1
|
Klein SM, Bozko M, Toennießen A, Rangno D, Bozko P. High p53 Protein Level Is a Negative Prognostic Marker for Pancreatic Adenocarcinoma. Int J Mol Sci 2024; 25:12307. [PMID: 39596373 PMCID: PMC11594790 DOI: 10.3390/ijms252212307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most aggressive types of cancer. Among different mechanisms generally believed to be important for the development of cancer, aberrant regulation of the p53 protein is a well-known and common feature for many cancer entities. Our work aims to analyze the impact of p53 deregulation and proteins encoded by p53 target genes on the survival of patients suffering from pancreatic adenocarcinoma. We, therefore, focused on the analysis of the selected collective for the TP53 mutation status, the p53 protein level, their correlation, and possible impacts on the prognosis/survival. We compared and analyzed a set of 123 patients. We have extracted information regarding the TP53 mutation status, p53 protein levels, the level of proteins encoded by prominent p53 target genes, and information on the overall survival. Survival analyses were displayed by Kaplan-Meier plots, using the log-rank test, in order to check for statistical significance. Protein levels were compared using the Mann-Whitney Test. We did not find any statistically significant correlation between the TP53 mutation status and the survival of the patients. Moreover, we have not found any significant correlation between the protein amount of prominent p53 target genes and the patients' survival. However, we see a significant correlation between the p53 protein level in cancer samples and the overall survival of pancreatic adenocarcinoma patients: patients having tumors with a p53 protein level within the upper quartile of all measured cases show a significantly reduced survival compared to the rest of the patients. Thus, in pancreatic adenocarcinoma, the p53 protein level is a relevant marker for prognosis, and cancers having a high p53 protein amount show a shortened patients' survival. In contrast, for this cancer entity, the TP53 mutation status or the protein amount of prominent p53 target genes on their own seems not to have a significant impact on survival.
Collapse
Affiliation(s)
- Sebastian M. Klein
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- M3 Research Institute, University Tübingen, 72076 Tübingen, Germany
| | - Maria Bozko
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 02-106 Warsaw, Poland
| | | | - Dennis Rangno
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Przemyslaw Bozko
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Universität Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- M3 Research Institute, University Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
2
|
Bodea IC, Ciocan A, Zaharie FV, Bodea R, Graur F, Ursu Ș, Ciocan RA, Al Hajjar N. HER2 Overexpression in Periampullary Tumors According to Anatomical and Histological Classification-A Systematic Review. J Pers Med 2024; 14:463. [PMID: 38793045 PMCID: PMC11122564 DOI: 10.3390/jpm14050463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Pancreatic cancer is one of the most aggressive, heterogeneous, and fatal types of human cancer; therefore, more effective therapeutic drugs are urgently needed. Human epidermal growth factor receptor 2 (HER2) overexpression and amplification have been identified as a cornerstone in this pathology. The aim of this review is to identify HER2 membrane overexpression in relation to pancreatic cancer pathways that can be used in order to develop a targeted therapy. After searching the keywords, 174 articles were found during a time span of 10 years, between 2013 and 2023, but only twelve scientific papers were qualified for this investigation. The new era of biomolecular research found a significant relationship between HER2 overexpression and pancreatic cancer cells in 25-30% of cases. The variables are dependent on tumor-derived cells, with differences in receptor overexpression between PDAC (pancreatic ductal adenocarcinoma), BTC (biliary tract cancer), ampullary carcinoma, and PNETs (pancreatic neuroendocrine tumors). HER2 overexpression is frequently encountered in human pancreatic carcinoma cell lines, and the ERBB family is one of the targets in the near future of therapy, with good results in phase I, II, and III studies evaluating downregulation and tumor downstaging, respectively.
Collapse
Affiliation(s)
- Ioan Cătălin Bodea
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Andra Ciocan
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Florin Vasile Zaharie
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Raluca Bodea
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Florin Graur
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Ștefan Ursu
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| | - Răzvan Alexandru Ciocan
- Department of Surgery-Practical Abilities, “Iuliu Hațieganu” University of Medicine and Pharmacy, Marinescu Street, No. 23, 400337 Cluj-Napoca, Romania;
| | - Nadim Al Hajjar
- Department of Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania; (I.C.B.); (F.V.Z.); (F.G.); (Ș.U.); (N.A.H.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, Croitorilor Street, No. 19–21, 400162 Cluj-Napoca, Romania;
| |
Collapse
|
3
|
Long T, Li J, Yin T, Liu K, Wang Y, Long J, Wang J, Cheng L. A genetic variant in gene NDUFAF4 confers the risk of non-small cell lung cancer by perturbing hsa-miR-215 binding. Mol Carcinog 2024; 63:145-159. [PMID: 37787384 DOI: 10.1002/mc.23642] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023]
Abstract
Hsa-microRNA-215 (hsa-miR-215) plays multiple roles in carcinogenesis through regulating its target genes. Genetic variants in hsa-miR-215 target sites thus may affect hsa-miR-215-mRNA interactions, result in altered expression of target genes and even influence cancer susceptibility. This study aimed to investigate the associations of genetic variants which located in the binding sites of hsa-miR-215 with non-small cell lung cancer (NSCLC) susceptibility in the Chinese population and reveal the potential regulatory mechanism of functional variants in NSCLC development. The candidate genetic variants were predicted and screened through bioinformatics analysis based on the degree of complementarity of hsa-miR-215 sequences. The potential effects of genetic variants on the binding ability of hsa-miR-215 and target genes were also predicted. A case-control study with 932 NSCLC patients and 1036 healthy controls was conducted to evaluate the association of candidate genetic variants with NSCLC susceptibility, and an independent case-control study with 552 NSCLC cases and 571 controls were used to further validate the promising associations. Dual luciferase reporter gene assay was applied to explore the regulation of the genetic variants on transcription activity of target gene. Cell phenotyping experiments in vitro and RNA sequencing (RNA-seq) were then carried out to preliminarily explore the potential regulatory mechanisms of the target genes in NSCLC. A total of five candidate genetic variants located in the binding sites of hsa-miR-215 were screened. The two-stage case-control study showed that a variant rs1854268 A > T, which located in the 3' untranslated (3'UTR) region of NDUFAF4 gene, was associated with decreased risk of NSCLC (additive model, odds ratio [OR] = 0.83, 95% confidence interval [CI]: 0.75-0.92, p < 0.001). Functional annotation displayed that rs1854268 A > T might downregulate the expression of NDUFAF4 by enhancing the binding affinity of hsa-miR-215-5p to NDUFAF4 mRNA. Additionally, transient knockdown of the NDUFAF4 could inhibit lung cancer cell migration and promote lung cancer cell apoptosis. Further RNA-seq analysis revealed that the knockdown of NDUFAF4 may affect NSCLC development by downregulating the nuclear factor kappa B (NF-κB) and phosphoinositide 3 kinase-AKT (PI3K-AKT) signaling pathways. Moreover, the overexpression of CCND1 could partially attenuate the effects of NDUFAF4 knock down on lung cancer cell migration and apoptosis, indicating that CCND1 may be involved in the tumor-promoting effects of NDUFAF4 as a downstream molecule of NDUFAF4 gene. In conclusion, the genetic variant rs1854268 (A > T) on NDUFAF4 confers NSCLC susceptibility by altering the binding affinity of hsa-miR-215-5p, thus regulating the expression of NDUFAF4 and subsequently influencing downstream tumor molecules and pathways such as CCND1, NF kappa B, and PI3K-AKT signaling pathways.
Collapse
Affiliation(s)
- Tingting Long
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaoyuan Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tongxin Yin
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Liu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jieyi Long
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianing Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Inkoom A, Ndemazie NB, Smith T, Frimpong E, Bulusu R, Poku R, Zhu X, Han B, Trevino J, Agyare E. Biological evaluation of novel gemcitabine analog in patient-derived xenograft models of pancreatic cancer. BMC Cancer 2023; 23:435. [PMID: 37179357 PMCID: PMC10182601 DOI: 10.1186/s12885-023-10928-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023] Open
Abstract
Gemcitabine (Gem) has been a standard first-line drug for pancreatic cancer (PCa) treatment; however, Gem's rapid metabolism and systemic instability (short half-life) limit its clinical outcome. The objective of this study was to modify Gem into a more stable form called 4-(N)-stearoyl-gemcitabine (4NSG) and evaluate its therapeutic efficacy in patient-derived xenograft (PDX) models from PCa of Black and White patients.Methods 4NSG was synthesized and characterized using nuclear magnetic resonance (NMR), elemental analysis, and high-performance liquid chromatography (HPLC). 4NSG-loaded solid lipid nanoparticles (4NSG-SLN) were developed using the cold homogenization technique and characterized. Patient-derived pancreatic cancer cell lines labeled Black (PPCL-192, PPCL-135) and White (PPCL-46, PPCL-68) were used to assess the in vitro anticancer activity of 4NSG-SLN. Pharmacokinetics (PK) and tumor efficacy studies were conducted using PDX mouse models bearing tumors from Black and White PCa patients.Results 4NSG was significantly stable in liver microsomal solution. The effective mean particle size (hydrodynamic diameter) of 4NSG-SLN was 82 ± 6.7 nm, and the half maximal inhibitory concentration (IC50) values of 4NSG-SLN treated PPCL-192 cells (9 ± 1.1 µM); PPCL-135 (11 ± 1.3 µM); PPCL-46 (12 ± 2.1) and PPCL-68 equaled to 22 ± 2.6 were found to be significantly lower compared to Gem treated PPCL-192 (57 ± 1.5 µM); PPCL-135 (56 ± 1.5 µM); PPCL-46 (56 ± 1.8 µM) and PPCL-68 (57 ± 2.4 µM) cells. The area under the curve (AUC), half-life, and pharmacokinetic clearance parameters for 4NSG-SLN were 3-fourfold higher than that of GemHCl. For in-vivo studies, 4NSG-SLN exhibited a two-fold decrease in tumor growth compared with GemHCl in PDX mice bearing Black and White PCa tumors.Conclusion 4NSG-SLN significantly improved the Gem's pharmacokinetic profile, enhanced Gem's systemic stability increased its antitumor efficacy in PCa PDX mice bearing Black and White patient tumors.
Collapse
Affiliation(s)
- Andriana Inkoom
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Nkafu Bechem Ndemazie
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Taylor Smith
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Esther Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Rosemary Poku
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Xue Zhu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Bo Han
- Department of Surgery, Keck School of Medicine University of Southern California, Los Angeles, California, 90033, USA
| | - Jose Trevino
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- Department of Surgery, College of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA.
| |
Collapse
|
5
|
Kuo YC, Kou HW, Hsu CP, Lo CH, Hwang TL. Identification and Clinical Significance of Pancreatic Cancer Stem Cells and Their Chemotherapeutic Drug Resistance. Int J Mol Sci 2023; 24:ijms24087331. [PMID: 37108495 PMCID: PMC10138402 DOI: 10.3390/ijms24087331] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Pancreatic cancer ranks in the 10th-11th position among cancers affecting men in Taiwan, besides being a rather difficult-to-treat disease. The overall 5-year survival rate of pancreatic cancer is only 5-10%, while that of resectable pancreatic cancer is still approximately 15-20%. Cancer stem cells possess intrinsic detoxifying mechanisms that allow them to survive against conventional therapy by developing multidrug resistance. This study was conducted to investigate how to overcome chemoresistance and its mechanisms in pancreatic cancer stem cells (CSCs) using gemcitabine-resistant pancreatic cancer cell lines. Pancreatic CSCs were identified from human pancreatic cancer lines. To determine whether CSCs possess a chemoresistant phenotype, the sensitivity of unselected tumor cells, sorted CSCs, and tumor spheroid cells to fluorouracil (5-FU), gemcitabine (GEM), and cisplatin was analyzed under stem cell conditions or differentiating conditions. Although the mechanisms underlying multidrug resistance in CSCs are poorly understood, ABC transporters such as ABCG2, ABCB1, and ABCC1 are believed to be responsible. Therefore, we measured the mRNA expression levels of ABCG2, ABCB1, and ABCC1 by real-time RT-PCR. Our results showed that no significant differences were found in the effects of different concentrations of gemcitabine on CSCs CD44+/EpCAM+ of various PDAC cell line cultures (BxPC-3, Capan-1, and PANC-1). There was also no difference between CSCs and non-CSCs. Gemcitabine-resistant cells exhibited distinct morphological changes, including a spindle-shaped morphology, the appearance of pseudopodia, and reduced adhesion characteristics of transformed fibroblasts. These cells were found to be more invasive and migratory, and showed increased vimentin expression and decreased E-cadherin expression. Immunofluorescence and immunoblotting experiments demonstrated increased nuclear localization of total β-catenin. These alterations are hallmarks of epithelial-to-mesenchymal transition (EMT). Resistant cells showed activation of the receptor protein tyrosine kinase c-Met and increased expression of the stem cell marker cluster of differentiation (CD) 24, CD44, and epithelial specific antigen (ESA). We concluded that the expression of the ABCG2 transporter protein was significantly higher in CD44+ and EpCAM+ CSCs of PDAC cell lines. Cancer stem-like cells exhibited chemoresistance. Gemcitabine-resistant pancreatic tumor cells were associated with EMT, a more aggressive and invasive phenotype of numerous solid tumors. Increased phosphorylation of c-Met may also be related to chemoresistance, and EMT and could be used as an attractive adjunctive chemotherapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Yu-Chi Kuo
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Hao-Wei Kou
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Chih-Po Hsu
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Chih-Hong Lo
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Tsann-Long Hwang
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| |
Collapse
|
6
|
Zhu X, Yu R, Peng Y, Miao Y, Jiang K, Li Q. Identification of genomic instability related lncRNA signature with prognostic value and its role in cancer immunotherapy in pancreatic cancer. Front Genet 2022; 13:990661. [PMID: 36118868 PMCID: PMC9481284 DOI: 10.3389/fgene.2022.990661] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/08/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Increasing evidence suggested the critical roles of lncRNAs in the maintenance of genomic stability. However, the identification of genomic instability-related lncRNA signature (GILncSig) and its role in pancreatic cancer (PC) remains largely unexplored. Methods: In the present study, a systematic analysis of lncRNA expression profiles and somatic mutation profiles was performed in PC patients from The Cancer Genome Atlas (TCGA). We then develop a risk score model to describe the characteristics of the model and verify its prediction accuracy. ESTIMATE algorithm, single-sample gene set enrichment analysis (ssGSEA), and CIBERSORT analysis were employed to reveal the correlation between tumor immune microenvironment, immune infiltration, immune checkpoint blockade (ICB) therapy, and GILncSig in PC. Results: We identified 206 GILnc, of which five were screened to develop a prognostic GInLncSig model. Multivariate Cox regression analysis and stratified analysis revealed that the prognostic value of the GILncSig was independent of other clinical variables. Receiver operating characteristic (ROC) analysis suggested that GILncSig is better than the existing lncRNA-related signatures in predicting survival. Additionally, the prognostic performance of the GILncSig was also found to be favorable in patients carrying wild-type KRAS, TP53, and SMAD4. Besides, a nomogram exhibited appreciable reliability for clinical application in predicting the prognosis of patients. Finally, the relationship between the GInLncSig model and the immune landscape in PC reflected its application value in clinical immunotherapy. Conclusion: In summary, the GILncSig identified by us may serve as novel prognostic biomarkers, and could have a crucial role in immunotherapy decisions for PC patients.
Collapse
Affiliation(s)
- Xiaole Zhu
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rong Yu
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yunpeng Peng
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Miao
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kuirong Jiang
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu, China
- *Correspondence: Kuirong Jiang, ; Qiang Li,
| | - Qiang Li
- Pancreas Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Pancreas Institute, Nanjing Medical University, Nanjing, Jiangsu, China
- *Correspondence: Kuirong Jiang, ; Qiang Li,
| |
Collapse
|
7
|
The E3 Ubiquitin Ligase Fbxo4 Functions as a Tumor Suppressor: Its Biological Importance and Therapeutic Perspectives. Cancers (Basel) 2022; 14:cancers14092133. [PMID: 35565262 PMCID: PMC9101129 DOI: 10.3390/cancers14092133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Fbxo4 is an E3 ubiquitin ligase that requires the formation of a complex with S-phase kinase-associated protein 1 and Cullin1 to catalyze the ubiquitylation of its substrates. Moreover, Fbxo4 depends on the existence of posttranslational modifications and/or co-factor to be activated to perform its biological functions. The well-known Fbxo4 substrates have oncogenic or oncogene-like activities, for example, cyclin D1, Trf1/Pin2, p53, Fxr1, Mcl-1, ICAM-1, and PPARγ; therefore, Fbxo4 is defined as a tumor suppressor. Biologically, Fbxo4 regulates cell cycle progression, DNA damage response, tumor metabolism, cellular senescence, metastasis and tumor cells’ response to chemotherapeutic compounds. Clinicopathologically, the expression of Fbxo4 is associated with patients’ prognosis depending on different tumor types. Regarding to its complicated regulation, more in-depth studies are encouraged to dissect the detailed molecular mechanisms to facilitate developing new treatment through targeting Fbxo4. Abstract Fbxo4, also known as Fbx4, belongs to the F-box protein family with a conserved F-box domain. Fbxo4 can form a complex with S-phase kinase-associated protein 1 and Cullin1 to perform its biological functions. Several proteins are identified as Fbxo4 substrates, including cyclin D1, Trf1/Pin2, p53, Fxr1, Mcl-1, ICAM-1, and PPARγ. Those factors can regulate cell cycle progression, cell proliferation, survival/apoptosis, and migration/invasion, highlighting their oncogenic or oncogene-like activities. Therefore, Fbxo4 is defined as a tumor suppressor. The biological functions of Fbxo4 make it a potential candidate for developing new targeted therapies. This review summarizes the gene and protein structure of Fbxo4, the mechanisms of how its expression and activity are regulated, and its substrates, biological functions, and clinicopathological importance in human cancers.
Collapse
|
8
|
Ghareyazi A, Mohseni A, Dashti H, Beheshti A, Dehzangi A, Rabiee HR, Alinejad-Rokny H. Whole-Genome Analysis of De Novo Somatic Point Mutations Reveals Novel Mutational Biomarkers in Pancreatic Cancer. Cancers (Basel) 2021; 13:4376. [PMID: 34503185 PMCID: PMC8431675 DOI: 10.3390/cancers13174376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
It is now known that at least 10% of samples with pancreatic cancers (PC) contain a causative mutation in the known susceptibility genes, suggesting the importance of identifying cancer-associated genes that carry the causative mutations in high-risk individuals for early detection of PC. In this study, we develop a statistical pipeline using a new concept, called gene-motif, that utilizes both mutated genes and mutational processes to identify 4211 3-nucleotide PC-associated gene-motifs within 203 significantly mutated genes in PC. Using these gene-motifs as distinguishable features for pancreatic cancer subtyping results in identifying five PC subtypes with distinguishable phenotypes and genotypes. Our comprehensive biological characterization reveals that these PC subtypes are associated with different molecular mechanisms including unique cancer related signaling pathways, in which for most of the subtypes targeted treatment options are currently available. Some of the pathways we identified in all five PC subtypes, including cell cycle and the Axon guidance pathway are frequently seen and mutated in cancer. We also identified Protein kinase C, EGFR (epidermal growth factor receptor) signaling pathway and P53 signaling pathways as potential targets for treatment of the PC subtypes. Altogether, our results uncover the importance of considering both the mutation type and mutated genes in the identification of cancer subtypes and biomarkers.
Collapse
Affiliation(s)
- Amin Ghareyazi
- Bioinformatics and Computational Biology Laboratory, Sharif University of Technology, Tehran 11365, Iran; (A.G.); (A.M.); (H.D.)
| | - Amir Mohseni
- Bioinformatics and Computational Biology Laboratory, Sharif University of Technology, Tehran 11365, Iran; (A.G.); (A.M.); (H.D.)
| | - Hamed Dashti
- Bioinformatics and Computational Biology Laboratory, Sharif University of Technology, Tehran 11365, Iran; (A.G.); (A.M.); (H.D.)
| | - Amin Beheshti
- Department of Computing, Macquarie University, Sydney, NSW 2109, Australia;
| | - Abdollah Dehzangi
- Department of Computer Science, Rutgers University, Camden, NJ 08102, USA;
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA
| | - Hamid R. Rabiee
- Bioinformatics and Computational Biology Laboratory, Sharif University of Technology, Tehran 11365, Iran; (A.G.); (A.M.); (H.D.)
| | - Hamid Alinejad-Rokny
- BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
- UNSW Data Science Hub, The University of New South Wales, Sydney, NSW 2052, Australia
- Health Data Analytics Program, AI-Enabled Processes (AIP) Research Centre, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
9
|
Zhang Y, Wang D, Lv B, Hou X, Liu Q, Liao C, Xu R, Zhang Y, Xu F, Zhang P. Oleic Acid and Insulin as Key Characteristics of T2D Promote Colorectal Cancer Deterioration in Xenograft Mice Revealed by Functional Metabolomics. Front Oncol 2021; 11:685059. [PMID: 34434893 PMCID: PMC8381473 DOI: 10.3389/fonc.2021.685059] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers with high mortality worldwide. Type 2 diabetes mellitus (T2D), known as a risk factor of CRC, can promote the deterioration of CRC, but the underlying mechanism is elusive. In this study, we aimed to reveal the relationship between CRC and T2D from the perspective of small-molecule metabolism. First, a list of common dysregulated metabolites in CRC and T2D was obtained by retrieving existing metabolomics publications. Among these metabolites, oleic acid (OA) was found to be able to promote the proliferation and migration of colon carcinoma cell HCT116. Further experiments proved that insulin could significantly strengthen this promotion and showed a synergistic effect with OA. Mechanism study found that OA and insulin acted synergistically through the extracellular signal-regulated kinase (ERK)1/2/c-Myc/cyclin D1 pathway. In addition, the combination of ERK1/2 inhibitor SCH772984 and cyclin-dependent kinase (CDK)4/6 inhibitor palbociclib showed a remarkable inhibitory effect on tumor growth in vivo. Taken together, the current study found that OA plays an important role in CRC development by using a functional metabolomics approach. More importantly, insulin and OA were confirmed to synergistically promote the deterioration of CRC in vitro and in vivo via ERK1/2/c-Myc/cyclin D1 pathway. Our findings may shed light on CRC treatment among the T2D population.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Di Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Bo Lv
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Xiaoying Hou
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Qiwei Liu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Chuyao Liao
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Ruijie Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Yuxin Zhang
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
10
|
MicroRNA-1252-5p, regulated by Myb, inhibits invasion and epithelial-mesenchymal transition of pancreatic cancer cells by targeting NEDD9. Aging (Albany NY) 2021; 13:18924-18945. [PMID: 34314382 PMCID: PMC8351675 DOI: 10.18632/aging.203344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/23/2021] [Indexed: 01/14/2023]
Abstract
MicroRNAs (miRNAs) are known to be involved in the development and progression of pancreatic cancer (PAC). The expression levels and roles of miR-1252-5p in PAC remain unclear. Quantitative real-time PCR and in situ hybridization were used to detect miR-1252-5p expression in PAC cells and human tissues. We studied the gain and loss of function of miR-1252-5p in the PAC cell lines in vitro and in vivo. The direct targets of miR-1252-5p were analyzed using public databases and a dual-luciferase reporter assay. Expression levels of miR-1252-5p are downregulated in PAC cell lines and tissue samples, and its expression is negatively associated with adverse clinical features and poor prognosis. In vitro and in vivo experiments show that miR-1252-5p overexpression inhibits the proliferation, migration, invasion, and epithelial-mesenchymal transition of PAC cells, and miR-1252-5p knockdown enhances these biological behaviors. MiR-1252-5p negatively regulates neural precursor cell expressed, developmentally downregulated 9 (NEDD9) by directly binding its 3'- untranslated region. Further mechanism research revealed that the SRC/STAT3 pathway is involved in miR-1252-5p/NEDD9 mediation of PAC's biological behaviors. We also verified that Myb inhibited miR-1252-5p by directly binding at its promoter. MiR-1252-5p may act as a tumor-suppressing miRNA in PAC and may be a potential therapeutic target for PAC patients.
Collapse
|
11
|
Lan HR, Wu ZQ, Zhang LH, Jin KT, Wang SB. Nanotechnology Assisted Chemotherapy for Targeted Cancer Treatment: Recent Advances and Clinical Perspectives. Curr Top Med Chem 2021; 20:2442-2458. [PMID: 32703133 DOI: 10.2174/1568026620666200722110808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/05/2019] [Accepted: 12/20/2019] [Indexed: 12/30/2022]
Abstract
Nanotechnology has recently provided exciting platforms in the field of anticancer research with promising potentials for improving drug delivery efficacy and treatment outcomes. Nanoparticles (NPs) possess different advantages over the micro and bulk therapeutic agents, including their capability to carry high payloads of drugs, with prolonged half-life, reduced toxicity of the drugs, and increased targeting efficiency. The wide variety of nanovectors, coupled with different conjugation and encapsulation methods available for different theranostic agents provide promising opportunities to fine-tune the pharmacological properties of these agents for more effective cancer treatment methods. This review discusses applications of NPs-assisted chemotherapy in preclinical and clinical settings and recent advances in design and synthesis of different nanocarriers for chemotherapeutic agents. Moreover, physicochemical properties of different nanocarriers, their impacts on different tumor targeting strategies and effective parameters for efficient targeted drug delivery are discussed. Finally, the current approved NPs-assisted chemotherapeutic agents for clinical applications and under different phases of clinical trials are discussed.
Collapse
Affiliation(s)
- Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, China
| | - Zhi-Qiang Wu
- Department of Pharmacy, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, China
| | - Li-Hua Zhang
- Department of Colorectal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, China
| | - Ke-Tao Jin
- Department of Colorectal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, China
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, China
| |
Collapse
|
12
|
Xiong Y, Wang Y, Li T, Yu X, Zeng Y, Xiao G, Zhou F, Zhou Y. A novel function for cyclin D1 as a transcriptional role in oncogenesis and tumor development by ChIP-Seq and RNA-Seq. J Cancer 2021; 12:5181-5192. [PMID: 34335935 PMCID: PMC8317529 DOI: 10.7150/jca.52909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 06/19/2021] [Indexed: 12/28/2022] Open
Abstract
Introduction: Aberrations in cell cycle control is defined as one of the hallmarks of cancer, while cyclin D1 is an essential protein to cell cycle which promote G1 phase into S phase, and frequently overexpressed in many human cancers. However, new functions have been identified in transgenic mice models, including the transcription of genome, the development of chromosome instability and DNA repair. In this research, our aim is to find the function of cyclin D1 in transcription in human cancers. Methods: The correlation of the cyclin D1 expression levels and prognosis of cervical cancer patients were analyzed in tissue microarray (TMA) cohort. We chose C33A as our main research object. Using chromatin immunoprecipitation sequencing (ChIP-seq) coupled with RNA sequencing (RNA-seq), to find out the genes differentially expressed in C33A, cyclin D1 knock-in C33A and cyclin D1 knock-down C33A. Results: We found that upregulation of cyclin D1 was associated with shorter overall survival (OS) and disease-free survival (DFS). Functionally, we identified 422 genes differentially expressed through analysis of the results of ChIP-seq and RNA-seq. These genes are highly enriched in Gene Ontology categories and involve in diverse cellular functions via KEGG classification, including replication and repair, signal transduction, cell growth and death. Conclusion: These findings suggested that the expression of cyclin D1 was associated with the prognosis of patients with cervical cancer. Cyclin D1 can serve both to activate and downregulate gene expression as a transcriptional role directly binding with genome DNA, which means that cyclin D1 may be a key protein during oncogenesis and tumor development.
Collapse
Affiliation(s)
- Yudi Xiong
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yuan Wang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Tianqi Li
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xiaoyan Yu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yangyang Zeng
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Guohui Xiao
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fuxiang Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Radiation Oncology and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Ahmad RS, Eubank TD, Lukomski S, Boone BA. Immune Cell Modulation of the Extracellular Matrix Contributes to the Pathogenesis of Pancreatic Cancer. Biomolecules 2021; 11:biom11060901. [PMID: 34204306 PMCID: PMC8234537 DOI: 10.3390/biom11060901] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/07/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival rate of only 9%. PDAC is characterized by a dense, fibrotic stroma composed of extracellular matrix (ECM) proteins. This desmoplastic stroma is a hallmark of PDAC, representing a significant physical barrier that is immunosuppressive and obstructs penetration of cytotoxic chemotherapy agents into the tumor microenvironment (TME). Additionally, dense ECM promotes hypoxia, making tumor cells refractive to radiation therapy and alters their metabolism, thereby supporting proliferation and survival. In this review, we outline the significant contribution of fibrosis to the pathogenesis of pancreatic cancer, with a focus on the cross talk between immune cells and pancreatic stellate cells that contribute to ECM deposition. We emphasize the cellular mechanisms by which neutrophils and macrophages, specifically, modulate the ECM in favor of PDAC-progression. Furthermore, we investigate how activated stellate cells and ECM influence immune cells and promote immunosuppression in PDAC. Finally, we summarize therapeutic strategies that target the stroma and hinder immune cell promotion of fibrogenesis, which have unfortunately led to mixed results. An enhanced understanding of the complex interactions between the pancreatic tumor ECM and immune cells may uncover novel treatment strategies that are desperately needed for this devastating disease.
Collapse
Affiliation(s)
- Ramiz S. Ahmad
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA;
| | - Timothy D. Eubank
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Slawomir Lukomski
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Brian A. Boone
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA;
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
- Correspondence:
| |
Collapse
|
14
|
Abstract
The introduction of cyclin-dependent kinase 4/6 inhibitors (CKIs) has marked a major development in the standard treatment of advanced breast cancer. Extensive preclinical, translational and clinical research efforts into CKI agents are ongoing, and clinical application of this class of systemic anti-cancer therapy is anticipated to expand beyond metastatic breast cancer treatment. Emerging evidence indicates that mechanisms by which CKI agents exert their therapeutic effect transcend their initially expected impacts on cell cycle control into the realms of cancer immunology and metabolism. The recent expansion in our understanding of the multifaceted impact of CKIs on tumour biology has the potential to improve clinical study design, therapeutic strategies and ultimately patient outcomes. This review contextualises the current status of CKI therapy by providing an overview of the original and emerging insights into mechanisms of action and the evidence behind their current routine use in breast cancer management. Recent preclinical and clinical studies into CKIs across tumour types are discussed, including a synthesis of the more than 300 clinical trials of CKI-combination treatments registered as of November 2020. Key challenges and opportunities anticipated in the 2020s are explored, including treatment resistance, combination therapy strategies and potential biomarker development.
Collapse
|
15
|
Attiyeh MA, Amini A, Chung V, Melstrom LG. Multidisciplinary management of locally advanced pancreatic adenocarcinoma: Biology is King. J Surg Oncol 2021; 123:1395-1404. [PMID: 33831247 DOI: 10.1002/jso.26415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 12/21/2022]
Abstract
The annual incidence of pancreatic cancer is nearly 50,000 patients. The 5-year overall survival is only 9%, and there remains a great need for better therapy. A subset of these patients presents with locally advanced disease. Multidisciplinary therapy has evolved to include some combination of systemic chemotherapy, locoregional radiation, and surgery in select patients with excellent biology. This review will address the thoughtful evidence-based and individualized approach to these patients.
Collapse
Affiliation(s)
- Marc A Attiyeh
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - Arya Amini
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - Vincent Chung
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California, USA
| | - Laleh G Melstrom
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
16
|
McGuigan AJ, Coleman HG, McCain RS, Kelly PJ, Johnston DI, Taylor MA, Turkington RC. Immune cell infiltrates as prognostic biomarkers in pancreatic ductal adenocarcinoma: a systematic review and meta-analysis. J Pathol Clin Res 2021; 7:99-112. [PMID: 33481339 PMCID: PMC7869931 DOI: 10.1002/cjp2.192] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/15/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022]
Abstract
Immune cell infiltration has been identified as a prognostic biomarker in several cancers. However, no immune based biomarker has yet been validated for use in pancreatic ductal adenocarcinoma (PDAC). We undertook a systematic review and meta-analysis of immune cell infiltration, measured by immunohistochemistry (IHC), as a prognostic biomarker in PDAC. All other IHC prognostic biomarkers in PDAC were also summarised. MEDLINE, EMBASE and Web of Science were searched between 1998 and 2018. Studies investigating IHC biomarkers and prognosis in PDAC were included. REMARK score and Newcastle-Ottawa scale were used for qualitative analysis. Random-effects meta-analyses were used to pool results, where possible. Twenty-six articles studied immune cell infiltration IHC biomarkers and PDAC prognosis. Meta-analysis found high infiltration with CD4 (hazard ratio [HR] = 0.65, 95% confidence interval [CI] = 0.51-0.83.) and CD8 (HR = 0.68, 95% CI = 0.55-0.84.) T-lymphocytes associated with better disease-free survival. Reduced overall survival was associated with high CD163 (HR = 1.62, 95% CI = 1.03-2.56). Infiltration of CD3, CD20, FoxP3 and CD68 cells, and PD-L1 expression was not prognostic. In total, 708 prognostic biomarkers were identified in 1101 studies. In summary, high CD4 and CD8 infiltration are associated with better disease-free survival in PDAC. Increased CD163 is adversely prognostic. Despite the publication of 708 IHC prognostic biomarkers in PDAC, none has been validated for clinical use. Further research should focus on reproducibility of prognostic biomarkers in PDAC in order to achieve this.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Biomarkers/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/pathology
- Disease-Free Survival
- Humans
- Immunohistochemistry
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/pathology
- Prognosis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Reproducibility of Results
- Pancreatic Neoplasms
Collapse
Affiliation(s)
- Andrew J McGuigan
- The Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastBelfastUK
| | - Helen G Coleman
- The Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastBelfastUK
- Centre for Public HealthQueen's University BelfastBelfastUK
| | - R Stephen McCain
- Centre for Public HealthQueen's University BelfastBelfastUK
- Department of Hepatobiliary SurgeryMater Hospital, Belfast Health and Social Care TrustBelfastUK
| | - Paul J Kelly
- Department of Tissue PathologyRoyal Victoria Hospital, Belfast Health and Social Care TrustBelfastUK
| | - David I Johnston
- Northern Ireland Cancer CentreBelfast Health and Social Care TrustBelfastUK
| | - Mark A Taylor
- Department of Hepatobiliary SurgeryMater Hospital, Belfast Health and Social Care TrustBelfastUK
| | - Richard C Turkington
- The Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastBelfastUK
| |
Collapse
|
17
|
Son M, Kim H, Han D, Kim Y, Huh I, Han Y, Hong SM, Kwon W, Kim H, Jang JY, Kim Y. A Clinically Applicable 24-Protein Model for Classifying Risk Subgroups in Pancreatic Ductal Adenocarcinomas using Multiple Reaction Monitoring-Mass Spectrometry. Clin Cancer Res 2021; 27:3370-3382. [PMID: 33593883 DOI: 10.1158/1078-0432.ccr-20-3513] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 01/12/2021] [Accepted: 02/12/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) subtypes have been identified using various methodologies. However, it is a challenge to develop classification system applicable to routine clinical evaluation. We aimed to identify risk subgroups based on molecular features and develop a classification model that was more suited for clinical applications. EXPERIMENTAL DESIGN We collected whole dissected specimens from 225 patients who underwent surgery at Seoul National University Hospital [Seoul, Republic of Korea (South)], between October 2009 and February 2018. Target proteins with potential relevance to tumor progression or prognosis were quantified with robust quality controls. We used hierarchical clustering analysis to identify risk subgroups. A random forest classification model was developed to predict the identified risk subgroups, and the model was validated using transcriptomic datasets from external cohorts (N = 700), with survival analysis. RESULTS We identified 24 protein features that could classify the four risk subgroups associated with patient outcomes: stable, exocrine-like; activated, and extracellular matrix (ECM) remodeling. The "stable" risk subgroup was characterized by proteins that were associated with differentiation and tumor suppressors. "Exocrine-like" tumors highly expressed pancreatic enzymes. Two high-risk subgroups, "activated" and "ECM remodeling," were enriched in terms such as cell cycle, angiogenesis, immunocompetence, tumor invasion metastasis, and metabolic reprogramming. The classification model that included these features made prognoses with relative accuracy and precision in multiple cohorts. CONCLUSIONS We proposed PDAC risk subgroups and developed a classification model that may potentially be useful for routine clinical implementations, at the individual level. This clinical system may improve the accuracy of risk prediction and treatment guidelines.See related commentary by Thakur and Singh, p. 3272.
Collapse
Affiliation(s)
- Minsoo Son
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Hongbeom Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Dohyun Han
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (South)
| | - Yoseop Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Iksoo Huh
- College of Nursing and Research Institute of Nursing Science, Seoul National University, Seoul, Republic of Korea (South)
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea (South)
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Haeryoung Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea (South).
| | - Youngsoo Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, Republic of Korea (South).
| |
Collapse
|
18
|
Machine-Learning Provides Patient-Specific Prediction of Metastatic Risk Based on Innovative, Mechanobiology Assay. Ann Biomed Eng 2021; 49:1774-1783. [PMID: 33483841 DOI: 10.1007/s10439-020-02720-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022]
Abstract
Cancer mortality is mostly related to metastasis. Metastasis is currently prognosed via histopathology, disease-statistics, or genetics; those are potentially inaccurate, not rapidly available and require known markers. We had developed a rapid (~ 2 h) mechanobiology-based approach to provide early prognosis of the clinical likelihood for metastasis. Specifically, invasive cell-subsets seeded on impenetrable, physiological-stiffness polyacrylamide gels forcefully indent the gels, while non-invasive/benign cells do not. The number of indenting cells and their attained depths, the mechanical invasiveness, accurately define the metastatic risk of tumors and cell-lines. Utilizing our experimental database, we compare the capacity of several machine learning models to predict the metastatic risk. Models underwent supervised training on individual experiments using classification from literature and commercial-sources for established cell-lines and clinical histopathology reports for tumor samples. We evaluated 2-class models, separating invasive/non-invasive (e.g. benign) samples, and obtained sensitivity and specificity of 0.92 and 1, respectively; this surpasses other works. We also introduce a novel approach, using 5-class models (i.e. normal, benign, cancer-metastatic-non/low/high) that provided average sensitivity and specificity of 0.69 and 0.91. Combining our rapid, mechanical invasiveness assay with machine learning classification can provide accurate and early prognosis of metastatic risk, to support choice of treatments and disease management.
Collapse
|
19
|
Xu XT, Chen J, Ren X, Ma YR, Wang X, Ma YY, Zhao DG, Zhou RP, Zhang K, Goodin S, Li DL, Zheng X. Effects of atorvastatin in combination with celecoxib and tipifarnib on proliferation and apoptosis in pancreatic cancer sphere-forming cells. Eur J Pharmacol 2020; 893:173840. [PMID: 33359649 DOI: 10.1016/j.ejphar.2020.173840] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/21/2022]
Abstract
Cancer stem cell (CSC) plays an important role in pancreatic cancer pathogenesis and treatment failure. CSCs are characterized by their ability to form tumor spheres in serum-free medium and expression of CSC related markers. In the present study, we investigated the effect atorvastatin, celecoxib and tipifarnib in combination on proliferation and apoptosis in Panc-1 sphere-forming cells. The sphere-forming cells were isolated from Panc-1 cells by sphere-forming method. These sphere-forming cells showed CSC properties. The levels of CD44, CD133 and ALDH1A1 in the sphere-forming cells were increased. Moreover, Panc-1 sphere-forming cells were resistant to chemotherapeutic drug gemcitabine. Combined atorvastatin with celecoxib and tipifarnib synergistically decreased the sphere forming ability of Panc-1 cells and the drug combination also strongly inhibited cell proliferation and promoted apoptosis in the sphere-forming cells. The effects of the drug combination on the Panc-1 sphere-forming cells were associated with decreases in the levels of CD44, CD133 and ALDH1A1, and suppression of Akt and NF-κB activation. Results of the present study indicate that the combination of atorvastatin, celecoxib and tipifarnib may represent an effective approach for inhibiting pancreatic CSCs.
Collapse
Affiliation(s)
- Xue-Tao Xu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529020, PR China
| | - Jie Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Xiang Ren
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Yu-Ran Ma
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Xiao Wang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China
| | - Yan-Yan Ma
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529020, PR China
| | - Den-Gao Zhao
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529020, PR China
| | - Ren-Ping Zhou
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529020, PR China
| | - Susan Goodin
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Dong-Li Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen, 529020, PR China
| | - Xi Zheng
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
20
|
Gulliver C, Hoffmann R, Baillie GS. The enigmatic helicase DHX9 and its association with the hallmarks of cancer. Future Sci OA 2020; 7:FSO650. [PMID: 33437516 PMCID: PMC7787180 DOI: 10.2144/fsoa-2020-0140] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Much interest has been expended lately in characterizing the association between DExH-Box helicase 9 (DHX9) dysregulation and malignant development, however, the enigmatic nature of DHX9 has caused conflict as to whether it regularly functions as an oncogene or tumor suppressor. The impact of DHX9 on malignancy appears to be cell-type specific, dependent upon the availability of binding partners and activation of inter-connected signaling pathways. Realization of DHX9's pivotal role in the development of several hallmarks of cancer has boosted the enzyme's potential as a cancer biomarker and therapeutic target, opening up novel avenues for exploring DHX9 in precision medicine applications. Our review discusses the ascribed functions of DHX9 in cancer, explores its enigmatic nature and potential as an antineoplastic target.
Collapse
Affiliation(s)
- Chloe Gulliver
- Institute of Cardiovascular & Medical Science, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Ralf Hoffmann
- Institute of Cardiovascular & Medical Science, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
- Philips Research Europe, High Tech Campus, Eindhoven, The Netherlands
| | - George S Baillie
- Institute of Cardiovascular & Medical Science, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
21
|
Abstract
Objective: We aimed to define preoperative clinical and molecular characteristics that would allow better patient selection for operative resection. Background: Although we use molecular selection methods for systemic targeted therapies, these principles are not applied to surgical oncology. Improving patient selection is of vital importance for the operative treatment of pancreatic cancer (pancreatic ductal adenocarcinoma). Although surgery is the only chance of long-term survival, 80% still succumb to the disease and approximately 30% die within 1 year, often sooner than those that have unresected local disease. Method: In 3 independent pancreatic ductal adenocarcinoma cohorts (total participants = 1184) the relationship between aberrant expression of prometastatic proteins S100A2 and S100A4 and survival was assessed. A preoperative nomogram based on clinical variables available before surgery and expression of these proteins was constructed and compared to traditional measures, and a postoperative nomogram. Results: High expression of either S100A2 or S100A4 was independent poor prognostic factors in a training cohort of 518 participants. These results were validated in 2 independent patient cohorts (Glasgow, n = 198; Germany, n = 468). Aberrant biomarker expression stratified the cohorts into 3 distinct prognostic groups. A preoperative nomogram incorporating S100A2 and S100A4 expression predicted survival and nomograms derived using postoperative clinicopathological variables. Conclusions: Of those patients with a poor preoperative nomogram score, approximately 50% of patients died within a year of resection. Nomograms have the potential to improve selection for surgery and neoadjuvant therapy, avoiding surgery in aggressive disease, and justifying more extensive resections in biologically favorable disease.
Collapse
|
22
|
Holm M, Saraswat M, Joenväärä S, Seppänen H, Renkonen R, Haglund C. Label-free proteomics reveals serum proteins whose levels differ between pancreatic ductal adenocarcinoma patients with short or long survival. Tumour Biol 2020; 42:1010428320936410. [PMID: 32586207 DOI: 10.1177/1010428320936410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is the most common and aggressive type of pancreatic cancer, with a 5-year survival rate that is less than 10%. New biomarkers to aid in predicting the prognosis of pancreatic ductal adenocarcinoma patients are needed. Previous proteomic studies have to a great extent focused on finding proteins of value for the diagnosis of pancreatic ductal adenocarcinoma. There is a lack of studies that have profiled the serum or plasma proteome in order to discover candidates for new prognostic biomarkers. In this study, we have used ultra-performance liquid chromatography-ultra-definition mass spectrometry to analyze the serum samples of 21 pancreatic ductal adenocarcinoma patients with short or long survival. Statistical analysis discovered 31 proteins whose expression differed significantly between pancreatic ductal adenocarcinoma patients with short or long survival. Pathway analysis discovered multiple canonical pathways enriched in this data set, with several pathways having roles in inflammation and lipid metabolism. The serum proteins identified here, which include complement components and several enzymes, could be of value as candidates for new noninvasive prognostic markers.
Collapse
Affiliation(s)
- Matilda Holm
- Department of Surgery, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Department of Pathology, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mayank Saraswat
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland.,HUSLAB, Helsinki University Hospital, Helsinki, Finland.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Sakari Joenväärä
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland.,HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Hanna Seppänen
- Department of Surgery, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Risto Renkonen
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland.,HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Cui SJ, Tang TY, Zou XW, Su QM, Feng L, Gong XY. Role of imaging biomarkers for prognostic prediction in patients with pancreatic ductal adenocarcinoma. Clin Radiol 2020; 75:478.e1-478.e11. [PMID: 32037002 DOI: 10.1016/j.crad.2019.12.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive tumours. PDAC has a poor prognosis; therefore, it is necessary to perform further risk stratification. Identifying prognostic factors before treatment might help to implement suitable and personalised treatment for individuals and avoid side effects. Conventional staging systems and tumour biomarkers are fundamental to establish prognosis; however, they have obvious limitations. Novel imaging biomarkers extracted from advanced imaging techniques offer opportunities to evaluate underlying tumour physiological characteristics, such as mutational status, cellular composition, local microenvironment, tumour metabolism, and biological behaviour. Thus, imaging biomarkers might help the decision making of oncologists and surgeons. The present review discusses the functions of imaging biomarkers for prognostic prediction in patients with PDAC and their potential value for further translation in clinical practice.
Collapse
Affiliation(s)
- S-J Cui
- The Second Clinical Medical College, Zhejiang Chinese Medical University, 310053, Hangzhou, China; Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 310013, Hangzhou, China
| | - T-Y Tang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - X-W Zou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Q-M Su
- Department of General Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - L Feng
- Department of Nuclear Medicine, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - X-Y Gong
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 310013, Hangzhou, China; Institute of Artificial Intelligence and Remote Imaging, Hangzhou Medical College, 310000, Hangzhou, China.
| |
Collapse
|
24
|
Cyclin D degradation by E3 ligases in cancer progression and treatment. Semin Cancer Biol 2020; 67:159-170. [PMID: 32006569 DOI: 10.1016/j.semcancer.2020.01.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/20/2020] [Accepted: 01/27/2020] [Indexed: 12/15/2022]
Abstract
D cyclins include three isoforms: D1, D2, and D3. D cyclins heterodimerize with cyclin-dependent kinase 4/6 (CDK4/6) to form kinase complexes that can phosphorylate and inactivate Rb. Inactivation of Rb triggers the activation of E2F transcription factors, which in turn regulate the expression of genes whose products drive cell cycle progression. Because D-type cyclins function as mitogenic sensors that link growth factor signaling directly with G1 phase progression, it is not surprising that D cyclin accumulation is dysregulated in a variety of human tumors. Elevated expression of D cyclins results from gene amplification, increased gene transcription and protein translation, decreased microRNA levels, and inefficiency or loss of ubiquitylation-mediated protein degradation. This review focuses on the clinicopathological importance of D cyclins, how dysregulation of Ubiquitin-Proteasome System (UPS) contributes to the overexpression of D cyclins, and the therapeutic potential through targeting D cyclin-related machinery in human tumors.
Collapse
|
25
|
Zhou L, Jiao X, Peng X, Yao X, Liu L, Zhang L. MicroRNA-628-5p inhibits invasion and migration of human pancreatic ductal adenocarcinoma via suppression of the AKT/NF-kappa B pathway. J Cell Physiol 2020; 235:8141-8154. [PMID: 31957029 DOI: 10.1002/jcp.29468] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022]
Abstract
The biological function and underlying mechanism of microRNA-628-5p (miR-628-5p) remains to be clarified in the growth and progression of pancreatic ductal adenocarcinoma (PDAC). Here, the expression levels of miR-628-5p in PDAC tissues and cells were detected by quantitative reverse transcriptase polymerase chain reaction and in situ hybridization. The relationship between miR-628-5p expression and clinicopathologic characteristics was examined in human PDAC tissue samples. Gain- and loss-of-function and the putative targets of miR-628-5p were evaluated in PDAC cell lines. The upstream and downstream signals of miR-628-5p in PDAC were also examined. MiR-628-5p was lowly expressed in PDAC tissues and cell lines, and low miR-628-5p expression in PDAC tissues was associated with poor clinicopathological characteristics and shorter overall survival. Functionally, restoration of miR-628-5p expression decreased PDAC cell proliferation, migration, invasion, and promoted cell apoptosis, whereas miR-628-5p silencing abolished these biological behaviors. MiR-628-5p was found to target and negatively regulate phospholipid scramblase 1 and insulin receptor substrate 1 expression, which resulted in the inhibition of the AKT/NF-κB signaling pathway. MYC knockdown led to miR-628-5p upregulation, whereas MYC overexpression repressed miR-628-5p expression. These findings indicate that miR-628-5p functions as a tumor-suppressive microRNA in PDAC and implicate miR-628-5p as a potential therapeutic target for PDAC patients.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoxiao Jiao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoqian Peng
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaomeng Yao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Liu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lianfeng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
26
|
Huang J, Liu J, Qiu L. Transient receptor potential vanilloid 1 promotes EGFR ubiquitination and modulates EGFR/MAPK signalling in pancreatic cancer cells. Cell Biochem Funct 2020; 38:401-408. [PMID: 31907951 DOI: 10.1002/cbf.3483] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022]
Abstract
Transient receptor potential vanilloid-1 (TRPV1) was first identified in sensory neurons, where it was suggested as a therapeutic target for pain relief. Here, we show that TRPV1 is expressed in the pancreatic cancer cell line, PANC-1; that epidermal growth factor receptor (EGFR) expression is downregulated by overexpression or agonist-induced activation of TRPV1; and conversely, that EGFR expression is increased after silencing TRPV1. Furthermore, TRPV1 overexpression inhibits cell proliferation and significantly reduces the mRNA levels of two oncogenes, KRAS and AKT2. More importantly, TRPV1 downregulates EGFR levels by inducing EGFR ubiquitination and degradation, which modulate EGFR/MAPK signalling in pancreatic cancer cells. These results illustrate the regulation and mechanism of TRPV1 on EGFR in pancreatic cancer cells and may provide new ideas for the design of novel antitumor drugs targeting EGFR. SIGNIFICANCE OF THE STUDY: We investigated the effect and mechanism of TRPV1 on EGFR-mediated proliferation and transformation of pancreatic cancer cells, with the aim of providing new ideas and experimental evidence for the application of strategies that promote EGFR degradation to treat pancreatic cancer.
Collapse
Affiliation(s)
- Jin Huang
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingxue Liu
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,College of Pharmacy, The Second Military Medical University, Shanghai, China
| | - Lei Qiu
- College of Pharmacy, The Second Military Medical University, Shanghai, China
| |
Collapse
|
27
|
Lv K, Yang J, Sun J, Guan J. Identification of key candidate genes for pancreatic cancer by bioinformatics analysis. Exp Ther Med 2019; 18:451-458. [PMID: 31281439 PMCID: PMC6580103 DOI: 10.3892/etm.2019.7619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/15/2019] [Indexed: 12/24/2022] Open
Abstract
Although pancreatic cancer has the highest mortality rate among all neoplasms worldwide, its exact mechanism remains poorly understood. In the present study, three Gene Expression Omnibus (GEO) datasets were integrated to elucidate the potential genes and pathways that contribute to the development of pancreatic cancer. Initially, a total of 226 differentially expressed genes (DEGs) were identified in the three GEO datasets, containing 179 upregulated and 47 downregulated DEGs. Furthermore, function and pathway enrichment analyses were performed to explore the function and pathway of these genes, and the results indicated that the DEGs participated in extracellular matrix (ECM) processes. In addition, a protein-protein interaction network was constructed and 163 genes of the 229 DEGs were filtered into the network, resulting in a network complex of 163 nodes and 438 edges. Finally, 24 hub genes were identified in the network, and the top 2 most significant modules were selected for function and pathway analysis. The hub genes were involved in several processes, including activation of matrix, degradation of ECM and ECM organization. Taken collectively, the data demonstrated potential key genes and pathways in pancreatic cancer, which may provide novel insights to the mechanism of pancreatic cancer. In addition, these hub genes and pathways may be considered as targets for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Kui Lv
- Department of Emergency Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Jianying Yang
- Department of Emergency Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Junfeng Sun
- Department of Emergency Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Jianguo Guan
- Department of Emergency Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
- Correspondence to: Dr Jianguo Guan, Department of Emergency Medicine, Anhui No. 2 Provincial People's Hospital, 1868 Danshan Road, Hefei, Anhui 230041, P.R. China, E-mail:
| |
Collapse
|
28
|
Tunçel D, Bayol NÜ. Pankreas duktal adenokarsinomunda NF-Kappa B ekspresyonu. CUKUROVA MEDICAL JOURNAL 2019. [DOI: 10.17826/cumj.481396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
29
|
Abstract
Cancers that appear morphologically similar often have dramatically different clinical features, respond variably to therapy and have a range of outcomes. Compelling evidence now demonstrates that differences in the molecular pathology of otherwise indistinguishable cancers substantially impact the clinical characteristics of the disease. Molecular subtypes now guide preclinical and clinical therapeutic development and treatment in many cancer types. The ability to predict optimal therapeutic strategies ahead of treatment improves overall patient outcomes, minimizing treatment-related morbidity and cost. Although clinical decision making based on histopathological criteria underpinned by robust data is well established in many cancer types, subtypes of pancreatic cancer do not currently inform treatment decisions. However, accumulating molecular data are defining subgroups in pancreatic cancer with distinct biology and potential subtype-specific therapeutic vulnerabilities, providing the opportunity to define a de novo clinically applicable molecular taxonomy. This Review summarizes current knowledge concerning the molecular subtyping of pancreatic cancer and explores future strategies for using a molecular taxonomy to guide therapeutic development and ultimately routine therapy with the overall goal of improving outcomes for this disease.
Collapse
Affiliation(s)
| | - Peter Bailey
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, Scotland, UK
| | - David K Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, Scotland, UK
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, Scotland, UK.
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
- South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, Australia.
| |
Collapse
|
30
|
Andleeb F, Hafeezullah, Atiq A, Atiq M, Malik S. Attenuated total reflectance spectroscopy to diagnose skin cancer and to distinguish different metastatic potential of melanoma cell. Cancer Biomark 2018; 23:373-380. [PMID: 30248045 DOI: 10.3233/cbm-181393] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Farah Andleeb
- Biophotoics Research Group, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
- Govt Sadiq College, Women University of Bahawalpur, Bahawalpur, Pakistan
| | - Hafeezullah
- Biophotoics Research Group, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Atia Atiq
- Biophotoics Research Group, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Maria Atiq
- Biophotoics Research Group, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Sadia Malik
- Biophotoics Research Group, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
31
|
Cen M, Yao Y, Cui L, Yang G, Lu G, Fang L, Bao Z, Zhou J. Honokiol induces apoptosis of lung squamous cell carcinoma by targeting FGF2-FGFR1 autocrine loop. Cancer Med 2018; 7:6205-6218. [PMID: 30515999 PMCID: PMC6308115 DOI: 10.1002/cam4.1846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/24/2018] [Accepted: 09/28/2018] [Indexed: 12/27/2022] Open
Abstract
Lung squamous cell carcinoma (SCC) accounts for a considerable proportion of lung cancer cases, but there is still a lack of effective therapies. FGFR1 amplification is generally considered a promising therapeutic target. Honokiol is a chemical compound that has been proven to be effective against various malignancies and whose analog has been reported to target the mitogen‐activated protein kinase family, members of a downstream signaling pathway of FGFR1. This was an explorative study to determine the mechanism of honokiol in lung SCC. We found that honokiol induced apoptosis and cell cycle arrest in lung SCC cell lines in a time‐ and dose‐dependent manner. Honokiol also restricted cell migration in lung SCC cell lines. Moreover, the expression of FGF2 and the activation of FGFR1 were both downregulated by honokiol. Pharmacological inhibition and siRNA knockdown of FGFR1 induced apoptosis in lung SCC cells. Our in vivo study indicated that honokiol could suppress the growth of xenograft tumors, and this effect was associated with the inhibition of the FGF2‐FGFR1 signaling pathway. In conclusion, honokiol induced cell apoptosis in lung SCC by targeting the FGF2‐FGFR1 autocrine loop.
Collapse
Affiliation(s)
- Mengyuan Cen
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yinan Yao
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Luyun Cui
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Guangdie Yang
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Guohua Lu
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Liangjie Fang
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhang Bao
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Diseases, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
32
|
Cerium Oxide Nanoparticles Sensitize Pancreatic Cancer to Radiation Therapy through Oxidative Activation of the JNK Apoptotic Pathway. Cancers (Basel) 2018. [DOI: 10.10.3390/cancers10090303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Side effects of radiation therapy (RT) remain the most challenging issue for pancreatic cancer treatment. Cerium oxide nanoparticles (CONPs) are currently being tested in pre-clinical trials as an adjuvant to sensitize pancreatic cancer cells to RT and protect normal tissues from the harmful side effects. CONPs were not able to significantly affect RT-induced DNA damage in cancer cells, thereby ruling out sensitization through increased mitotic catastrophe. However, activation of c-Jun terminal kinase (JNK), a key driver of RT-induced apoptosis, was significantly enhanced by co-treatment with CONPs and RT in pancreatic cancer cells in vitro and human pancreatic tumors in nude mice in vivo compared to CONPs or RT treatment alone. Further, CONP-driven increase in RT-induced JNK activity was associated with a marked increase in Caspase 3/7 activation, indicative of apoptosis. We have previously shown that CONPs increase reactive oxygen species (ROS) production in cancer cells. ROS has been shown to drive the oxidation of thioredoxin 1 (TRX1) which results in the activation of apoptosis signaling kinase 1 (ASK1). The increase in ASK1 activation following the co-treatment with CONPs followed by RT suggests that the increased JNK activation is the result of increased TRX1 oxidation. The ability of CONPs to sensitize pancreatic cancer cells to RT was mitigated when the TRX1 oxidation was prevented by mutagenesis of a cysteine residue or when the JNK activation was blocked by an inhibitor. Taken together, these data demonstrate an important mechanism for CONPs in specifically killing cancer cells and provide novel insights into the utilization of CONPs as a radiosensitizer and therapeutic agent for pancreatic cancer.
Collapse
|
33
|
Wason MS, Lu H, Yu L, Lahiri SK, Mukherjee D, Shen C, Das S, Seal S, Zhao J. Cerium Oxide Nanoparticles Sensitize Pancreatic Cancer to Radiation Therapy through Oxidative Activation of the JNK Apoptotic Pathway. Cancers (Basel) 2018; 10:cancers10090303. [PMID: 30200491 PMCID: PMC6162528 DOI: 10.3390/cancers10090303] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 08/30/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
Side effects of radiation therapy (RT) remain the most challenging issue for pancreatic cancer treatment. Cerium oxide nanoparticles (CONPs) are currently being tested in pre-clinical trials as an adjuvant to sensitize pancreatic cancer cells to RT and protect normal tissues from the harmful side effects. CONPs were not able to significantly affect RT-induced DNA damage in cancer cells, thereby ruling out sensitization through increased mitotic catastrophe. However, activation of c-Jun terminal kinase (JNK), a key driver of RT-induced apoptosis, was significantly enhanced by co-treatment with CONPs and RT in pancreatic cancer cells in vitro and human pancreatic tumors in nude mice in vivo compared to CONPs or RT treatment alone. Further, CONP-driven increase in RT-induced JNK activity was associated with a marked increase in Caspase 3/7 activation, indicative of apoptosis. We have previously shown that CONPs increase reactive oxygen species (ROS) production in cancer cells. ROS has been shown to drive the oxidation of thioredoxin 1 (TRX1) which results in the activation of apoptosis signaling kinase 1 (ASK1). The increase in ASK1 activation following the co-treatment with CONPs followed by RT suggests that the increased JNK activation is the result of increased TRX1 oxidation. The ability of CONPs to sensitize pancreatic cancer cells to RT was mitigated when the TRX1 oxidation was prevented by mutagenesis of a cysteine residue or when the JNK activation was blocked by an inhibitor. Taken together, these data demonstrate an important mechanism for CONPs in specifically killing cancer cells and provide novel insights into the utilization of CONPs as a radiosensitizer and therapeutic agent for pancreatic cancer.
Collapse
Affiliation(s)
- Melissa S Wason
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL 32827, USA.
| | - Heng Lu
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL 32827, USA.
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | - Lin Yu
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL 32827, USA.
| | - Satadru K Lahiri
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL 32827, USA.
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Debarati Mukherjee
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL 32827, USA.
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Chao Shen
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL 32827, USA.
- Department of Microbiology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China.
| | - Soumen Das
- Department of Mechanical, Materials and Aerospace Engineering, Advanced Materials Processing and Analysis Center; Nanoscience and Nanotechnology Center, University of Central Florida, Orlando, FL 32816, USA.
| | - Sudipta Seal
- Department of Mechanical, Materials and Aerospace Engineering, Advanced Materials Processing and Analysis Center; Nanoscience and Nanotechnology Center, University of Central Florida, Orlando, FL 32816, USA.
| | - Jihe Zhao
- Burnett School of Biomedical Sciences University of Central Florida College of Medicine, Orlando, FL 32827, USA.
| |
Collapse
|
34
|
Sikdar N, Saha G, Dutta A, Ghosh S, Shrikhande SV, Banerjee S. Genetic Alterations of Periampullary and Pancreatic Ductal Adenocarcinoma: An Overview. Curr Genomics 2018; 19:444-463. [PMID: 30258276 PMCID: PMC6128383 DOI: 10.2174/1389202919666180221160753] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic Ductal AdenoCarcinoma (PDAC) is one of the most lethal malignancies of all solid cancers. Precancerous lesions for PDAC include PanIN, IPMNs and MCNs. PDAC has a poor prognosis with a 5-year survival of approximately 6%. Whereas Periampulary AdenoCarcinoma (PAC) having four anatomic subtypes, pancreatic, Common Bile Duct (CBD), ampullary and duodenum shows relative better prognosis. The highest incidence of PDAC has been reported with black with respect to white population. Similarly, incidence rate of PAC also differs with different ethnic populations. Several lifestyle, environmental and occupational exposures including long-term diabetes, obesity, and smoking, have been linked to PDAC, however, for PAC the causal risk factors were poorly described. It is now clear that PDAC and PAC are a multi-stage process resulting from the accumulation of genomic alterations in the somatic DNA of normal cells as well as inherited mutations. Approximately 10% of PDAC have a familial inheritance. Germline mutations in CDKN2A, BRCA2, STK11, PALB2, PRSS1, etc., as well as certain syndromes have been well associated with predisposition to PDAC. KRAS, CDKN2A, TP53 and SMAD4 are the 4 "mountains" (high-frequency driver genes) which have been known to earliest somatic alterations for PDAC while relatively less frequent in PAC. Our understanding of the molecular carcinogenesis has improved in the last few years due to extensive research on PDAC which was not well explored in case of PAC. The genetic alterations that have been identified in PDAC and different subgroups of PAC are important implications for the development of genetic screening test, early diagnosis, and prognostic genetic markers. The present review will provide a brief overview of the incidence and prevalence of PDAC and PAC, mainly, increased risk in India, the several kinds of risk factors associated with the diseases as well as required genetic alterations for disease initiation and progression.
Collapse
Affiliation(s)
- Nilabja Sikdar
- Address correspondence to this author at the Human Genetics Unit, Indian Statistical Institute, 203, B.T. Road Kolkata 700108, India; Tel (1): +91-33
-25773240 (L); (2): +91-9830780397 (M); Fax: +91 33 35773049;, E-mail:
| | | | | | | | | | | |
Collapse
|
35
|
Roger M, Martínez J, Peiró G, Aparicio JR, Ruiz F, Compañy L, Casellas JA. EUS-FNA cytological material from pancreatic lesions: the expression of cathepsins and its predictive value of malignancy. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2018; 110:446-450. [PMID: 29893579 DOI: 10.17235/reed.2018.4200/2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIMS To assess the expression of cathepsins in pancreatic samples obtained by endoscopic ultrasonography and fine needle aspiration (EUS-FNA) and to investigate their relationship with the staging of the pancreatic ductal adenocarcinoma (PDAC). METHODS We prospectively included patients with solid pancreatic masses, in which EUS-FNA were performed. Cathepsins B, L, S and H expression was determined in FNA samples. RESULTS Seventeen FNA were performed. All cytological material was from PDAC. Expression of cathepsins was predominantly low (B 65%, L 23%, S 76%, and H 41%). We found no correlation between the expression levels and the extension of the neoplasm. CONCLUSION Expression of cathepsins in the cytological material of PDAC is diverse but still poor to be useful in the pre-operative diagnosis. There is no correlation between the expression levels of cathepsins and the extension of the PDAC.
Collapse
Affiliation(s)
- Manuela Roger
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante, España
| | - Juan Martínez
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante
| | - Gloria Peiró
- Servicio de Anatomía Patológica, Hospital General Universitario de Alicante
| | | | - Francisco Ruiz
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante, España
| | - Luís Compañy
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante, España
| | | |
Collapse
|
36
|
Knudsen ES, Balaji U, Mannakee B, Vail P, Eslinger C, Moxom C, Mansour J, Witkiewicz AK. Pancreatic cancer cell lines as patient-derived avatars: genetic characterisation and functional utility. Gut 2018; 67:508-520. [PMID: 28073890 PMCID: PMC5868284 DOI: 10.1136/gutjnl-2016-313133] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) is a therapy recalcitrant disease with the worst survival rate of common solid tumours. Preclinical models that accurately reflect the genetic and biological diversity of PDAC will be important for delineating features of tumour biology and therapeutic vulnerabilities. DESIGN 27 primary PDAC tumours were employed for genetic analysis and development of tumour models. Tumour tissue was used for derivation of xenografts and cell lines. Exome sequencing was performed on the originating tumour and developed models. RNA sequencing, histological and functional analyses were employed to determine the relationship of the patient-derived models to clinical presentation of PDAC. RESULTS The cohort employed captured the genetic diversity of PDAC. From most cases, both cell lines and xenograft models were developed. Exome sequencing confirmed preservation of the primary tumour mutations in developed cell lines, which remained stable with extended passaging. The level of genetic conservation in the cell lines was comparable to that observed with patient-derived xenograft (PDX) models. Unlike historically established PDAC cancer cell lines, patient-derived models recapitulated the histological architecture of the primary tumour and exhibited metastatic spread similar to that observed clinically. Detailed genetic analyses of tumours and derived models revealed features of ex vivo evolution and the clonal architecture of PDAC. Functional analysis was used to elucidate therapeutic vulnerabilities of relevance to treatment of PDAC. CONCLUSIONS These data illustrate that with the appropriate methods it is possible to develop cell lines that maintain genetic features of PDAC. Such models serve as important substrates for analysing the significance of genetic variants and create a unique biorepository of annotated cell lines and xenografts that were established simultaneously from same primary tumour. These models can be used to infer genetic and empirically determined therapeutic sensitivities that would be germane to the patient.
Collapse
Affiliation(s)
- Erik S Knudsen
- University of Arizona Department of Medicine, University of Arizona, Tucson, Arizona, USA,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA,McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Uthra Balaji
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Brian Mannakee
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Paris Vail
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Cody Eslinger
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher Moxom
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John Mansour
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Agnieszka K Witkiewicz
- University of Arizona Department of Medicine, University of Arizona, Tucson, Arizona, USA,University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA,McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA,University of Arizona Department of Pathology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
37
|
Dayot S, Speisky D, Couvelard A, Bourgoin P, Gratio V, Cros J, Rebours V, Sauvanet A, Bedossa P, Paradis V, Ruszniewski P, Couvineau A, Voisin T. In vitro, in vivo and ex vivo demonstration of the antitumoral role of hypocretin-1/orexin-A and almorexant in pancreatic ductal adenocarcinoma. Oncotarget 2018; 9:6952-6967. [PMID: 29467942 PMCID: PMC5805528 DOI: 10.18632/oncotarget.24084] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 01/02/2018] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is still the poorest prognostic tumor of the digestive system. We investigated the antitumoral role of orexin-A and almorexant in PDAC. We analyzed the orexin receptor type 1 (OX1R) expression by immunohistochemistry in human normal pancreas, PDAC and its precursor dysplastic intraepithelial lesions. We used PDAC-derived cell lines and fresh tissue slices to study the apoptotic role of hypocretin-1/orexin-A and almorexant in vitro and ex vivo. We analyzed in vivo the hypocretin-1/orexin-A and almorexant effect on tumor growth in mice xenografted with PDAC cell lines expressing, or not, OX1R. Ninety-six percent of PDAC expressed OX1R, while adjacent normal exocrine pancreas did not. OX1R was expressed in pre-cancerous lesions. In vitro, under hypocretin-1/orexin-A and almorexant, the OX1R-positive AsPC-1 cells underwent apoptosis, abolished by the tyrosine phosphatase SHP2 inhibitor, NSC-87877, whereas the OX1R-negative HPAF-II cell line did not. These effects were mediated by phosphorylation of OX1R and recruitment of SHP2. Ex vivo, caspase-3 positive tumor cells were significantly higher in fresh tumour slices treated 48h with hypocretin-1/orexin-A, as compared to control, whereas cellular proliferation, assessed by Ki-67 index, was not modified. In vivo, when AsPC-1 cells or patient-derived cells were xenografted in nude mice, hypocretin-1/orexin-A or almorexant, administrated both starting the day of cell line inoculation or after tumoral development, strongly slowed tumor growth. Hypocretin-1/orexin-A and almorexant induce, through OX1R, the inhibition of PDAC cellular growth by apoptosis. Hypocretins/orexins and almorexant might be powerful candidates for the treatment of PDAC.
Collapse
Affiliation(s)
- Stéphanie Dayot
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
| | - Daniela Speisky
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
| | - Anne Couvelard
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
- 2 Département de Pathologie Beaujon-Bichat, AP-HP, Hôpital Bichat, Huchard, 75018 Paris, France
| | - Pierre Bourgoin
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
| | - Valérie Gratio
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
| | - Jérôme Cros
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
- 4 Département de Pathologie Beaujon-Bichat, AP-HP, Hôpital Beaujon, 92118 Clichy, France
| | - Vinciane Rebours
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
- 3 Service de Pancréatologie-Gastroentérologie PMAD, Pôle des Maladies de l’Appareil Digestif, AP-HP, Hôpital Beaujon, 92118 Clichy, France
| | - Alain Sauvanet
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
- 3 Service de Pancréatologie-Gastroentérologie PMAD, Pôle des Maladies de l’Appareil Digestif, AP-HP, Hôpital Beaujon, 92118 Clichy, France
| | - Pierre Bedossa
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
- 4 Département de Pathologie Beaujon-Bichat, AP-HP, Hôpital Beaujon, 92118 Clichy, France
| | - Valérie Paradis
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
- 4 Département de Pathologie Beaujon-Bichat, AP-HP, Hôpital Beaujon, 92118 Clichy, France
| | - Philippe Ruszniewski
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
- 3 Service de Pancréatologie-Gastroentérologie PMAD, Pôle des Maladies de l’Appareil Digestif, AP-HP, Hôpital Beaujon, 92118 Clichy, France
| | - Alain Couvineau
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
| | - Thierry Voisin
- 1 INSERM UMR1149 Centre de Recherche sur l’Inflammation (CRI), Université Paris-Diderot, Sorbonne Paris Cité, DHU UNITY, Faculté de Médecine Xavier Bichat, Huchard, 75018 Paris, France
| |
Collapse
|
38
|
Wang L, Xiong L, Wu Z, Miao X, Liu Z, Li D, Zou Q, Yang Z. Expression of UGP2 and CFL1 expression levels in benign and malignant pancreatic lesions and their clinicopathological significance. World J Surg Oncol 2018; 16:11. [PMID: 29347944 PMCID: PMC5774110 DOI: 10.1186/s12957-018-1316-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 01/10/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND This study investigated UGP2 (uridine diphosphate-glucose pyrophosphorylase-2) and CFL1 (cofilin-1) expression in pancreatic ductal carcinoma (PDC), paracancerous tissue (PT), benign lesions (BL), and normal tissue (NT) and their clinicopathological significance. METHODS Surgical specimens, which were collected from 106 cases of pancreatic ductal carcinoma, 35 cases of paracancerous tissues, 55 cases of benign lesions and 13 cases of normal pancreatic tissues, were fixed with 4% formaldehyde to prepare conventional paraffin-embedded sections. EnVision immunohistochemical was used to stain for UGP2 and CFL1. Kaplan-Meier survival analysis was performed to assess the correlation of expression pattern with survival. RESULTS We found that positive UGP2 and CFL1 expression in PDC were significantly higher than those in PT, BL, and NT. In PT and BL with positive UGP2 and CFL1 expression, mild to severe atypical hyperplasia or intraepithelial neoplasia of grades II-III was observed in ductal epithelium. Positive UGP2 and CFL1 expression in cases with high differentiation, no lymph node metastasis, no surrounding invasion, and TNM (tumor-node-metastasis) staging I or/and II were significantly lower than those in cases with poor differentiation, lymph node metastasis, surrounding invasion, and TNM stage III and/or IV. Positive UGP2 expression in male patients was significantly lower than that in female patients. UGP2 and CFL1 expression in PDC were positively correlated. Kaplan-Meier survival analysis showed the degree of differentiation, tumor maximal diameter, TNM stage, lymph node metastasis, and surrounding invasion, and UGP2 and CFL1 expression were closely related to the average survival time of patients with PDC. The survival time of patients with positive UGP2 and CFL1 expression was significantly shorter than that of patients with negative expression. Cox multivariate analysis showed that poor differentiation, tumor maximal diameter ≥ 3 cm, TNM stage III or IV, lymph node metastasis, surrounding invasion, and positive UGP2 and CFL1 expression was negatively correlated with the postoperative survival rate and positively correlated with the mortality of patients with PDC. CONCLUSION Positive expression of UGP2 and CFL1 can serve a valuable prognostic factor in pancreatic cancer.
Collapse
Affiliation(s)
- Lingxiang Wang
- General Surgery Department, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.,Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Li Xiong
- General Surgery Department, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.,Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Zhengchun Wu
- General Surgery Department, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.,Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Xiongying Miao
- General Surgery Department, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.,Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Ziru Liu
- General Surgery Department, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.,Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Daiqiang Li
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China
| | - Qiong Zou
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zhulin Yang
- General Surgery Department, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China. .,Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, People's Republic of China.
| |
Collapse
|
39
|
Couvineau A, Dayot S, Nicole P, Gratio V, Rebours V, Couvelard A, Voisin T. The Anti-tumoral Properties of Orexin/Hypocretin Hypothalamic Neuropeptides: An Unexpected Therapeutic Role. Front Endocrinol (Lausanne) 2018; 9:573. [PMID: 30319552 PMCID: PMC6170602 DOI: 10.3389/fendo.2018.00573] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/10/2018] [Indexed: 02/05/2023] Open
Abstract
Orexins (OxA and OxB) also termed hypocretins are hypothalamic neuropeptides involved in central nervous system (CNS) to control the sleep/wake process which is mediated by two G protein-coupled receptor subtypes, OX1R, and OX2R. Beside these central effects, orexins also play a role in various peripheral organs such as the intestine, pancreas, adrenal glands, kidney, adipose tissue and reproductive tract.In the past few years, an unexpected anti-tumoral role of orexins mediated by a new signaling pathway involving the presence of two immunoreceptor tyrosine-based inhibitory motifs (ITIM) in both orexin receptors subtypes, the recruitment of the phosphotyrosine phosphatase SHP2 and the induction of mitochondrial apoptosis has been elucidated. In the present review, we will discuss the anti-tumoral effect of orexin/OXR system in colon, pancreas, prostate and other cancers, and its interest as a possible therapeutic target.
Collapse
|
40
|
Using Attenuated Total Reflection-Fourier Transform Infra-Red (ATR-FTIR) spectroscopy to distinguish between melanoma cells with a different metastatic potential. Sci Rep 2017; 7:4381. [PMID: 28663552 PMCID: PMC5491518 DOI: 10.1038/s41598-017-04678-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/18/2017] [Indexed: 11/08/2022] Open
Abstract
The vast majority of cancer related deaths are caused by metastatic tumors. Therefore, identifying the metastatic potential of cancer cells is of great importance both for prognosis and for determining the correct treatment. Infrared (IR) spectroscopy of biological cells is an evolving research area, whose main aim is to find the spectral differences between diseased and healthy cells. In the present study, we demonstrate that Attenuated Total Reflection Fourier Transform IR (ATR-FTIR) spectroscopy may be used to determine the metastatic potential of cancer cells. Using the ATR-FTIR spectroscopy, we can identify spectral alterations that are a result of hydration or molecular changes. We examined two murine melanoma cells with a common genetic background but a different metastatic level, and similarly, two human melanoma cells. Our findings revealed that higher metastatic potential correlates with membrane hydration level. Measuring the spectral properties of the cells allows us to determine the membrane hydration levels. Thus, ATR-FTIR spectroscopy has the potential to help in cancer metastasis prognosis.
Collapse
|
41
|
Wu ZC, Xiong L, Wang LX, Miao XY, Liu ZR, Li DQ, Zou Q, Liu KJ, Zhao H, Yang ZL. Comparative study of ROR2 and WNT5a expression in squamous/adenosquamous carcinoma and adenocarcinoma of the gallbladder. World J Gastroenterol 2017; 23:2601-2612. [PMID: 28465645 PMCID: PMC5394524 DOI: 10.3748/wjg.v23.i14.2601] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/01/2017] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the expression and clinical pathological significance of ROR2 and WNT5a in gallbladder squamous/adenosquamous carcinoma (SC/ASC) and adenocarcinoma (AC). METHODS EnVision immunohistochemistry was used to stain for ROR2 and WNT5a in 46 SC/ASC patients and 80 AC patients. RESULTS Poorly differentiated AC among AC patients aged > 45 years were significantly more frequent compared with SC/ASC patients, while tumors with a maximal diameter > 3 cm in the SC/ASC group were significantly more frequent compared with the AC group. Positive ROR2 and WNT5a expression was significantly lower in SC/ASC or AC with a maximal mass diameter ≤ 3 cm, a TNM stage of I + II, no lymph node metastasis, no surrounding invasion, and radical resection than in patients with a maximal mass diameter > 3 cm, TNM stage IV, lymph node metastasis, surrounding invasion, and no resection. Positive ROR2 expression in patients with highly differentiated SC/ASC was significantly lower than in patients with poorly differentiated SC/ASC. Positive ROR2 and WNT5a expression levels in highly differentiated AC were significantly lower than in poorly differentiated AC. Kaplan-Meier survival analysis showed that differentiation degree, maximal mass diameter, TNM stage, lymph node metastasis, surrounding invasion, surgical procedure and the ROR2 and WNT5a expression levels were closely related to average survival of SC/ASC or AC. The survival of SC/ASC or AC patients with positive expression of ROR2 and WNT5a was significantly shorter than that of patients with negative expression results. Cox multivariate analysis revealed that poor differentiation, a maximal diameter of the mass ≥ 3 cm, TNM stage III or IV, lymph node metastasis, surrounding invasion, unresected surgery and positive ROR2 or WNT5a expression in the SC/ASC or AC patients were negatively correlated with the postoperative survival rate and positively correlated with mortality, which are risk factors and independent prognostic predictors. CONCLUSION SC/ASC or AC patients with positive ROR2 or WNT5a expression generally have a poor prognosis.
Collapse
|
42
|
Watanabe M, Iizumi Y, Sukeno M, Iizuka-Ohashi M, Sowa Y, Sakai T. The pleiotropic regulation of cyclin D1 by newly identified sesaminol-binding protein ANT2. Oncogenesis 2017; 6:e311. [PMID: 28368390 PMCID: PMC5520487 DOI: 10.1038/oncsis.2017.10] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 02/03/2017] [Accepted: 02/12/2017] [Indexed: 12/14/2022] Open
Abstract
The expression of cyclin D1 is upregulated in various cancer cells by diverse mechanisms, such as increases in mRNA levels, the promotion of the translation by mammalian target of rapamycin complex 1 (mTORC1) signaling and the protein stabilization. We here show that sesaminol, a sesame lignan, reduces the expression of cyclin D1 with decreasing mRNA expression levels, inhibiting mTORC1 signaling and promoting proteasomal degradation. We subsequently generated sesaminol-immobilized FG beads to newly identify sesaminol-binding proteins. As a consequence, we found that adenine nucleotide translocase 2 (ANT2), the inner mitochondrial membrane protein, directly bound to sesaminol. Consistent with the effects of sesaminol, the depletion of ANT2 caused a reduction in cyclin D1 with decreases in its mRNA levels, mTORC1 inhibition and the proteasomal degradation of its protein, suggesting that sesaminol negatively regulates the function of ANT2. Furthermore, we screened other ANT2-binding compounds and found that the proliferator-activated receptor-γ agonist troglitazone also reduced cyclin D1 expression in a multifaceted manner, analogous to that of the sesaminol treatment and ANT2 depletion. Therefore, the chemical biology approach using magnetic FG beads employed in the present study revealed that sesaminol bound to ANT2, which may pleiotropically upregulate cyclin D1 expression at the mRNA level and protein level with mTORC1 activation and protein stabilization. These results suggest the potential of ANT2 as a target against cyclin D1-overexpressing cancers.
Collapse
Affiliation(s)
- M Watanabe
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Y Iizumi
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - M Sukeno
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - M Iizuka-Ohashi
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Division of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Y Sowa
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - T Sakai
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
43
|
Yang Z, Li D, Liu Z, Miao X, Yang L, Zou Q, Yuan Y. BIRC7 and KLF4 expression in benign and malignant lesions of pancreas and their clinicopathological significance. Cancer Biomark 2017; 17:437-444. [PMID: 27802195 DOI: 10.3233/cbm-160660] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study investigated the KLF4 and BIRC7 protein expression in malignant and benign pancreatic tissues by immunohistochemical staining and the clinical and pathological significance of KLF4 and BIRC7 expression in PDAC. KLF4 expression was significantly lower, whereas BIRC7 expression was significantly higher in PDAC than that in peritumoral tissue, benign pancreatic lesions, and normal pancreatic tissue (P < 0.01). The percentage of positive BIRC7 and negative KLF4 expression was significantly lower in PDAC patients with well differentiated tumors, maximum tumor size < 3 cm, no lymph node metastasis, no invasion to the surrounding tissues and organs, and TNM stage I/II stage disease than in patients with poorly differentiated tumor, maximum tumor size > 5 cm, lymph node metastasis, invasion to surrounding tissues and organs, and TNM stage III/IV disease (P < 0.05 or P < 0.01). Kaplan-Meier survival analysis showed that the differentiation, maximum tumor size, TNM stage, lymph node metastasis, invasion, negative KLF4 expression, and positive BIRC7 expression were significantly associated with the short survival of patients with PDAC (P < 0.05 or P < 0.01). Cox multivariate analysis revealed that positive BIRC7 expression and negative KLF4 expression were independent poor prognosis factors in PDAC patients. In conclusions, positive BIRC7 expression and negative KLF4 expression are associated with the progression of PDAC and poor prognosis in patients with PDAC.
Collapse
Affiliation(s)
- Zhulin Yang
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Daiqiang Li
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziru Liu
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiongying Miao
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Leping Yang
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiong Zou
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuan Yuan
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
44
|
Rezaei M, Hosseini A, Nikeghbalian S, Ghaderi A. Establishment and characterization of a new human acinar cell carcinoma cell line, Faraz-ICR, from pancreas. Pancreatology 2017; 17:303-309. [PMID: 28215484 DOI: 10.1016/j.pan.2017.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/25/2017] [Accepted: 02/06/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Basic research in the field of acinar cell carcinoma (ACC) as a rare neoplasm of the pancreas is dependent on the availability of pragmatic model such as new pancreatic cancer cell lines. Thus, establishment and characterization of new pancreatic cancer cell lines from ACC origin are deemed important. METHODS Faraz-ICR cell line was derived from a 58-years old woman with pancreatic acinar cell carcinoma by the collagenase digestion protocol. We characterized the cell line by examining its morphology and cytostructural and functional profile. RESULTS Faraz-ICR has a doubling time of 35 hours and grows in soft agar with a colony-forming efficiency of 25%. The cell had nearly normal pattern of chromosomes in karyotype analysis and Comparative Genomic Hybridization (CGH) array analysis. Evaluation of cells by flowcytometry showed that Faraz-ICR is negative for EpCAM and mesenchymal markers in different passages, and has epithelial nature. Immunofluorescence staining revealed that cells were strongly positive for vimentin, desmin, ezrin, S100, nestin and they were negative for pan-cytokeratins, chromogranin and alpha smooth muscle actin. CONCLUSIONS We were able to establish a new pancreatic carcinoma cell line with partial aspects of Epithelial-mesenchymal transition and aggressiveness. This cell line might be suitable for studying various anticancer drugs and protein profile aiming to see any possible tumor associated marker for ACC.
Collapse
Affiliation(s)
- Marzieh Rezaei
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Hosseini
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saman Nikeghbalian
- Department of Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
45
|
Dai B, Roife D, Kang Y, Gumin J, Rios Perez MV, Li X, Pratt M, Brekken RA, Fueyo-Margareto J, Lang FF, Fleming JB. Preclinical Evaluation of Sequential Combination of Oncolytic Adenovirus Delta-24-RGD and Phosphatidylserine-Targeting Antibody in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2017; 16:662-670. [PMID: 28138026 DOI: 10.1158/1535-7163.mct-16-0526] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 12/01/2016] [Accepted: 12/15/2016] [Indexed: 12/24/2022]
Abstract
Delta-24-RGD (DNX-2401) is a conditional replication-competent oncolytic virus engineered to preferentially replicate in and lyse tumor cells with abnormality of p16/RB/E2F pathway. In a phase I clinical trial, Delta-24-RGD has shown favorable safety profile and promising clinical efficacy in brain tumor, which prompted us to evaluate its anticancer activity in pancreatic ductal adenocarcinoma (PDAC), which also has high frequency of homozygous deletion and promoter methylation of CDKN2A encoding the p16 protein. Our results demonstrate that Delta-24-RGD can induce dramatic cytotoxicity in a subset of PDAC cell lines with high cyclin D1 expression. Induction of autophagy and apoptosis by Delta-24-RGD in sensitive PDAC cells was confirmed with LC3B-GFP autophagy reporter and acridine orange staining as well as Western blotting analysis of LC3B-II expression. Notably, we found that Delta-24-RGD induced phosphatidylserine exposure in infected cells independent of cells' sensitivity to Delta-24-RGD, which renders a rationale for combination of Delta-24-RGD viral therapy and phosphatidylserine targeting antibody for PDAC. In a mouse PDAC model derived from a liver metastatic pancreatic cancer cell line, Delta-24-RGD significantly inhibited tumor growth compared with control (P < 0.001), and combination of phosphatidylserine targeting antibody 1N11 further enhanced its anticancer activity (P < 0.01) possibly through inducing synergistic anticancer immune responses. Given that these 2 agents are currently in clinical evaluation, our study warrants further clinical evaluation of this novel combination strategy in pancreatic cancer therapy. Mol Cancer Ther; 16(4); 662-70. ©2016 AACR.
Collapse
Affiliation(s)
- Bingbing Dai
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Roife
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ya'an Kang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mayrim V Rios Perez
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xinqun Li
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Pratt
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Juan Fueyo-Margareto
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
46
|
Qie S, Diehl JA. Cyclin D1, cancer progression, and opportunities in cancer treatment. J Mol Med (Berl) 2016; 94:1313-1326. [PMID: 27695879 PMCID: PMC5145738 DOI: 10.1007/s00109-016-1475-3] [Citation(s) in RCA: 478] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/15/2022]
Abstract
Mammalian cells encode three D cyclins (D1, D2, and D3) that coordinately function as allosteric regulators of cyclin-dependent kinase 4 (CDK4) and CDK6 to regulate cell cycle transition from G1 to S phase. Cyclin expression, accumulation, and degradation, as well as assembly and activation of CDK4/CDK6 are governed by growth factor stimulation. Cyclin D1 is more frequently dysregulated than cyclin D2 or D3 in human cancers, and as such, it has been more extensively characterized. Overexpression of cyclin D1 results in dysregulated CDK activity, rapid cell growth under conditions of restricted mitogenic signaling, bypass of key cellular checkpoints, and ultimately, neoplastic growth. This review discusses cyclin D1 transcriptional, translational, and post-translational regulations and its biological function with a particular focus on the mechanisms that result in its dysregulation in human cancers.
Collapse
Affiliation(s)
- Shuo Qie
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St, Charleston, SC, 29425, USA
| | - J Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St, Charleston, SC, 29425, USA.
| |
Collapse
|
47
|
Huang SF, Yang ZL, Li DQ, Liu ZY, Wang CW, Miao XY, Zou Q, Yuan Y. Jagged1 and DLL4 expressions in benign and malignant pancreatic lesions and their clinicopathological significance. Hepatobiliary Pancreat Dis Int 2016; 15:640-646. [PMID: 27919854 DOI: 10.1016/s1499-3872(16)60110-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is characterized by a poor prognosis. Despite intensive research, markers for the early diagnosis, prognosis, and targeting therapy of PDAC are not available. This study aimed to investigate the protein expressions of Jagged1 and DLL4 in PDAC tumor, benign pancreatic and normal pancreatic tissues, and analyze the associations of the two proteins with the clinical and pathological characteristics of PDAC. METHODS A total of 106 PDAC tumor tissues and 35 peritumoral tissues were collected from January 2000 to December 2011 at our hospitals. Thirteen normal pancreatic tissues and 55 benign pancreatic specimens were collected at the same period. Immunohistochemical staining was used to measure Jagged1 and DLL4 protein expressions in these tissues. RESULTS The percentage of positive Jagged1 and DLL4 was significantly higher in PDAC than in normal pancreatic tissues, benign pancreatic tissues, and peritumoral tissues (P<0.01). The higher Jagged1 and DLL4 expressions in PDAC were significantly associated with poor differentiation, maximum tumor size >5 cm, invasion, regional lymph node metastasis, and TNM III/IV disease (P<0.05). In PDAC, Jagged1 expression positively correlated with DLL4 expression. Univariate Kaplan-Meier analysis showed that positive Jagged1 and DLL4 expressions were significantly associated with shorter survival in patients with PDAC. Multivariate Cox regression analysis showed that positive Jagged1 and DLL4 expressions were independent prognostic factors for poor prognosis of patients with PDAC. CONCLUSION Positive Jagged1 and DLL4 expression is closely correlated with severe clinicopathological characteristics and poor prognosis in patients with PDAC.
Collapse
Affiliation(s)
- Sheng-Fu Huang
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Martínez JF, Aparicio JR, Peiró G, Cabezas A, Roger M, Ruiz F, Compañy L, Casellas JA. Study of the expression of cathepsins in histological material from pancreatic lesions. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2016; 108:780-784. [PMID: 27855482 DOI: 10.17235/reed.2016.3749/2015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS To assess the expression levels of cathepsins in malignant and premalignant lesions. METHODS We retrospectively included patients who underwent pancreatic surgery on pancreatic solid or cystic masses. The expression of cathepsin H, L, B and S was determined in both types of samples. Lesions were divided into three categories: malignant (pancreatic adenocarcinoma and malignant mucinous neoplasms), premalignant (mucinous neoplasms) and benign (other lesions). RESULTS Thirty-one surgical resection samples were studied. The expression of cathepsins was significantly higher in malignant lesions than in premalignant and benign lesions (H 75%, 27%, 37% p = 0.05; L 92%, 36%, 37% p = 0.011; B 83%, 36%, 62% p = 0.069; S 92%, 36%, 25% p = 0.004, respectively). CONCLUSIONS Cathepsins are overexpressed in histological samples of malignant lesions compared to premalignant and benign lesions. However, the expression of cathepsins is similar in both premalignant and benign lesions.
Collapse
Affiliation(s)
- Juan F Martínez
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante, España
| | - José Ramón Aparicio
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante, España
| | - Gloria Peiró
- Servicio de Anatomía Patológica, Hospital General Universitario de Alicante, España
| | - Antonio Cabezas
- Servicio de Anatomía Patológica, Hospital General Universitario de Alicante, España
| | - Manuela Roger
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante, España
| | - Francisco Ruiz
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante, España
| | - Luís Compañy
- Unidad de Endoscopia Digestiva, Hospital General Universitario de Alicante, España
| | | |
Collapse
|
49
|
McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, Libra M, Nicoletti F, Scalisi A, Torino F, Fenga C, Neri LM, Marmiroli S, Cocco L, Martelli AM. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2942-2976. [PMID: 27612668 DOI: 10.1016/j.bbamcr.2016.09.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that participates in an array of critical cellular processes. GSK-3 was first characterized as an enzyme that phosphorylated and inactivated glycogen synthase. However, subsequent studies have revealed that this moon-lighting protein is involved in numerous signaling pathways that regulate not only metabolism but also have roles in: apoptosis, cell cycle progression, cell renewal, differentiation, embryogenesis, migration, regulation of gene transcription, stem cell biology and survival. In this review, we will discuss the roles that GSK-3 plays in various diseases as well as how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, Wnt/beta-catenin, hedgehog, Notch and TP53. Mutations that occur in these and other pathways can alter the effects that natural GSK-3 activity has on regulating these signaling circuits that can lead to cancer as well as other diseases. The novel roles that microRNAs play in regulation of the effects of GSK-3 will also be evaluated. Targeting GSK-3 and these other pathways may improve therapy and overcome therapeutic resistance.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Steve L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP-Catania, Catania 95100, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Concettina Fenga
- Department of Biomedical, Odontoiatric, Morphological and Functional Images, Occupational Medicine Section - Policlinico "G. Martino" - University of Messina, Messina 98125, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
50
|
Peng W, Jiang A. Long noncoding RNA CCDC26 as a potential predictor biomarker contributes to tumorigenesis in pancreatic cancer. Biomed Pharmacother 2016; 83:712-717. [PMID: 27470572 DOI: 10.1016/j.biopha.2016.06.059] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is the fourth most common cancer worldwide and has the least patient survival rate of any cancer. Emerging studies have demonstrated that long noncoding RNAs (lncRNAs) were present in cancer patients and have shown great potential as powerful markers and therapeutic targets. However, little is known about the role of lncRNAs in PC. The present study aimed to investigate the expression pattern, clinical significance and biological function of lncRNA CCDC26 (CCDC26) in PC. With quantitative real-time PCR, we analyzed CCDC26 expression levels in 40 PC patients. We found that the CCDC26 expression was significantly higher in PC tissues than in normal tissues. CCDC26 levels were correlated with tumor size, tumor number, and reduced overall survival (OS). Univariate and multivariate analysis showed that CCDC26 expression is an independent prognostic factor of OS in patients with PC. Additionally, ROCAUC of CCDC26 was up to 0.663, implicating that CCDC26 could be a diagnostic marker for distinguishing PC from normal. Knockdown of CCDC26 expression by small interfering RNA significantly promoted growth arrest and apoptosis. Moreover, we found that the expression of CCDC26 was positively correlated with PCNA and Bcl2. Our data suggest that CCDC26 may be identified as a novel oncogene in PC, and responsible for growth and apoptosis of cancer cell, partly by regulating the PCNA and Bcl2 expression. This work provides a novel biomarker and therapeutic target of PC for cancer clinic in future.
Collapse
Affiliation(s)
- Wei Peng
- Department of Medical Oncology, Jiangsu Cancer Hospital affiliated to Nanjing Medical University, Nanjing, China.
| | - Airen Jiang
- Research Management Office, Jiangsu Cancer Hospital affiliated to Nanjing Medical University, Nanjing, China.
| |
Collapse
|