancer cells
  • Oct 17, 2005
  • The American Journal of Surgery
  • Min Li + 5 more

Effect of cyclophilin A on gene expression in human pancreatic cancer cells

  • Research Article
  • Cite Count Icon 77
  • 10.1016/j.bbrc.2010.04.100
Identification of sites subjected to serine/threonine phosphorylation by SGK1 affecting N-myc downstream-regulated gene 1 (NDRG1)/Cap43-dependent suppression of angiogenic CXC chemokine expression in human pancreatic cancer cells
  • Apr 21, 2010
  • Biochemical and Biophysical Research Communications
  • Yuichi Murakami + 8 more

Identification of sites subjected to serine/threonine phosphorylation by SGK1 affecting N-myc downstream-regulated gene 1 (NDRG1)/Cap43-dependent suppression of angiogenic CXC chemokine expression in human pancreatic cancer cells

More from: Arhiv za bioloske nauke
  • Research Article
  • 10.2298/abs250214008k
Korean red ginseng extracts alleviate skin damage in heat-stimulated skin cells by suppressing NF-κB activation
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Jeeyoung Kim + 4 more

  • Research Article
  • 10.2298/abs250702021b
Genetic association of dopamine beta-hydroxylase (DBH) rs72393728 and D-amino acid oxidase activator (DAOA) rs3918342 polymorphisms with bipolar disorder in northeastern Algeria
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Amina Bernou + 7 more

  • Open Access Icon
  • Research Article
  • 10.2298/abs250324010m
Diversity of myxobacteria isolated from Weizhou Island, Guangxi, and their potential biological activities
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Kangting Meng + 5 more

  • Research Article
  • 10.2298/abs250425016j
Antitumor activity of Coptis chinensis rhizome extract against PANC-1 cells
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Ji Jeong + 1 more

  • Research Article
  • 10.2298/abs250626025n
Methyl jasmonate elicitation improves antioxidant and antibacterial activities in Portulaca oleracea
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Laavanya Nyanasaigran + 3 more

  • Open Access Icon
  • Research Article
  • Cite Count Icon 1
  • 10.2298/abs250208006g
Assessment of facial and nasal phenotypes: Implications in forensic facial reconstruction
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Ankita Guleria + 2 more

  • Open Access Icon
  • Research Article
  • 10.2298/abs250114012i
The role of ADAM10 in the prediction of neoadjuvant chemoradiotherapy response in patients with locally advanced rectal cancer
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Andjelka Isakovic + 6 more

  • Open Access Icon
  • Research Article
  • 10.2298/abs241126001j
The bioherbicidal potential of hemlock water extracts
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Vladan Jovanovic + 6 more

  • Research Article
  • 10.2298/abs250903028a
Molecular characterization of a novel betanucleorhabdovirus infecting sugar beet in Iran
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Javad Abkhoo + 2 more

  • Research Article
  • 10.2298/abs250410015s
Diversity of fungi isolated from the carapace of the European pond turtle (Emys orbicularis, L. 1758) in South Banat, Serbia
  • Jan 1, 2025
  • Arhiv za bioloske nauke
  • Milos Stupar + 3 more

Save Icon
Up Arrow
Open/Close
  • Ask R Discovery Star icon
  • Chat PDF Star icon

AI summaries and top papers from 250M+ research sources.

Search IconWhat is the difference between bacteria and viruses?
Open In New Tab Icon
Search IconWhat is the function of the immune system?
Open In New Tab Icon
Search IconCan diabetes be passed down from one generation to the next?
Open In New Tab Icon