1
|
Liu J, Pan Y, Liu Y, Wei W, Hu X, Xin W, Chen N. The regulation of PTEN: Novel insights into functions as cancer biomarkers and therapeutic targets. J Cell Physiol 2023; 238:1693-1715. [PMID: 37334436 DOI: 10.1002/jcp.31053] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023]
Abstract
This review summarizes the implications of the primary tumor suppressor protein phosphatase and tensin homolog (PTEN) in aggressive cancer development. PTEN interacts with other cellular proteins or factors suggesting the existence of an intricate molecular network that regulates their oncogenic function. Accumulating evidence has shown that PTEN exists and plays a role in the cytoplasmic organelles and in the nucleus. PTEN blocks phosphoinositide 3-kinases (PI3K)-protein kinase B-mammalian target of rapamycin signaling pathway by dephosphorylating phosphatidylinositol (PI)-3,4,5-triphosphate to PI-4,5-bisphosphate thus counteracting PI3K function. Studies have shown that PTEN expression is tightly regulated at transcriptional, posttranscriptional, and posttranslational levels (including protein-protein interactions and posttranslational modifications). Despite recent advances in PTEN research, the regulation and function of the PTEN gene remain largely unknown. How mutation or loss of specific exons in the PTEN gene occurs and involves in cancer development is not clear. This review illustrates the regulatory mechanisms of PTEN expression and discusses how PTEN participates in tumor development and/or suppression. Future prospects for the clinical applications are also highlighted.
Collapse
Affiliation(s)
- Jie Liu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yongli Pan
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Yuheng Liu
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Wei Wei
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaoping Hu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wenqiang Xin
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Nan Chen
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
2
|
Coronel-Hernández J, Delgado-Waldo I, Cantú de León D, López-Camarillo C, Jacobo-Herrera N, Ramos-Payán R, Pérez-Plasencia C. HypoxaMIRs: Key Regulators of Hallmarks of Colorectal Cancer. Cells 2022; 11:1895. [PMID: 35741024 PMCID: PMC9221210 DOI: 10.3390/cells11121895] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 01/27/2023] Open
Abstract
Hypoxia in cancer is a thoroughly studied phenomenon, and the logical cause of the reduction in oxygen tension is tumor growth itself. While sustained hypoxia leads to death by necrosis in cells, there is an exquisitely regulated mechanism that rescues hypoxic cells from their fatal fate. The accumulation in the cytoplasm of the transcription factor HIF-1α, which, under normoxic conditions, is marked for degradation by a group of oxygen-sensing proteins known as prolyl hydroxylases (PHDs) in association with the von Hippel-Lindau anti-oncogene (VHL) is critical for the cell, as it regulates different mechanisms through the genes it induces. A group of microRNAs whose expression is regulated by HIF, collectively called hypoxaMIRs, have been recognized. In this review, we deal with the hypoxaMIRs that have been shown to be expressed in colorectal cancer. Subsequently, using data mining, we analyze a panel of hypoxaMIRs expressed in both normal and tumor tissues obtained from TCGA. Finally, we assess the impact of these hypoxaMIRs on cancer hallmarks through their target genes.
Collapse
Affiliation(s)
- Jossimar Coronel-Hernández
- Genomics Laboratory, The National Cancer Institute of México, Tlalpan, Mexico City 14080, Mexico; (I.D.-W.); (D.C.d.L.)
- Functional Genomics Laboratory, Biomedicine Unit, FES-IZTACALA, UNAM, Tlalnepantla 54090, Mexico
| | - Izamary Delgado-Waldo
- Genomics Laboratory, The National Cancer Institute of México, Tlalpan, Mexico City 14080, Mexico; (I.D.-W.); (D.C.d.L.)
| | - David Cantú de León
- Genomics Laboratory, The National Cancer Institute of México, Tlalpan, Mexico City 14080, Mexico; (I.D.-W.); (D.C.d.L.)
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City 03100, Mexico;
| | - Nadia Jacobo-Herrera
- Biochemistry Unit, Institute of Medical Sciences and Nutrition, Salvador Zubirán, Tlalpan, Mexico City 14080, Mexico;
| | - Rosalío Ramos-Payán
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacan City 80030, Mexico;
| | - Carlos Pérez-Plasencia
- Genomics Laboratory, The National Cancer Institute of México, Tlalpan, Mexico City 14080, Mexico; (I.D.-W.); (D.C.d.L.)
- Functional Genomics Laboratory, Biomedicine Unit, FES-IZTACALA, UNAM, Tlalnepantla 54090, Mexico
| |
Collapse
|
3
|
Estaras M, Martinez R, García A, Ortiz-Placin C, Iovanna JL, Santofimia-Castaño P, Gonzalez A. Melatonin modulates metabolic adaptation of pancreatic stellate cells subjected to hypoxia. Biochem Pharmacol 2022; 202:115118. [DOI: 10.1016/j.bcp.2022.115118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022]
|
4
|
Dexmedetomidine Mitigates Microglial Activation Associated with Postoperative Cognitive Dysfunction by Modulating the MicroRNA-103a-3p/VAMP1 Axis. Neural Plast 2022; 2022:1353778. [PMID: 35494481 PMCID: PMC9042642 DOI: 10.1155/2022/1353778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Surgery-induced microglial activation is critical in mediating postoperative cognitive dysfunction (POCD) in elderly patients, where the important protective effect of dexmedetomidine has been indicated. However, the mechanisms of action of dexmedetomidine during the neuroinflammatory response that underlies POCD remain largely unknown. We found that lipopolysaccharide (LPS) induced substantial inflammatory responses in primary and BV2 microglial cells. The screening of differentially expressed miRNAs revealed that miR-103a-3p was downregulated in these cell culture models. Overexpression of miR-103a-3p mimics and inhibitors suppressed and enhanced the release of inflammatory factors, respectively. VAMP1 expression was upregulated in LPS-treated primary and BV-2 microglial cells, and it was validated as a downstream target of miR-103-3p. VAMP1-knockdown significantly inhibited the LPS-induced inflammatory response. Dexmedetomidine treatment markedly inhibited LPS-induced inflammation and the expression of VAMP1, and miR-103a-3p expression reversed this inhibition. Moreover, dexmedetomidine mitigated microglial activation and the associated inflammatory response in a rat model of surgical trauma that mimicked POCD. In this model, dexmedetomidine reversed miR-103a-3p and VAMP1 expression; this effect was abolished by miR-103a-3p overexpression. Taken together, the data show that miR-103a-3p/VAMP1 is critical for surgery-induced microglial activation of POCD.
Collapse
|
5
|
Ye H, Kang L, Yan X, Li S, Huang Y, Mu R, Duan X, Chen L. MiR-103a-3p Promotes Zika Virus Replication by Targeting OTU Deubiquitinase 4 to Activate p38 Mitogen-Activated Protein Kinase Signaling Pathway. Front Microbiol 2022; 13:862580. [PMID: 35317262 PMCID: PMC8934420 DOI: 10.3389/fmicb.2022.862580] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/18/2022] [Indexed: 12/21/2022] Open
Abstract
Background MicroRNAs (miRNAs) play critical roles in regulating virus infection and replication. However, the mechanism by which miRNA regulates Zika virus (ZIKV) replication remains elusive. We aim to explore how the differentially expressed miR-103a-3p regulates ZIKV replication and to clarify the underlying molecular mechanism. Methods Small RNA sequencing (RNA-Seq) was performed to identify differentially expressed miRNAs in A549 cells with or without ZIKV infection and some of the dysregulated miRNAs were validated by quantitative real time PCR (qRT-PCR). The effect of miR-103a-3p on ZIKV replication was examined by transfecting miR-103a-3p mimic or negative control (NC) into A549 cells with or without p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 and expression levels of ZIKV NS5 mRNA and NS1 protein were detected by qRT-PCR and Western blot, respectively. The potential target genes for miR-103a-3p were predicted by four algorithms and further validated by mutation analysis through luciferase reporter assay. The predicated target gene OTU deubiquitinase (DUB) 4 (OTUD4) was over-expressed by plasmid transfection or silenced by siRNA transfection into cells prior to ZIKV infection. Activation status of p38 MAPK signaling pathway was revealed by looking at the phosphorylation levels of p38 (p-p38) and HSP27 (p-HSP27) by Western blot. Results Thirty-five differentially expressed miRNAs in ZIKV-infected A549 cells were identified by RNA-Seq analysis. Five upregulated and five downregulated miRNAs were further validated by qRT-PCR. One of the validated upregulated miRNAs, miR-103a-3p significantly stimulated ZIKV replication both at mRNA (NS5) and protein (NS1) levels. We found p38 MAPK signaling was activated following ZIKV infection, as demonstrated by the increased expression of the phosphorylation of p38 MAPK and HSP27. Blocking p38 MAPK signaling pathway using SB203580 inhibited ZIKV replication and attenuated the stimulating effect of miR-103a-3p on ZIKV replication. We further identified OTUD4 as a direct target gene of miR-103a-3p. MiR-103a-3p over-expression or OTUD4 silencing activated p38 MAPK signaling and enhanced ZIKV replication. In contrast, OTUD4 over-expression inhibited p38 MAPK activation and decreased ZIKV replication. In addition, OTUD4 over-expression attenuated the stimulating effect of miR-103a-3p on ZIKV replication and activation of p38 MAPK signaling. Conclusion Zika virus infection induced the expression of miR-103a-3p, which subsequently activated p38 MAPK signaling pathway by targeting OTUD4 to facilitate ZIKV replication.
Collapse
Affiliation(s)
- Haiyan Ye
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Lan Kang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Xipeng Yan
- The Joint Laboratory on Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
| | - Shilin Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Yike Huang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Rongrong Mu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Xiaoqiong Duan
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
- *Correspondence: Xiaoqiong Duan,
| | - Limin Chen
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
- The Joint Laboratory on Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
- Limin Chen,
| |
Collapse
|
6
|
LncRNA-miRNA-mRNA regulatory axes in endometrial cancer: a comprehensive overview. Arch Gynecol Obstet 2022; 306:1431-1447. [PMID: 35182183 DOI: 10.1007/s00404-022-06423-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Recent research on tumorigenesis and progression has opened up an array of novel molecular mechanisms in the form of interactions between cellular non-coding RNAs (long non-coding RNA[lncRNA]/microRNA [miRNA]) and coding transcripts that regulate health and disease. Endometrial cancer (EC) is a prominent gynecological malignancy with a high incidence rate and poorly known etiology and prognostic factors that hinder the success of disease management. The emerging role of lncRNA-miRNA-mRNA interactions and their dysregulation in the pathophysiology of EC has been elucidated in many recent studies. METHODS A thorough literature review was conducted to explore information about lncRNA-miRNA-mRNA axes in EC. RESULTS Several lncRNAs act as molecular sponges that sequester various tumor suppressor miRNAs to inhibit their function, leading to the dysregulation of their target mRNA transcripts that contribute to the EC regulation. CONCLUSIONS This review summarizes these networks of molecular mechanisms and their contribution to different aspects of endometrial carcinogenesis, leading to a better conceptualization of the molecular pathways that underlie the disease and helping establish novel diagnostic biomarkers and therapeutic intervention points to aid the curative intent of EC.
Collapse
|
7
|
The promotional effect of microRNA-103a-3p in cervical cancer cells by regulating the ubiquitin ligase FBXW7 function. Hum Cell 2022; 35:472-485. [PMID: 35094292 PMCID: PMC8866291 DOI: 10.1007/s13577-021-00649-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/13/2021] [Indexed: 02/08/2023]
Abstract
MicroRNAs (miRNAs) have been reported to be involved in the initiation and progression of human tumors including cervical cancer (CC). However, the mechanisms underlying of their actions in CC remain to be fully elucidated. Herein, the differentially expressed miRNAs that were screened based on GSE55940 microarray data retrieved from Gene Expression Omnibus (GEO), and miR-103a-3p was significantly upregulated in CC tissues which was selected as the target miRNA for further research. We also found that high expression of miR-103a-3p was closely associated with histological grades, FIGO stage and distant metastasis as well as reflected poor overall survival. Moreover, miR-103a-3p inhibition decreased the growth capacity of SiHa and HeLa cells by inducing cell apoptosis. And F-box and WD repeat-domain containing protein 7 (FBXW7), a well-known tumor suppressor in many cancer types, was identified as a direct target of miR-103a-3p. It was further found that FBXW7 was significantly downregulated in CC tissues, and it was inversely correlated with miR-103a-3p expression levels. Further investigation demonstrated that FBXW7 upregulation could simulate the roles of miR-103a-3p knockdown in cell viability and apoptosis. Moreover, FBXW7 knockdown efficiently abrogated the influences of CC cells proliferation caused by miR-103a-3p inhibition. Notably, miR-103a-3p could block FBXW7 mediated the downstream transcription factor pathways. Taken together, these findings suggest that miR-103a-3p functions as an oncogene in CC by targeting FBXW7.
Collapse
|
8
|
Zhang C, Gao J, Zhu S. Hypoxia-inducible factor-1α promotes proliferation of airway smooth muscle cells through miRNA-103-mediated signaling pathway under hypoxia. In Vitro Cell Dev Biol Anim 2021; 57:944-952. [PMID: 34888746 DOI: 10.1007/s11626-021-00607-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/06/2021] [Indexed: 10/19/2022]
Abstract
The hypoxia-inducible factor-1α (HIF-1α) activated during asthma development plays a causative role in the abnormal proliferation of airway smooth muscle (ASM) cells and consequential airway remodeling. Although the underlying mechanisms of HIF-1α activity have not been fully revealed, HIF-1α-regulated miRNA signaling is considered important for disrupted differentiation and proliferation of local cells in various tissues under inflammation. We aimed to identify the key miRNA signaling involved in HIF-1α regulation of the proliferation of ASM cells. This study was based on primary ASM cells isolated from adult male rats. Three percent O2 and 21% O2 were set as hypoxic and normoxic condition for ASM cell treatment, respectively. Knockdown of HIF-1α was performed through transfection of pSUPER-shHIF-1α plasmid. Overexpression and knockdown of miRNA-103 were performed through transfection of miRNA-103 mimic or inhibitor, respectively. Levels of HIF-1α, PTEN, and PCNA were determined with Western blot and RT-qPCR. Hypoxia increased HIF-1α and miRNA-103 expression and proliferation in ASM cells. Knockdown of HIF-1α suppressed hypoxia-induced upregulation of proliferation and miRNA-103 expression in ASM cells. Knockdown of miRNA-103 displayed similar effects as knockdown of HIF-1α in ASM cells under hypoxia, while overexpression of miRNA-103 played the opposite role. Additionally, increased or decreased expression of PTEN was also detected when HIF-1α/miRNA-103 was knocked down under hypoxia or miRNA-103 was overexpressed under normoxia, respectively. Our results suggest that HIF-1α promotes the proliferation of ASM cells via upregulating miRNA-103 expression under hypoxia, and PTEN is involved in the miRNA-103-mediated signaling pathway.
Collapse
Affiliation(s)
- Cantang Zhang
- Department of Respiration, The Affiliated Hospital of Xuzhou Medical University, 89 Huaihai West Road, Xuzhou, 221000, Jiangsu, China
| | - Jin Gao
- Department of Cell Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shuyang Zhu
- Department of Respiration, The Affiliated Hospital of Xuzhou Medical University, 89 Huaihai West Road, Xuzhou, 221000, Jiangsu, China.
| |
Collapse
|
9
|
Valentino M, Dejana E, Malinverno M. The multifaceted PDCD10/CCM3 gene. Genes Dis 2021; 8:798-813. [PMID: 34522709 PMCID: PMC8427250 DOI: 10.1016/j.gendis.2020.12.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
The programmed cell death 10 (PDCD10) gene was originally identified as an apoptosis-related gene, although it is now usually known as CCM3, as the third causative gene of cerebral cavernous malformation (CCM). CCM is a neurovascular disease that is characterized by vascular malformations and is associated with headaches, seizures, focal neurological deficits, and cerebral hemorrhage. The PDCD10/CCM3 protein has multiple subcellular localizations and interacts with several multi-protein complexes and signaling pathways. Thus PDCD10/CCM3 governs many cellular functions, which include cell-to-cell junctions and cytoskeleton organization, cell proliferation and apoptosis, and exocytosis and angiogenesis. Given its central role in the maintenance of homeostasis of the cell, dysregulation of PDCD10/CCM3 can result in a wide range of altered cell functions. This can lead to severe diseases, including CCM, cognitive disability, and several types of cancers. Here, we review the multifaceted roles of PDCD10/CCM3 in physiology and pathology, with a focus on its functions beyond CCM.
Collapse
Affiliation(s)
| | - Elisabetta Dejana
- The FIRC Institute of Molecular Oncology (IFOM), Milan, 16 20139, Italy.,Department of Oncology and Haemato-Oncology, University of Milan, Milan, 7 20122, Italy.,Vascular Biology, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, SE-751 05, Sweden
| | - Matteo Malinverno
- The FIRC Institute of Molecular Oncology (IFOM), Milan, 16 20139, Italy
| |
Collapse
|
10
|
MicroRNA-103a-3p Promotes Cell Proliferation and Invasion in Non-Small-Cell Lung Cancer Cells through Akt Pathway by Targeting PTEN. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7590976. [PMID: 34307670 PMCID: PMC8279842 DOI: 10.1155/2021/7590976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/26/2021] [Indexed: 01/01/2023]
Abstract
Background Increasing evidence has suggested that microRNA- (miR-) 103a-3p is crucial for cancer progression. However, the specific mechanism of miR-103a-3p in non-small-cell lung cancer (NSCLC) remains unclear until now. So, it is particularly urgent to clarify the mechanism between them. Methods qRT-PCR and western blot were used to measure the expression of miR-103a-3p, PTEN, Akt, and p-Akt. Cell biology experiment was applied to detect the biological function of miR-103a-3p in NSCLC cell lines. Moreover, bioinformatics analysis, luciferase reporter assay, and functional complementation analysis were carried out to investigate the target gene. Results miR-103a-3p was highly expressed in primary NSCLC samples and cell lines. miR-103a-3p mimics promoted the proliferation and invasion of NSCLC cells; miR-103a-3p inhibitor had the opposite effect. A double luciferase reporter gene experiment revealed that miR-103a-3p directly targets the PTEN mRNA 3'UTR region. siPTEN inhibited the proliferation and invasion of NSCLC cells. Further mechanistic studies showed that both overexpression of miR-103a-3p and PTEN knockdown reduced the expression of the p-Akt protein. Overexpression of PTEN partially reversed the cancer-promoting effect of miR-103a-3p. Conclusion miR-103a-3p promotes the progression of NSCLC via Akt signaling by targeting PTEN, highlighting the role of miR-103a-3p/PTEN/Akt signaling and suggesting miR-103a-3p as a novel therapeutic target for NSCLC.
Collapse
|
11
|
Fodor A, Lazar AL, Buchman C, Tiperciuc B, Orasan OH, Cozma A. MicroRNAs: The Link between the Metabolic Syndrome and Oncogenesis. Int J Mol Sci 2021; 22:ijms22126337. [PMID: 34199293 PMCID: PMC8231835 DOI: 10.3390/ijms22126337] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic syndrome (MetS) represents a cluster of disorders that increase the risk of a plethora of conditions, in particular type two diabetes, cardiovascular diseases, and certain types of cancers. MetS is a complex entity characterized by a chronic inflammatory state that implies dysregulations of adipokins and proinflammatory cytokins together with hormonal and growth factors imbalances. Of great interest is the implication of microRNA (miRNA, miR), non-coding RNA, in cancer genesis, progression, and metastasis. The adipose tissue serves as an important source of miRs, which represent a novel class of adipokines, that play a crucial role in carcinogenesis. Altered miRs secretion in the adipose tissue, in the context of MetS, might explain their implication in the oncogenesis. The interplay between miRs expressed in adipose tissue, their dysregulation and cancer pathogenesis are still intriguing, taking into consideration the fact that miRNAs show both carcinogenic and tumor suppressor effects. The aim of our review was to discuss the latest publications concerning the implication of miRs dysregulation in MetS and their significance in tumoral signaling pathways. Furthermore, we emphasized the role of miRNAs as potential target therapies and their implication in cancer progression and metastasis.
Collapse
Affiliation(s)
- Adriana Fodor
- Department of Diabetes and Nutrtion, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence: (A.F.); (A.L.L.); (C.B.)
| | - Andrada Luciana Lazar
- Department of Dermatology, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence: (A.F.); (A.L.L.); (C.B.)
| | - Cristina Buchman
- Department of Oncology, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence: (A.F.); (A.L.L.); (C.B.)
| | - Brandusa Tiperciuc
- Department of Pharmaceutical Chemistry, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Olga Hilda Orasan
- Internal Medicine Department, 4th Medical Clinic “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (O.H.O.); (A.C.)
| | - Angela Cozma
- Internal Medicine Department, 4th Medical Clinic “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (O.H.O.); (A.C.)
| |
Collapse
|
12
|
Wang L, Xu B, Sun S, Wang B. Overexpression of long non-coding RNA H19 relieves hypoxia-induced injury by down-regulating microRNA-107 in neural stem cells. Neurosci Lett 2021; 753:135855. [PMID: 33785379 DOI: 10.1016/j.neulet.2021.135855] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Neonatal hypoxia-ischemia (HI) is one of the commonest conditions which seriously influences the development of infants' nervous system and causes series of neurological sequelaes. The aim of the present study was to analyze the potential regulatory mechanism of long non-coding (lnc) RNA H19 under hypoxia conditions. METHODS Neural stem cells (NSCs) were incubated in hypoxic conditions for 8 h to induce hypoxia injury. qRT-PCR was performed to detect H19 or micro (miR)-107 expression. Cell Counting Kit-8 (CCK-8) assay and Annexin V-FITC/PI staining assay were employed to detect the effects of hypoxia on cell viability and apoptosis, respectively. Moreover, NSCs were transfected with H19 overexpressing plasmid or shRNA-H19 and then subjected to hypoxia treatment. The effects of H19/miR-107 on NSC cell biological behaviors were confirmed. Furthermore, the signaling pathways involved in HI were analyzed using western blot. RESULTS Hypoxia treatment restrained cell viability and induced cell apoptosis in NSCs. Overexpression of lncRNA H19 attenuated hypoxia-induced NSCs injury, while knockdown of lncRNA H19 aggravated NSCs injury. Further experiments suggested that miR-107 up-regulation reversed the effects of lncRNA H19 overexpression on NSCs. Moreover, the activation of Wnt/β-catenin and PI3K/AKT pathways triggered by H19 were reversed by miR-107 up-regulation in hypoxia-treated NSCs. CONCLUSION LncRNA H19 overexpression attenuated hypoxia-induced NSCs injury and promoted activation of Wnt/β-catenin and PI3K/AKT pathways through downregulating miR-107.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pediatrics, Liaocheng Second People's Hospital, The Second Hospital of Liaocheng Affiliated to Shandong First Medical University, Liaocheng, 252600, Shandong, China
| | - Bin Xu
- Department of Pediatrics, Liaocheng Second People's Hospital, The Second Hospital of Liaocheng Affiliated to Shandong First Medical University, Liaocheng, 252600, Shandong, China
| | - Shuying Sun
- Department of Cardiology, Liaocheng Second People's Hospital, The Second Hospital of Liaocheng Affiliated to Shandong First Medical University, Liaocheng, 252600, Shandong, China
| | - Bin Wang
- Department of Children Rehabilitation, Liaocheng Second People's Hospital, The Second Hospital of Liaocheng Affiliated to Shandong First Medical University, Liaocheng, 252600, Shandong, China.
| |
Collapse
|
13
|
Bertolazzi G, Cipollina C, Benos PV, Tumminello M, Coronnello C. miR-1207-5p Can Contribute to Dysregulation of Inflammatory Response in COVID-19 via Targeting SARS-CoV-2 RNA. Front Cell Infect Microbiol 2020; 10:586592. [PMID: 33194826 PMCID: PMC7658538 DOI: 10.3389/fcimb.2020.586592] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
The present study focuses on the role of human miRNAs in SARS-CoV-2 infection. An extensive analysis of human miRNA binding sites on the viral genome led to the identification of miR-1207-5p as potential regulator of the viral Spike protein. It is known that exogenous RNA can compete for miRNA targets of endogenous mRNAs leading to their overexpression. Our results suggest that SARS-CoV-2 virus can act as an exogenous competing RNA, facilitating the over-expression of its endogenous targets. Transcriptomic analysis of human alveolar and bronchial epithelial cells confirmed that the CSF1 gene, a known target of miR-1207-5p, is over-expressed following SARS-CoV-2 infection. CSF1 enhances macrophage recruitment and activation and its overexpression may contribute to the acute inflammatory response observed in severe COVID-19. In summary, our results indicate that dysregulation of miR-1207-5p-target genes during SARS-CoV-2 infection may contribute to uncontrolled inflammation in most severe COVID-19 cases.
Collapse
Affiliation(s)
- Giorgio Bertolazzi
- Department of Economics, Business and Statistics, University of Palermo, Palermo, Italy
- Fondazione Ri.MED, Palermo, Italy
| | - Chiara Cipollina
- Fondazione Ri.MED, Palermo, Italy
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Panayiotis V. Benos
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michele Tumminello
- Department of Economics, Business and Statistics, University of Palermo, Palermo, Italy
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | | |
Collapse
|
14
|
Konoshenko M, Sagaradze G, Orlova E, Shtam T, Proskura K, Kamyshinsky R, Yunusova N, Alexandrova A, Efimenko A, Tamkovich S. Total Blood Exosomes in Breast Cancer: Potential Role in Crucial Steps of Tumorigenesis. Int J Mol Sci 2020; 21:E7341. [PMID: 33027894 PMCID: PMC7582945 DOI: 10.3390/ijms21197341] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/19/2020] [Accepted: 09/30/2020] [Indexed: 12/15/2022] Open
Abstract
Exosomes are crucial players in cell-to-cell communication and are involved in tumorigenesis. There are two fractions of blood circulating exosomes: free and cell-surface-associated. Here, we compared the effect of total blood exosomes (contain plasma exosomes and blood cell-surface-associated exosomes) and plasma exosomes from breast cancer patients (BCPs, n = 43) and healthy females (HFs, n = 35) on crucial steps of tumor progression. Exosomes were isolated by ultrafiltration, followed by ultracentrifugation, and characterized by cryo-electron microscopy (cryo-EM), nanoparticle tracking analysis, and flow cytometry. Cryo-EM revealed a wider spectrum of exosome morphology with lipid bilayers and vesicular internal structures in the HF total blood in comparison with plasma. No differences in the morphology of both exosomes fractions were detected in BCP blood. The plasma exosomes and total blood exosomes of BCPs had different expression levels of tumor-associated miR-92a and miR-25-3p, induced angiogenesis and epithelial-to-mesenchymal transition (EMT), and increased the number of migrating pseudo-normal breast cells and the total migration path length of cancer cells. The multidirectional effects of HF total blood exosomes on tumor dissemination were revealed; they suppress the angiogenesis and total migration path length of MCF10A, but stimulate EMT and increase the number of migrating MCF10A and the total path length of SKBR3 cells. In addition, HF plasma exosomes enhance the metastasis-promoting properties of SKBR3 cells and stimulate angiogenesis. Both cell-free and blood cell-surface-associated exosomes are involved in the crucial stages of carcinogenesis: the initiation of EMT and the stimulation of proliferation, cell migration, and angiogenesis. Thus, for the estimation of the diagnostic/prognostic significance of circulating exosomes in the blood of cancer patients more correctly, the total blood exosomes, which consist of plasma exosomes and blood cell-surface-associated exosomes should be used.
Collapse
Affiliation(s)
- Maria Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.K.); (K.P.)
| | - Georgy Sagaradze
- Medical Research and Education Center, Lomonosov Moscow State University, 119991 Moscow, Russia; (G.S.); (A.E.)
| | - Evgeniya Orlova
- N.N. Blokhin Cancer Research Center” of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (E.O.); (A.A.)
| | - Tatiana Shtam
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Center “Kurchatov Institute”, 188300 Gatchina, Russia;
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia;
| | - Ksenia Proskura
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.K.); (K.P.)
- Novosibirsk Regional Clinical Oncological Dispensary, 630108 Novosibirsk, Russia
| | - Roman Kamyshinsky
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia;
- Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Moscow region, Russia
- Shubnikov Institute of Crystallography of Federal Scientific Research Centre, “Crystallography and Photonics” of Russian Academy of Sciences, 119333 Moscow, Russia
| | - Natalia Yunusova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Science, 634050 Tomsk, Russia;
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Biology, Siberian State Medical University, 634050 Tomsk, Russia
| | - Antonina Alexandrova
- N.N. Blokhin Cancer Research Center” of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (E.O.); (A.A.)
| | - Anastasia Efimenko
- Medical Research and Education Center, Lomonosov Moscow State University, 119991 Moscow, Russia; (G.S.); (A.E.)
| | - Svetlana Tamkovich
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (M.K.); (K.P.)
- Department of Molecular Biology and Biotechnology, Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
15
|
Saberinia A, Alinezhad A, Jafari F, Soltany S, Akhavan Sigari R. Oncogenic miRNAs and target therapies in colorectal cancer. Clin Chim Acta 2020; 508:77-91. [DOI: 10.1016/j.cca.2020.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022]
|
16
|
Sellars E, Gabra M, Salmena L. The Complex Landscape of PTEN mRNA Regulation. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036236. [PMID: 31871240 DOI: 10.1101/cshperspect.a036236] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a key tumor suppressor in the development and progression of different tumor types. Emerging data indicate that small reductions in PTEN protein levels can promote cancer. PTEN protein levels are tightly controlled by a plethora of mechanisms beginning with epigenetic and transcriptional regulation and ending with control of protein synthesis and stability. PTEN messenger RNA (mRNA) is also subject to exquisite regulation by microRNAs, coding and long noncoding RNAs, and RNA-binding proteins. Additionally, PTEN mRNA is markedly influenced by alternative splicing and variable polyadenylation. Herein we provide a synoptic description of the current understanding of the complex regulatory landscape of PTEN mRNA regulation including several specific processes that modulate its stability and expression, in the context of PTEN loss-associated cancers.
Collapse
Affiliation(s)
- Erin Sellars
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Martino Gabra
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Leonardo Salmena
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C1, Canada
| |
Collapse
|
17
|
Abou-Fadel J, Qu Y, Gonzalez EM, Smith M, Zhang J. Emerging roles of CCM genes during tumorigenesis with potential application as novel biomarkers across major types of cancers. Oncol Rep 2020; 43:1945-1963. [PMID: 32186778 PMCID: PMC7160551 DOI: 10.3892/or.2020.7550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/14/2020] [Indexed: 12/31/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are microvascular anomalies in the brain that result in increased susceptibility to stroke. Three genes have been identified as causes of CCMs: cerebral cavernous malformations 1 [(CCM1) also termed Krev interaction trapped 1 (KRIT1)], cerebral cavernous malformation 2 [(CCM2) also termed MGC4607] and cerebral cavernous malformation 3 [(CCM3) also termed programmed cell death 10 (PDCD10)]. It has been demonstrated that both CCM1 and CCM3 bind to CCM2 to form a CCM signaling complex (CSC) with which to modulate multiple signaling cascades. CCM proteins have been reported to play major roles in microvascular angiogenesis in human and animal models. However, CCM proteins are ubiquitously expressed in all major tissues, suggesting an unseen broader role of the CSC in biogenesis. Recent evidence suggests the possible involvement of the CSC complex during tumorigenesis; however, studies concerning this aspect are limited. This is the first report to systematically investigate the expression patterns of CCM proteins in major human tumors using real‑time quantitative PCR, RNA‑fluorescence in situ hybridization, immunohistochemistry and multicolor immunofluorescence imaging. Our data demonstrated that differential expression patterns of the CSC complex are correlated with certain types and grades of major human cancers, indicating the potential application of CCM genes as molecular biomarkers for clinical oncology. Our data strongly suggest that more efforts are needed to elucidate the role of the CSC complex in tumorigenesis, which may have enormous clinical potential for cancer diagnostic, prognostic and therapeutic applications.
Collapse
Affiliation(s)
- Johnathan Abou-Fadel
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| | - Yanchun Qu
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| | - Elias M. Gonzalez
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| | - Mark Smith
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
18
|
Johnson TS, Li S, Franz E, Huang Z, Dan Li S, Campbell MJ, Huang K, Zhang Y. PseudoFuN: Deriving functional potentials of pseudogenes from integrative relationships with genes and microRNAs across 32 cancers. Gigascience 2019; 8:5480571. [PMID: 31029062 PMCID: PMC6486473 DOI: 10.1093/gigascience/giz046] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/13/2018] [Accepted: 03/29/2019] [Indexed: 12/14/2022] Open
Abstract
Background Long thought “relics” of evolution, not until recently have pseudogenes been of medical interest regarding regulation in cancer. Often, these regulatory roles are a direct by-product of their close sequence homology to protein-coding genes. Novel pseudogene-gene (PGG) functional associations can be identified through the integration of biomedical data, such as sequence homology, functional pathways, gene expression, pseudogene expression, and microRNA expression. However, not all of the information has been integrated, and almost all previous pseudogene studies relied on 1:1 pseudogene–parent gene relationships without leveraging other homologous genes/pseudogenes. Results We produce PGG families that expand beyond the current 1:1 paradigm. First, we construct expansive PGG databases by (i) CUDAlign graphics processing unit (GPU) accelerated local alignment of all pseudogenes to gene families (totaling 1.6 billion individual local alignments and >40,000 GPU hours) and (ii) BLAST-based assignment of pseudogenes to gene families. Second, we create an open-source web application (PseudoFuN [Pseudogene Functional Networks]) to search for integrative functional relationships of sequence homology, microRNA expression, gene expression, pseudogene expression, and gene ontology. We produce four “flavors” of CUDAlign-based databases (>462,000,000 PGG pairwise alignments and 133,770 PGG families) that can be queried and downloaded using PseudoFuN. These databases are consistent with previous 1:1 PGG annotation and also are much more powerful including millions of de novo PGG associations. For example, we find multiple known (e.g., miR-20a-PTEN-PTENP1) and novel (e.g., miR-375-SOX15-PPP4R1L) microRNA-gene-pseudogene associations in prostate cancer. PseudoFuN provides a “one stop shop” for identifying and visualizing thousands of potential regulatory relationships related to pseudogenes in The Cancer Genome Atlas cancers. Conclusions Thousands of new PGG associations can be explored in the context of microRNA-gene-pseudogene co-expression and differential expression with a simple-to-use online tool by bioinformaticians and oncologists alike.
Collapse
Affiliation(s)
- Travis S Johnson
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, USA.,Department of Medicine, Indiana University School of Medicine, 545 Barnhill Drive, Indianapolis, IN 46202, USA
| | - Sihong Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, USA
| | - Eric Franz
- Ohio Supercomputer Center, 1224 Kinnear Road, Columbus, OH 43212, USA
| | - Zhi Huang
- School of Electrical and Computer Engineering, Purdue University, 465 Northwestern Avenue, West Lafayette, IN 47907, USA.,Department of Medicine, Indiana University School of Medicine, 545 Barnhill Drive, Indianapolis, IN 46202, USA
| | - Shuyu Dan Li
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Moray J Campbell
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, 500 West 12 th Avenue, Columbus, OH 43210, USA
| | - Kun Huang
- Department of Medicine, Indiana University School of Medicine, 545 Barnhill Drive, Indianapolis, IN 46202, USA.,Regenstrief Institute, Indiana University, 1101 West 10 th Street, Indianapolis, IN 46262, USA
| | - Yan Zhang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 1800 Cannon Drive, Columbus, OH 43210, USA.,The Ohio State University Comprehensive Cancer Center (OSUCCC - James), 460 West 10 th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
19
|
Soleimani A, Rahmani F, Ferns GA, Ryzhikov M, Avan A, Hassanian SM. Role of Regulatory Oncogenic or Tumor Suppressor miRNAs of PI3K/AKT Signaling Axis in the Pathogenesis of Colorectal Cancer. Curr Pharm Des 2019; 24:4605-4610. [PMID: 30636581 DOI: 10.2174/1381612825666190110151957] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is the leading cause of cancer death worldwide and its incidence is increasing. In most patients with CRC, the PI3K/AKT signaling axis is over-activated. Regulatory oncogenic or tumor suppressor microRNAs (miRNAs) for PI3K/AKT signaling regulate cell proliferation, migration, invasion, angiogenesis, as well as resistance to chemo-/radio-therapy in colorectal cancer tumor tissues. Thus, regulatory miRNAs of PI3K/AKT/mTOR signaling represent novel biomarkers for new patient diagnosis and obtaining clinically invaluable information from post-treatment CRC patients for improving therapeutic strategies. This review summarizes the current knowledge of miRNAs' regulatory roles of PI3K/AKT signaling in CRC pathogenesis.
Collapse
Affiliation(s)
- Atena Soleimani
- Department of Medical Biochemistry, faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Department of Medical Biochemistry, faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, United Kingdom
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, MO, United States
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of M edical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Siveen KS, Raza A, Ahmed EI, Khan AQ, Prabhu KS, Kuttikrishnan S, Mateo JM, Zayed H, Rasul K, Azizi F, Dermime S, Steinhoff M, Uddin S. The Role of Extracellular Vesicles as Modulators of the Tumor Microenvironment, Metastasis and Drug Resistance in Colorectal Cancer. Cancers (Basel) 2019; 11:cancers11060746. [PMID: 31146452 PMCID: PMC6628238 DOI: 10.3390/cancers11060746] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide, with high morbidity and mortality rates. A number of factors including modulation of the tumor microenvironment, high metastatic capability, and resistance to treatment have been associated with CRC disease progression. Recent studies have documented that tumor-derived extracellular vesicles (EVs) play a significant role in intercellular communication in CRC via transfer of cargo lipids, proteins, DNA and RNAs to the recipient tumor cells. This transfer influences a number of immune-related pathways leading to activation/differentiation/expression of immune cells and modulation of the tumor microenvironment that plays a significant role in CRC progression, metastasis, and drug resistance. Furthermore, tumor-derived EVs are secreted in large amounts in biological fluids of CRC patients and as such the expression analysis of EV cargoes have been associated with prognosis or response to therapy and may be a source of therapeutic targets. This review aims to provide a comprehensive insight into the role of EVs in the modulation of the tumor microenvironment and its effects on CRC progression, metastasis, and drug resistance. On the other hand, the potential role of CRC derived EVs as a source of biomarkers of response and therapeutic targets will be discussed in detail to understand the dynamic role of EVs in CRC diagnosis, treatment, and management.
Collapse
Affiliation(s)
- Kodappully S Siveen
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Afsheen Raza
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Eiman I Ahmed
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Abdul Q Khan
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Kirti S Prabhu
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Shilpa Kuttikrishnan
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Jericha M Mateo
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Hatem Zayed
- College of Health Sciences, Department of Biomedical Sciences, Qatar University, Doha P.O. Box 2713, Qatar.
| | - Kakil Rasul
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Fouad Azizi
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Said Dermime
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| | - Martin Steinhoff
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
- Department of Dermatology Venereology, Hamad Medical Corporation, Doha, P.O. Box 3050, Qatar.
- Weill Cornell-Medicine, Doha P.O. Box 24811, Qatar.
- Weill Cornell University, New York, NY 10065, USA.
| | - Shahab Uddin
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar.
| |
Collapse
|
21
|
Zheng P, Bin H, Chen W. Inhibition of microRNA-103a inhibits the activation of astrocytes in hippocampus tissues and improves the pathological injury of neurons of epilepsy rats by regulating BDNF. Cancer Cell Int 2019; 19:109. [PMID: 31049031 PMCID: PMC6482545 DOI: 10.1186/s12935-019-0821-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
Background The aim of this study is to explore the effect of microRNA-103a (miR-103a) on astrocytes activation and hippocampal neuron injury in epilepsy rats by targeting brain-derived neurotrophic factor (BDNF). Methods The epilepsy rat model was induced by intraperitoneal injection of lithium chloride-pilocarpine. Successful modeled rats were intralateroventricularly microinjected with miR-103a inhibitors, inhibitors negative control (NC), siRNA-NC and BDNF-siRNA, respectively. The RT-qPCR and western blot analysis were used to detect the expression of miR-103a, BDNF and glial fibrillary acidic protein (GFAP) in hippocampus tissues of rats. TUNEL staining was used to detect the apoptosis of hippocampal neurons. The RT-PCR and ELISA was used to detect the levels of TNF-α and IL-6 in hippocampal tissues and in serum, respectively. Results Increased expression of miR-103a, GFAP, and number of apoptotic neurons, decreased expression of BDNF and number of surviving neurons were found in hippocampus tissues of epilepsy rats. After miR-103a inhibitors interfered with epilepsy rats, there showed decreased expression of miR-103a and GFAP, increased expression of BDNF and decreased number of apoptotic neuron as well as increased number of surviving neurons. Compared with miR-103a inhibitors alone, epilepsy rats treated with BDNF-siRNA combined with miR-103a inhibitors significantly increased expression of GFAP in hippocampal tissues of epilepsy rats, increased number of apoptotic neurons and significantly decreased the number of surviving neurons. Conclusion Our study provides evidence that the inhibition of miR-103a can inhibit the activation of astrocytes in hippocampus tissues and improve the pathological injury of neurons of epilepsy rats by regulating BDNF gene. Electronic supplementary material The online version of this article (10.1186/s12935-019-0821-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ping Zheng
- 1Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, No 490, South Chuanhuan Road, Shanghai, 201299 People's Republic of China
| | - He Bin
- 1Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, No 490, South Chuanhuan Road, Shanghai, 201299 People's Republic of China
| | - Wei Chen
- 1Department of Neurosurgery, Shanghai Pudong New Area People's Hospital, No 490, South Chuanhuan Road, Shanghai, 201299 People's Republic of China.,2Department of Neurosurgery, First affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
22
|
Ji J, Dai X, Yeung SCJ, He X. The role of long non-coding RNA GAS5 in cancers. Cancer Manag Res 2019; 11:2729-2737. [PMID: 31114330 PMCID: PMC6497482 DOI: 10.2147/cmar.s189052] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/15/2019] [Indexed: 12/31/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have shown potential as a biomarker in the diagnosis and prognosis in multiple cancers. LncRNAs are dysregulated in various cancers, playing either oncogenic or tumor suppressive roles. Emerging evidences have proved that the growth arrest-specific 5 (GAS5) lncRNA can function as a tumor suppressor in several cancers. LncRNA GAS5 is downregulated in many types of cancer, regulating cellular processes such as cell proliferation, apoptosis and invasion. The low level of GAS5 expression often elevates capacity of proliferation and predicts poorer prognosis in some cancers. This review aims to summarize the recent published literature on the biogenesis, regulation mechanism and function of GAS5 in different types of cancers and explore its potential for cancer diagnosis, prognosis and treatment.
Collapse
Affiliation(s)
- Jiali Ji
- Department of Medical Oncology, The 2nd Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaolan Dai
- Department of Pharmacy, School of Medicine, Shantou University, Shantou, Guangdong, People’s Republic of China
| | - Sai-Ching Jim Yeung
- Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuexin He
- Department of Medical Oncology, The 2nd Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
23
|
Xu L, Yuan X, Ni J, Shen L, Cai M, Jiang D. Gain of microRNA-103 triggers metastatic behavior by targeting ubiquitin specific peptidase 10 in pancreatic cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1214-1223. [PMID: 31933936 PMCID: PMC6947054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 01/13/2019] [Indexed: 06/10/2023]
Abstract
Emerging studies have shown that microRNAs (miRNAs) play key roles in regulating progression of pancreatic cancer (PaCa). miR-103 has been reported to serve as an oncomiR in hepatocellular carcinoma and colorectal cancer. However, litter is known regarding the function and molecular mechanism of miR-103 in PaCa. Here, we observed that miR-103 was markedly highly expressed in PaCa tissues and cell lines. Up-regulation of miR-103 expression was associated with advanced TNM stage, positive lymph node metastasis, and poor prognosis. Furthermore, knock-down of miR-103 by its inhibitor resulted in inhibited cell invasion and migration. Using a dual-luciferase reporter assay, we identified that USP10 (Ubiquitin specific peptidase 10) was a directly target of miR-103. In addition, by using qRT-PCR assay and western blotting analysis, we found that miR-103 down-regulated the expression of USP10 in PaCa tissues and cell lines. Taken together, the present study demonstrates that up-regulation of miR-103 is associated with tumor metastasis and poor prognosis in PaCa patients. Our data further indicates that miR-103 is an upregulated oncomiR and promotes cell metastasis by targeting USP10, suggesting miR-103 may be a potential prognosis and treatment target for PaCa.
Collapse
Affiliation(s)
- Liu Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jiaxing UniversityJiaxing, P. R. China
| | - Xiangfei Yuan
- Tianjin Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin Nankai HospitalTianjin, P. R. China
| | - Jianqi Ni
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jiaxing UniversityJiaxing, P. R. China
| | - Lan Shen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jiaxing UniversityJiaxing, P. R. China
| | - Min Cai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jiaxing UniversityJiaxing, P. R. China
| | - Dawei Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jiaxing UniversityJiaxing, P. R. China
| |
Collapse
|
24
|
Subramanian VS, Sabui S, Marchant JS, Said HM. MicroRNA-103a regulates sodium-dependent vitamin C transporter-1 expression in intestinal epithelial cells. J Nutr Biochem 2019; 65:46-53. [PMID: 30616065 PMCID: PMC6420349 DOI: 10.1016/j.jnutbio.2018.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/04/2018] [Accepted: 12/03/2018] [Indexed: 12/31/2022]
Abstract
Intestinal absorption of ascorbic acid (AA) occurs via a Na+-dependent carrier-mediated process facilitated through the human sodium-dependent vitamin C transporters-1 &-2 (hSVCT1 and hSVCT2). Many studies have shown that hSVCT1 (product of the SLC23A1 gene) is expressed on the apical membrane of polarized enterocytes where it mediates AA absorption. hSVCT1 expression levels are therefore an important determinant of physiological vitamin C homeostasis. However, little is known about posttranscriptional mechanisms that regulate hSVCT1 expression in intestinal epithelia. In this study, we investigated regulation of hSVCT1 by microRNA (miRNA). A pmirGLO-SLC23A1-3'-UTR construct transfected into human intestinal cell lines (Caco-2 and NCM460 cells) showed markedly reduced luciferase activity. Bioinformatic analysis of the SLC23A1-3'-UTR predicted five miRNA binding sites (miR-103a, miR-107, miR-328, miR-384, and miR-499-5p) in the 3'-UTR. Expression of mature miR-103a was markedly higher compared to the other four putative miRNA regulators in both intestinal cell lines and mouse jejunal mucosa. Addition of a miR-103a mimic, but not a miR-103a mutant construct, markedly reduced the luminescence of the pmirGLO-SLC23A1-3'-UTR reporter. Reciprocally, addition of a miR-103a inhibitor significantly increased luciferase reporter activity. Addition of the miR-103a mimic led to a significant inhibition in AA uptake, associated with decreased hSVCT1 mRNA and protein expression in Caco-2 cells. In contrast, the miR-103a inhibitor increased AA uptake, associated with increased levels of hSVCT1 mRNA and protein. These findings provide the first evidence for posttranscriptional regulation of hSVCT1 by miRNA in intestinal epithelial cells.
Collapse
Affiliation(s)
- Veedamali S Subramanian
- Department of Medicine, University of California, Irvine, CA 92697; Department of Physiology/Biophysics, University of California, Irvine, CA 92697; VA Medical Center, Long Beach, CA 90822.
| | - Subrata Sabui
- Department of Medicine, University of California, Irvine, CA 92697; Department of Physiology/Biophysics, University of California, Irvine, CA 92697; VA Medical Center, Long Beach, CA 90822
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, WI 53226
| | - Hamid M Said
- Department of Medicine, University of California, Irvine, CA 92697; Department of Physiology/Biophysics, University of California, Irvine, CA 92697; VA Medical Center, Long Beach, CA 90822
| |
Collapse
|
25
|
Qian T, Fan C, Liu Q, Yi S. Systemic functional enrichment and ceRNA network identification following peripheral nerve injury. Mol Brain 2018; 11:73. [PMID: 30558654 PMCID: PMC6297964 DOI: 10.1186/s13041-018-0421-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/05/2018] [Indexed: 12/28/2022] Open
Abstract
Peripheral nerve injury is a worldwide clinical issue that impacts patients' quality of life and causes huge society and economic burden. Injured peripheral nerves are able to regenerate by themselves. However, for severe peripheral nerve injury, the regenerative abilities are very limited and the regenerative effects are very poor. A better understanding of the mechanisms following peripheral nerve injury will benefit its clinical treatment. In this study, we systematically explored the dynamic changes of mRNAs and long non-coding RNAs (lncRNAs) in the injured sciatic nerve segments after nerve crush, identified significantly involved Gene ontology (GO) terms and Kyoto Enrichment of Genes and Genomes (KEGG) pathways, and innovatively analyzed the correlation of differentially expressed mRNAs and lncRNAs. After the clustering of co-expressed mRNAs and lncRNAs, we performed functional analysis, selected GO term "negative regulation of cell proliferation", and constructed a competing endogenous RNA (ceRNA) network of LIF and HMOX1 gene in this GO term. This study is the first to provide a systematic dissection of mRNA-microRNA (miRNA)-lncRNA ceRNA network following peripheral nerve injury and thus lays a foundation for further investigations of the regulating mechanisms of non-coding RNAs in peripheral nerve repair and regeneration.
Collapse
Affiliation(s)
- Tianmei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province People’s Republic of China 226001
| | - Chunlin Fan
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu China
| | - Qianyan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province People’s Republic of China 226001
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province People’s Republic of China 226001
| |
Collapse
|
26
|
Li W, Jiang LJ, Zhou XJ, Lu XZ, Liu LF, Wang S. Association of the invasiveness of colon cancer with the expression of C/EBPα. Oncol Lett 2018; 17:1974-1979. [PMID: 30675263 DOI: 10.3892/ol.2018.9755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/05/2017] [Indexed: 12/22/2022] Open
Abstract
The present study aimed to investigate the association of the invasiveness of colon cancer (CC) with the expression of CCAAT/enhancer binding protein α (C/EBPα). Immunohistochemistry was performed to determine the expression of C/EBPα in the cancer and adjacent tissue samples from 48 patients with CC. A pCDGFP-C/EBPα eukaryotic expression vector was constructed, and a wound-healing assay was performed to observe the effect of transfection on the migration of SW480 cells. In addition, the expression levels of tumor invasion-associated proteins, including Kruppel-like factor 5 (KLF5), matrix metallopeptidase (MMP)-2, MMP-9, and E-cadherin (ECD) were detected subsequent to transfection. Immunohistochemistry analysis demonstrated that the rate of low C/EBPα expression in normal tissue was 6.25%, whereas the rate in CC tissues was 68.75%; this difference was statistically significant (P<0.05). The patients with lower C/EBPα expression exhibited statistically larger tumor diameters, more advanced tumor-node-metastasis (TMN) stages and a greater likelihood of lymph node metastasis. The overexpression of C/EBPα significantly reduced the mobility of SW480 cells, and the expression of KLF5, MMP-2 and MMP-9 was reduced, whereas the expression of ECD was increased. In conclusion, C/EBPα was downregulated in CC tissue samples, and associated with the TMN stage and metastasis of CC; in addition, the overexpression of C/EBPα significantly reduced the invasiveness of CC cells. This may be significant for the diagnosis and treatment of CC in the future.
Collapse
Affiliation(s)
- Wei Li
- Department of General Surgery, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421002, P.R. China
| | - Liang-Jun Jiang
- Department of Gastroenterology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421002, P.R. China
| | - Xiao-Jun Zhou
- Department of General Surgery, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421002, P.R. China
| | - Xian-Zhou Lu
- Department of General Surgery, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421002, P.R. China
| | - Long-Fei Liu
- Department of General Surgery, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421002, P.R. China
| | - Song Wang
- Department of General Surgery, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421002, P.R. China
| |
Collapse
|
27
|
Jia Z, Peng J, Yang Z, Chen J, Liu L, Luo D, He P. Long non-coding RNA TP73‑AS1 promotes colorectal cancer proliferation by acting as a ceRNA for miR‑103 to regulate PTEN expression. Gene 2018; 685:222-229. [PMID: 30472379 DOI: 10.1016/j.gene.2018.11.072] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 01/10/2023]
Abstract
There is an increasing evidence that long non-coding RNAs (lncRNAs) play an important role in tumorigenesis and cancer progression. This study focused on the functional role of P73 antisense RNA 1T (TP73‑AS1), a lncRNA, in colorectal cancer (CRC). We found that TP73‑AS1 expression was significantly low in CRC tissues and cells, and high TP73‑AS1 expression was negatively associated with TNM stage, prognosis, overall survival, and disease-free survival in the CRC patients. Moreover, TP73‑AS1 overexpression dramatically inhibited CRC cell growth, promoted apoptosis, downregulated Bcl‑2 levels, and increased caspase‑3 expression. Furthermore, TP73‑AS1 expression levels were positively associated with PTEN levels in clinical CRC samples. As expected, TP73‑AS1 could upregulate PTEN expression in CRC cells. Mechanistically, PTEN was shown to be the target of miR‑103. Interestingly, TP73‑AS1 overexpression could increase PTEN expression through competitive binding to miR‑103. Functionally, our data show that such TP73‑AS1-induced PTEN expression through binding to miR‑103 facilitated CRC cell proliferation. Thus, we showed that TP73‑AS1 inhibits CRC cell growth by functioning as a ceRNA (competing endogenous RNAs) to regulate PTEN levels. Our findings provide new insights into the underlying molecular mechanisms of TP73‑AS1-mediated CRC.
Collapse
Affiliation(s)
- Zeming Jia
- Department of Colorectal & Anal Surgery, Hepatobiliary & Enteric Surgery Research Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Jian Peng
- Department of Colorectal & Anal Surgery, Hepatobiliary & Enteric Surgery Research Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhi Yang
- Department of Colorectal & Anal Surgery, Hepatobiliary & Enteric Surgery Research Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jie Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ling Liu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Dongren Luo
- Department of Colorectal & Anal Surgery, Hepatobiliary & Enteric Surgery Research Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Panxiang He
- Department of Colorectal & Anal Surgery, Hepatobiliary & Enteric Surgery Research Center of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
28
|
Zhang H, Zhu M, Shan X, Zhou X, Wang T, Zhang J, Tao J, Cheng W, Chen G, Li J, Liu P, Wang Q, Zhu W. A panel of seven-miRNA signature in plasma as potential biomarker for colorectal cancer diagnosis. Gene 2018; 687:246-254. [PMID: 30458288 DOI: 10.1016/j.gene.2018.11.055] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/02/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022]
Abstract
Colorectal cancer (CRC) has been one of the most commonly diagnosed cancers in global. The differential expression profiles of microRNAs (miRNAs) in CRC plasma of patients have the potential to serve as a diagnostic biomarker. We conducted a four-stage study to identify the potential plasma miRNAs for CRC detection. In the initial screening phase, Exiqon panel (miRCURY-Ready-to-Use-PCR-Human-panel-I + II-V1.M) including 3 CRC pools and 1 normal controls (NCs) pool were applied to acquire miRNA profiles. In the training stage (30 CRC VS. 30 NCs) and testing stage (79 CRC VS. 76 NCs), quantitative real-time polymerase chain reaction (qRT-PCR) was utilized to conduct candidate miRNA profiles. Then the identified miRNAs were verified in external validation stage (30 CRC VS. 26 NCs). Expression levels of identified miRNAs were assessed in tissue samples (24 pairs) and plasma exosomes (18 CRC VS. 18 NCs). Receiver operating characteristic (ROC) curves were constructed to evaluate the diagnostic accuracy. Seven miRNAs (miR-103a-3p, miR-127-3p, miR-151a-5p, miR-17-5p, miR-181a-5p, miR-18a-5p and miR-18b-5p) were significantly overexpressed in CRC compared with NCs. Area under the ROC curve of the seven-miRNA signature was 0.762, 0.824 and 0.895 for the training, testing and the external validation stages, respectively. Additionally, miR-103a-3p, miR-127-3p, miR-17-5p and miR-18a-5p were discovered significantly up-regulated in CRC tissues; while miR-17-5p, miR-181a-5p, miR-18a-5p and miR-18b-5p were significantly elevated in CRC plasma exosomes. In conclusion, we established a seven-miRNA signature in the peripheral plasma for CRC detection.
Collapse
Affiliation(s)
- Huo Zhang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Mingxia Zhu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China; Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Xia Shan
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China; Department of Respiration, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Xin Zhou
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Tongshan Wang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jinying Zhang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jinsong Tao
- Department of Cardiology, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin 214400, PR China
| | - Wenfang Cheng
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Gang Chen
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China; Department of Oncology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 210000, PR China
| | - Jian Li
- Department of Cardiology, The Affiliated Jiangyin Hospital of Southeast University, Jiangyin 214400, PR China
| | - Ping Liu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China.
| | - Qiang Wang
- Department of General Surgery, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215000, PR China.
| | - Wei Zhu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China; Department of Oncology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215000, PR China.
| |
Collapse
|
29
|
Wang X, Lin Y, Peng L, Sun R, Gong X, Du J, Zhang X. MicroRNA-103 Promotes Proliferation and Inhibits Apoptosis in Spinal Osteosarcoma Cells by Targeting p57. Oncol Res 2018; 26:933-940. [PMID: 29295723 PMCID: PMC7844680 DOI: 10.3727/096504017x15144741233346] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma is one of the most aggressive malignancies with poor prognosis rates. Many studies have demonstrated that miRNAs were involved in osteosarcoma, but the role of miR-103a in osteosarcoma remains elusive. In this study, we detected the expression levels of miR-103 in osteosarcoma and non-osteosarcoma tissues and cell lines. The binding effect of miR-103 on p57 was detected by luciferase reporter assay. After altering expressions of miR-103 or p57, viability, migration, invasion, and apoptosis of MG63 cells and expressions of proteins related with the JNK/STAT and mTOR pathways were all detected. We found the higher expression of miR-103 in osteosarcoma tissues and cell lines compared with non-osteosarcoma tissues and cell lines. miR-103 overexpression promoted survival, migration, and invasion of MG63 cells. Knockdown of miR-103a inhibited cell survival, migration, and invasion by upregulating the expression of p57, which was a target of miR-103. Moreover, miR-103a overexpression activated the JNK/STAT and mTOR pathways probably through inhibiting p57 expression. In conclusion, miR-103a acted as an oncogene in osteosarcoma, probably through activating the JNK/STAT and mTOR pathways by inhibiting p57 expression.
Collapse
Affiliation(s)
- Xuesong Wang
- *Department of Spinal, Qingdao Central Hospital, Qingdao, P.R. China
| | - Yong Lin
- †Department of Spinal, Qingdao Municipal Hospital, Qingdao, P.R. China
| | - Lei Peng
- ‡Library of Qingdao Central Hospital, Qingdao, P.R. China
| | - Ruifu Sun
- *Department of Spinal, Qingdao Central Hospital, Qingdao, P.R. China
| | - Xiaojin Gong
- *Department of Spinal, Qingdao Central Hospital, Qingdao, P.R. China
| | - Jinlong Du
- *Department of Spinal, Qingdao Central Hospital, Qingdao, P.R. China
| | - Xiugong Zhang
- *Department of Spinal, Qingdao Central Hospital, Qingdao, P.R. China
| |
Collapse
|
30
|
Hu X, Miao J, Zhang M, Wang X, Wang Z, Han J, Tong D, Huang C. miRNA-103a-3p Promotes Human Gastric Cancer Cell Proliferation by Targeting and Suppressing ATF7 in vitro. Mol Cells 2018; 41:390-400. [PMID: 29754469 PMCID: PMC5974616 DOI: 10.14348/molcells.2018.2078] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 02/28/2018] [Accepted: 03/15/2018] [Indexed: 12/11/2022] Open
Abstract
Studies have revealed that miR-103a-3p contributes to tumor growth in several human cancers, and high miR-103a-3p expression is associated with poor prognosis in advanced gastric cancer (GC) patients. Moreover, bioinformatics analysis has shown that miR-103a-3p is upregulated in The Cancer Genome Atlas (TCGA) stomach cancer cohort. These results suggest that miR-103a-3p may function as an oncogene in GC. The present study aimed to investigate the role of miR-103a-3p in human GC. miR-103a-3p expression levels were increased in 33 clinical GC specimens compared with adjacent nontumor stomach tissues. Gain- and loss-of-function studies were performed to identify the correlation between miR-103a-3p and tumorigenesis in human GC. Inhibiting miR-103a-3p suppressed GC cell proliferation and blocked the S-G2/M transition in MKN-45/SGC-7901 cells, whereas miR-103a-3p overexpression improved GC cell proliferation and promoted the S-G2/M transition in vitro. Bioinformatics and dual-luciferase reporter assays confirmed that ATF7 is a direct target of miR-103a-3p. Analysis of the TCGA stomach cancer cohort further revealed that miR-103a-3p expression was inversely correlated with ATF7 expression. Notably, silencing ATF7 showed similar cellular and molecular effects as miR-103a-3p overexpression, namely, increased GC cell proliferation, improved CDK2 expression and decreased P27 expression. ATF7 overexpression eliminated the effects of miR-103a-3p expression. These findings indicate that miR-103a-3p promotes the proliferation of GC cell by targeting and suppressing ATF7 in vitro.
Collapse
Affiliation(s)
- Xiaoyi Hu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
- Department of Oral Maxillofacial Surgery, Stomatological Hospital, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
| | - Jiyu Miao
- Key Laboratory of Environment and Genes Related to Diseases, College of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
| | - Min Zhang
- College of Life Science, Yanan University, Yan’an, Shaanxi,
China
| | - Xiaofei Wang
- Key Laboratory of Environment and Genes Related to Diseases, College of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
| | - Zhenzhen Wang
- Key Laboratory of Environment and Genes Related to Diseases, College of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
| | - Jia Han
- Key Laboratory of Environment and Genes Related to Diseases, College of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
| | - Dongdong Tong
- Key Laboratory of Environment and Genes Related to Diseases, College of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
| | - Chen Huang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
- Key Laboratory of Environment and Genes Related to Diseases, College of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi,
China
| |
Collapse
|
31
|
Fasihi A, M. Soltani B, Atashi A, Nasiri S. Introduction of
hsa‐miR‐103a
and
hsa‐miR‐1827
and
hsa‐miR‐137
as new regulators of Wnt signaling pathway and their relation to colorectal carcinoma. J Cell Biochem 2018; 119:5104-5117. [DOI: 10.1002/jcb.26357] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/15/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Ali Fasihi
- Molecular Genetics DepartmentFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Bahram M. Soltani
- Molecular Genetics DepartmentFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Amir Atashi
- Hematology DepartmentFaculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Shirzad Nasiri
- Tehran University of Medical SciencesShariati HospitalTehranIran
| |
Collapse
|
32
|
Wilczyński M, Żytko E, Danielska J, Szymańska B, Dzieniecka M, Nowak M, Malinowski J, Owczarek D, Wilczyński JR. Clinical significance of miRNA-21, -103, -129, -150 in serous ovarian cancer. Arch Gynecol Obstet 2018; 297:741-748. [DOI: 10.1007/s00404-018-4660-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 01/09/2018] [Indexed: 02/07/2023]
|
33
|
Nogueira I, Dias F, Teixeira AL, Medeiros R. miRNAs as potential regulators of mTOR pathway in renal cell carcinoma. Pharmacogenomics 2018; 19:249-261. [PMID: 29334302 DOI: 10.2217/pgs-2017-0160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is the most commonly occurring solid cancer of the adult kidney with the majority of RCC cases being detected accidentally. The most aggressive subtype is clear cell RCC (ccRCC). miRNAs, a family of small noncoding RNAs regulating gene expression have been identified as key biological modulators. The von Hippel-Lindau pathway is one of the signaling pathways involved in the pathophysiology of ccRCC. Another oncogenic mechanism involves the activation of PI3K/AKT/mTOR signaling and serves as a central regulator of cell metabolism, proliferation and survival. Several studies have described the involvement of miRNA dysregulation in the pathogenesis and progression of ccRCC. These molecules can be considered as potential diagnostic and prognostic biomarkers, allowing response to therapy to be monitored.
Collapse
Affiliation(s)
- Inês Nogueira
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,ICBAS, Abel Salazar Institute for the Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.,Research Department, LPCC-Portuguese League, Against Cancer (NRNorte), 4200-172 Porto, Portugal.,CEBIMED, Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
| |
Collapse
|
34
|
Targeting PTEN in Colorectal Cancers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1110:55-73. [DOI: 10.1007/978-3-030-02771-1_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
35
|
MicroRNA-103 suppresses glioma cell proliferation and invasion by targeting the brain-derived neurotrophic factor. Mol Med Rep 2017; 17:4083-4089. [PMID: 29257320 DOI: 10.3892/mmr.2017.8282] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/06/2017] [Indexed: 11/05/2022] Open
Abstract
Glioma is the most common and aggressive of malignant brain tumours. MicroRNAs (miRNAs/miRs) are involved in tumour development of various human cancers, including glioma. Therefore, miRNAs may have potential tumour diagnostic, prognostic and therapeutic values in human glioma. miR‑103 is abnormally expressed in various human cancer types. However, the detailed expression pattern, biological functions and underlying molecular mechanism of miR‑103 in glioma remain unclear. Therefore, the present study aimed to investigate the expression, biological roles and underlying mechanisms of miR‑103 in glioma. Results of the present study demonstrated that miR‑103 was significantly down‑regulated in glioma tissues and cell lines. Functional experiments demonstrated that miR‑103 overexpression inhibited the proliferation and invasion of glioma cells in vitro. Additionally, brain‑derived neurotrophic factor (BDNF) was identified as a direct functional target of miR‑103 in glioma. Furthermore, mRNA and protein expression levels of BDNF were highly upregulated in glioma tissues compared with normal brain tissues. Spearman's correlation analysis indicated a negative association between miR‑103 and BDNF mRNA expression levels in glioma tissues. Furthermore, rescue experiments demonstrated that BDNF up‑regulation reversed the suppressive effects of miR‑103 on glioma cell proliferation and invasion. Therefore, the authors of the present study hypothesized that the interaction between miR‑103 and BDNF serves a role in glioma progression and, in the future, may serve as a therapeutic target for glioma treatment.
Collapse
|
36
|
Kent MS, Zwingenberger A, Westropp JL, Barrett LE, Durbin-Johnson BP, Ghosh P, Vinall RL. MicroRNA profiling of dogs with transitional cell carcinoma of the bladder using blood and urine samples. BMC Vet Res 2017; 13:339. [PMID: 29141625 PMCID: PMC5688639 DOI: 10.1186/s12917-017-1259-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 11/07/2017] [Indexed: 12/16/2022] Open
Abstract
Background Early signs of canine transitional cell carcinoma (TCC) are frequently assumed to be caused by other lower urinary tract diseases (LUTD) such as urinary tract infections, resulting in late diagnosis of TCC which could be fatal. The development of a non-invasive clinical test for TCC could dramatically reduce mortality. To determine whether microRNAs (miRNAs) can be used as non-invasive diagnostic biomarkers, we assessed miRNA expression in blood and/or urine from dogs with clinically normal bladders (n = 28), LUTD (n = 25), and TCC (n = 17). Expression levels of 5 miRNA associated with TCC pathophysiology (miR-34a, let-7c, miR-16, miR-103b, and miR-106b) were assessed by quantitative real-time PCR. Results Statistical analyses using ranked ANOVA identified significant differences in miR-103b and miR-16 levels between urine samples from LUTD and TCC patients (miR-103b, p = 0.002; and miR-16, p = 0.016). No statistically significant differences in miRNA levels were observed between blood samples from LUTD versus TCC patients. Expression levels of miR-34a trended with miR-16, let-7c, and miR-103b levels in individual normal urine samples, however, this coordination was completely lost in TCC urine samples. In contrast, co-ordination of miR-34a, miR-16, let-7c, and miR-103b expression levels was maintained in blood samples from TCC patients. Conclusions Our combined data indicate a potential role for miR-103b and miR-16 as diagnostic urine biomarkers for TCC, and that further investigation of miR-103b and miR-16 in the dysregulation of coordinated miRNA expression in bladder carcinogenesis is warranted. Electronic supplementary material The online version of this article (10.1186/s12917-017-1259-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michael S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Allison Zwingenberger
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Jodi L Westropp
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Laura E Barrett
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Blythe P Durbin-Johnson
- Department of Public Health Sciences, University of California Davis, Davis, California, 95616, USA
| | - Paramita Ghosh
- Department of Urology, University of California, Davis, School of Medicine, Sacramento, CA, USA. .,Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, CA, USA. .,VA Northern California Health Care System, Sacramento, CA, USA.
| | - Ruth L Vinall
- Department of Urology, University of California, Davis, School of Medicine, Sacramento, CA, USA. .,Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, CA, USA. .,Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA, USA.
| |
Collapse
|
37
|
Xiong J, Wang D, Wei A, Lu H, Tan C, Li A, Tang J, Wang Y, He S, Liu X, Hu W. Deregulated expression of miR-107 inhibits metastasis of PDAC through inhibition PI3K/Akt signaling via caveolin-1 and PTEN. Exp Cell Res 2017; 361:316-323. [PMID: 29111166 DOI: 10.1016/j.yexcr.2017.10.033] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/23/2017] [Accepted: 10/26/2017] [Indexed: 02/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) displays a highly aggressive malignancy and is considered to be an incurable and rapidly lethal disease. MicroRNAs (miRNAs) are small non-coding RNAs of approximately nucleotides that regulate several aspects of tumors pathogenesis, including migration, invasion, metastasis and epithelial-mesenchymal transition. We have found that miR-107 was significantly high expression in PDAC tissues and cells. High miR-107 expression is associated with poor clinicopathological parameters and prognosis in PDAC patients. Deregulated expression of miR-107 in PDAC cells (AsPC-1 and Panc-1) is sufficient to reduce cell migration and invasion, and to induce upregulation of epithelial markers (β-catenin, ZO-1 and E-cadherin) and a decrease of mesenchymal marker expression (ZEB-1 and vimentin). We also found that the caveolin-1, PTEN and p-Akt expression are modulated by miR-107 in PDAC cells. Moreover, our study clearly demonstrated that deregulated expression of miR-107 inhibited cell migration and invasion and EMT by up-regulation of caveolin-1 and PTEN, and inhibition of PI3K/Akt signaling in PDAC cells. Our study suggested that miR‑107 expression might both be a useful indicator of the metastatic potential and provided a new potential therapeutic target in PDAC.
Collapse
Affiliation(s)
- Junjie Xiong
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ailin Wei
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu 610041, China; Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huimin Lu
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Chunlu Tan
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ang Li
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jie Tang
- State Key Laboratory of biotherapy, Sichuan University, Chengdu, China
| | - Yichao Wang
- Department of Thyroid Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Sirong He
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xubao Liu
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Weiming Hu
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
38
|
Berania I, Cardin GB, Clément I, Guertin L, Ayad T, Bissada E, Nguyen-Tan PF, Filion E, Guilmette J, Gologan O, Soulieres D, Rodier F, Wong P, Christopoulos A. Four PTEN-targeting co-expressed miRNAs and ACTN4- targeting miR-548b are independent prognostic biomarkers in human squamous cell carcinoma of the oral tongue. Int J Cancer 2017; 141:2318-2328. [PMID: 28779483 DOI: 10.1002/ijc.30915] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/31/2017] [Accepted: 07/10/2017] [Indexed: 02/04/2023]
Abstract
The purpose of this study was to determine the prognostic value and oncogenic pathways associated to miRNA expression in squamous cell carcinoma of the oral tongue and to link these miRNA candidates with potential gene targets. We performed a miRNA screening within our institutional cohort (n = 58 patients) and reported five prognostic targets including a cluster of four co-expressed miRNAs (miR-18a, miR-92a, miR-103, and miR-205). Multivariate analysis showed that expression of miR-548b (p = 0.007) and miR-18a (p = 0.004, representative of co-expressed miRNAs) are independent prognostic markers for squamous cell carcinoma of the oral tongue. These findings were validated in The Cancer Genome Atlas (TCGA) cohort (n = 131) for both miRNAs (miR-548b: p = 0.027; miR-18a: p = 0.001). Bioinformatics analysis identified PTEN and ACTN4 as direct targets of the four co-expressed miRNAs and miR-548b, respectively. Correlations between the five identified miRNAs and their respective targeted genes were validated in the two merged cohorts and were concordantly significant (miR-18a/PTEN: p < 0.0001; miR-92a/PTEN: p = 0.0008; miR-103/PTEN: p = 0.008; miR-203/PTEN: p = 0.019; miR-548b/ACTN4: p = 0.009).
Collapse
Affiliation(s)
- Ilyes Berania
- CRCHUM and Institut du cancer de Montréal, Montreal, QC, Canada.,Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | | | | | - Louis Guertin
- Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Tareck Ayad
- Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Eric Bissada
- Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Phuc Felix Nguyen-Tan
- Department of Radiation Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Edith Filion
- Department of Radiation Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Julie Guilmette
- Department of Pathology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Department of Pathology, Massachusetts General Hospital/Massachusetts Eye and Ear Infirmary, Boston, MA
| | - Olguta Gologan
- Department of Pathology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Denis Soulieres
- Department of Medicine, Service of Hemato-Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Francis Rodier
- CRCHUM and Institut du cancer de Montréal, Montreal, QC, Canada.,Département de radiologie, radio-oncologie et medicine nucléaire, Université de Montréal, Montreal, QC, Canada
| | - Philip Wong
- CRCHUM and Institut du cancer de Montréal, Montreal, QC, Canada.,Department of Radiation Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Apostolos Christopoulos
- CRCHUM and Institut du cancer de Montréal, Montreal, QC, Canada.,Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
39
|
Yang D, Wang JJ, Li JS, Xu QY. miR-103 Functions as a Tumor Suppressor by Directly Targeting Programmed Cell Death 10 in NSCLC. Oncol Res 2017; 26:519-528. [PMID: 28734041 PMCID: PMC7844823 DOI: 10.3727/096504017x15000757094686] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancer cases. Absence of miR-103 has recently been identified to be associated with metastatic capacity of primary lung tumors. However, the exact role of miR-103 in NSCLC and the molecular mechanism are unclear. In the present study, we showed that miR-103 expression was reduced in NSCLC tissues and cells. miR-103 expression was negatively correlated with tumor size and stage. The overall survival was longer in patients with higher miR-103 level than in those with lower miR-103 expression. miR-103 inhibited cell proliferation in A549 cells, decreased tumor weight and volume, and prolonged survival of tumor-implanted nude mice. miR-103 increased apoptotic cell death in A549 cells. Furthermore, miR-103 decreased the invasion and migration abilities in A549 cells, as evidenced by Transwell and wound healing results. Downregulation of miR-103 significantly reduced the level of programmed cell death 10 (PDCD10). We found a significant decrease in the relative luciferase activity of the reporter gene in A549 cells cotransfected with the miR-103 mimic and pGL3-PDCD10 WT 3′-UTR, but not pGL3-PDCD10 mut 3′-UTR. We showed that overexpression of PDCD10 significantly inhibited miR-103-induced inhibition of cell proliferation, increased apoptosis, and decreased invasion and migration in A549 cells. Moreover, we found that PDCD10 expression was increased in NSCLC tissues and cells. PDCD10 expression was positively correlated with tumor size and stage. Overexpression of PDCD10 increased cell proliferation and inhibited apoptosis in A549 cells. The data demonstrated that dysregulation of the miR-103/PDCD10 signal may be a novel therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Dong Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Jian-Jun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Jin-Song Li
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Qian-Yu Xu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| |
Collapse
|
40
|
Colorectal Cancer: From the Genetic Model to Posttranscriptional Regulation by Noncoding RNAs. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7354260. [PMID: 28573140 PMCID: PMC5442347 DOI: 10.1155/2017/7354260] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/16/2017] [Indexed: 12/11/2022]
Abstract
Colorectal cancer is the third most common form of cancer in developed countries and, despite the improvements achieved in its treatment options, remains as one of the main causes of cancer-related death. In this review, we first focus on colorectal carcinogenesis and on the genetic and epigenetic alterations involved. In addition, noncoding RNAs have been shown to be important regulators of gene expression. We present a general overview of what is known about these molecules and their role and dysregulation in cancer, with a special focus on the biogenesis, characteristics, and function of microRNAs. These molecules are important regulators of carcinogenesis, progression, invasion, angiogenesis, and metastases in cancer, including colorectal cancer. For this reason, miRNAs can be used as potential biomarkers for diagnosis, prognosis, and efficacy of chemotherapeutic treatments, or even as therapeutic agents, or as targets by themselves. Thus, this review highlights the importance of miRNAs in the development, progression, diagnosis, and therapy of colorectal cancer and summarizes current therapeutic approaches for the treatment of colorectal cancer.
Collapse
|
41
|
miR-103 Promotes Proliferation and Metastasis by Targeting KLF4 in Gastric Cancer. Int J Mol Sci 2017; 18:ijms18050910. [PMID: 28445396 PMCID: PMC5454823 DOI: 10.3390/ijms18050910] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 04/19/2017] [Accepted: 04/23/2017] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) play important roles in the cancer development and progression; overexpression of miR-103 has been identified in various tumors. However, its biological function and regulatory mechanism involved in modulation of human gastric cancer (GC) remain largely unknown. This study aimed to confirm clinical significance of miR-103 and investigate its biological role and underlying mechanism in GC. Real-time quantitative PCR (qRT-PCR) revealed miR-103 was highly expressed in GC tissues and cell lines. miR-103 expression was correlated closely with tumor size, Lauren’s classification, and lymph node metastasis. Importantly, Kaplan-Meier analysis revealed that high expression of miR-103 was significantly associated with poor overall survival and disease-free survival of GC patients. Downregulation of miR-103 by transfecting with miR-103 inhibitor significantly suppressed cell proliferation, induced apoptosis, inhibited migration and invasion in vitro and in vivo. Furthermore, miRNA target databases and luciferase reporter assay confirmed that Krüppel-like Factor-4 (KLF4) was a direct target of miR-103 in GC, and there was a significant inverse correlation between miR-103 and KLF4 expression in GC tissues. Moreover, KLF4 downregulation could rescue miR-103’s oncogenic effect on GC cell proliferation, apoptosis, migration, and invasion. Therefore, these results suggested that miR-103 overexpression could contribute to tumor progression by suppressing KLF4, and it might serve as a promising candidate for the prognosis of GC patients.
Collapse
|
42
|
Kim SW. [The Role of MicroRNAs in Colorectal Cancer]. THE KOREAN JOURNAL OF GASTROENTEROLOGY = TAEHAN SOHWAGI HAKHOE CHI 2017; 69:206-211. [PMID: 28449421 DOI: 10.4166/kjg.2017.69.4.206] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer related deaths in the world. Many oncogenes and tumor suppressor genes are involved in the development of CRC. MicroRNAs (miRNAs) are a class of small, non-coding, endogenous RNAs in animals and plants. Recent studies have shown that miRNAs are associated with the mediation process of tumorigenesis, including inflammation, cell cycle, stress response, differentiation, apoptosis, migration, and invasion in cancer. These miRNAs have been linked to the development of CRC and recently studied as new potential biomarkers in the diagnosis and treatment for CRC. Specific miRNAs expression patterns help distinguish CRC from other colon-related diseases, and miRNAs can target the oncogenes and regulatory molecular pathways. Recent studies have demonstrated the restoration of tumor suppressive miRNAs and inhibition of oncogenic miRNAs for CRC treatment. Herein, we describe the diagnostic and therapeutic roles of miRNAs in CRC.
Collapse
Affiliation(s)
- Sang Wook Kim
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School, Jeonju, Korea
| |
Collapse
|
43
|
Xiong B, Lei X, Zhang L, Fu J. miR-103 regulates triple negative breast cancer cells migration and invasion through targeting olfactomedin 4. Biomed Pharmacother 2017; 89:1401-1408. [PMID: 28320108 DOI: 10.1016/j.biopha.2017.02.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 02/01/2017] [Accepted: 02/09/2017] [Indexed: 01/15/2023] Open
Abstract
Our previous study showed olfactomedin 4 (OLFM4) suppressed triple-negative breast cancer cells migration, invasion and metastasis-associated protein MMP 9 expression. OLFM4 was identified as a potential target of miR-103 according to microRNA target databases and published studies. The aim of this study is to validate the relationship between miR-103 and OLFM4, and explore the function and clinical significance of miR-103 in triple-negative breast cancer patients. In our results, miR-103 negatively regulated OLFM4 expression by directly targeting its 3'-UTR. OLFM4 was a functional target of miR-103 to regulate triple-negative breast cancer cells migration, invasion and MMP 9 expression. Moreover, miR-103 overexpression was observed in triple-negative breast cancer tissues and cell lines, and associated with lymph node metastasis, distant metastasis and clinical stage. Univariate and multivariate analyses suggested that miR-103 overexpression was a poor independent prognostic factor for triple-negative breast cancer patients. In conclusion, miR-103 acts as an oncogene miRNA to promote triple-negative breast cancer cells migration and invasion through targeting OLFM4.
Collapse
Affiliation(s)
- Bin Xiong
- Surgery Teaching and Research Section, Clinical Medical School, Jining Medical University, NO. 16 Hehua Road, Jining, Shandong 272067, China
| | - Xuefeng Lei
- Surgery Teaching and Research Section, Clinical Medical School, Jining Medical University, NO. 16 Hehua Road, Jining, Shandong 272067, China
| | - Lei Zhang
- Surgery Teaching and Research Section, Clinical Medical School, Jining Medical University, NO. 16 Hehua Road, Jining, Shandong 272067, China
| | - Jia Fu
- Academy of Basic Medicine, Jining Medical University, NO. 16 Hehua Road, Jining, Shandong 272067, China.
| |
Collapse
|
44
|
Kfir-Erenfeld S, Haggiag N, Biton M, Stepensky P, Assayag-Asherie N, Yefenof E. miR-103 inhibits proliferation and sensitizes hemopoietic tumor cells for glucocorticoid-induced apoptosis. Oncotarget 2017; 8:472-489. [PMID: 27888798 PMCID: PMC5352135 DOI: 10.18632/oncotarget.13447] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/12/2016] [Indexed: 11/25/2022] Open
Abstract
Glucocorticoid (GC) hormones are an important ingredient of leukemia therapy since they are potent inducers of lymphoid cell apoptosis. However, the development of GC resistance remains an obstacle in GC-based treatment. In the present investigation we found that miR-103 is upregulated in GC-sensitive leukemia cells treated by the hormone. Transfection of GC resistant cells with miR-103 sensitized them to GC induced apoptosis (GCIA), while miR-103 sponging of GC sensitive cells rendered them partially resistant. miR-103 reduced the expression of cyclin dependent kinase (CDK2) and its cyclin E1 target, thereby leading to inhibition of cellular proliferation. miR-103 is encoded within the fifth intron of PANK3 gene. We demonstrate that the GC receptor (GR) upregulates miR-103 by direct interaction with GC response element (GRE) in the PANK3 enhancer. Consequently, miR-103 targets the c-Myc activators c-Myb and DVL1, thereby reducing c-Myc expression. Since c-Myc is a transcription factor of the miR-17~92a poly-cistron, all six miRNAs of the latter are also downregulated. Of these, miR-18a and miR-20a are involved in GCIA, as they target GR and BIM, respectively. Consequently, GR and BIM expression are elevated, thus advancing GCIA. Altogether, this study highlights miR-103 as a useful prognostic biomarker and drug for leukemia management in the future.
Collapse
Affiliation(s)
- Shlomit Kfir-Erenfeld
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Noa Haggiag
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Moshe Biton
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Polina Stepensky
- Pediatric Hemato-Oncology and Bone Marrow Transplantation Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nathalie Assayag-Asherie
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eitan Yefenof
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
45
|
Abstract
MicroRNAs (miRNAs) are a class of endogenous, evolutionarily conserved small non-coding RNAs, which play a vital role in tumour formation, development, metastasis and recurrence by inducing DNA methylation, changing tumor microenvironment and regulating signal pathways such as Wnt/β-catenin, phosphoinositide3-kinase (PI3K), K-RAS, epithelial mesenchymal transitions (EMT) and so on. Recent studies have found that the expression of many miRNAs is dyregulated in colorectal cancer, and they participate in and control the formation and development of colorectal cancer. Thus, understanding the roles and mechanisms of action of miRNAs in colorectal cancer can provide a new avenue for its early diagnosis, clinical staging, treatment and prognosis evaluation.
Collapse
|
46
|
Ma ZL, Zhang BJ, Wang DT, Li X, Wei JL, Zhao BT, Jin Y, Li YL, Jin YX. Tanshinones suppress AURKA through up-regulation of miR-32 expression in non-small cell lung cancer. Oncotarget 2016; 6:20111-20. [PMID: 26036635 PMCID: PMC4652991 DOI: 10.18632/oncotarget.3933] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/04/2015] [Indexed: 01/09/2023] Open
Abstract
Tanshinone is the liposoluble constituent of Salia miltiorrhiza, a root used in traditional herbal medicine which is known to possess certain health benefits. Although it is known that tanshinones, including tanshinone I (T1), tanshinone IIA (T2A), and cryptotanshinone (CT), can inhibit the growth of lung cancer cells in vitro, the mechanism under which they act is still unclear. AURKA, an oncogene, encodes a serine-threonine kinase which regulates mitotic processes in mammalian cells. Here, we reported that tanshinones mediate AURKA suppression partly through up-regulating the expression of miR-32. We found that tanshinones could inhibit cell proliferation, promote apoptosis, and impede cell-cycle progression, thus performing an antineoplastic function in non-small cell lung cancer (NSCLC). Additionally, we demonstrated that tanshinones attained these effects in part by down-regulating AURKA, corroborating previous reports. Our results showed that in NSCLC, similar effects were obtained with knock-down of the AURKA gene by siRNA. We also verified that AURKA was the direct target of miR-32. Collectively, our results demonstrated that tanshinones could inhibit NSCLC by suppressing AURKA via up-regulating the expressions of miR-32 and other related miRNAs.
Collapse
Affiliation(s)
- Zhong-Liang Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Bing-Jie Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - D Tao Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xue Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jia-Li Wei
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Bo-Tao Zhao
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yan Jin
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yan-Li Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - You-Xin Jin
- School of Life Sciences, Shanghai University, Shanghai 200444, China.,State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
47
|
Zheng YB, Xiao K, Xiao GC, Tong SL, Ding Y, Wang QS, Li SB, Hao ZN. MicroRNA-103 promotes tumor growth and metastasis in colorectal cancer by directly targeting LATS2. Oncol Lett 2016; 12:2194-2200. [PMID: 27602163 DOI: 10.3892/ol.2016.4814] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/18/2016] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) has become the third most common cancer worldwide and leads to a high mortality rate. Although colorectal cancer has been studied widely, the underlying molecular mechanism remains unclear. Increasing evidence shows that the abnormal expression of microRNAs (miRNAs) is involved in tumorigenesis. Previous studies have reported that miRNA-103 (miR-103) is dysregulated in CRC; however, the expression, function and mechanism of miR-103 in CRC are not well known. The present study showed that miR-103 was overexpressed in the primary tumor tissues of patients with CRC and was significantly associated with a more aggressive phenotype of CRC in patients. Survival rate analysis demonstrated that CRC patients with high miR-103 expression had a poorer overall survival compared with CRC patients with low miR-103 expression. In CRC cell lines, miR-103 inhibition significantly decreased the proliferation, invasion and migration of the cells in vitro. Furthermore, miR-103 repressed large tumor suppressor kinase 2 (LATS2) expression by directly binding to the LATS2-3'-untranslated region, and an inverse correlation was identified between the expression of miR-103 and LATS2 messenger RNA in primary CRC tissues. In addition, the restoration of LATS2 led to suppressed proliferation, invasion and migration of CRC cells. In vivo, miR-103 promotes tumor growth in nude mice. In summary, miR-103 performs a critical role in the promotion of the invasive and metastatic capacities of CRC, possibly by directly targeting LATS2. This miRNA may be involved in the development and progression of CRC.
Collapse
Affiliation(s)
- Yong-Bin Zheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Kuang Xiao
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Gao-Chun Xiao
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shi-Lun Tong
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yu Ding
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qiu-Shuang Wang
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sheng-Bo Li
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhi-Nan Hao
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
48
|
Fu X, Zhang W, Su Y, Lu L, Wang D, Wang H. MicroRNA-103 suppresses tumor cell proliferation by targeting PDCD10 in prostate cancer. Prostate 2016; 76:543-51. [PMID: 26771762 DOI: 10.1002/pros.23143] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/15/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND It is known that microRNAs (miRNAs) are a class of small, non-coding RNAs that act as key regulators in various physiological and pathological processes. However, the regulatory mechanisms involving miRNAs in prostate cancer remain largely unknown. Here, we found that miR-103 is down-regulated in prostate cancer and closely associated with tumor proliferation and migration. Our objective was to explore the role of the miR-103 in prostate cancer. METHODS In this study, we measured miR-103 level using real-time polymerase chain reaction in the human prostate cancer cell lines, including PC-3, LNCap, 22Rv1, DU145, and the normal prostate epithelium cell line RWPE-1, a total of 25 pairs of primary prostate cancer tissues and adjacent non-cancerous tissues (NCTs) were measured also. In addition, over-expression of miR-103 in prostate cancer cell lines to determine the role of miR-103 in prostate cancer. RESULTS We found that miR-103 is down-regulated in prostate cancer and closely associated with tumor proliferation and migration. In addition, over-expression of miR-103 apparently inhibits prostate cancer cell proliferation and migration in vitro. Gain-of-function in vitro experiments further show that miR-103 mimics significantly inhibited prostate cancer cell proliferation, invasion and increase the cell cycle in G1 phase, while promoted cell apoptosis. Subsequent dual-luciferase reporter assay identified one of the proto-oncogene PDCD10 as direct target of miR-103. CONCLUSIONS Therefore, our data collectively demonstrate that miR-103 is a proto-oncogene miRNA that can suppress prostate cancer proliferation and migration by down-regulating the oncogene PDCD10, indicating that miR-103 may represent a new potential diagnostic and therapeutic target for prostate cancer treatment.
Collapse
Affiliation(s)
- Xiaoliang Fu
- Departmentof Urology, Xi'an Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Zhang
- Departmentof Urology, Xi'an Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yansheng Su
- The Chinese People's Liberation Army 323 hospital, Xi'an, China
| | - Lu Lu
- Armed Police Hospital of Shaanxi, Xi'an, China
| | - Dong Wang
- Departmentof Urology, Xi'an Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - He Wang
- Departmentof Urology, Xi'an Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
49
|
Guo C, Song WQ, Sun P, Jin L, Dai HY. LncRNA-GAS5 induces PTEN expression through inhibiting miR-103 in endometrial cancer cells. J Biomed Sci 2015; 22:100. [PMID: 26511107 PMCID: PMC4625733 DOI: 10.1186/s12929-015-0213-4] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/16/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Growth arrest-specific 5 (GAS5) was reported to be implicated and aberrantly express in multiple cancers. However, the expression and mechanism of action of GAS5 were largely poor understood in endometrial carcinoma. RESULTS According to the result of real-time reverse-transcriptase polymerase chain reaction (RT-PCR) and flow cytometry analysis, we identified that GAS5 was down-regulated in endometrial cancer cells and stimulated the apoptosis of endometrial cancer cells. To investigate the expression of GAS5, PTEN and miR-103, RT-PCR was performed. And we found that the expression of PTEN was up-regulated when endometrial cancer cells overexpressed GAS5. The prediction of bioinformatics online revealed that GAS5 could bind to miR-103, which was further found to be regulated by GAS5. Finally, we found that miR-103 mimic could decrease the mRNA and protein levels of PTEN through luciferase reporter assay and western blotting, and GAS5 plasmid may reverse this regulation effect in endometrial cancer cells. CONCLUSION In summary, we demonstrate that GAS5 acts as an tumor suppressor lncRNA in endometrial cancer. Through inhibiting the expression of miR-103, GAS5 significantly enhanced the expression of PTEN to promote cancer cell apoptosis, and, thus, could be an important mediator in the pathogenesis of endometrial cancer.
Collapse
Affiliation(s)
- Chen Guo
- Department of Obstetrics & Gynaecology, Affiliated Hospital of inner Mongolia University For The Nationalities, Huolinhe Avenue East NO. 1742, Tongliao, 028000, China.
| | - Wei-Qi Song
- Department of Obstetrics & Gynaecology, Affiliated Hospital of inner Mongolia University For The Nationalities, Huolinhe Avenue East NO. 1742, Tongliao, 028000, China
| | - Ping Sun
- Department of Obstetrics & Gynaecology, Affiliated Hospital of inner Mongolia University For The Nationalities, Huolinhe Avenue East NO. 1742, Tongliao, 028000, China
| | - Lian Jin
- Department of Obstetrics & Gynaecology, Affiliated Hospital of inner Mongolia University For The Nationalities, Huolinhe Avenue East NO. 1742, Tongliao, 028000, China
| | - Hong-Yan Dai
- Department of Obstetrics & Gynaecology, Affiliated Hospital of inner Mongolia University For The Nationalities, Huolinhe Avenue East NO. 1742, Tongliao, 028000, China
| |
Collapse
|
50
|
Huang YK, Yu JC. Circulating microRNAs and long non-coding RNAs in gastric cancer diagnosis: An update and review. World J Gastroenterol 2015; 21:9863-9886. [PMID: 26379393 PMCID: PMC4566381 DOI: 10.3748/wjg.v21.i34.9863] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 05/15/2015] [Accepted: 07/18/2015] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is the fourth most common cancer and the third leading cause of cancer mortality worldwide. MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are the most popular non-coding RNAs in cancer research. To date, the roles of miRNAs and lncRNAs have been extensively studied in GC, suggesting that miRNAs and lncRNAs represent a vital component of tumor biology. Furthermore, circulating miRNAs and lncRNAs are found to be dysregulated in patients with GC compared with healthy individuals. Circulating miRNAs and lncRNAs may function as promising biomarkers to improve the early detection of GC. Multiple possibilities for miRNA secretion have been elucidated, including active secretion by microvesicles, exosomes, apoptotic bodies, high-density lipoproteins and protein complexes as well as passive leakage from cells. However, the mechanism underlying lncRNA secretion and the functions of circulating miRNAs and lncRNAs have not been fully illuminated. Concurrently, to standardize results of global investigations of circulating miRNAs and lncRNAs biomarker studies, several recommendations for pre-analytic considerations are put forward. In this review, we summarize the known circulating miRNAs and lncRNAs for GC diagnosis. The possible mechanism of miRNA and lncRNA secretion as well as methodologies for identification of circulating miRNAs and lncRNAs are also discussed. The topics covered here highlight new insights into GC diagnosis and screening.
Collapse
|