1
|
Kalra A, Meltzer SJ. The Role of DNA Methylation in Gastrointestinal Disease: An Expanded Review of Malignant and Nonmalignant Gastrointestinal Diseases. Gastroenterology 2025; 168:245-266. [PMID: 38971197 PMCID: PMC11698954 DOI: 10.1053/j.gastro.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Esophageal, colorectal, pancreatic, hepatocellular, and gastric cancer together impact millions of patients worldwide each year, with high overall mortality rates, and are increasing in incidence. Additionally, premalignant gastrointestinal diseases, such as Barrett's esophagus and inflammatory bowel disease, are also increasing in incidence. However, involvement of aberrant DNA methylation in these diseases is incompletely understood, especially given recent research advancements in this field. Here, we review knowledge of this epigenetic mechanism in gastrointestinal preneoplasia and neoplasia, considering mechanisms of action, genetic and environmental factors, and 5'-C-phosphate-G-3' island methylator phenotype. We also highlight developments in translational research, focusing on genomic-wide data, methylation-based biomarkers and diagnostic tests, machine learning, and therapeutic epigenetic strategies.
Collapse
Affiliation(s)
- Andrew Kalra
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Stephen J Meltzer
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
2
|
Corradi C, Gentiluomo M, Adsay V, Sainz J, Camisa PR, Wlodarczyk B, Crippa S, Tavano F, Capurso G, Campa D. Multi-omic markers of intraductal papillary mucinous neoplasms progression into pancreatic cancer. Semin Cancer Biol 2025; 109:25-43. [PMID: 39733817 DOI: 10.1016/j.semcancer.2024.12.005] [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: 10/23/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal and common form of pancreatic cancer, it has no specific symptoms, and most of the patients are diagnosed when the disease is already at an advanced stage. Chemotherapy typically has only a modest effect, making surgery the most effective treatment option. However, only a small percentage of patients are amenable to surgery. One viable strategy to reduce PDAC death burden associated with the disease is to focus on precursor lesions and identify markers able to predict who will evolve into PDAC. While most PDACs are believed to be preceded by pancreatic intraepithelial neoplasms (PanINs), 5-10 % arise from Intraductal papillary mucinous neoplasms (IPMNs), which are mass-forming cystic lesions that are very common in the general population. IPMNs offer an invaluable model of pancreatic carcinogenesis for researchers to analyse, as well as a target population for PDAC early detection by clinicians. The evolution of IPMN into cancer is a complex and multistep process, therefore the identification of individual markers will not be the solution. In recent years, multiple omics technologies have been instrumental to identify possible biomarkers of IPMN progression and carcinogenesis. The only foreseeable strategy will be to integrate multi-omics data, alongside clinical and morphological features, into a progression score or signature using either standard epidemiologic tools or artificial intelligence. The aim of this manuscript is to review the current knowledge on genetic biomarkers and to briefly mention also additional omics, such as metabolomics, the exposome, the miRNome and epigenomics of IPMNs.
Collapse
Affiliation(s)
| | | | - Volkan Adsay
- Department of Pathology, Koç University School of Medicine and Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Juan Sainz
- Department of Biochemistry and Molecular Biology, University of Granada, Granada, Spain
| | - Paolo Riccardo Camisa
- Division of Pancreatic Surgery and Transplantation, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Barbara Wlodarczyk
- Department of Digestive Tract Diseases, Medical University of Lodz, Lodz, Poland
| | - Stefano Crippa
- Division of Pancreatic Surgery and Transplantation, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Francesca Tavano
- Division of Gastroenterology and Research Laboratory, Fondazione IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Italy
| | - Gabriele Capurso
- Vita-Salute San Raffaele University, Milan, Italy; Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy.
| |
Collapse
|
3
|
Przybyszewski O, Mik M, Nowicki M, Kusiński M, Mikołajczyk-Solińska M, Śliwińska A. Using microRNAs Networks to Understand Pancreatic Cancer-A Literature Review. Biomedicines 2024; 12:1713. [PMID: 39200178 PMCID: PMC11351910 DOI: 10.3390/biomedicines12081713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic cancer is a severe disease, challenging to diagnose and treat, and thereby characterized by a poor prognosis and a high mortality rate. Pancreatic ductal adenocarcinoma (PDAC) represents approximately 90% of pancreatic cancer cases, while other cases include neuroendocrine carcinoma. Despite the growing knowledge of the pathophysiology of this cancer, the mortality rate caused by it has not been effectively reduced. Recently, microRNAs have aroused great interest among scientists and clinicians, as they are negative regulators of gene expression, which participate in many processes, including those related to the development of pancreatic cancer. The aim of this review is to show how microRNAs (miRNAs) affect key signaling pathways and related cellular processes in pancreatic cancer development, progression, diagnosis and treatment. We included the results of in vitro studies, animal model of pancreatic cancer and those performed on blood, saliva and tumor tissue isolated from patients suffering from PDAC. Our investigation identified numerous dysregulated miRNAs involved in KRAS, JAK/STAT, PI3/AKT, Wnt/β-catenin and TGF-β signaling pathways participating in cell cycle control, proliferation, differentiation, apoptosis and metastasis. Moreover, some miRNAs (miRNA-23a, miRNA-24, miRNA-29c, miRNA-216a) seem to be engaged in a crosstalk between signaling pathways. Evidence concerning the utility of microRNAs in the diagnosis and therapy of this cancer is poor. Therefore, despite growing knowledge of the involvement of miRNAs in several processes associated with pancreatic cancer, we are beginning to recognize and understand their role and usefulness in clinical practice.
Collapse
Affiliation(s)
- Oskar Przybyszewski
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland
| | - Michał Mik
- Department of General and Colorectal Surgery, Medical University of Lodz, 113 Stefana Żeromskiego St., 90-549 Lodz, Poland; (M.M.); (M.N.)
| | - Michał Nowicki
- Department of General and Colorectal Surgery, Medical University of Lodz, 113 Stefana Żeromskiego St., 90-549 Lodz, Poland; (M.M.); (M.N.)
| | - Michał Kusiński
- Department of Endocrinological, General and Oncological Surgery, Medical University of Lodz, 62 Pabianicka St., 93-513 Lodz, Poland;
| | - Melania Mikołajczyk-Solińska
- Department of Internal Medicine, Diabetology and Clinical Pharmacology, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland;
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska St., 92-213 Lodz, Poland
| |
Collapse
|
4
|
Zhang H, Han B, Tian S, Gong Y, Chen L, Liu L. HOXC4 promotes proliferation of pancreatic cancer cells by increasing LDHA-mediated glycolysis. Aging (Albany NY) 2024; 16:11103-11116. [PMID: 38990159 PMCID: PMC11272123 DOI: 10.18632/aging.206008] [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: 10/30/2023] [Accepted: 05/03/2024] [Indexed: 07/12/2024]
Abstract
Homeobox C4 (HOXC4) is a member of homeobox family and acts as a transcription factor in regulating morphological development. The current study aimed to determine its role in pancreatic cancer (PC). Bioinformatics analysis was employed to assess the expression and clinical significance of HOXC4 in PC, while the expression of HOXC4 was further confirmed in PC tissues through quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). The impact of HOXC4 on PC cell proliferation was evaluated using various assays including Cell Counting Kit-8, colony formation, apoptosis detection, cell cycle analysis, and subcutaneous tumorigenesis. Extracellular acidification rate, glucose uptake, and lactate production measurements were detected to examine the impact of HOXC4 on glycolysis. The relationship between HOXC4 and lactate dehydrogenase A (LDHA) was investigated using CHIP assay, luciferase reporter assay, and western blot. Notably, there was a substantial increase in HOXC4 expression in PC, and patients with elevated HOXC4 levels exhibited shorter survival durations. HOXC4 knockdown resulted in significantly reduced proliferation and colony formation in PC cells, accompanied by increased apoptosis and G1 phase arrest. The overexpression of HOXC4 resulted in contrasting effects. In vivo, the proliferation of PC cells was diminished upon the knockdown of HOXC4. HOXC4 exhibited an increase in LDHA expression by binding to its promoter. The suppressive effects of HOXC4 knockdown on PC cells were counteracted upon the restoration of LDHA. In conclusion, HOXC4 promoted the proliferation of PC cells by increasing LDHA-mediated glycolysis. HOXC4 can act as a target for PC therapy.
Collapse
Affiliation(s)
- Hao Zhang
- College of Clinical Medicine, Guizhou Medical University, Guiyang, China
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bing Han
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - She Tian
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yongjun Gong
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Liwen Chen
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Li Liu
- School of Public Health, Guizhou Medical University, Guiyang, China
| |
Collapse
|
5
|
Silva LGDO, Lemos FFB, Luz MS, Rocha Pinheiro SL, Calmon MDS, Correa Santos GL, Rocha GR, de Melo FF. New avenues for the treatment of immunotherapy-resistant pancreatic cancer. World J Gastrointest Oncol 2024; 16:1134-1153. [PMID: 38660642 PMCID: PMC11037047 DOI: 10.4251/wjgo.v16.i4.1134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/26/2024] [Accepted: 03/04/2024] [Indexed: 04/10/2024] Open
Abstract
Pancreatic cancer (PC) is characterized by its extremely aggressive nature and ranks 14th in the number of new cancer cases worldwide. However, due to its complexity, it ranks 7th in the list of the most lethal cancers worldwide. The pathogenesis of PC involves several complex processes, including familial genetic factors associated with risk factors such as obesity, diabetes mellitus, chronic pancreatitis, and smoking. Mutations in genes such as KRAS, TP53, and SMAD4 are linked to the appearance of malignant cells that generate pancreatic lesions and, consequently, cancer. In this context, some therapies are used for PC, one of which is immunotherapy, which is extremely promising in various other types of cancer but has shown little response in the treatment of PC due to various resistance mechanisms that contribute to a drop in immunotherapy efficiency. It is therefore clear that the tumor microenvironment (TME) has a huge impact on the resistance process, since cellular and non-cellular elements create an immunosuppressive environment, characterized by a dense desmoplastic stroma with cancer-associated fibroblasts, pancreatic stellate cells, extracellular matrix, and immunosuppressive cells. Linked to this are genetic mutations in TP53 and immunosuppressive factors that act on T cells, resulting in a shortage of CD8+ T cells and limited expression of activation markers such as interferon-gamma. In this way, finding new strategies that make it possible to manipulate resistance mechanisms is necessary. Thus, techniques such as the use of TME modulators that block receptors and stromal molecules that generate resistance, the use of genetic manipulation in specific regions, such as microRNAs, the modulation of extrinsic and intrinsic factors associated with T cells, and, above all, therapeutic models that combine these modulation techniques constitute the promising future of PC therapy. Thus, this study aims to elucidate the main mechanisms of resistance to immunotherapy in PC and new ways of manipulating this process, resulting in a more efficient therapy for cancer patients and, consequently, a reduction in the lethality of this aggressive cancer.
Collapse
Affiliation(s)
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Samuel Luca Rocha Pinheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Mariana dos Santos Calmon
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Gabriel Lima Correa Santos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Gabriel Reis Rocha
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
6
|
Izdebska WM, Daniluk J, Niklinski J. Microbiome and MicroRNA or Long Non-Coding RNA-Two Modern Approaches to Understanding Pancreatic Ductal Adenocarcinoma. J Clin Med 2023; 12:5643. [PMID: 37685710 PMCID: PMC10488817 DOI: 10.3390/jcm12175643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of humans' most common and fatal neoplasms. Nowadays, a number of PDAC studies are being conducted in two different fields: non-coding RNA (especially microRNA and long non-coding RNA) and microbiota. It has been recently discovered that not only does miRNA affect particular bacteria in the gut microbiome that can promote carcinogenesis in the pancreas, but the microbiome also has a visible impact on the miRNA. This suggests that it is possible to use the combined impact of the microbiome and noncoding RNA to suppress the development of PDAC. Nevertheless, insufficient research has focused on bounding both approaches to the diagnosis, treatment, and prevention of pancreatic ductal adenocarcinoma. In this article, we summarize the recent literature on the molecular basis of carcinogenesis in the pancreas, the two-sided impact of particular types of non-coding RNA and the pancreatic cancer microbiome, and possible medical implications of the discovered phenomenon.
Collapse
Affiliation(s)
- Wiktoria Maria Izdebska
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Jaroslaw Daniluk
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Jacek Niklinski
- Department of Clinical Molecular Biology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
7
|
Shenoy US, Adiga D, Kabekkodu SP, Hunter KD, Radhakrishnan R. Molecular implications of HOX genes targeting multiple signaling pathways in cancer. Cell Biol Toxicol 2022; 38:1-30. [PMID: 34617205 PMCID: PMC8789642 DOI: 10.1007/s10565-021-09657-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/10/2021] [Indexed: 11/17/2022]
Abstract
Homeobox (HOX) genes encode highly conserved homeotic transcription factors that play a crucial role in organogenesis and tissue homeostasis. Their deregulation impacts the function of several regulatory molecules contributing to tumor initiation and progression. A functional bridge exists between altered gene expression of individual HOX genes and tumorigenesis. This review focuses on how deregulation in the HOX-associated signaling pathways contributes to the metastatic progression in cancer. We discuss their functional significance, clinical implications and ascertain their role as a diagnostic and prognostic biomarker in the various cancer types. Besides, the mechanism of understanding the theoretical underpinning that affects HOX-mediated therapy resistance in cancers has been outlined. The knowledge gained shall pave the way for newer insights into the treatment of cancer.
Collapse
Affiliation(s)
- U Sangeetha Shenoy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Keith D Hunter
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S10 2TA, UK
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
8
|
Malaab M, Renaud L, Takamura N, Zimmerman KD, da Silveira WA, Ramos PS, Haddad S, Peters-Golden M, Penke LR, Wolf B, Hardiman G, Langefeld CD, Medsger TA, Feghali-Bostwick CA. Antifibrotic factor KLF4 is repressed by the miR-10/TFAP2A/TBX5 axis in dermal fibroblasts: insights from twins discordant for systemic sclerosis. Ann Rheum Dis 2022; 81:268-277. [PMID: 34750102 PMCID: PMC8758541 DOI: 10.1136/annrheumdis-2021-221050] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/29/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVES Systemic sclerosis (SSc) is a complex disease of unknown aetiology in which inflammation and fibrosis lead to multiple organ damage. There is currently no effective therapy that can halt the progression of fibrosis or reverse it, thus studies that provide novel insights into disease pathogenesis and identify novel potential therapeutic targets are critically needed. METHODS We used global gene expression and genome-wide DNA methylation analyses of dermal fibroblasts (dFBs) from a unique cohort of twins discordant for SSc to identify molecular features of this pathology. We validated the findings using in vitro, ex vivo and in vivo models. RESULTS Our results revealed distinct differentially expressed and methylated genes, including several transcription factors involved in stem cell differentiation and developmental programmes (KLF4, TBX5, TFAP2A and homeobox genes) and the microRNAs miR-10a and miR-10b which target several of these deregulated genes. We show that KLF4 expression is reduced in SSc dFBs and its expression is repressed by TBX5 and TFAP2A. We also show that KLF4 is antifibrotic, and its conditional knockout in fibroblasts promotes a fibrotic phenotype. CONCLUSIONS Our data support a role for epigenetic dysregulation in mediating SSc susceptibility in dFBs, illustrating the intricate interplay between CpG methylation, miRNAs and transcription factors in SSc pathogenesis, and highlighting the potential for future use of epigenetic modifiers as therapies.
Collapse
Affiliation(s)
- Maya Malaab
- Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ludivine Renaud
- Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Naoko Takamura
- Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kip D Zimmerman
- Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Willian A da Silveira
- School of Biological Sciences, Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| | - Paula S Ramos
- Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sandra Haddad
- Science, Bay Path University, Longmeadow, Massachusetts, USA
| | - Marc Peters-Golden
- Internal Medicine, University of Michigan Michigan Medicine, Ann Arbor, Michigan, USA
| | - Loka R Penke
- Internal Medicine, University of Michigan Michigan Medicine, Ann Arbor, Michigan, USA
| | - Bethany Wolf
- Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gary Hardiman
- School of Biological Sciences, Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| | - Carl D Langefeld
- Biostatistical Sciences and Center for Public Health Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Thomas A Medsger
- Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
9
|
Palamaris K, Felekouras E, Sakellariou S. Epithelial to Mesenchymal Transition: Key Regulator of Pancreatic Ductal Adenocarcinoma Progression and Chemoresistance. Cancers (Basel) 2021; 13:cancers13215532. [PMID: 34771695 PMCID: PMC8582651 DOI: 10.3390/cancers13215532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma’s (PDAC) dismal prognosis is associated with its aggressive biological behavior and resistance to chemotherapy. Epithelial to mesenchymal transition (EMT) has been recognized as a key driver of PDAC progression and development of drug resistance. EMT is a transient and reversible process leading to transdifferentiation of epithelial cells into a more mesenchymal phenotype. It is regulated by multiple signaling pathways that control the activity of a transcription factors network. Activation of EMT in pre-invasive stages of PDAC has been accused for early dissemination. Furthermore, it contributes to the development of intratumoral heterogeneity and drug resistance. This review summarizes the available data regarding signaling networks regulating EMT and describes the integral role of EMT in different aspects of PDAC pathogenesis. Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies, characterized by aggressive biological behavior and a lack of response to currently available chemotherapy. Emerging evidence has identified epithelial to mesenchymal transition (EMT) as a key driver of PDAC progression and a central regulator in the development of drug resistance. EMT is a reversible transdifferentiation process controlled by complex interactions between multiple signaling pathways such as TGFb, Wnt, and Notch, which converge to a network of specific transcription factors. Activation of EMT transcriptional reprogramming converts cancer cells of epithelial differentiation into a more mesenchymal phenotypic state. EMT occurrence in pre-invasive pancreatic lesions has been implicated in early PDAC dissemination. Moreover, cancer cell phenotypic plasticity driven by EMT contributes to intratumoral heterogeneity and drug tolerance and is mechanistically associated with the emergence of cells exhibiting cancer stem cells (CSCs) phenotype. In this review we summarize the available data on the signaling cascades regulating EMT and the molecular isnteractions between pancreatic cancer and stromal cells that activate them. In addition, we provide a link between EMT, tumor progression, and chemoresistance in PDAC.
Collapse
Affiliation(s)
- Kostas Palamaris
- 1ST Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Evangelos Felekouras
- 1ST Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Stratigoula Sakellariou
- 1ST Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Correspondence:
| |
Collapse
|
10
|
Smolarz B, Durczyński A, Romanowicz H, Hogendorf P. The Role of microRNA in Pancreatic Cancer. Biomedicines 2021; 9:biomedicines9101322. [PMID: 34680441 PMCID: PMC8533140 DOI: 10.3390/biomedicines9101322] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are small ribonucleic acid molecules that play a key role in regulating gene expression. The increasing number of studies undertaken on the functioning of microRNAs in the tumor formation clearly indicates their important potential in oncological therapy. Pancreatic cancer is one of the deadliest cancers. The expression of miRNAs released into the bloodstream appears to be a good indicator of progression and evaluation of the aggressiveness of pancreatic cancer, as indicated by studies. The work reviewed the latest literature on the importance of miRNAs for pancreatic cancer development.
Collapse
Affiliation(s)
- Beata Smolarz
- Laboratory of Cancer Genetics, Department of Pathology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
- Correspondence: ; Tel.: +48-42-271-1290
| | - Adam Durczyński
- Department of General and Transplant Surgery, N. Barlicki Memorial Clinical Hospital, Medical University of Lodz, 90-153 Lodz, Poland; (A.D.); (P.H.)
| | - Hanna Romanowicz
- Laboratory of Cancer Genetics, Department of Pathology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
| | - Piotr Hogendorf
- Department of General and Transplant Surgery, N. Barlicki Memorial Clinical Hospital, Medical University of Lodz, 90-153 Lodz, Poland; (A.D.); (P.H.)
| |
Collapse
|
11
|
Guo H, Li C, Su X, Huang X. A Five-mRNA Expression Signature to Predict Survival in Oral Squamous Cell Carcinoma by Integrated Bioinformatic Analyses. Genet Test Mol Biomarkers 2021; 25:517-527. [PMID: 34406843 PMCID: PMC8403201 DOI: 10.1089/gtmb.2021.0066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objectives: This study was designed to identify a messenger RNA (mRNA) expression signature to predict survival in patients with oral squamous cell carcinoma (OSCC). Methods: mRNA expression profiles were integrated with clinical data from 280 samples, including 19 normal tissues and 261 OSCC tissues in The Cancer Genome Atlas. We identified differentially expressed mRNAs (DEmRNAs) between the OSCC and normal tissue samples and developed a novel mRNA-focused expression signature using a Cox regression analysis and other bioinformatic methods. The prognostic value of this signature was evaluated by Kaplan–Meier analysis, multivariable COX regression, and receiver operating characteristic (ROC) curve analysis. Protein–protein interaction (PPI) network, gene ontology, and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed to predict the function of the DEmRNAs. Signature-related mRNAs were analyzed by gene set enrichment analyses (GSEA) and validated by quantitative real-time polymerase chain reaction (qRT-PCR) in 20 paired OSCC and adjacent healthy tissues. Results: We identified a novel 5-mRNA expression signature (HOXA1, CELSR3, HIST1H3J, ZFP42, and ASCL4) that could predict patient outcomes in OSCC. The risk score based on the signature was able to separate OSCC patients into high- and low-risk groups that showed significantly different overall survival (p < 0.001, log-rank test). The signature was further validated as an effective independent prognostic predictor of OSCC by multivariate Cox regression analysis (hazard ratio = 3.747, confidence interval: 2.279–5.677, p < 0.001) and ROC curve of the third year (area under the curve = 0.733). Functional analysis demonstrated that the key hub genes in the PPI network were mainly enriched in cell division, cell proliferation, and the p53 signaling pathway. GSEA results showed that the 5 mRNAs were significantly enriched in mismatch repair, DNA replication, and the NOTCH signaling pathway. Finally, qRT-PCR results showed that the 5 mRNAs were upregulated in OSCC tissue in agreement with the predictions from our bioinformatics analysis. Conclusions: We identified a novel 5-mRNA signature that could predict the survival of patients with OSCC and may be a promising biomarker for personalized cancer treatments.
Collapse
Affiliation(s)
- Hejia Guo
- Guangxi Medical University College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Guangxi Key Laboratory of the Rehabilitation and Reconstruction of Oral and Maxillofacial Research, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Guangxi Colleges and Universities Key Laboratory of Treatment and Research for Oral and Maxillofacial Surgery Disease, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Medical Scientific Research Center, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Department of Oral and Maxillofacial Surgery, the Affiliated Stomatology Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Cuiping Li
- Guangxi Medical University College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Guangxi Key Laboratory of the Rehabilitation and Reconstruction of Oral and Maxillofacial Research, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Guangxi Colleges and Universities Key Laboratory of Treatment and Research for Oral and Maxillofacial Surgery Disease, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Medical Scientific Research Center, College of Stomatology, Guangxi Medical University, Nanning, P.R. China
| | - Xiaoping Su
- Guangxi Medical University College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Guangxi Key Laboratory of the Rehabilitation and Reconstruction of Oral and Maxillofacial Research, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Guangxi Colleges and Universities Key Laboratory of Treatment and Research for Oral and Maxillofacial Surgery Disease, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Medical Scientific Research Center, College of Stomatology, Guangxi Medical University, Nanning, P.R. China
| | - Xuanping Huang
- Guangxi Medical University College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Guangxi Key Laboratory of the Rehabilitation and Reconstruction of Oral and Maxillofacial Research, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Guangxi Colleges and Universities Key Laboratory of Treatment and Research for Oral and Maxillofacial Surgery Disease, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Medical Scientific Research Center, College of Stomatology, Guangxi Medical University, Nanning, P.R. China.,Department of Oral and Maxillofacial Surgery, the Affiliated Stomatology Hospital of Guangxi Medical University, Nanning, P.R. China
| |
Collapse
|
12
|
Yang W, Chen L, Xu L, Bilotta AJ, Yao S, Liu Z, Cong Y. MicroRNA-10a Negatively Regulates CD4 + T Cell IL-10 Production through Suppression of Blimp1. THE JOURNAL OF IMMUNOLOGY 2021; 207:985-995. [PMID: 34301843 DOI: 10.4049/jimmunol.2100017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/27/2021] [Indexed: 11/19/2022]
Abstract
An uncontrolled CD4+ T cell response is a critical hallmark of autoimmune diseases. IL-10, which can be produced by both effector and regulatory CD4+ T cells, plays an essential role in the inhibition of autoimmunity. MicroRNAs are key molecules involved in regulating immune responses. However, how miR-10a regulates CD4+ T cell function in the pathogenesis of intestinal immune responses is not fully understood. In this study, we show that the mice with deficient miR-10a in CD4+ T cells were more resistant to intestinal inflammation upon inflammatory insult. miR-10a-deficient CD4+CD45Rbhi T cells were less colitogenic in Rag -/- mice, in which CD4+ T cell production of IL-10 was increased. miR-10a-deficient CD4+ T cells expressed a higher expression of IL-10 in vitro. Blocking the IL-10/IL-10R pathway in vivo aggravated colitis induced by miR-10a-deficient CD4+CD45Rbhi T cells. Mechanically, miR-10a suppressed CD4+ T cell production of IL-10 through targeting Prdm1, which encodes Blimp1. We further show that that CD4+ T cells lacking Blimp1 produced lower levels of IL-10 and induced more severe colitis in Rag -/- mice. These data thus establish the role of miR-10a in the inhibition of IL-10 production in CD4+ T cells to regulate intestinal homeostasis.
Collapse
Affiliation(s)
- Wenjing Yang
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX
| | - Liang Chen
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Shanghai, China; and
| | - Leiqi Xu
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX
| | - Anthony J Bilotta
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX
| | - Suxia Yao
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX
| | - Zhanju Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Shanghai, China; and
| | - Yingzi Cong
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, Galveston, TX; .,Department of Pathology, The University of Texas Medical Branch at Galveston, Galveston, TX
| |
Collapse
|
13
|
Donovan PD, McHale NM, Venø MT, Prehn JHM. tsRNAsearch: A pipeline for the identification of tRNA and ncRNA fragments from small RNA-sequencing data. Bioinformatics 2021; 37:4424-4430. [PMID: 34255836 DOI: 10.1093/bioinformatics/btab515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 05/27/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022] Open
Abstract
MOTIVATION tRNAs were originally considered uni-functional RNA molecules involved in the delivery of amino acids to growing peptide chains on the ribosome. More recently, the liberation of tRNA fragments from tRNAs via specific enzyme cleavage has been characterized. Detection of tRNA fragments in sequencing data is difficult due to tRNA sequence redundancy and the short length of both tRNAs and their fragments. RESULTS Here we introduce tsRNAsearch, a Nextflow pipeline for the identification of differentially abundant tRNA fragments and other non-coding RNAs from small RNA-sequencing data. tsRNAsearch is intended for use when comparing two groups of datasets, such as control and treatment groups. tsRNAsearch comparatively searches for tRNAs and ncRNAs with irregular read distribution profiles (a proxy for RNA cleavage) using a combined score made up of four novel methods and a differential expression analysis, and reports the top ranked results in simple PDF and TEXT files. In this study, we used publicly available small RNA-seq data to replicate the identification of tsRNAs from chronic hepatitis-infected liver tissue data. In addition, we applied tsRNAsearch to pancreatic ductal adenocarcinoma (PDAC) and matched healthy pancreatic tissue small RNA-sequencing data. Our results support the identification of miR135b from the original study as a potential biomarker of PDAC and identify other potentially stronger miRNA biomarkers of PDAC. AVAILABILITY https://github.com/GiantSpaceRobot/tsRNAsearch. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Paul D Donovan
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, St Stephen's Green, Dublin, Ireland
| | - Natalie M McHale
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, St Stephen's Green, Dublin, Ireland
| | | | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland, St Stephen's Green, Dublin, Ireland
| |
Collapse
|
14
|
Mortoglou M, Tabin ZK, Arisan ED, Kocher HM, Uysal-Onganer P. Non-coding RNAs in pancreatic ductal adenocarcinoma: New approaches for better diagnosis and therapy. Transl Oncol 2021; 14:101090. [PMID: 33831655 PMCID: PMC8042452 DOI: 10.1016/j.tranon.2021.101090] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/14/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies with a 5-year survival rate less than 8%, which has remained unchanged over the last 50 years. Early detection is particularly difficult due to the lack of disease-specific symptoms and a reliable biomarker. Multimodality treatment including chemotherapy, radiotherapy (used sparingly) and surgery has become the standard of care for patients with PDAC. Carbohydrate antigen 19-9 (CA 19-9) is the most common diagnostic biomarker; however, it is not specific enough especially for asymptomatic patients. Non-coding RNAs are often deregulated in human malignancies and shown to be involved in cancer-related mechanisms such as cell growth, differentiation, and cell death. Several micro, long non-coding and circular RNAs have been reported to date which are involved in PDAC. Aim of this review is to discuss the roles and functions of non-coding RNAs in diagnosis and treatments of PDAC.
Collapse
Affiliation(s)
- Maria Mortoglou
- Cancer Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK.
| | - Zoey Kathleen Tabin
- Cancer Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK.
| | - E Damla Arisan
- Institution of Biotechnology, Gebze Technical University, Gebze, Turkey.
| | - Hemant M Kocher
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University London, London EC1M 6BQ, UK.
| | - Pinar Uysal-Onganer
- Cancer Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK.
| |
Collapse
|
15
|
Hai X, Zhao G, Li Z, Wu J, Xu X, Yang Y. Effects of microRNA-103 on the Proliferation and Apoptosis of Pancreatic Cancer Cells via Targeting Phosphatase and Tensin Homolog Deleted on Chromosome Ten (PTEN) and Activating Phosphoinositide 3-Kinase/A Serine/Threonine Kinase (PI3K/Akt) Signaling Pathway. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objective: To investigate whether micro ribonucleic acid (miR)-103 affects pancreatic cancer (PaCa) cells via PTEN-activated PI3K/Akt signaling pathway. Methods: Differences in miR-103 expression in 35 pairs of PaCa tissues and cell lines (SW1990 and PATU8988S) were detected
by RT-qPCR. miR-103 inhibitor was transfected into PaCa PATU8988S cell followed by analysis of proliferation and apoptosis of PaCa cells by MTT assay and flow cytometry, respectively. Results: MiR-103 exhibited a significantly high expression in PaCa tissues and cell lines (p
< 0.05). Besides, the exogenous inhibition of miR-103 expression in PATU8988S cells significantly inhibited cell proliferation and migration but increased apoptosis activity (p < 0.05). According to the prediction of TargetScan biological database, miR-103 could bind PTEN 3′
untranslated region (3′UTR) and miR-103 was confirmed to suppress PTEN expression in a targeted way (p<0.05). Furthermore, down-regulation of PTEN activated PI3K/Akt signaling to affect the proliferation and apoptosis of PaCa cells (p < 0.05 or p <0.01). Conclusion:
MiR-103 displays a significantly increased expression in PaCa cells and targets PTEN to activate PI3K/Akt signaling pathway, thus promoting malignant phenotype formation.
Collapse
Affiliation(s)
- Xiaoyu Hai
- Department of General Surgery, Ningxia Fifth People’s Hospital, Shizuishan, 753000, Ningxia, China
| | - Guozhong Zhao
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750000, Ningxia, China
| | - Zhaolong Li
- Department of General Surgery, Ningxia Fifth People’s Hospital, Shizuishan, 753000, Ningxia, China
| | - Junli Wu
- Department of Pancreatic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 753000, Jiangsu, China
| | - Xiangzhao Xu
- Department of Anesthesiology, Ningxia Fifth People’s Hospital, Shizuishan, 753000, Ningxia, China
| | - Yaowen Yang
- Department of Radiology, Fifth People’s Hospital of Ningxia, Shizuishan, 753000, Ningxia, China
| |
Collapse
|
16
|
Qu S, Niu K, Wang J, Dai J, Ganguly A, Gao C, Tian Y, Lin Z, Yang X, Zhang X, Liu Z, Li H. LINC00671 suppresses cell proliferation and metastasis in pancreatic cancer by inhibiting AKT and ERK signaling pathway. Cancer Gene Ther 2021; 28:221-233. [PMID: 32801328 DOI: 10.1038/s41417-020-00213-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/25/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022]
Abstract
Long noncoding RNAs (lncRNAs) represent an emerging field of tumor biology, playing essential roles in cancer cell proliferation, invasion, and metastasis. However, the overall functional and clinical significance of most lncRNAs in pancreatic cancer is not thoroughly understood. Here, we described most of the lncRNAs with aberrant expression patterns in pancreatic cancer as detected by microarray. Quantitative real-time polymerase chain reaction further verified that the expression of LINC00671 was decreased in pancreatic cancer cell lines and patient samples. Furthermore, lower LINC00671 expression was associated with reduced tumor differentiation, aggressiveness, and poor prognosis. Functionally, LINC00671 overexpression inhibited pancreatic cancer cell proliferation, invasion, and migration in vitro, and reduced tumor growth in vivo. LINC00671 is mainly located in the cytoplasm. RNA sequencing and bioinformatics analyses indicated that LINC00671 binds to multiple miRNAs and therefore could be involved in multiple tumor-associated pathways, such as the AMPK signaling pathway and PI3K-Akt signaling pathway. Western blotting and immunohistochemistry further confirmed that LINC00671 overexpression suppressed the AKT, ERK, and epithelial-mesenchymal transition pathways. Overall, these results indicated that LINC00671 acts as a novel tumor suppressor in pancreatic cancer. Our findings may provide a new potential target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Shibin Qu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kunwei Niu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianlin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jimin Dai
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Anutosh Ganguly
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Chao Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Science, Bejing Institute of Lifeomics, Beijing, China
| | - Yuzi Tian
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhibin Lin
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xisheng Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xuan Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhengcai Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Haimin Li
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
17
|
MiR-10a in Pancreatic Juice as a Biomarker for Invasive Intraductal Papillary Mucinous Neoplasm by miRNA Sequencing. Int J Mol Sci 2021; 22:ijms22063221. [PMID: 33809988 PMCID: PMC8004614 DOI: 10.3390/ijms22063221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023] Open
Abstract
New biomarkers are needed to further stratify the risk of malignancy in intraductal papillary mucinous neoplasm (IPMN). Although microRNAs (miRNAs) are expected to be stable biomarkers, they can vary owing to a lack of definite internal controls. To identify universal biomarkers for invasive IPMN, we performed miRNA sequencing using tumor-normal paired samples. A total of 19 resected tissues and 13 pancreatic juice samples from 32 IPMN patients were analyzed for miRNA expression by next-generation sequencing with a two-step normalization of miRNA sequence data. The miRNAs involved in IPMN associated with invasive carcinoma were identified from this tissue analysis and further verified with the pancreatic juice samples. From the tumor-normal paired tissue analysis of the expression levels of 2792 miRNAs, 20 upregulated and 17 downregulated miRNAs were identified. In IPMN associated with invasive carcinoma (INV), miR-10a-5p and miR-221-3p were upregulated and miR-148a-3p was downregulated when compared with noninvasive IPMN. When these findings were further validated with pancreatic juice samples, miR-10a-5p was found to be elevated in INV (p = 0.002). Therefore, three differentially expressed miRNAs were identified in tissues with INV, and the expression of miR-10a-5p was also elevated in pancreatic juice samples with INV. MiR-10a-5p is a promising additional biomarker for invasive IPMN.
Collapse
|
18
|
Li H, Wang X, Zhang M, Wang M, Zhang J, Ma S. Identification of HOXA1 as a Novel Biomarker in Prognosis of Head and Neck Squamous Cell Carcinoma. Front Mol Biosci 2021; 7:602068. [PMID: 33763449 PMCID: PMC7982851 DOI: 10.3389/fmolb.2020.602068] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/12/2020] [Indexed: 12/24/2022] Open
Abstract
Hox genes, a highly conserved homolog in most animals, play vital functions in cell development and organ formation. In recent years, researchers have discovered that it can act as a tumor regulator, and its members can participate in tumorigenesis by regulating receptor signaling, cell differentiation, apoptosis, migration, EMT, and angiogenesis. Hox genes and which major members play a vital role in the progress of head and neck squamous cell carcinoma (HNSCC) is still unclear. After analyzing the expression differences and prognostic value of all Hox genes through the TCGA-HNSC database, we use histochemistry stains in 52 pairs of HNSCC slices to verify the expression level of the key member-HOXA1. In correlation analysis, we found that high HOXA1 expression is related to poor pathological grade (p = 0.0077), advanced T stage (p = 0.021) and perineural invasion (PNI) (p = 0.0019). Furthermore, we used Cox univariate and multivariate regression analysis to confirm the independent predictive power of HOXA1 expression. To explore the underlying mechanisms behind HOXA1, we ran GSVA and GSEA and found fourteen mutual signaling pathways, including neuroprotein secretion and transport, tumor-associated signaling pathways, cell adhere junction and metabolic reprogramming. Finally, we found that the high expression of HOXA1 is significantly related to the decrease of CD8+ T cell infiltration and the decline of DNA methylation level. Our findings demonstrated that HOXA1, as a notable member of the HOX family, maybe an independent prognostic indicator in HNSCC.
Collapse
Affiliation(s)
- Hui Li
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaomin Wang
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mingjie Zhang
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mengjun Wang
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Junjie Zhang
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shiyin Ma
- Department of Otolaryngology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
19
|
Nie HX, Zhang L, He T, Wang L, Wan BS, Wang XQ, Han F. lncRNA-XLOC_012370 Promotes the Development of Pancreatic Cancer and Inactivates the NF-κB Pathway Through miR-140-5p. Front Oncol 2021; 10:620550. [PMID: 33708618 PMCID: PMC7940521 DOI: 10.3389/fonc.2020.620550] [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: 10/23/2020] [Accepted: 12/17/2020] [Indexed: 12/26/2022] Open
Abstract
Pancreatic cancer is a high incidence, high degree of malignancy, and high mortality in the digestive system tumor. The incidence of pancreatic cancer in China has increased nearly six folds in the past 20 years, ranking fifth in the mortality rate of malignant tumors, so it is particularly important to actively explore clinical indicators with better diagnostic significance for pancreatic cancer. LncRNA performs an essential regulatory function in the occurrence, development, and metastasis of many kinds of tumors, playing both a carcinogenic role and a tumor suppressor gene. Here, we demonstrated the function and mechanism of LncRNA-XLOC_012370 in the development of pancreatic cancer. In our research, the abnormal upregulation of XLOC_012370 was observed in pancreatic cancer patients’ tumor tissues. XLOC_012370 was related to tumor stage, lymph node metastasis, and overall survival. Silencing of XLOC_012370 prevented the proliferation, migration, and invasion via the NF-κB signal pathway. Further, miR-140-5p was identified as the target and downstream of XLOC_012370 and involved in pancreatic cancer progression. In vivo, knockdown of XLOC_012370 inhibited tumor growth via the NF-κB signal pathway. In conclusion, lncRNA-XLOC_012370 is closely related to some malignant clinicopathological features and prognosis of pancreatic cancer. Thus the miR-140-5p/NF-κB signal pathway might represent a promising treatment strategy to combat pancreatic cancer.
Collapse
Affiliation(s)
- Han-Xiao Nie
- Department of Hepatopancreatobiliary Surgery, Henan Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Ling Zhang
- Department of Hepatopancreatobiliary Surgery, Henan Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Tao He
- Department of Hepatopancreatobiliary Surgery, Henan Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Li Wang
- Department of Hepatopancreatobiliary Surgery, Henan Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Bai-Shun Wan
- Department of Hepatopancreatobiliary Surgery, Henan Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xiao-Qian Wang
- Department of Hepatopancreatobiliary Surgery, Henan Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Feng Han
- Department of Hepatopancreatobiliary Surgery, Henan Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Paço A, Aparecida de Bessa Garcia S, Leitão Castro J, Costa-Pinto AR, Freitas R. Roles of the HOX Proteins in Cancer Invasion and Metastasis. Cancers (Basel) 2020; 13:E10. [PMID: 33375038 PMCID: PMC7792759 DOI: 10.3390/cancers13010010] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Invasion and metastasis correspond to the foremost cause of cancer-related death, and the molecular networks behind these two processes are extremely complex and dependent on the intra- and extracellular conditions along with the prime of the premetastatic niche. Currently, several studies suggest an association between the levels of HOX genes expression and cancer cell invasion and metastasis, which favour the formation of novel tumour masses. The deregulation of HOX genes by HMGA2/TET1 signalling and the regulatory effect of noncoding RNAs generated by the HOX loci can also promote invasion and metastasis, interfering with the expression of HOX genes or other genes relevant to these processes. In this review, we present five molecular mechanisms of HOX deregulation by which the HOX clusters products may affect invasion and metastatic processes in solid tumours.
Collapse
Affiliation(s)
- Ana Paço
- BLC3—Biomassa Lenho-Celulósica de 3ª Geração, Campus of Technology and Innovation, 3405-169 Oliveira do Hospital, Portugal
| | - Simone Aparecida de Bessa Garcia
- I3S—Institute for Innovation & Health Research, University of Porto, 4200-135 Porto, Portugal; (S.A.d.B.G.); (J.L.C.); (A.R.C.-P.); (R.F.)
| | - Joana Leitão Castro
- I3S—Institute for Innovation & Health Research, University of Porto, 4200-135 Porto, Portugal; (S.A.d.B.G.); (J.L.C.); (A.R.C.-P.); (R.F.)
| | - Ana Rita Costa-Pinto
- I3S—Institute for Innovation & Health Research, University of Porto, 4200-135 Porto, Portugal; (S.A.d.B.G.); (J.L.C.); (A.R.C.-P.); (R.F.)
| | - Renata Freitas
- I3S—Institute for Innovation & Health Research, University of Porto, 4200-135 Porto, Portugal; (S.A.d.B.G.); (J.L.C.); (A.R.C.-P.); (R.F.)
- ICBAS—Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
21
|
Wang J, Liu Y, Li Y, Zheng X, Gan J, Wan Z, Zhang J, Liu Y, Wang Y, Hu W, Li Y, Liu Y. Exosomal‑miR‑10a derived from colorectal cancer cells suppresses migration of human lung fibroblasts, and expression of IL‑6, IL‑8 and IL‑1β. Mol Med Rep 2020; 23:84. [PMID: 33236127 PMCID: PMC7716406 DOI: 10.3892/mmr.2020.11723] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/21/2020] [Indexed: 01/10/2023] Open
Abstract
MicroRNAs (miRs) carried in exosomes serve an important role in the pre‑metastatic microenvironment and in intercellular interactions. However, the function of exosomal‑miR‑10a derived from primary colorectal cancer (CRC) cells on fibroblasts in the lung metastatic microenvironment of patients with CRC remains unclear. Reverse transcription‑quantitative PCR was performed using samples from patients with CRC, and demonstrated that the expression levels of miR‑10a were significantly lower in serum and cancer tissue samples from patients with CRC compared with in serum from healthy individuals and paired non‑cancerous tissues, respectively. In addition, the expression levels of miR‑10a were inversely associated with the invasion depth of CRC. Exosomal‑miR‑10a derived from CRC cells reduced the proliferative and migratory activities of primary normal human lung fibroblasts (NHLFs), and the expression levels of IL‑6, IL‑8 and IL‑1β in NHLFs. The present study provided insight into the phenotypic alterations of NHLFs induced by exosomal‑miR‑10a derived from CRC cells, which may aid understanding of the mechanism underlying the process of CRC lung metastasis.
Collapse
Affiliation(s)
- Jian Wang
- Department of Gastrointestinal Surgery, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Yuanting Liu
- Department of Gastrointestinal Surgery, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Ying Li
- Nuclear Medicine Clinical Laboratory, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Xuan Zheng
- Nuclear Medicine Clinical Laboratory, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Jianhui Gan
- Department of Anesthesiology, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Zhaoyuan Wan
- The Cancer Institute, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Jun Zhang
- The Cancer Institute, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Yan Liu
- The Cancer Institute, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Yaqi Wang
- Department of Breast Surgery, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Wanning Hu
- The Cancer Institute, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Yufeng Li
- The Cancer Institute, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Yankun Liu
- The Cancer Institute, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| |
Collapse
|
22
|
Induction of multiple myeloma bone marrow stromal cell apoptosis by inhibiting extracellular vesicle miR-10a secretion. Blood Adv 2020; 3:3228-3240. [PMID: 31698453 DOI: 10.1182/bloodadvances.2019000403] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/22/2019] [Indexed: 02/08/2023] Open
Abstract
Bone marrow stromal cells (BMSCs) interact with multiple myeloma (MM) cells in the bone marrow and create a permissive microenvironment for MM cell proliferation and survival. In this study, we investigated the role of extracellular vesicles (EVs) from BMSCs derived from patients with MM (MM-BMSCs). EV-encapsulated miR-10a expression was high while intracellular miR-10a was low in MM-BMSCs. We therefore hypothesized that miR-10a was packaged into EVs that were actively released into the extracellular space. Inhibition of EV release resulted in accumulation of intracellular miR-10a, inhibition of cell proliferation, and induction of apoptosis in MM-BMSCs. In contrast, proliferation and apoptosis of BMSCs derived from healthy individuals were unaffected by inhibition of EV release. Furthermore, miR-10a derived from MM-BMSCs was transferred into MM cells via EVs and enhanced their proliferation. These results suggest that inhibition of EV release induced apoptosis in MM-BMSCs and inhibited MM cell proliferation, indicating a possible role for MM-BMSC-targeted therapy.
Collapse
|
23
|
Wang C, Yin W, Liu H. MicroRNA-10a promotes epithelial-to-mesenchymal transition and stemness maintenance of pancreatic cancer stem cells via upregulating the Hippo signaling pathway through WWC2 inhibition. J Cell Biochem 2020; 121:4505-4521. [PMID: 32542845 DOI: 10.1002/jcb.29716] [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: 11/07/2019] [Accepted: 01/14/2020] [Indexed: 12/29/2022]
Abstract
MicroRNAs (miRNAs)-mediated cancer stem cells (CSCs) have drawn wide attention. This study aimed to probe the role of miR-10a in epithelial-mesenchymal transition (EMT) and stemness maintenance of pancreatic CSCs (PCSCs). Differentially expressed miRs and genes in pancreatic cancer (PC) were predicted via an online database, and the miR-10a and WW and C2 domain containing 2 (WWC2) expression were identified via a comparative study in PC and pancreatitis tissues. PCNCs were isolated and identified, and then the functional roles of miR-10a and WWC2 in proliferation, invasion, migration, self-renewal, colony formation abilities, EMT, and stemness maintenance of PCNCs were determined. The effects of miR-10a on tumor growth in vivo were studied by performing a xenograft tumor in nude mice. Consequently, miR-10a was highly expressed while WWC2 was lowly expressed in PC tissues. miR-10a could target WWC2 expression. miR-10a inhibition reduced EMT and stemness maintenance of PCSCs via enhancing WWC2 expression. The in vitro results were reproduced in in vivo studies. miR-10a promoted EMT and stemness maintenance of PCSCs via activating the Hippo signaling pathway. Our study provided evidence that miR-10a inhibition reduced EMT and stemness maintenance of PCSCs via upregulating WWC2 expression and inhibiting the Hippo signaling pathway.
Collapse
Affiliation(s)
- Caiyan Wang
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Wen Yin
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Hui Liu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| |
Collapse
|
24
|
Brotto DB, Siena ÁDD, de Barros II, Carvalho SDCES, Muys BR, Goedert L, Cardoso C, Plaça JR, Ramão A, Squire JA, Araujo LF, Silva WAD. Contributions of HOX genes to cancer hallmarks: Enrichment pathway analysis and review. Tumour Biol 2020; 42:1010428320918050. [PMID: 32456563 DOI: 10.1177/1010428320918050] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Homeobox genes function as master regulatory transcription factors during development, and their expression is often altered in cancer. The HOX gene family was initially studied intensively to understand how the expression of each gene was involved in forming axial patterns and shaping the body plan during embryogenesis. More recent investigations have discovered that HOX genes can also play an important role in cancer. The literature has shown that the expression of HOX genes may be increased or decreased in different tumors and that these alterations may differ depending on the specific HOX gene involved and the type of cancer being investigated. New studies are also emerging, showing the critical role of some members of the HOX gene family in tumor progression and variation in clinical response. However, there has been limited systematic evaluation of the various contributions of each member of the HOX gene family in the pathways that drive the common phenotypic changes (or "hallmarks") and that underlie the transformation of normal cells to cancer cells. In this review, we investigate the context of the engagement of HOX gene targets and their downstream pathways in the acquisition of competence of tumor cells to undergo malignant transformation and tumor progression. We also summarize published findings on the involvement of HOX genes in carcinogenesis and use bioinformatics methods to examine how their downstream targets and pathways are involved in each hallmark of the cancer phenotype.
Collapse
Affiliation(s)
- Danielle Barbosa Brotto
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Ádamo Davi Diógenes Siena
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Isabela Ichihara de Barros
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Simone da Costa E Silva Carvalho
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Bruna Rodrigues Muys
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Lucas Goedert
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil.,Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cibele Cardoso
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Jessica Rodrigues Plaça
- National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Anelisa Ramão
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Jeremy Andrew Squire
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Luiza Ferreira Araujo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil
| | - Wilson Araújo da Silva
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute of Science and Technology in Stem Cell and Cell Therapy (INCT/CNPq) and Center for Cell-Based Therapy, CEPID/FAPESP, Ribeirão Preto, Brazil.,Center for Integrative System Biology (CISBi), NAP/USP, University of São Paulo, Ribeirão Preto, Brazil.,Center for Medical Genomics, Clinics Hospital, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
25
|
Gong R, Jiang Y. Non-coding RNAs in Pancreatic Ductal Adenocarcinoma. Front Oncol 2020; 10:309. [PMID: 32257946 PMCID: PMC7089935 DOI: 10.3389/fonc.2020.00309] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are reported to be expressed in human cancers, including pancreatic ductal adenocarcinoma (PDAC). These ncRNAs affect the growth, migration and invasion of tumor cells by regulating cell cycle and apoptosis, as well as playing important roles in epigenetic processes, transcription and post-transcriptional regulation. It is still unclear whether alterations in ncRNAs influence PDAC development and progression. Because of this, analysis based on existing data on ncRNAs, which are crucial for modulating pancreatic tumorigenesis, will be important for future research on PDAC. Here, we summarize ncRNAs with tumor-promoting functions: HOTAIR, HOTTIP, MALAT1, lncRNA H19, lncRNA PVT1, circ-RNA ciRS-7, circ-0030235, circ-RNA_100782, circ-LDLRAD3, circ-0007534, circRHOT1, circZMYM2, circ-IARS, circ-RNA PDE8A, miR-21, miR-155, miR-221/222, miR-196b, miR-10a. While others including GAS5, MEG3, and lncRNA ENST00000480739, has_circ_0001649, miR-34a, miR-100, miR-217, miR-143 inhibit the proliferation and invasion of PDAC. Hence, we summarize the functions of ncRNAs in the occurrence, development and metastasis of PDAC, with the goal to provide guidance in the clinical diagnosis and treatment of PDAC.
Collapse
Affiliation(s)
- Ruining Gong
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yueping Jiang
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
26
|
Cohen SJ, Papoulas M, Graubardt N, Ovdat E, Loewenstein S, Kania-Almog J, Pasmanik-Chor M, Brazowski E, Cagnano E, Nachmany I, Lahat G, Klausner JM, Lubezky N. Micro-RNA Expression Patterns Predict Metastatic Spread in Solid Pseudopapillary Neoplasms of the Pancreas. Front Oncol 2020; 10:328. [PMID: 32232006 PMCID: PMC7082878 DOI: 10.3389/fonc.2020.00328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/25/2020] [Indexed: 12/17/2022] Open
Abstract
Solid pseudopapillary neoplasm (SPN) of pancreas is a rare pancreatic neoplasm with a low metastatic potential. Up to 10% of patients with localized disease at presentation will develop systemic metastases, usually in the peritoneum or the liver. Due to the rarity of SPNs and the overall excellent prognosis, reliable prognostic factors to predict malignant biological behavior remain undetermined. Therefore, we aimed to define clinical, histological, and microRNA patterns that are associated with metastatic disease. We conducted a retrospective single center study on all patients operated for SPN of pancreas between 1995 and 2018. Clinical and pathological data were collected, and expression patterns of 2,578 human microRNAs were analyzed using microRNA array (Affimetrix 4.1) in normal pancreases (NPs), localized tumors (LTs), and metastatic tumors (MTs). The diagnosis of SPN was confirmed in 35 patients who included 28 females and 3 males, with a mean age of 33.8 ± 13.9 years. The only clinical factor associated with metastases was tumor size (mean tumor size 5.20 ± 3.78 in LT vs. 8.13± 1.03 in MT, p < 0.012). Microscopic features of malignancy were not associated with metastases, nor were immunohistochemical stains, including the proliferative index KI67. Higher expressions of miR-184, miR-10a, and miR-887, and lower expressions of miR-375, miR-217, and miR-200c were observed in metastatic tissues on microarray, and validated by real-time polymerase chain reaction. Hierarchal clustering demonstrated that the microRNA expression pattern of MTs was significantly different from that of LTs. The only clinical factor associated with metastases of SPN of pancreas was tumor size. Histological features and immunohistological staining were not predictive of metastases. A panel of six microRNAs was differentially expressed in MTs, and these findings could potentially be used to predict tumor behavior. Validation of these results is needed in larger series.
Collapse
Affiliation(s)
- Shmuel Jaffe Cohen
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Michail Papoulas
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nadine Graubardt
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Esther Ovdat
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shelly Loewenstein
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Juliane Kania-Almog
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Eli Brazowski
- Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Institute of Pathology, Tel-Aviv University, Tel-Aviv, Israel
| | - Emanuela Cagnano
- Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Institute of Pathology, Tel-Aviv University, Tel-Aviv, Israel
| | - Ido Nachmany
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Guy Lahat
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Joseph M Klausner
- Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nir Lubezky
- Surgical Division Research Laboratory, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Department of Surgery, Tel-Aviv Sourasky Medical Center Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
27
|
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
|
28
|
Chhatriya B, Mukherjee M, Ray S, Sarkar P, Chatterjee S, Nath D, Das K, Goswami S. Comparison of tumour and serum specific microRNA changes dissecting their role in pancreatic ductal adenocarcinoma: a meta-analysis. BMC Cancer 2019; 19:1175. [PMID: 31795960 PMCID: PMC6891989 DOI: 10.1186/s12885-019-6380-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is considered as one of the most aggressive cancers lacking efficient early detection biomarkers. Circulating miRNAs are now being considered to have potency to be used as diagnostic and prognostic biomarkers in different diseases as well as cancers. In case of cancer, a fraction of the circulating miRNAs is actually derived from the tumour tissue. This fraction would function as stable biomarker for the disease and also would contribute to the understanding of the disease development. There are not many studies exploring this aspect in pancreatic cancer and even there is not much overlap of results between existing studies. Methods In order to address that gap, we performed a miRNA microarray analysis to identify differentially expressed circulating miRNAs between PDAC patients and normal healthy individuals and also found two more similar datasets to perform a meta-analysis using a total of 182 PDAC patients and 170 normal, identifying a set of miRNAs significantly altered in patient serum. Next, we found five datasets studying miRNA expression profile in tumour tissues of PDAC patients as compared to normal pancreas and performed a second meta-analysis using data from a total of 183 pancreatic tumour and 47 normal pancreas to detect significantly deregulated miRNAs in pancreatic carcinoma. Comparison of these two lists and subsequent search for their target genes which were also deregulated in PDAC in inverse direction to miRNAs was done followed by investigation of their role in disease development. Results We identified 21 miRNAs altered in both pancreatic tumour tissue and serum. While deciphering the functions of their target genes, we characterized key miR-Gene interactions perturbing the biological pathways. We identified important cancer related pathways, pancreas specific pathways, AGE-RAGE signaling, prolactin signaling and insulin resistance signaling pathways among the most affected ones. We also reported the possible involvement of crucial transcription factors in the process. Conclusions Our study identified a unique meta-signature of 21 miRNAs capable of explaining pancreatic carcinogenesis and possibly holding the potential to act as biomarker for the disease detection which could be explored further.
Collapse
Affiliation(s)
| | - Moumita Mukherjee
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Sukanta Ray
- School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, West Bengal, India
| | - Piyali Sarkar
- Present Address: Tata Medical Centre, Kolkata, West Bengal, India
| | | | - Debashis Nath
- Indira Gandhi Memorial Hospital, Agartala, Tripura, India
| | - Kshaunish Das
- School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, West Bengal, India
| | - Srikanta Goswami
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India.
| |
Collapse
|
29
|
Horii R, Honda M, Shirasaki T, Shimakami T, Shimizu R, Yamanaka S, Murai K, Kawaguchi K, Arai K, Yamashita T, Sakai Y, Yamashita T, Okada H, Nakamura M, Mizukoshi E, Kaneko S. MicroRNA-10a Impairs Liver Metabolism in Hepatitis C Virus-Related Cirrhosis Through Deregulation of the Circadian Clock Gene Brain and Muscle Aryl Hydrocarbon Receptor Nuclear Translocator-Like 1. Hepatol Commun 2019; 3:1687-1703. [PMID: 31832575 PMCID: PMC6887665 DOI: 10.1002/hep4.1431] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022] Open
Abstract
The circadian rhythm of the liver plays an important role in maintaining its metabolic homeostasis. We performed comprehensive expression analysis of microRNAs (miRNAs) using TaqMan polymerase chain reaction of liver biopsy tissues to identify the miRNAs that are significantly up‐regulated in advanced chronic hepatitis C (CHC). We found miR‐10a regulated various liver metabolism genes and was markedly up‐regulated by hepatitis C virus infection and poor nutritional conditions. The expression of miR‐10a was rhythmic and down‐regulated the expression of the circadian rhythm gene brain and muscle aryl hydrocarbon receptor nuclear translocator‐like 1 (Bmal1) by directly suppressing the expression of RA receptor‐related orphan receptor alpha (RORA). Overexpression of miR‐10a in hepatocytes blunted circadian rhythm of Bmal1 and inhibited the expression of lipid synthesis genes (sterol regulatory element binding protein [SREBP]1, fatty acid synthase [FASN], and SREBP2), gluconeogenesis (peroxisome proliferator‐activated receptor gamma coactivator 1 alpha [PGC1α]), protein synthesis (mammalian target of rapamycin [mTOR] and ribosomal protein S6 kinase [S6K]) and bile acid synthesis (liver receptor homolog 1 [LRH1]). The expression of Bmal1 was significantly correlated with the expression of mitochondrial biogenesis‐related genes and reduced Bmal1 was associated with increased serum alanine aminotransferase levels and progression of liver fibrosis in CHC. Thus, impaired circadian rhythm expression of Bmal1 by miR‐10a disturbs metabolic adaptations, leading to liver damage, and is closely associated with the exacerbation of abnormal liver metabolism in patients with advanced CHC. In patients with hepatitis C‐related liver cirrhosis, liver tissue miR‐10a levels were significantly associated with hepatic reserve, fibrosis markers, esophageal varix complications, and hepatitis C‐related hepatocellular carcinoma recurrence. Conclusion: MiRNA‐10a is involved in abnormal liver metabolism in cirrhotic liver through down‐regulation of the expression of the circadian rhythm gene Bmal1. Therefore, miR‐10a is a possible useful biomarker for estimating the prognosis of liver cirrhosis.
Collapse
Affiliation(s)
- Rika Horii
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Masao Honda
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan.,Department of Laboratory Medicine Kanazawa University Graduate School of Health Medicine Kanazawa Japan
| | - Takayoshi Shirasaki
- Department of Laboratory Medicine Kanazawa University Graduate School of Health Medicine Kanazawa Japan
| | - Tetsuro Shimakami
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Ryogo Shimizu
- Department of Laboratory Medicine Kanazawa University Graduate School of Health Medicine Kanazawa Japan
| | - Souma Yamanaka
- Department of Laboratory Medicine Kanazawa University Graduate School of Health Medicine Kanazawa Japan
| | - Kazuhisa Murai
- Department of Laboratory Medicine Kanazawa University Graduate School of Health Medicine Kanazawa Japan
| | - Kazunori Kawaguchi
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Kuniaki Arai
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Tatsuya Yamashita
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Yoshio Sakai
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Taro Yamashita
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Hikari Okada
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Mikiko Nakamura
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Eishiro Mizukoshi
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| | - Shuichi Kaneko
- Department of Gastroenterology Kanazawa University Graduate School of Medicine Kanazawa Japan
| |
Collapse
|
30
|
Rawat M, Kadian K, Gupta Y, Kumar A, Chain PSG, Kovbasnjuk O, Kumar S, Parasher G. MicroRNA in Pancreatic Cancer: From Biology to Therapeutic Potential. Genes (Basel) 2019; 10:genes10100752. [PMID: 31557962 PMCID: PMC6827136 DOI: 10.3390/genes10100752] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer is one of the most aggressive malignancies, accounting for more than 45,750 deaths annually in the U.S. alone. The aggressive nature and late diagnosis of pancreatic cancer, coupled with the limitations of existing chemotherapy, present the pressing need for the development of novel therapeutic strategies. Recent reports have demonstrated a critical role of microRNAs (miRNAs) in the initiation, progression, and metastasis of cancer. Furthermore, aberrant expressions of miRNAs have often been associated with the cause and consequence of pancreatic cancer, emphasizing the possible use of miRNAs in the effective management of pancreatic cancer patients. In this review, we provide a brief overview of miRNA biogenesis and its role in fundamental cellular process and miRNA studies in pancreatic cancer patients and animal models. Subsequent sections narrate the role of miRNA in, (i) cell cycle and proliferation; (ii) apoptosis; (iii) invasions and metastasis; and (iv) various cellular signaling pathways. We also describe the role of miRNA's in pancreatic cancer; (i) diagnosis; (ii) prognosis and (iii) therapeutic intervention. Conclusion section describes the gist of review with future directions.
Collapse
Affiliation(s)
- Manmeet Rawat
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| | - Kavita Kadian
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand 263001, India.
| | - Yash Gupta
- Department of Internal Medicine, Loyola University Medical Center, Chicago, IL 60153, USA.
| | - Anand Kumar
- Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - Patrick S G Chain
- Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - Olga Kovbasnjuk
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | - Gulshan Parasher
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| |
Collapse
|
31
|
Elevated microRNA-145 inhibits the development of oral squamous cell carcinoma through inactivating ERK/MAPK signaling pathway by down-regulating HOXA1. Biosci Rep 2019; 39:BSR20182214. [PMID: 31138758 PMCID: PMC6591566 DOI: 10.1042/bsr20182214] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/19/2019] [Accepted: 05/08/2019] [Indexed: 01/18/2023] Open
Abstract
Background: Oral cancer is one of the most frequent solid cancers worldwide, and oral squamous cell carcinoma (OSCC) constitutes approximately 90% of oral cancers. The discovery of reliable prognostic indicators would be a potential strategy for OSCC treatment. In the present study, we aim to explore the underlying mechanism by which microRNA-145 (miR-145) affected OSCC. Methods: Forty-eight patients diagnosed with OSCC were enrolled to obtain the OSCC tissues and adjacent normal tissues. The targeting relationship between miR-145 and Homeobox A1 (HOXA1) was verified. In order to assess the effects of miR-145 in OSCC and the detailed regulatory mechanism, the SCC-9 cell line was adopted, in which expression of miR-145 and HOXA1 were altered by transfection. Then, a series of in vitro and in vivo experiments were performed to evaluate the cell viability, migration, invasion, and tumor growth. Results: miR-145 was poorly expressed and HOXA1 was highly expressed in OSCC. HOXA1 was verified as a target of miR-145 to mediate the activation of the extracellular signal-regulated kinase/mitogen activated protein kinase (ERK/MAPK) signaling pathway. In the circumstance of miR-145 elevation or HOXA1 depletion, the SCC-9 cell line manifested with inhibited cell viability, invasion, and migration in vitro, coupled with reduced tumor growth in vivo, with a decreased expression of ERK/MAPK signaling pathway-related genes/proteins. Conclusion: These findings suggested that miR-145 can inhibit HOXA1 to inactivate the ERK/MAPK signaling pathway, thereby suppressing OSCC cell proliferation, migration, and invasion to further inhibit the development of OSCC, highlighting a novel therapeutic target for the OSCC treatment.
Collapse
|
32
|
Kuo TL, Cheng KH, Chen LT, Hung WC. Deciphering The Potential Role of Hox Genes in Pancreatic Cancer. Cancers (Basel) 2019; 11:cancers11050734. [PMID: 31137902 PMCID: PMC6562939 DOI: 10.3390/cancers11050734] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023] Open
Abstract
The Hox gene family plays an important role in organogenesis and animal development. Currently, 39 Hox genes that are clustered in four chromosome regions have been identified in humans. Emerging evidence suggests that Hox genes are involved in the development of the pancreas. However, the expression of Hox genes in pancreatic tumor tissues has been investigated in only a few studies. In addition, whether specific Hox genes can promote or suppress cancer metastasis is not clear. In this article, we first review the recent progress in studies on the role of Hox genes in pancreatic cancer. By comparing the expression profiles of pancreatic cancer cells isolated from genetically engineered mice established in our laboratory with three different proliferative and metastatic abilities, we identified novel Hox genes that exhibited tumor-promoting activity in pancreatic cancer. Finally, a potential oncogenic mechanism of the Hox genes was hypothesized.
Collapse
Affiliation(s)
- Tzu-Lei Kuo
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| | - Kuang-Hung Cheng
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
33
|
Tao C, Sun H, Sang W, Li S. miRNA-99a inhibits cell invasion and migration in liver cancer by directly targeting HOXA1. Oncol Lett 2019; 17:5108-5114. [PMID: 31186723 PMCID: PMC6507307 DOI: 10.3892/ol.2019.10199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/14/2019] [Indexed: 12/18/2022] Open
Abstract
Liver cancer is a malignant tumor that threatens human health worldwide. It has poor prognosis rates and ineffective therapeutic options. Recently, various miRNAs have been proven to exert promoting or inhibiting functions in different malignancies. However, the definitive mechanisms of miR-99a in liver cancer remain unclear. In the current study, we explored the relationships between the expression of miR-99a and HOXA1 in liver cancer tissues and cells to explore their combined effects on the occurrence and metastasis of liver cancer. The expression of miR-99a and HOXA1 in liver cancer tissue samples and cells was measured by RT-qPCR. Following transfection, transwell assays were conducted to assess the invasion and migration capacities of liver cancer cells. Subsequently, western blots and luciferase reporter assays were performed in liver cancer cells to identify the target of miR-99a. The data indicated that miRNA-99a expression was significantly reduced in both liver cancer tissue samples and cells compared with normal tissues and normal liver cells respectively. By contrast, the HOXA1 expression levels in liver cancer tissues and cells were significantly increased in contrast to the control group. The findings also revealed that the miR-99a expression was negatively correlated with HOXA1 expression in liver cancer tissue samples and miR-99a could suppress cell invasion and migration by targeting HOXA1 in liver cancer.
Collapse
Affiliation(s)
- Changming Tao
- Department of Hepatology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Huiling Sun
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Weiwei Sang
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Shanshan Li
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
34
|
Wang Z, Lv J, Zou X, Huang Z, Zhang H, Liu Q, Jiang L, Zhou X, Zhu W. A three plasma microRNA signature for papillary thyroid carcinoma diagnosis in Chinese patients. Gene 2019; 693:37-45. [PMID: 30684524 DOI: 10.1016/j.gene.2019.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/17/2018] [Accepted: 01/11/2019] [Indexed: 01/07/2023]
Abstract
Whether plasma miRNAs could be used as novel non-invasive biomarkers in diagnosing papillary thyroid carcinoma (PTC) remains unknown. In this study, we designed a four-phase study to identify differentially expressed plasma miRNAs in Chinese PTC patients. Exiqon panel was initially utilized to conduct plasma miRNA profile (3 PTC pools VS. 1 healthy control (HC) pool; each 10 samples were pooled as 1 sample). The dysregulated miRNAs were then analyzed in the training (30 PTC VS. 30 HCs), testing (57 PTC VS. 54 HCs) and external validation phases (33 PTC VS. 30HCs). The identified miRNAs were further affirmed in benign nodules (2 nodular goiter (NG) pool VS. 1 HC pool). We also verified the expression of identified miRNAs in 17 matched malignant and normal tissue samples, NG plasma samples (29 PTC VS. 29 NG) and plasma exosomes (25 PTC VS. 25 HCs). Receiver operating characteristic (ROC) curves were constructed to evaluate the diagnostic value of the identified miRNAs. As a result, the screening phase demonstrated 30 dysregulated plasma miRNAs in PTC patients compared with HCs. After multiphase experiment processes, miR-346, miR-10a-5p and miR-34a-5p were found significantly elevated in PTC plasma samples relative to HCs. The areas under the ROC curve (AUC) of the three-miRNA panel for the training, testing and validation phases were 0.926, 0.811 and 0.816, separately. The panel could also differentiate PTC from NG with the AUC of 0.877. MiR-346 and miR-34a-5p but not miR-10a-5p were up-regulated in PTC tissues. And the three miRNAs showed consistently up-regulation in PTC plasma exosomes. In conclusion, our study established a three-miRNA panel in plasma with considerable clinical value in discriminating PTC from HC or NG.
Collapse
Affiliation(s)
- Zhiyan Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China; Department of Pediatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, PR China
| | - Jinru Lv
- Department of Emergency, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China
| | - Xuan Zou
- First Clinical College of Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, PR China
| | - Zebo Huang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China; Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi 214062, Jiangsu Province, PR China
| | - Huo Zhang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China
| | - Qingxie Liu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China
| | - Lin Jiang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, PR China.
| | - Xin Zhou
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China.
| | - Wei Zhu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, PR China; Department of Oncology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, No.1399 West Road, Shengze Town, Wujiang District, Suzhou 215000, China.
| |
Collapse
|
35
|
Balatti V, Oghumu S, Bottoni A, Maharry K, Cascione L, Fadda P, Parwani A, Croce C, Iwenofu OH. MicroRNA Profiling of Salivary Duct Carcinoma Versus Her2/Neu Overexpressing Breast Carcinoma Identify miR-10a as a Putative Breast Related Oncogene. Head Neck Pathol 2018; 13:344-354. [PMID: 30259272 PMCID: PMC6684709 DOI: 10.1007/s12105-018-0971-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022]
Abstract
Salivary duct carcinomas (SDC) and Her2/Neu3-overexpressing invasive breast carcinomas (HNPIBC/IBC) are histologically indistinguishable. We investigated whether common histopathologic and immunophenotypic features of SDC and IBC are mirrored by a similar microRNA (miRNA) profile. MiRNA profiling of 5 SDCs, 6 IBCs Her2/Neu3+, and 5 high-grade ductal breast carcinoma in situ (DCIS) was performed by NanoString platform. Selected miRNAs and HOXA1 gene were validated by RT-PCR. We observed similar miRNA expression profiles between IBC and SDC with the exception of 2 miRNAs, miR-10a and miR-142-3p, which were higher in IBC tumors. DCIS tumors displayed increased expression of miR-10a, miR-99a, miR-331-3p and miR-335, and decreased expression of miR-15a, miR-16 and miR-19b compared to SDC. The normal salivary gland and breast tissues also showed similar expression profiles. Interestingly, miR-10a was selectively increased in both IBC and normal breast tissue compared to SDC and normal salivary gland tissue. Moreover, our NanoString and RT-PCR data confirmed that miR-10a was upregulated in IBC and DCIS compared to SDC. Finally, we show downregulation of HOXA1, a miR-10 target, in IBC tumors compared to normal breast tissue. Taken together, our data demonstrates that, based on miRNA profiling, SDC is closely related to HNPIBC. Our results also suggest that miR-10a is differentially expressed in IBC compared to SDC and may have potential utility as a diagnostic biomarker in synchronous or metachronous malignant epithelial malignancies involving both organs. In addition, miR-10a could be playing an important role as a mammary-specific oncogene, involved in breast cancer initiation (DCIS) and progression (IBC), through mechanisms that include modulation of HOXA1 gene expression.
Collapse
Affiliation(s)
- Veronica Balatti
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, USA
| | - Steve Oghumu
- Department of Pathology and Laboratory Medicine, The Ohio State University, Columbus, USA
| | - Arianna Bottoni
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, USA
| | - Kati Maharry
- Department of Epidemiology, College of Public Health, The Ohio State University, Columbus, USA
| | - Luciano Cascione
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, USA ,Institute of Oncology Research, Bellinzona, Switzerland
| | - Paolo Fadda
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, USA
| | - Anil Parwani
- Department of Pathology and Laboratory Medicine, The Ohio State University, Columbus, USA
| | - Carlo Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, USA
| | - O. Hans Iwenofu
- Department of Pathology and Laboratory Medicine, The Ohio State University, Columbus, USA
| |
Collapse
|
36
|
Luo L, Yu ZP, Qin H, Zhu ZX, Liao MH, Liao HT, Yuan KF, Zeng Y. Exosomal MicroRNA-10a Is Associated with Liver Regeneration in Rats through Downregulation of EphA4. Chin Med J (Engl) 2018; 131:454-460. [PMID: 29451151 PMCID: PMC5830831 DOI: 10.4103/0366-6999.225057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background: MicroRNAs (miRNAs) have been reported to play vital roles in liver regeneration. Previous studies mainly focused on the functions of intracellular miRNAs, while the functions of circulating exosomal miRNAs in liver regeneration remain largely unknown. The aim of this study was to identify the key exosomal miRNA that played vital roles in liver regeneration. Methods: The Sprague–Dawley male rats were assigned to 70% partially hepatectomized group (n = 6) and sham surgery group (n = 6). The peripheral blood of both groups was collected 24 h after surgery. The exosomal miRNAs were extracted, and microarray was used to find out the key miRNA implicated in liver regeneration. Adenovirus was used to overexpress the key miRNA in rats, and proliferating cell nuclear antigen (PCNA) staining was applied to study the effect of key miRNA overexpression on liver regeneration. Western blotting was used to validate the predicted target of the key miRNA. Results: Exosomal miR-10a was upregulated more than nine times in hepatectomized rats. The level of miR-10a was increased in the early phase of liver regeneration, reached the top at 72 h postsurgery, and decreased to perioperative level 168 h after surgery. Moreover, enforced expression of miR-10a by adenovirus facilitated the process of liver regeneration as evidenced by immunohistochemical staining of PCNA. Erythropoietin-producing hepatocellular receptor A4 (EphA4) has been predicted to be a target of miR-10a. The protein level of EphA4 was decreased in the early phase of liver regeneration, reached the bottom at 72 h postsurgery, and rose to perioperative level 168 h after surgery, which was negatively correlated with miR-10a, confirming that EphA4 served as a downstream target of miR-10a. Moreover, inhibition of EphA4 by rhynchophylline could promote the proliferation of hepatocytes by regulating the cell cycle. Conclusion: Exosomal miR-10a might accelerate liver regeneration through downregulation of EphA4.
Collapse
Affiliation(s)
- Lin Luo
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ze-Ping Yu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Han Qin
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ze-Xin Zhu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ming-Heng Liao
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hao-Tian Liao
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ke-Fei Yuan
- Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yong Zeng
- Department of Liver Surgery, Liver Transplantation Division, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
37
|
Botti G, De Chiara A, Di Bonito M, Cerrone M, Malzone MG, Collina F, Cantile M. Noncoding RNAs within the
HOX
gene network in tumor pathogenesis and progression. J Cell Physiol 2018; 234:395-413. [DOI: 10.1002/jcp.27036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Gerardo Botti
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Anna De Chiara
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Maurizio Di Bonito
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Margherita Cerrone
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Maria Gabriella Malzone
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Francesca Collina
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| | - Monica Cantile
- Department of Support for Oncological Pathways Diagnostic Area, Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale” Napoli Italy
| |
Collapse
|
38
|
Masalha M, Sidi Y, Avni D. The contribution of feedback loops between miRNAs, cytokines and growth factors to the pathogenesis of psoriasis. Exp Dermatol 2018; 27:603-610. [DOI: 10.1111/exd.13520] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Moamen Masalha
- Laboratory of Molecular Cell Biology; Center for Cancer Research and Department of Medicine C; Sheba Medical Center; Tel Hashomer Israel
- Faculty of Medicine; Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Yechezkel Sidi
- Laboratory of Molecular Cell Biology; Center for Cancer Research and Department of Medicine C; Sheba Medical Center; Tel Hashomer Israel
- Faculty of Medicine; Sackler School of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Dror Avni
- Laboratory of Molecular Cell Biology; Center for Cancer Research and Department of Medicine C; Sheba Medical Center; Tel Hashomer Israel
| |
Collapse
|
39
|
Xiong G, Huang H, Feng M, Yang G, Zheng S, You L, Zheng L, Hu Y, Zhang T, Zhao Y. MiR-10a-5p targets TFAP2C to promote gemcitabine resistance in pancreatic ductal adenocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:76. [PMID: 29615098 PMCID: PMC5883523 DOI: 10.1186/s13046-018-0739-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/18/2018] [Indexed: 12/22/2022]
Abstract
Background By regulating target genes, microRNAs play essential roles in carcinogenesis and drug resistance in human pancreatic ductal adenocarcinoma (PDAC). Previous studies have shown that microRNA-10a-5p (miR-10a-5p) is overexpressed in PDAC and acts as an oncogene to promote the metastatic behavior of PDAC cells. However, the role of miR-10a-5p in PDAC chemoresistance remains unclear. Methods The effects of miR-10a-5p on biological behaviors were analyzed. MiR-10a-5p and TFAP2C levels in tissues were detected, and the clinical value was evaluated. Results We found that miR-10a-5p is up-regulated in gemcitabine-resistant PDAC cells and enhances PDAC cell gemcitabine resistance in vitro and vivo. Meanwhile, we also determined that miR-10a-5p promotes the migratory and invasive ability of PDAC cells. Next, we confirmed that transcription factor activating protein 2 gamma (TFAP2C) is a target of miR-10a-5p, and TFAP2C overexpression resensitizes PDAC cells to gemcitabine, which is initiated by miR-10a-5p. Further studies revealed that TFAP2C also decreased PDAC cell migration and invasion capability. Finally, survival analysis demonstrated that high miR-10a-5p expression levels and low TFAP2C expression levels were both independent adverse prognostic factors in patients with PDAC. Conclusion Together, these results indicate that miR-10a-5p/TFAP2C may be new therapeutic target and prognostic marker in PDAC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0739-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guangbing Xiong
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.,Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Hua Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Suli Zheng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ya Hu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China. .,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
| |
Collapse
|
40
|
|
41
|
Duell EJ, Lujan-Barroso L, Sala N, McElyea SD, Overvad K, Tjonneland A, Olsen A, Weiderpass E, Busund LT, Moi L, Muller D, Vineis P, Aune D, Matullo G, Naccarati A, Panico S, Tagliabue G, Tumino R, Palli D, Kaaks R, Katzke VA, Boeing H, Bueno-de-Mesquita H, Peeters PH, Trichopoulou A, Lagiou P, Kotanidou A, Travis RC, Wareham N, Khaw KT, Quiros JR, Rodríguez-Barranco M, Dorronsoro M, Chirlaque MD, Ardanaz E, Severi G, Boutron-Ruault MC, Rebours V, Brennan P, Gunter M, Scelo G, Cote G, Sherman S, Korc M. Plasma microRNAs as biomarkers of pancreatic cancer risk in a prospective cohort study. Int J Cancer 2017; 141:905-915. [PMID: 28542740 PMCID: PMC5536971 DOI: 10.1002/ijc.30790] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/27/2017] [Accepted: 04/19/2017] [Indexed: 02/06/2023]
Abstract
Noninvasive biomarkers for early pancreatic ductal adenocarcinoma (PDAC) diagnosis and disease risk stratification are greatly needed. We conducted a nested case-control study within the Prospective Investigation into Cancer and Nutrition (EPIC) cohort to evaluate prediagnostic microRNAs (miRs) as biomarkers of subsequent PDAC risk. A panel of eight miRs (miR-10a, -10b, -21-3p, -21-5p, -30c, -106b, -155 and -212) based on previous evidence from our group was evaluated in 225 microscopically confirmed PDAC cases and 225 controls matched on center, sex, fasting status and age/date/time of blood collection. MiR levels in prediagnostic plasma samples were determined by quantitative RT-PCR. Logistic regression was used to model levels and PDAC risk, adjusting for covariates and to estimate area under the receiver operating characteristic curves (AUC). Plasma miR-10b, -21-5p, -30c and -106b levels were significantly higher in cases diagnosed within 2 years of blood collection compared to matched controls (all p-values <0.04). Based on adjusted logistic regression models, levels for six miRs (miR-10a, -10b, -21-5p, -30c, -155 and -212) overall, and for four miRs (-10a, -10b, -21-5p and -30c) at shorter follow-up time between blood collection and diagnosis (≤5 yr, ≤2 yr), were statistically significantly associated with risk. A score based on the panel showed a linear dose-response trend with risk (p-value = 0.0006). For shorter follow-up (≤5 yr), AUC for the score was 0.73, and for individual miRs ranged from 0.73 (miR-212) to 0.79 (miR-21-5p).
Collapse
Affiliation(s)
- Eric J. Duell
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Leila Lujan-Barroso
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Núria Sala
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Samantha Deitz McElyea
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kim Overvad
- Aarhus University, Department of Public Health, Section for Epidemiology, Aarhus C, Denmark
| | | | - Anja Olsen
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elisabete Weiderpass
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Community Medicine, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
- Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
| | - Lill-Tove Busund
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Line Moi
- Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - David Muller
- School of Public Health, Epidemiology & Biostatistics, Imperial College London, London, United Kingdom
| | - Paolo Vineis
- School of Public Health, Epidemiology & Biostatistics, Imperial College London, London, United Kingdom
| | - Dagfinn Aune
- School of Public Health, Epidemiology & Biostatistics, Imperial College London, London, United Kingdom
| | - Giuseppe Matullo
- Human Genetics Foundation (HuGeF), Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Giovanna Tagliabue
- Lombardy Cancer Registry Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civic - M.P. Arezzo" Hospital, ASP Ragusa, Italy
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Cancer Research and Prevention Institute – ISPO, Florence- Italy
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena A. Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany
| | - H.B(as) Bueno-de-Mesquita
- Dt. for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Dt. of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom
- Dt. of Social & Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Petra H. Peeters
- Dept of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- MRC-PHE Centre for Environment and Health, Dept of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Dept. of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Pagona Lagiou
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Dept. of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Greece
- Department of Epidemiology, Harvard School of Public Health, Boston, USA
| | - Anastasia Kotanidou
- Hellenic Health Foundation, Athens, Greece
- Department of Critical Care Medicine & Pulmonary Services, University of Athens Medical School, Evangelismos Hospital, Athens, Greece
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Kay-Tee Khaw
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - Miguel Rodríguez-Barranco
- Andalusian School of Public Health, Research Insititute Biosanitary Granada, University Hospital Granada/University of Granada, Granada
- CIBER Epidemiology and Public Health (CIBERESP), Madrid
| | - Miren Dorronsoro
- CIBER Epidemiology and Public Health (CIBERESP), Madrid
- Basque Regional Health Department, San Sebatian, Spain
| | - María-Dolores Chirlaque
- CIBER Epidemiology and Public Health (CIBERESP), Madrid
- Department of Epidemiology, Murcia Regional Health Authority, Murcia, Spain
| | - Eva Ardanaz
- CIBER Epidemiology and Public Health (CIBERESP), Madrid
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Gianluca Severi
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
- Gustave Roussy, Villejuif, France
| | | | - Vinciane Rebours
- Pancreatology Unit, Beaujon Hospital, Clichy, France
- INSERM, University Paris, France
| | - Paul Brennan
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Marc Gunter
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Greg Cote
- Medical University of South Carolina, Charleston, USA
| | - Stuart Sherman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Murray Korc
- Departments of Medicine and Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, USA
- Pancreatic Cancer Signature Center, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, USA
| |
Collapse
|
42
|
Arai T, Okato A, Kojima S, Idichi T, Koshizuka K, Kurozumi A, Kato M, Yamazaki K, Ishida Y, Naya Y, Ichikawa T, Seki N. Regulation of spindle and kinetochore-associated protein 1 by antitumor miR-10a-5p in renal cell carcinoma. Cancer Sci 2017; 108:2088-2101. [PMID: 28746769 PMCID: PMC5623743 DOI: 10.1111/cas.13331] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/20/2017] [Accepted: 07/23/2017] [Indexed: 12/11/2022] Open
Abstract
Analysis of our original microRNA (miRNA) expression signature of patients with advanced renal cell carcinoma (RCC) showed that microRNA‐10a‐5p (miR‐10a‐5p) was significantly downregulated in RCC specimens. The aims of the present study were to investigate the antitumor roles of miR‐10a‐5p and the novel cancer networks regulated by this miRNA in RCC cells. Downregulation of miR‐10a‐5p was confirmed in RCC tissues and RCC tissues from patients treated with tyrosine kinase inhibitors (TKI). Ectopic expression of miR‐10a‐5p in RCC cell lines (786‐O and A498 cells) inhibited cancer cell migration and invasion. Spindle and kinetochore‐associated protein 1 (SKA1) was identified as an antitumor miR‐10a‐5p target by genome‐based approaches, and direct regulation was validated by luciferase reporter assays. Knockdown of SKA1 inhibited cancer cell migration and invasion in RCC cells. Overexpression of SKA1 was observed in RCC tissues and TKI‐treated RCC tissues. Moreover, analysis of The Cancer Genome Atlas database demonstrated that low expression of miR‐10a‐5p and high expression of SKA1 were significantly associated with overall survival in patients with RCC. These findings showed that downregulation of miR‐10a‐5p and overexpression of the SKA1 axis were highly involved in RCC pathogenesis and resistance to TKI treatment in RCC.
Collapse
Affiliation(s)
- Takayuki Arai
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Okato
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Satoko Kojima
- Department of Urology, Teikyo University Chiba Medical Center, Ichihara, Japan
| | - Tetsuya Idichi
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Keiichi Koshizuka
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Akira Kurozumi
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Mayuko Kato
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kazuto Yamazaki
- Department of Pathology, Teikyo University Chiba Medical Center, Ichihara, Japan
| | - Yasuo Ishida
- Department of Pathology, Teikyo University Chiba Medical Center, Ichihara, Japan
| | - Yukio Naya
- Department of Urology, Teikyo University Chiba Medical Center, Ichihara, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naohiko Seki
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
43
|
Kraft S, Moore JB, Muzikansky A, Scott KL, Duncan LM. Differential UBE2C and HOXA1 expression in melanocytic nevi and melanoma. J Cutan Pathol 2017; 44:843-850. [DOI: 10.1111/cup.12997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 06/16/2017] [Accepted: 06/22/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Stefan Kraft
- Pathology Service and Dermatopathology Unit; Massachusetts General Hospital and Harvard Medical School; Boston Massachusetts
| | - Johanna B. Moore
- Pathology Service and Dermatopathology Unit; Massachusetts General Hospital and Harvard Medical School; Boston Massachusetts
- Department of Dermatopathology; Western Pathology Inc; San Luis Obispo California
| | - Alona Muzikansky
- Biostatistics Center; Massachusetts General Hospital and Harvard Medical School; Boston Massachusetts
| | - Kenneth L. Scott
- Department of Molecular and Human Genetics; Baylor College of Medicine; Houston Texas
| | - Lyn M. Duncan
- Pathology Service and Dermatopathology Unit; Massachusetts General Hospital and Harvard Medical School; Boston Massachusetts
| |
Collapse
|
44
|
XRN2 promotes EMT and metastasis through regulating maturation of miR-10a. Oncogene 2017; 36:3925-3933. [PMID: 28319071 DOI: 10.1038/onc.2017.39] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 01/05/2017] [Accepted: 01/31/2017] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) have been proposed as critical regulatory molecules in the epithelial-mesenchymal transition (EMT) program. However, the roles of mature miRNA biogenesis during EMT process needs to be defined. Here we determined that increased expression of XRN2 induced EMT and promoted metastasis in vitro and in vivo. Furthermore, we uncovered that XRN2 functions as pro-metastatic gene, which accelerates miR-10a maturation by binding pre-miR-10a in a DICER-independent manner. These findings suggest that XRN2 is a novel regulator of EMT that contributes to the metastatic processes in lung cancer through a novel miRNA regulatory mechanism.
Collapse
|
45
|
Zou D, Zhou Q, Wang D, Guan L, Yuan L, Li S. The Downregulation of MicroRNA-10b and its Role in Cervical Cancer. Oncol Res 2017; 24:99-108. [PMID: 27296950 PMCID: PMC7838698 DOI: 10.3727/096504016x14611963142173] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
It has been demonstrated that microRNAs (miRNAs) act as oncogenes or tumor suppressors in a variety of cancers. Our previous work suggested that miR-10a/b functioned as a tumor suppressor in gastric cancer, and miR-10b was also reported to be significantly downregulated in advanced stage cervical cancer tissues. However, the aberrant expression of miR-10b in cervical cancer and its possible role in cervical carcinogenesis was largely unknown. In this study, we investigated the expression of miR-10b in cervical cancer tissues, carcinoma in situ tissues, mild dysplasia, moderate dysplasia, severe dysplasia tissues, and normal controls. We found that miR-10b was significantly downregulated during cervical cancer progression, and the lower level of miR-10b in cervical cancer was significantly associated with a more aggressive tumor phenotype. Moreover, overexpression of miR-10b in cervical cancer cells could inhibit the cell proliferation and invasion, and the further mechanism study suggested that its role was possibly through directly targeting HOXA1. These results suggested that the downregulation of miR-10b and the resulting elevated HOXA1 level in cervical cancer tissues might play critical roles in cervical cancer progression.
Collapse
Affiliation(s)
- Dongling Zou
- Department of Radiological Medicine, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
46
|
Wang D, Zhai G, Ji Y, Jing H. microRNA-10a Targets T-box 5 to Inhibit the Development of Cardiac Hypertrophy. Int Heart J 2017; 58:100-106. [PMID: 28100873 DOI: 10.1536/ihj.16-020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The mechanism of cardiac hypertrophy involving microRNAs (miRNAs) is attracting increasing attention. Our study aimed to investigate the role of miR-10a in cardiac hypertrophy development and the underlying regulatory mechanism.Transverse abdominal aortic constriction (TAAC) surgery was performed to establish a cardiac hypertrophy rat model, and angiotensin II (AngII) was used to induce cardiac hypertrophy in cultured neonatal rat cardiomyocytes. Expression of T-box 5 (TBX5) and miR-10a was altered by cell transfection of siRNA or miRNA mimic/inhibitor. Leucine incorporation assay, histological and cytological examination, quantitative real-time PCR (qRT-PCR), and Western blot were performed to detect the effects of miR-10a and TBX5 on cardiac hypertrophy. Dual-luciferase reporter assay was conducted to verify the regulation of TBX5 by miR-10a.miR-10a was down-regulated, and TBX5 was up-regulated in the rat model and AngII-stimulated cardiomyocytes. miR-10a inhibited TBX5 expression by directly targeting the binding site in Tbx5 3'UTR. Overexpression of miR-10a in AngII-treated cardiomyocytes decreased relative cell area, and significantly reduced the mRNA levels of natriuretic peptide A (Nppa), myosin heavy chain 7 cardiac muscle beta (Myh7), and leucine incorporation (P < 0.01 or P < 0.001). Knockdown of Tbx5 had similar effects on AngII-induced cardiomyocytes.Our findings indicate that miR-10a may inhibit cardiac hypertrophy via targeting Tbx5. Thus, miR-10a provides promising therapeutic strategies for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Dan Wang
- Fifth Department of Cardiology, Zhengzhou Central Hospital
| | | | | | | |
Collapse
|
47
|
Azarnezhad A, Mehdipour P. Cancer Genetics at a Glance: The Comprehensive Insights. CANCER GENETICS AND PSYCHOTHERAPY 2017:79-389. [DOI: 10.1007/978-3-319-64550-6_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
48
|
Lu Y, Wei G, Liu L, Mo Y, Chen Q, Xu L, Liao R, Zeng D, Zhang K. Direct targeting of MAPK8IP1 by miR-10a-5p is a major mechanism for gastric cancer metastasis. Oncol Lett 2016; 13:1131-1136. [PMID: 28454224 PMCID: PMC5403407 DOI: 10.3892/ol.2016.5544] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 10/24/2016] [Indexed: 12/28/2022] Open
Abstract
MicroRNA (miRNA) are endogenous non-coding RNAs that suppress gene expression at the transcriptional, post-transcriptional or translational level by targeting the 3'-UTRs of specific mRNAs. miR-10a has been frequently reported to be aberrantly overexpressed in human tumors. In gastric cancer (GC), miR-10a has an important role in the metastasis from primary GC to lymph nodes. However, the role and relevant pathways of miR-10a in GC metastasis remain largely unknown. The present study was performed using 41 GC and 20 normal gastric mucosa tissues. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis demonstrated that MAPK8IP1 was significant downregulated in GC tissue. A statistically significant inverse correlation was detected between miR-10a and MAPK8IP1 mRNA expression levels in GC specimens. Luciferase reporter assay and qPCR results suggested that MAPK8IP1 was a direct target of miR-10a in GC cells. Matrigel invasion assay and wound-healing assay results showed that MAPK8IP1 overexpression rescued the increased migration ability of miR-10a effectors in MKN45 cells. Furthermore, the underlying mechanism of miR-10a functions in GC was explored. The findings indicated that miR-10a-5p directly targets MAPK8IP1, as a major mechanism for gastric cancer metastasis. The results of the present study suggested that miR-10a may be a potential target for the treatment of GC in the future.
Collapse
Affiliation(s)
- Yaoyong Lu
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Ganbao Wei
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Liangbo Liu
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Yichao Mo
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Qingsheng Chen
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Lufei Xu
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Rongwei Liao
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Dehao Zeng
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| | - Kunqiang Zhang
- Department of Radiation Oncology, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, P.R. China
| |
Collapse
|
49
|
A SNP in pri-miR-10a is associated with recurrent spontaneous abortion in a Han-Chinese population. Oncotarget 2016; 7:8208-22. [PMID: 26824181 PMCID: PMC4884987 DOI: 10.18632/oncotarget.7002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/01/2016] [Indexed: 11/25/2022] Open
Abstract
MicroRNA-10a (miR-10a) has a wide range of functions in nearly all mammalian tissues and is involved in the occurrence of many diseases. However, it remains unknown whether miR-10a is associated with human recurrent spontaneous abortion (RSA). In this study, we found that rs3809783 A > T in miR-10a coding region was significantly associated with the increase of the risk of human unexplained RSA (URSA) acquisition in a Han-Chinese population. The T allele of rs3809783 hindered the production of mature miR-10a. A to T substitution in miR-10a rs3809783 repressed cell proliferation and migratory capacity. Further investigation discovered that Bcl-2-interacting mediator (Bim) was the functional target of miR-10a and inversely regulated Bim expression. Dual-luciferase assay indicated that A allele in miR-10a rs3809783 could more effectively suppress Bim expression than T allele. In addition, A to T substitution in miR-10a rs3809783 attenuated the sensibility of cells to progesterone and its antagonist mifepristone. Collectively, our data suggest that rs3809783 A > T in pri-miR-10a may be conductive to the genetic predisposition to RSA by disrupting the production of mature miR-10a and reinforcing the expression of Bim.
Collapse
|
50
|
Wang D, Zhu ZM, Tu YL, Dou CQ, Xu Y, Tan XL, Han MM, Yang ZJ, Jin X, Zhang B, Cai S, Liu ZW. Identfication of key miRNAs in pancreatitis using bioinformatics analysis of microarray data. Mol Med Rep 2016; 14:5451-5460. [PMID: 27840954 PMCID: PMC5355656 DOI: 10.3892/mmr.2016.5928] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 09/13/2016] [Indexed: 12/27/2022] Open
Abstract
Pancreatitis is a type of inflammation in the pancreas, which frequently occurs due to alcohol and gallstones. The present study aimed to identify pancreatitis-associated microRNAs (miRNAs) by analyzing the microarray of GSE24279. GSE24279 was downloaded from the Gene Expression Omnibus, composed of a collective of 27 pancreatitis and 22 normal control samples. The differentially expressed miRNAs (DE-miRNAs) in pancreatitis samples were screened using the Limma package in Bioconductor. Subsequently, target genes of the DE-miRNAs were predicted using the miRecords and miRWalk databases. Their potential functions were analyzed by functional and pathway enrichment analysis using the Database for Annotation, Visualization and Integrated Discovery online tool. Finally, pancreatitis-associated genes among the target genes identified were searched using the Comparative Toxicogenomics Database, and a regulatory network of pancreatitis-associated genes and their target miRNAs were constructed using Cytoscape software. A total 14 upregulated and 39 downregulated miRNAs were identified in pancreatitis samples compared with control samples and 290 target genes of DE-miRNAs were determined. Cyclin D1 (CCND1), v-akt murine thymoma viral oncogene homolog 2 (AKT2), cyclin-dependent kinase 6 (CDK6) and SMAD family member 2 (SMAD2) were involved in the pathway of pancreatic cancer. Among the target genes, 279 genes were pancreatitis-associated genes, which in turn were targeted by 37 miRNAs in the regulatory network. Hsa-miR-15a, hsa-miR-16, hsa-miR-155, hsa-miR-375 and hsa-miR-429 in particular may be involved in pancreatitis by targeting genes in the regulatory network, including hsa-miR-15a→CCND1, hsa-miR-16→CCND1, hsa-miR-155→CCND1/SMAD2, hsa-miR-375→AKT2/CDK6 and hsa-miR-429→CCND1. The above miRNAs and their targets may contribute to the pathogenesis of pancreatitis; therefore, they may be potential therapeutic targets.
Collapse
Affiliation(s)
- Dadong Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Zi-Man Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Yu-Liang Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Chun-Qing Dou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Yong Xu
- Department of Second Surgical Oncology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Xiang-Long Tan
- Department of Second Surgical Oncology, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Ming-Ming Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Zhuang-Jie Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Xin Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Bao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Shouwang Cai
- Department of Hepatobiliary Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Zhi-Wei Liu
- Department of Hepatobiliary Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|