1
|
Fang Y, Tan C, Zheng Z, Yang J, Tang J, Guo R, Silli EK, Chen Z, Chen J, Ge R, Liu Y, Wen X, Liang J, Zhu Y, Jin Y, Li Q, Wang Y. The function of microRNA related to cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Biochem Pharmacol 2025; 236:116849. [PMID: 40056941 DOI: 10.1016/j.bcp.2025.116849] [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: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a poor prognosis. A prominent feature of PDAC is the rich and dense stroma present in the tumor microenvironment (TME), which significantly hinders drug penetration. Cancer-associated fibroblasts (CAFs), activated fibroblasts originating from various cell sources, including pancreatic stellate cells (PSCs) and mesenchymal stem cells (MSCs), play a critical role in PDAC progression and TME formation. MicroRNAs (miRNAs) are small, single-stranded non-coding RNA molecules that are frequently involved in tumorigenesis and progression, exhibiting either oncolytic or oncogenic activity. Increasing evidence suggests that aberrant expression of miRNAs can mediate interactions between cancer cells and CAFs, thereby providing novel therapeutic targets for PDAC treatment. In this review, we will focus on the potential roles of miRNAs that target CAFs or CAFs-derived exosomes in PDAC progression, highlighting the feasibility of therapeutic strategies aimed at restoring aberrantly expressed miRNAs associated with CAFs, offering new pathways for the clinical management of PDAC.
Collapse
Affiliation(s)
- Yaohui Fang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenjiang Zheng
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiali Tang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruizhe Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Epiphane K Silli
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhe Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jia Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruyu Ge
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yuquan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiuqi Wen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jingdan Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunfei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yutong Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qian Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
2
|
Sarkar R, Xu Z, Perera CJ, Apte MV. Emerging role of pancreatic stellate cell-derived extracellular vesicles in pancreatic cancer. Semin Cancer Biol 2023; 93:114-122. [PMID: 37225047 DOI: 10.1016/j.semcancer.2023.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/17/2023] [Accepted: 05/19/2023] [Indexed: 05/26/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer that is characterised by a prominent collagenous stromal reaction/desmoplasia surrounding tumour cells. Pancreatic stellate cells (PSCs) are responsible for the production of this stroma and have been shown to facilitate PDAC progression. Recently, extracellular vesicles (EVs), in particular, small extracellular vesicles (exosomes) have been a topic of interest in the field of cancer research for their emerging roles in cancer progression and diagnosis. EVs act as a form of intercellular communication by carrying their molecular cargo from one cell to another, regulating functions of the recipient cells. Although the knowledge of the bi-directional interactions between the PSCs and cancer cells that promote disease progression has advanced significantly over the past decade, studies on PSC-derived EVs in PDAC are currently rather limited. This review provides an overview of PDAC, pancreatic stellate cells and their interactions with cancer cells, as well as the currently known role of extracellular vesicles derived from PSCs in PDAC progression.
Collapse
Affiliation(s)
- Rohit Sarkar
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; Ingham Institute of Applied Medical Research, Sydney 2170, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; Ingham Institute of Applied Medical Research, Sydney 2170, Australia
| | - Chamini J Perera
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; Ingham Institute of Applied Medical Research, Sydney 2170, Australia.
| | - Minoti V Apte
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia; Ingham Institute of Applied Medical Research, Sydney 2170, Australia
| |
Collapse
|
3
|
Tushoski‐Alemán G, Davidson A, Herremans K, Forsmark C, Zhang W, Hughes S, Han S. A peek at the other side of the coin: Tumor-suppressor role of microRNAs expressed by pancreatic cancer-associated fibroblasts. Clin Transl Med 2023; 13:e1357. [PMID: 37539872 PMCID: PMC10401917 DOI: 10.1002/ctm2.1357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023] Open
Affiliation(s)
| | - Aaron Davidson
- Department of SurgeryCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Kelly Herremans
- Department of SurgeryCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Chris Forsmark
- Department of MedicineCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Weizhou Zhang
- Department of PathologyImmunology and Laboratory MedicineCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Steven Hughes
- Department of SurgeryCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Song Han
- Department of SurgeryCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
4
|
Sarcar B, Fang B, Izumi V, O Nunez Lopez Y, Tassielli A, Pratley R, Jeong D, Permuth JB, Koomen JM, Fleming JB, Stewart PA. A comparative Proteomics Analysis Identified Differentially Expressed Proteins in Pancreatic Cancer-Associated Stellate Cell Small Extracellular Vesicles. Mol Cell Proteomics 2022; 21:100438. [PMID: 36332889 PMCID: PMC9792568 DOI: 10.1016/j.mcpro.2022.100438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 10/03/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Human pancreatic stellate cells (HPSCs) are an essential stromal component and mediators of pancreatic ductal adenocarcinoma (PDAC) progression. Small extracellular vesicles (sEVs) are membrane-enclosed nanoparticles involved in cell-to-cell communications and are released from stromal cells within PDAC. A detailed comparison of sEVs from normal pancreatic stellate cells (HPaStec) and from PDAC-associated stellate cells (HPSCs) remains a gap in our current knowledge regarding stellate cells and PDAC. We hypothesized there would be differences in sEVs secretion and protein expression that might contribute to PDAC biology. To test this hypothesis, we isolated sEVs using ultracentrifugation followed by characterization by electron microscopy and Nanoparticle Tracking Analysis. We report here our initial observations. First, HPSC cells derived from PDAC tumors secrete a higher volume of sEVs when compared to normal pancreatic stellate cells (HPaStec). Although our data revealed that both normal and tumor-derived sEVs demonstrated no significant biological effect on cancer cells, we observed efficient uptake of sEVs by both normal and cancer epithelial cells. Additionally, intact membrane-associated proteins on sEVs were essential for efficient uptake. We then compared sEV proteins isolated from HPSCs and HPaStecs cells using liquid chromatography-tandem mass spectrometry. Most of the 1481 protein groups identified were shared with the exosome database, ExoCarta. Eighty-seven protein groups were differentially expressed (selected by 2-fold difference and adjusted p value ≤0.05) between HPSC and HPaStec sEVs. Of note, HPSC sEVs contained dramatically more CSE1L (chromosome segregation 1-like protein), a described marker of poor prognosis in patients with pancreatic cancer. Based on our results, we have demonstrated unique populations of sEVs originating from stromal cells with PDAC and suggest that these are significant to cancer biology. Further studies should be undertaken to gain a deeper understanding that could drive novel therapy.
Collapse
Affiliation(s)
- Bhaswati Sarcar
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Bin Fang
- Proteomics and Metabolomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Victoria Izumi
- Proteomics and Metabolomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | | | - Alexandra Tassielli
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Richard Pratley
- Translational Research Institute, Advent Health, Orlando, Florida, USA
| | - Daniel Jeong
- Department of Diagnostic and Interventional Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Jennifer B Permuth
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA; Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - John M Koomen
- Proteomics and Metabolomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.
| | - Paul A Stewart
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.
| |
Collapse
|
5
|
Effects of TP53 Mutations and miRs on Immune Responses in the Tumor Microenvironment Important in Pancreatic Cancer Progression. Cells 2022; 11:cells11142155. [PMID: 35883598 PMCID: PMC9318640 DOI: 10.3390/cells11142155] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 01/27/2023] Open
Abstract
Approximately 90% of pancreatic cancers are pancreatic ductal adenocarcinomas (PDAC). PDAC is the fourth leading cause of cancer death world-wide. Therapies for PDAC are largely ineffective due to the dense desmoplastic tumor microenvironment which prevents chemotherapeutic drugs and small molecule inhibitors from exerting effective anti-cancer effects. In this review, we will discuss the roles of TP53 and miRs on the PDAC tumor microenvironment and how loss of the normal functions of TP53 promote tumor progression. The TP53 gene is mutated in approximately 50% of pancreatic cancers. Often, these TP53 mutations are point mutations which confer additional functions for the TP53 proteins. These are called gain of function (GOF) mutations (mut). Another class of TP53 mutations are deletions which result in loss of the TP53 protein; these are referred to TP53-null mutations. We have organized this review into various components/properties of the PDAC microenvironment and how they may be altered in the presence of mutant TP53 and loss of certain miR expression.
Collapse
|
6
|
Sempere LF, Azmi AS, Moore A. microRNA-based diagnostic and therapeutic applications in cancer medicine. WILEY INTERDISCIPLINARY REVIEWS. RNA 2021; 12:e1662. [PMID: 33998154 PMCID: PMC8519065 DOI: 10.1002/wrna.1662] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 01/18/2023]
Abstract
It has been almost two decades since the first link between microRNAs and cancer was established. In the ensuing years, this abundant class of short noncoding regulatory RNAs has been studied in virtually all cancer types. This tremendously large body of research has generated innovative technological advances for detection of microRNAs in tissue and bodily fluids, identified the diagnostic, prognostic, and/or predictive value of individual microRNAs or microRNA signatures as potential biomarkers for patient management, shed light on regulatory mechanisms of RNA-RNA interactions that modulate gene expression, uncovered cell-autonomous and cell-to-cell communication roles of specific microRNAs, and developed a battery of viral and nonviral delivery approaches for therapeutic intervention. Despite these intense and prolific research efforts in preclinical and clinical settings, there are a limited number of microRNA-based applications that have been incorporated into clinical practice. We review recent literature and ongoing clinical trials that highlight most promising approaches and standing challenges to translate these findings into viable microRNA-based clinical tools for cancer medicine. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Lorenzo F. Sempere
- Department of Radiology, Precision Health ProgramMichigan State UniversityEast LansingMichiganUSA
| | - Asfar S. Azmi
- Department of OncologyWayne State University School of MedicineDetroitMichiganUSA
- Karmanos Cancer InstituteDetroitMichiganUSA
| | - Anna Moore
- Departments of Radiology and Physiology, Precision Health ProgramMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
7
|
Yadav P, Bandyopadhayaya S, Ford BM, Mandal C. Interplay between DNA Methyltransferase 1 and microRNAs During Tumorigenesis. Curr Drug Targets 2021; 22:1129-1148. [PMID: 33494674 DOI: 10.2174/1389450122666210120141546] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/16/2020] [Accepted: 10/18/2020] [Indexed: 01/18/2023]
Abstract
Cancer is a genetic disease resulting from genomic changes; however, epigenetic alterations act synergistically with these changes during tumorigenesis and cancer progression. Epigenetic variations are gaining more attention as an important regulator in tumor progression, metastasis and therapy resistance. Aberrant DNA methylation at CpG islands is a central event in epigeneticmediated gene silencing of various tumor suppressor genes. DNA methyltransferase 1 (DNMT1) predominately methylates at CpG islands on hemimethylated DNA substrates in proliferation of cells. DNMT1 has been shown to be overexpressed in various cancer types and exhibits tumor-promoting potential. The major drawbacks to DNMT1-targeted cancer therapy are the adverse effects arising from nucleoside and non-nucleoside based DNMT1 inhibitors. This paper focuses on the regulation of DNMT1 by various microRNAs (miRNAs), which may be assigned as future DNMT1 modulators, and highlights how DNMT1 regulates various miRNAs involved in tumor suppression. Importantly, the role of reciprocal inhibition between DNMT1 and certain miRNAs in tumorigenic potential is approached in this review. Hence, this review seeks to project an efficient and strategic approach using certain miRNAs in conjunction with conventional DNMT1 inhibitors as a novel cancer therapy. It has also been pinpointed to select miRNA candidates associated with DNMT1 regulation that may not only serve as potential biomarkers for cancer diagnosis and prognosis, but may also predict the existence of aberrant methylation activity in cancer cells.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh- 305817, Ajmer, Rajasthan, India
| | - Shreetama Bandyopadhayaya
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh- 305817, Ajmer, Rajasthan, India
| | - Bridget M Ford
- Department of Biology, University of the Incarnate Word, San Antonio, TX 78209, United States
| | - Chandi Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh- 305817, Ajmer, Rajasthan, India
| |
Collapse
|
8
|
Chen B, Hu C, Jiang L, Xiang Z, Zuo Z, Lin Y, Liu C. Exploring the significance of novel immune-related gene signatures in the prognosis and immune features of pancreatic adenocarcinoma. Int Immunopharmacol 2021; 92:107359. [PMID: 33465729 DOI: 10.1016/j.intimp.2020.107359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Immune-related genes (IRGs) are associated with the prognosis of different cancers and are helpful for the diagnosis and management of systematic treatment for cancer patients. However, there have been a few corresponding studies in pancreatic adenocarcinoma (PAAD). METHODS The data of PAAD patients were obtained from the TCGA, GEO, and ICGC databases. Additionally, the expression profiles of the normal pancreas from the GTEx database were used to screen differentially expressed immune-related genes (DEIRGs). Cox regression analyses were used to explore overall survival (OS)- and progression-free survival (PFS)-related DEIRGs and to establish two nomograms for PAAD prognosis. Finally, transcription factor (TF), immune infiltration, and unsupervised consensus analyses were performed to understand the potential mechanisms. RESULTS An OS-prognostic signature based on seven DEIRGs and a PFS-prognostic signature based on seven DEIRGs were generated, and their robust prognostic ability was confirmed by ROC curves (OS: 0.736 ~ 0.774, PFS: 0.732 ~ 0.840). According to the risk score, the OS and PFS of the high-risk group were poorer than those of the low-risk group in the training set and four external validation sets. In addition, two nomograms based on the signatures and clinical variables also showed excellent discrimination. And two hub regulatory pathways were successfully validated in several independent datasets. Discernable patterns of DEIRGs in unsupervised consensus analysis showed that patients with low expression of immune checkpoints had a favorable prognosis. CONCLUSION Two DEIRG-based signatures can be used as independent tools for the prognostic prediction of PAAD and to provide potential novel immunotherapy targets.
Collapse
Affiliation(s)
- Bo Chen
- The First Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Chuan Hu
- Department of Joint Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liqing Jiang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
| | - Zhouxia Xiang
- The First Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Ziyi Zuo
- The First Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Yangjun Lin
- The First Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Chuan Liu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
9
|
Yang X, Feng KX, Li H, Wang L, Xia H. MicroRNA-199a Inhibits Cell Proliferation, Migration, and Invasion and Activates AKT/mTOR Signaling Pathway by Targeting B7-H3 in Cervical Cancer. Technol Cancer Res Treat 2020; 19:1533033820942245. [PMID: 32856542 PMCID: PMC7457410 DOI: 10.1177/1533033820942245] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cervical cancer is a deadly disease. Some microRNAs are involved in tumor invasion and metastasis. Decreased expression of microRNA-199a has been correlated with tumorigenesis. In our study, the quantitative real-time polymerase chain reaction results indicated that microRNA-199a was expressed at lower levels in cervical cancer tissues, and the expression level of B7-H3 was significantly increased compared with that in the adjacent normal tissues, and the expression levels of B7-H3 and microRNA-199a in cervical cancer tissues and in adjacent normal tissues were inversely correlated. We also found that the expression of microRNA-199a was downregulated in cervical cancer cell lines when compared to immortalized cells. In this study, B7-H3 was identified as a novel target of microRNA-199a in cervical cancer. TargetScan (http://www.targetscan.org/) bioinformatics analysis was used to predict that the 3'-untranslated region of B7-H3 is a direct target of microRNA-199a. The result was also verified by the luciferase reporter assay. MicroRNA-199a could directly target the 3'-untranslated region of B7-H3, but the specific signaling pathways that were involved in regulating B7-H3 expression remained unclear. To clarify whether the suppressive effect of microRNA-199a was mediated through B7-H3, a series of experiments were performed. We found that the overexpression of microRNA-199a inhibited cell proliferation, migration, and invasion via direct binding to B7-H3. Epithelial-mesenchymal transition is a major factor involved in cervical cancer metastasis. Quantitative real-time polymerase chain reaction and western blot results indicated that microRNA-199a inhibits tumor progression in cervical cancer by targeting B7-H3. The microRNAs regulatory network is quite complex. We further examined the effect of microRNA-199a on the AKT/mTOR signaling pathway. We explored the regulatory role of microRNA-199a and first demonstrated that highly expressed microRNA-199a inhibits tumor growth and activates the AKT/mTOR signaling pathway by targeting B7-H3 in vivo and in vitro. Our findings not only provide a better understanding of the pathogenesis of cervical cancer but also provide novel findings and theoretical support for potential targeted therapeutic tools for cervical cancer.
Collapse
Affiliation(s)
- Xiang Yang
- Department of Gynecology, 477093Central Hospital of Panyu District, Shiqiao, Guangzhou, People's Republic of China
| | - Kai-Xun Feng
- Department of Gynecology, 477093Central Hospital of Panyu District, Shiqiao, Guangzhou, People's Republic of China
| | - Hu Li
- Department of Gynecology, 477093Central Hospital of Panyu District, Shiqiao, Guangzhou, People's Republic of China
| | - Li Wang
- Department of Gynecology, 477093Central Hospital of Panyu District, Shiqiao, Guangzhou, People's Republic of China
| | - Hong Xia
- Department of Gynecology, 477093Central Hospital of Panyu District, Shiqiao, Guangzhou, People's Republic of China
| |
Collapse
|
10
|
Kazemi Fard T, Tavakoli S, Ahmadi R, Moradi N, Fadaei R, Mohammadi A, Fallah S. Evaluation of IP10 and miRNA 296-a Expression Levels in Peripheral Blood Mononuclear Cell of Coronary Artery Disease Patients and Controls. DNA Cell Biol 2020; 39:1678-1684. [PMID: 32716219 DOI: 10.1089/dna.2020.5650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Coronary artery disease (CAD) is the main cause of death worldwide. Atherosclerosis, the leading underlying cause of CAD, is a progressive inflammatory disease. miRNAs play a substantial role in inflammation. The aim of this study was to investigate the associations of peripheral blood mononuclear cells (PBMCs) gene expression of IP10 and miRNA 296-a and serum levels of IP10 and serum inflammatory cytokines interleukin-6 (IL-6) in CAD patients and controls. This is a case-control study conducted on 82 angiography confirmed CAD patients and 82 controls. PBMC expressions of miR-296a and IP10 were evaluated by real-time method, and serum concentrations of IL-6 and TNF-α were evaluated by enzyme-linked immunosorbent assay in the study population. A significant increase was found for serum IP10, IL-6, and TNF-α levels, and PBMC expression of IP10 and miRNA 296-a genes expression of CAD as comparison with controls. No significant correlation was found between IP10 gene expression and miRNA 296-a. A significant positive correlation was found between PBMC gene expression level of IP10 and serum concentrations of IP10 and cytokines IL-6 and TNF-α levels. Taking together, in PBMC of CAD patients, the IP10 and 296-a miRNA genes expression levels were increased significantly than controls. IP10, IL-6, and TNF-α levels in CAD patients were more than those in controls significantly. Concerning positive relationship between miRNA 296-a gene expression level and serum concentrations of IL-6 and TNF-α in CAD patients, it is proposed that IL-6 and TNF-α inhibitor could be the main targets of miRNA 296a and, thereby the IL-6 and TNF-α levels were increased; however, further study is needed.
Collapse
Affiliation(s)
- Toktam Kazemi Fard
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samareh Tavakoli
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nariman Moradi
- Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Asghar Mohammadi
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soudabeh Fallah
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
van der Sijde F, Vietsch EE, Mustafa DAM, Li Y, van Eijck CHJ. Serum miR-338-3p and miR-199b-5p are associated with the absolute neutrophil count in patients with resectable pancreatic cancer. Clin Chim Acta 2020; 505:183-189. [PMID: 32145274 DOI: 10.1016/j.cca.2020.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Several peripheral blood cell counts and immune ratios, such as the systemic immune-inflammation index (SII = platelet x neutrophil count/lymphocyte count) have prognostic value in patients with resectable pancreatic cancer (PDAC). Circulating microRNAs (miRNAs) are involved in various aspects of cancer and inflammation. We aimed to identify measurable circulating miRNAs in PDAC patients correlating with systemic inflammation parameters. METHODS A total of 42 PDAC patients was included in this study: twelve in the discovery (n = 6 SII low; n = 6 SII high) and 30 patients in the validation cohort (n = 19 SII low, n = 11 SII high). MiRNAs isolated from preoperative serum samples were measured with a 352 miRNA panel in the discovery cohort and individual miRNA primers in the validation cohort, using RT-qPCR (ID3EAL assays, MiRXES). RESULTS Only in the discovery cohort miR-328-3p, miR-338-3p, miR-1258 and miR-199b-5p were upregulated in high compared to low SII patients (fold difference ≥ 2, P<0.05). In the total cohort (n = 42) correlations were found between miR-338-3p (r = 0.48, P = 0.002) and miR-199b-5p (r = 0.44, P = 0.005) and the absolute neutrophil count. CONCLUSION Circulating miR-338-3p and miR-199b-5p are correlated to the neutrophil count in the blood of PDAC patients, suggesting a potential role of circulating miRNAs in cancer immune evasion and systemic inflammation.
Collapse
Affiliation(s)
- Fleur van der Sijde
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Eveline E Vietsch
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Tumor Immuno-Pathology Laboratory, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Yunlei Li
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands.
| |
Collapse
|
12
|
Han S, Underwood P, Hughes SJ. From tumor microenvironment communicants to biomarker discovery: Selectively packaged extracellular vesicular cargoes in pancreatic cancer. Cytokine Growth Factor Rev 2020; 51:61-68. [PMID: 32005635 PMCID: PMC8711854 DOI: 10.1016/j.cytogfr.2020.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/30/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023]
Abstract
Virtually all cells release various types of vesicles into the extracellular environment. These extracellular vesicles (EVs) transport molecular cargoes, performing as communicants for information exchange both within the tumor microenvironment (TME) and to distant organs. Thus, understanding the selective packaging of EV cargoes and the mechanistic impact of those cargoes - including metabolites, lipids, proteins, and/or nucleic acids - offers an opportunity to increase our knowledge of cancer biology and identify EV cargoes that might serve as cancer biomarkers in blood, saliva, or urine samples. In this review, we collect and organize recent advances in this field with an emphasis on pancreatic cancer (pancreatic adenocarcinoma, PDAC) and the concept that cells selectively package cargo into EVs. These studies demonstrate PDAC EV cargoes signal to reprogram and remodel the TME and impact distant organs. EV cargoes identified as potential PDAC diagnostic and prognostic biomarkers are summarized.
Collapse
Affiliation(s)
- Song Han
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, United States.
| | - Patrick Underwood
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Steven J Hughes
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, United States
| |
Collapse
|
13
|
Ayres Pereira M, Chio IIC. Metastasis in Pancreatic Ductal Adenocarcinoma: Current Standing and Methodologies. Genes (Basel) 2019; 11:E6. [PMID: 31861620 PMCID: PMC7016631 DOI: 10.3390/genes11010006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/11/2019] [Accepted: 12/17/2019] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is an extremely aggressive disease with a high metastatic potential. Most patients are diagnosed with metastatic disease, at which the five-year survival rate is only 3%. A better understanding of the mechanisms that drive metastasis is imperative for the development of better therapeutic interventions. Here, we take the reader through our current knowledge of the parameters that support metastatic progression in pancreatic ductal adenocarcinoma, and the experimental models that are at our disposal to study this process. We also describe the advantages and limitations of these models to study the different aspects of metastatic dissemination.
Collapse
Affiliation(s)
| | - Iok In Christine Chio
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA;
| |
Collapse
|
14
|
Zhou J, Hui X, Mao Y, Fan L. Identification of novel genes associated with a poor prognosis in pancreatic ductal adenocarcinoma via a bioinformatics analysis. Biosci Rep 2019; 39:BSR20190625. [PMID: 31311829 PMCID: PMC6680377 DOI: 10.1042/bsr20190625] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/01/2019] [Accepted: 07/12/2019] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a class of the commonest malignant carcinomas. The present study aimed to elucidate the potential biomarker and prognostic targets in PDAC. The array data of GSE41368, GSE43795, GSE55643, and GSE41369 were downloaded from Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) and differentially expressed microRNAs (DEmiRNAs) in PDAC were obtained by using GEO2R, and overlapped DEGs were acquired with Venn Diagrams. Functional enrichment analysis of overlapped DEGs and DEmiRNAs was conducted with Metascape and FunRich, respectively. The protein-protein interaction (PPI) network of overlapped DEGs was constructed by STRING and visualized with Cytoscape. Overall survival (OS) of DEmiRNAs and hub genes were investigated by Kaplan-Meier (KM) plotter (KM plotter). Transcriptional data and correlation analyses among hub genes were verified through GEPIA and Human Protein Atlas (HPA). Additionally, miRNA targets were searched using miRTarBase, then miRNA-DEG regulatory network was visualized with Cytoscape. A total of 32 DEmiRNAs and 150 overlapped DEGs were identified, and Metascape showed that DEGs were significantly enriched in cellular chemical homeostasis and pathways in cancer, while DEmiRNAs were mainly enriched in signal transduction and Glypican pathway. Moreover, seven hub genes with a high degree, namely, V-myc avian myelocytomatosis viral oncogene homolog (MYC), solute carrier family 2 member 1 (SLC2A1), PKM, plasminogen activator, urokinase (PLAU), peroxisome proliferator activated receptor γ (PPARG), MET proto-oncogene, receptor tyrosine kinase (MET), and integrin subunit α 3 (ITGA3), were identified and found to be up-regulated between PDAC and normal tissues. miR-135b, miR-221, miR-21, miR-27a, miR-199b-5p, miR-143, miR-196a, miR-655, miR-455-3p, miR-744 and hub genes predicted poor OS of PDAC. An integrative bioinformatics analysis identified several hub genes that may serve as potential biomarkers or targets for early diagnosis and precision target treatment of PDAC.
Collapse
Affiliation(s)
- Jun Zhou
- Department of General Ward 1, Zhejiang Hospital of Lingyin District, Zhejiang, China
| | - Xiaoliang Hui
- Department of General Ward 1, Zhejiang Hospital of Lingyin District, Zhejiang, China
| | - Ying Mao
- Department of General Ward 1, Zhejiang Hospital of Lingyin District, Zhejiang, China
| | - Liya Fan
- Department of Gastroenterology, Zhejiang Hospital of Sandun District, Zhejiang, China
| |
Collapse
|
15
|
Maity G, Haque I, Ghosh A, Dhar G, Gupta V, Sarkar S, Azeem I, McGregor D, Choudhary A, Campbell DR, Kambhampati S, Banerjee SK, Banerjee S. The MAZ transcription factor is a downstream target of the oncoprotein Cyr61/CCN1 and promotes pancreatic cancer cell invasion via CRAF-ERK signaling. J Biol Chem 2018; 293:4334-4349. [PMID: 29414775 PMCID: PMC5868262 DOI: 10.1074/jbc.ra117.000333] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/01/2018] [Indexed: 01/18/2023] Open
Abstract
Myc-associated zinc-finger protein (MAZ) is a transcription factor with dual roles in transcription initiation and termination. Deregulation of MAZ expression is associated with the progression of pancreatic ductal adenocarcinoma (PDAC). However, the mechanism of action of MAZ in PDAC progression is largely unknown. Here, we present evidence that MAZ mRNA expression and protein levels are increased in human PDAC cell lines, tissue samples, a subcutaneous tumor xenograft in a nude mouse model, and spontaneous cancer in the genetically engineered PDAC mouse model. We also found that MAZ is predominantly expressed in pancreatic cancer stem cells. Functional analysis indicated that MAZ depletion in PDAC cells inhibits invasive phenotypes such as the epithelial-to-mesenchymal transition, migration, invasion, and the sphere-forming ability of PDAC cells. Mechanistically, we detected no direct effects of MAZ on the expression of K-Ras mutants, but MAZ increased the activity of CRAF-ERK signaling, a downstream signaling target of K-Ras. The MAZ-induced activation of CRAF-ERK signaling was mediated via p21-activated protein kinase (PAK) and protein kinase B (AKT/PKB) signaling cascades and promoted PDAC cell invasiveness. Moreover, we found that the matricellular oncoprotein cysteine-rich angiogenic inducer 61 (Cyr61/CCN1) regulates MAZ expression via Notch-1-sonic hedgehog signaling in PDAC cells. We propose that Cyr61/CCN1-induced expression of MAZ promotes invasive phenotypes of PDAC cells not through direct K-Ras activation but instead through the activation of CRAF-ERK signaling. Collectively, these results highlight key molecular players in PDAC invasiveness and may help inform therapeutic strategies to improve clinical management and outcomes of PDAC.
Collapse
Affiliation(s)
- Gargi Maity
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Pathology and Laboratory Medicine, and
| | - Inamul Haque
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Pathology and Laboratory Medicine, and
| | - Arnab Ghosh
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Gopal Dhar
- From the Cancer Research Unit, Veterans Affairs Medical Center
| | | | - Sandipto Sarkar
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Imaan Azeem
- From the Cancer Research Unit, Veterans Affairs Medical Center
| | - Douglas McGregor
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Department of Pathology and Laboratory Medicine, and
- the Pathology Department, Veterans Affairs Medical Center, Kansas City, Missouri 64128
| | - Abhishek Choudhary
- the Gastroenterology Department, Veterans Affairs Medical Center, Kansas City, Missouri 64128
| | - Donald R Campbell
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the University of Missouri Kansas City and Saint Luke's Hospital of Kansas City, Kansas City, Missouri, and
| | - Suman Kambhampati
- From the Cancer Research Unit, Veterans Affairs Medical Center
- the Sarah Cannon Cancer Center at HCA Midwest Health, Kansas City, Missouri 64131
| | - Sushanta K Banerjee
- From the Cancer Research Unit, Veterans Affairs Medical Center,
- the Department of Pathology and Laboratory Medicine, and
- the Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Snigdha Banerjee
- From the Cancer Research Unit, Veterans Affairs Medical Center,
- the Department of Pathology and Laboratory Medicine, and
| |
Collapse
|
16
|
Fu Y, Liu S, Zeng S, Shen H. The critical roles of activated stellate cells-mediated paracrine signaling, metabolism and onco-immunology in pancreatic ductal adenocarcinoma. Mol Cancer 2018; 17:62. [PMID: 29458370 PMCID: PMC5817854 DOI: 10.1186/s12943-018-0815-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/12/2018] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignant diseases worldwide. It is refractory to conventional treatments, and consequently has a documented 5-year survival rate as low as 7%. Increasing evidence indicates that activated pancreatic stellate cells (PSCs), one of the stromal components in tumor microenvironment (TME), play a crucial part in the desmoplasia, carcinogenesis, aggressiveness, metastasis associated with PDAC. Despite the current understanding of PSCs as a "partner in crime" to PDAC, detailed regulatory roles of PSCs and related microenvironment remain obscure. In addition to multiple paracrine signaling pathways, recent research has confirmed that PSCs-mediated tumor microenvironment may influence behaviors of PDAC via diverse mechanisms, such as rewiring metabolic networks, suppressing immune responses. These new activities are closely linked with treatment and prognosis of PDAC. In this review, we discuss the recent advances regarding new functions of activated PSCs, including PSCs-cancer cells interaction, mechanisms involved in immunosuppressive regulation, and metabolic reprogramming. It's clear that these updated experimental or clinical studies of PSCs may provide a promising approach for PDAC treatment in the near future.
Collapse
Affiliation(s)
- Yaojie Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Shanshan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
17
|
Han S, Gonzalo DH, Feely M, Rinaldi C, Belsare S, Zhai H, Kalra K, Gerber MH, Forsmark CE, Hughes SJ. Stroma-derived extracellular vesicles deliver tumor-suppressive miRNAs to pancreatic cancer cells. Oncotarget 2018; 9:5764-5777. [PMID: 29464032 PMCID: PMC5814172 DOI: 10.18632/oncotarget.23532] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/28/2017] [Indexed: 01/18/2023] Open
Abstract
The biology of tumor-associated stroma (TAS) in pancreatic ductal adenocarcinoma (PDAC) is not well understood. The paradoxical observation that stroma-depletion strategies lead to progression of PDAC reinforced the need to critically evaluate the functional contribution of TAS in the initiation and progression of PDAC. PDAC and TAS cells are unique in their expression of specific miRNAs, and this specific miRNA expression pattern alters host to tumor microenvironment interactions. Using primary human pancreatic TAS cells and primary xenograft PDAC cells co-culture, we provide evidence of miRNA trafficking and exchanging between TAS and PDAC cells, in a two-way, cell-contact independent fashion, via extracellular vesicles (EVs) transportation. Selective packaging of miRNAs into EVs led to enrichment of stromal specific miR-145 in EVs secreted by TAS cells. Exosomes, but not microvesicles, derived from human TAS cells demonstrated a tumor suppressive role by inducing PDAC cell apoptosis. This effect was mitigated by anti-miR-145 sequences. Our data suggest that TAS-derived miRNAs are delivered to adjacent PDAC cells via exosomes and suppress tumor cell growth. These data highlight that TAS cells secrete exosomes carrying tumor suppressive genetic materials, a possible anti-tumor capacity. Future work of the development of patient-derived exosomes could have therapeutic implications for unresectable PDAC.
Collapse
Affiliation(s)
- Song Han
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - David H. Gonzalo
- Department of Pathology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Michael Feely
- Department of Pathology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Carlos Rinaldi
- Department of Biomedical Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Sayali Belsare
- Department of Biomedical Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | | | | | - Michael H. Gerber
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christopher E. Forsmark
- Division of Gastroenterology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Steven J. Hughes
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
18
|
Karamitopoulou E, Haemmig S, Baumgartner U, Schlup C, Wartenberg M, Vassella E. MicroRNA dysregulation in the tumor microenvironment influences the phenotype of pancreatic cancer. Mod Pathol 2017; 30:1116-1125. [PMID: 28548126 DOI: 10.1038/modpathol.2017.35] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 01/18/2023]
Abstract
Cellular interactions in the tumor microenvironment influence neoplastic progression in pancreatic ductal adenocarcinoma. One underlying mechanism is the induction of the prognostically unfavorable epithelial-mesenchymal-transition-like tumor budding. Our aim is to explore the expression of microRNAs implicated in the regulation of tumor budding focusing on the microenvironment of the invasive front. To this end, RNA from laser-capture-microdissected material of the main tumor, tumor buds, juxta-tumoral stroma, tumor-remote stroma, and non-neoplastic pancreatic parenchyma from pancreatic cancer cases with (n=7) and without (n=6) tumor budding was analyzed by qRT-PCR for the expression of a panel of miRNAs that are known to be implicated in the regulation of epithelial-mesenchymal transition, including miR-21, miR-183, miR-200b, miR-200c, miR-203, miR-205, miR-210, and miR-217. Here we show that at the invasive front of pancreatic ductal adenocarcinoma, specific microRNAs, are differentially expressed between tumor buds and main tumor cells and between cases with and without tumor budding, indicating their involvement in the regulation of the budding phenotype. Notably, miR-200b and miR-200c were significantly downregulated in the tumor buds. Consistent with this finding, they negatively correlated with the expression of epithelial-mesenchymal-transition-associated E-cadherin repressors ZEB1 and ZEB2 in the budding cells (P<0.001). Interestingly, many microRNAs were also dysregulated in juxta-tumoral compared to tumor-remote stroma suggesting that juxta-tumoral stroma contributes to microRNA dysregulation. Notably, miR-200b and miR-200c were strongly downregulated while miR-210 and miR-21 were upregulated in the juxta-tumoral vs tumor-remote stroma in carcinomas with tumor budding. In conclusion, microRNA targeting in both tumor and stromal cells could represent a treatment option for aggressive pancreatic cancer.
Collapse
Affiliation(s)
- Eva Karamitopoulou
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Stefan Haemmig
- Molecular Pathology Unit, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Ulrich Baumgartner
- Molecular Pathology Unit, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Cornelia Schlup
- Molecular Pathology Unit, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Martin Wartenberg
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Erik Vassella
- Molecular Pathology Unit, Institute of Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Zhan Y, Zheng N, Teng F, Bao L, Liu F, Zhang M, Guo M, Guo W, Ding G, Wang Q. MiR-199a/b-5p inhibits hepatocellular carcinoma progression by post-transcriptionally suppressing ROCK1. Oncotarget 2017; 8:67169-67180. [PMID: 28978024 PMCID: PMC5620164 DOI: 10.18632/oncotarget.18052] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/26/2017] [Indexed: 01/18/2023] Open
Abstract
In this study, we explored the actions of miR-199a/b-5p during hepatocellular carcinoma (HCC) progression and its potential target genes. Through heatmap miRNA expression analysis of 15 matched HCC tumor and adjacent non-tumor liver tissues from the TCGA database, we detected 19 mRNAs that were upregulated and 13 that were downregulated specifically in HCC. Among these, miR-199 family members were downregulated in HCC tumors and cell lines, as compared to controls. Low miR-199a/b-5p expression was also associated with poor overall survival of HCC patients. miR-199a/b-5p overexpression in HCC cell lines inhibited cell proliferation, migration and invasion, both in vitro and in vivo. In addition, miR199-a/b-5p post-transcriptionally suppressed Rho-associated coiled-coil kinase 1 (ROCK1). This in turn led to inhibition of ROCK1/MLC and PI3K/Akt signaling, which is necessary for HCC proliferation and metastasis. These results indicate that miR-199a/b acts as tumor suppressors in HCC and represent promising therapeutic targets.
Collapse
Affiliation(s)
- Yangyang Zhan
- Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai 200433, China
| | - NanXin Zheng
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Fei Teng
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Leilei Bao
- Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai 200433, China.,Department of Pharmacy, No. 411 Hospital of PLA, Shanghai 200080, China
| | - Fang Liu
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Mingjian Zhang
- Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai 200433, China
| | - Meng Guo
- Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai 200433, China.,Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Wenyuan Guo
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Guoshan Ding
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Quanxing Wang
- Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
20
|
Zhu X, Luo W, Liang W, Tang F, Bei C, Ren Y, Qin L, Tan C, Zhang Y, Tan S. Overexpression and clinical significance of MYC-associated zinc finger protein in pancreatic carcinoma. Onco Targets Ther 2016; 9:7493-7501. [PMID: 28008270 PMCID: PMC5167488 DOI: 10.2147/ott.s124118] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
This study aimed to investigate the expression and clinical significance of MYC-associated zinc finger protein (MAZ) in pancreatic carcinoma (PC), and the biological functions of MAZ in PC cells. MAZ expression was detected in 57 PC tissues and 41 paired adjacent nontumor tissues by immunohistochemistry. Compared to the expression in adjacent nontumor tissues, MAZ was significantly higher expressed in PC tissues (P<0.0001). In addition, MAZ expression had a significant correlation with certain clinical characteristics of PC patients, such as age, tumor diameter, tumor number, and the serum level of CA199 (P<0.05). The survival analysis showed that the survival time of PC patients with high expression of MAZ was significantly lower than patients with low expression of MAZ (P=0.0365). After MAZ was knocked down in PANC-1 cells by RNA interference, the cells’ ability to proliferate, invade, and migrate was decreased significantly (P<0.01). Moreover, MAZ expression was found to be associated with Ki-67, a cell proliferation marker, in PC tissues, further supporting the idea that MAZ promotes PC cell proliferation. Our study clarifies an oncogenic role of MAZ in pathogenesis of PC and provides MAZ as a biomarker in the treatment and prognosis of PC.
Collapse
Affiliation(s)
- Xiaonian Zhu
- School of Public Health, Guilin Medical University, Guilin, People's Republic of China
| | - Wei Luo
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, People's Republic of China
| | - Wenjin Liang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, People's Republic of China
| | - Fen Tang
- Department of Hepatology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, People's Republic of China
| | - Chunhua Bei
- School of Public Health, Guilin Medical University, Guilin, People's Republic of China
| | - Yuan Ren
- School of Public Health, Guilin Medical University, Guilin, People's Republic of China
| | - Linyuan Qin
- School of Public Health, Guilin Medical University, Guilin, People's Republic of China
| | - Chao Tan
- School of Public Health, Guilin Medical University, Guilin, People's Republic of China
| | - Ying Zhang
- School of Public Health, Guilin Medical University, Guilin, People's Republic of China
| | - Shengkui Tan
- School of Public Health, Guilin Medical University, Guilin, People's Republic of China
| |
Collapse
|