1
|
Ramezani A, Rahnama M, Mahmoudian F, Shirazi F, Ganji M, Bakhshi S, Khalesi B, Hashemi ZS, Khalili S. Current Understanding of the Exosomes and Their Associated Biomolecules in the Glioblastoma Biology, Clinical Treatment, and Diagnosis. J Neuroimmune Pharmacol 2025; 20:48. [PMID: 40299204 DOI: 10.1007/s11481-025-10204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Glioblastoma is the most common and aggressive brain tumor with a low survival rate. Due to its heterogeneous composition, high invasiveness, and frequent recurrence after surgery, treatment success has been limited. In addition, due to the brain's unique immune status and the suppressor tumor microenvironment (TME), glioblastoma treatment has faced more challenges. Exosomes play a critical role in cancer metastasis by regulating cell-cell interactions that promote tumor growth, angiogenesis, metastasis, treatment resistance, and immunological regulation in the tumor microenvironment. This review explores the pivotal role of exosomes in the development of glioblastoma, with a focus on their potential as non-invasive biomarkers for prognosis, early detection and real-time monitoring of disease progression. Notably, exosome-based drug delivery methods hold promise for overcoming the blood-brain barrier (BBB) and developing targeted therapies for glioblastoma. Despite challenges in clinical translation, the potential for personalized exosome = -054321`therapies and the capacity to enhance therapeutic responses in glioblastoma, present intriguing opportunities for improving patient outcomes. It seems that getting a good and current grasp of the role of exosomes in the fight against glioblastoma would properly serve the scientific community to further their understanding of the related potentials of these biological moieties.
Collapse
Affiliation(s)
- Aghdas Ramezani
- Department of Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Rahnama
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Mahmoudian
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Shirazi
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Mahmoud Ganji
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shohreh Bakhshi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Education and Extension Organization, Razi Vaccine and Serum Research Institute, Agricultural Research, Karaj, 3197619751, Iran
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran.
| |
Collapse
|
2
|
Jiang X, Qu A, Zhang S, Jin S, Wang L, Zhang Y. RNA-seq profiling identified a three-lncRNA panel in serum as potential biomarker for muscle-invasive bladder cancer. Front Oncol 2024; 14:1451009. [PMID: 39737397 PMCID: PMC11683095 DOI: 10.3389/fonc.2024.1451009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Background Preoperative determination of muscular infiltration is crucial for appropriate treatment planning in patients with muscle-invasive bladder cancer (MIBC). We aimed to explore early diagnostic biomarkers in serum for MIBC in this study. Methods The expression profiles of long noncoding RNA (lncRNA) were initially screened by high-throughput sequencing and evaluation of potential lncRNAs were conducted by two phases of RT-qPCR assays using serum samples from 190 patients with MIBC and 190 non-muscle-invasive BC (NMIBC) patients. Multivariate logistic regression analysis was applied to establish a diagnostic signature with high accuracy and Fagan's nomogram was plotted to promote clinical application. Bioinformatics analysis was used to determine the potential miRNA-mRNA binding of candidate lncRNAs. Results We identified three differentially expressed lncRNAs (LINC00565, LINC00592 and NDUFA6-AS1) and established a 3-lncRNA panel which demonstrated high diagnostic accuracy for MIBC with an AUC of 0.903 (95% CI: 0.850-0.942) and 0.875 (95% CI: 0.802-0.928) in the training and validation set. Moreover, construction and assessment of Fagan'nomogram demonstrated that the 3-lncRNA panel could exhibit practical and helpful values for clinical use. Finally, a network map based on LINC00565 was constructed and we found that the expression of miR-143-5p and miR-4516 were significantly correlated with LINC00565 in MIBC. Conclusion Our findings indicated that the constructed 3-lncRNA panel in serum showed favorable diagnostic capacity and might serve as promising non-invasive biomarkers in the early diagnosis of MIBC.
Collapse
Affiliation(s)
- Xiumei Jiang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Ailin Qu
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Shoucai Zhang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Shuchao Jin
- Department of Urology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Lishui Wang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
3
|
Abedimanesh S, Safaralizadeh R, Jahanafrooz Z, Najafi S, Amini M, Nazarloo SS, Bahojb Mahdavi SZ, Baradaran B, Jebelli A, Mokhtarzadeh AA. Interaction of noncoding RNAs with hippo signaling pathway in cancer cells and cancer stem cells. Noncoding RNA Res 2024; 9:1292-1307. [PMID: 39045083 PMCID: PMC11263728 DOI: 10.1016/j.ncrna.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 07/25/2024] Open
Abstract
The Hippo signaling pathway has a regulatory function in the organogenesis process and cellular homeostasis, switching the cascade reactions of crucial kinases acts to turn off/on the Hippo pathway, altering the downstream gene expression and thereby regulating proliferation, apoptosis, or stemness. Disruption of this pathway can lead to the occurrence of various disorders and different types of cancer. Recent findings highlight the importance of ncRNAs, such as microRNA, circular RNA, and lncRNAs, in modulating the Hippo pathway. Defects in ncRNAs can disrupt Hippo pathway balance, increasing tumor cells, tumorigenesis, and chemotherapeutic resistance. This review summarizes ncRNAs' inhibitory or stimulatory role in - Hippo pathway regulation in cancer and stem cells. Identifying the relation between ncRNAs and the components of this pathway could pave the way for developing new biomarkers in the treatment and diagnosis of cancers.
Collapse
Affiliation(s)
- Saba Abedimanesh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zohreh Jahanafrooz
- Department of Biology, Faculty of Sciences, University of Maragheh, Maragheh, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Soltani Nazarloo
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asiyeh Jebelli
- Department of Biological Sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashid, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
4
|
Pan X, Shi X, Zhang H, Chen Y, Zhou J, Shen F, Wang J, Jiang R. Exosomal miR-4516 derived from ovarian cancer stem cells enhanced cisplatin tolerance in ovarian cancer by inhibiting GAS7. Gene 2024; 927:148738. [PMID: 38955306 DOI: 10.1016/j.gene.2024.148738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/05/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Ovarian cancer (OC) is a devastating disease for women, with chemotherapy resistance taking the lead. Cisplatin has been the first-line therapy for OC for a long time. However, the resistance of OC to cisplatin is an important impediment to its efficacy. Mounting studies showed that ovarian cancer stem cells (OCSCs) affected chemotherapy resistance by secreting exosomes. MicroRNAs (miRNAs) play important roles in exosomes secreted by OCSCs. Here, through the analysis of GEO database (GSE107155) combined with RT-qPCR of OC-related cells/clinical tissues, it was found that hsa-miR-4516 (miR-4516) was significantly up-regulated in OCSCs. Then, OCSCs-derived exosomes were isolated and identified, and it was observed the influence of exosomes on the chemoresistance in SKOV3/cisplatin (SKOV3/DDP) cells. These results manifested that OCSCs-mediated exosomes facilitated the chemoresistance of SKOV3/DDP cells by delivering miR-4516 into them. Growth arrest-specific 7 (GAS7), a downstream target of miR-4516, was determined by bioinformatics prediction combined with molecular biological detection. Next, we up-regulated GAS7 expression and discovered that the promotion of chemoresistance in SKOV3/DDP cells by OCSCs-derived exosomes was significantly impaired. Finally, the mice tumor model of SKOV3/DDP cells was built to estimate the effect of GAS7 over-expression on OC growth. The results showed that GAS7 inhibited the chemoresistance of OC in vivo. In conclusion, our experiments suggested that OCSCs-derived exosomes enhanced OC cisplatin resistance by suppressing GAS7 through the delivery of miR-4516. This study provides a possible target for the treatment of OC DDP resistance.
Collapse
Affiliation(s)
- Xin Pan
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - Xiu Shi
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - Hong Zhang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - YouGuo Chen
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - JinHua Zhou
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - FangRong Shen
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - Juan Wang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China
| | - Rong Jiang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215000, China.
| |
Collapse
|
5
|
Chong ZX. Roles of miRNAs in regulating ovarian cancer stemness. Biochim Biophys Acta Rev Cancer 2024; 1879:189191. [PMID: 39353485 DOI: 10.1016/j.bbcan.2024.189191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Ovarian cancer is one of the gynaecology malignancies with the highest mortality rate. Ovarian cancer stem cell (CSC) is a subpopulation of ovarian cancer cells with increased self-renewability, aggression, metastatic potentials, and resistance to conventional anti-cancer therapy. The emergence of ovarian CSC is a critical factor that promotes treatment resistance and frequent relapse among ovarian cancer patients, leading to poor clinical outcomes. MicroRNA (miRNA) is a short, non-protein-coding RNA that regulates ovarian CSC development. Although multiple original research articles have discussed the CSC-regulatory roles of different miRNAs in ovarian cancer, there is a deficiency of a review article that can summarize the findings from different research papers. To narrow the gap in the literature, this review aimed to provide an up-to-date summary of the CSC-regulatory roles of various miRNAs in modulating ovarian cancer cell stemness. This review will begin by giving an overview of ovarian CSC and the pathways responsible for driving its appearance. Next, the CSC-regulatory roles of miRNAs in controlling ovarian CSC development will be discussed. Overall, more than 60 miRNAs have been reported to play CSC-regulatory roles in the development and progression of ovarian cancer. By targeting various downstream targets, these miRNAs can control the signaling activities of PI3K/AKT, EGFR/ERK, WNT/ß-catenin, NF-kß, Notch, Hippo/YAP, EMT, and DNA repair pathways. Hence, these CSC-modulatory miRNAs have the potential to be used as prognostic biomarkers in predicting the clinical outcomes of ovarian cancer patients. Targeting CSC-promoting miRNAs or increasing the expressions of CSC-repressing miRNAs can help slow ovarian cancer progression. However, more in-depth functional and clinical trials must be carried out to evaluate the suitability, safety, sensitivity, and specificity of these CSC-regulating miRNAs as prognostic biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Zhi-Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, Jalan Broga, 43500 Semenyih, Selangor, Malaysia; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Centre for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599; Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, 14 Medical Drive, #12-01, Singapore 117599.
| |
Collapse
|
6
|
Sakaue T, Koga H, Iwamoto H, Nakamura T, Masuda A, Tanaka T, Suzuki H, Suga H, Hirai S, Hisaka T, Naito Y, Ohta K, Nakamura KI, Selvendiran K, Okabe Y, Torimura T, Kawaguchi T. Pancreatic Juice-Derived microRNA-4516 and microRNA-4674 as Novel Biomarkers for Pancreatic Ductal Adenocarcinoma. GASTRO HEP ADVANCES 2024; 3:761-772. [PMID: 39280916 PMCID: PMC11401553 DOI: 10.1016/j.gastha.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/24/2024] [Indexed: 09/18/2024]
Abstract
Background and Aims Precise diagnostic biomarkers are urgently required for pancreatic ductal adenocarcinoma (PDAC). Therefore, the aim of this study was to identify PDAC-specific exosomal microRNAs (Ex-miRs) from pancreatic juice (PJ) and evaluate their diagnostic potential. Methods Exosomes in PJ and serum were extracted using ultracentrifugation and confirmed morphologically and biochemically. PDAC-specific Ex-miRs were identified using our original miR arrays, in which "Ex-miRs derived from the PJ of patients with chronic pancreatitis (CP)" were subtracted from Ex-miRs commonly expressed in both "human PDAC cell lines" and "the PJ of patients with PDAC." We verified the expression of these miRs using quantitative real-time reverse transcription polymerase chain reaction. Changes in serum Ex-miR levels were assessed in 2 patients with PDAC who underwent curative resection. In situ hybridization was performed to directly visualize PDAC-specific miR expression in cancer cells. Results We identified novel Ex-miR-4516 and Ex-miR-4674 from the PJ of patients with PDAC, and they showed 80.0% and 81.8% sensitivity, 80.8% and 73.3% specificity, and 90.9% and 80.8% accuracy, respectively. The sensitivity, specificity, and accuracy of a triple assay of Ex-miR-4516/4674/PJ cytology increased to 93.3%, 81.8%, and 88.5%, respectively. In serum samples (n = 88), the sensitivity, specificity, and accuracy of Ex-miR-4516 were 97.5%, 34.3%, and 68%, respectively. Presurgical levels of serum-derived Ex-miR-4516 in 2 patients with relatively early disease stages declined after curative resection. In situ hybridization demonstrated that Ex-miR-4516 expression exclusively occurred in cancer cells. Conclusion Liquid assays using the in situ-proven Ex-miR-4516 may have a high potential for detecting relatively early-stage PDAC and monitoring its clinical course.
Collapse
Affiliation(s)
- Takahiko Sakaue
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Liver Cancer Research Division, Kurume University Research Center for Innovative Cancer Therapy, Kurume, Japan
- Division of Gynecologic Oncology, Department of Obstetrics/Gynecology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Liver Cancer Research Division, Kurume University Research Center for Innovative Cancer Therapy, Kurume, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Liver Cancer Research Division, Kurume University Research Center for Innovative Cancer Therapy, Kurume, Japan
| | - Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Liver Cancer Research Division, Kurume University Research Center for Innovative Cancer Therapy, Kurume, Japan
| | - Atsutaka Masuda
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Liver Cancer Research Division, Kurume University Research Center for Innovative Cancer Therapy, Kurume, Japan
| | - Toshimitsu Tanaka
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Liver Cancer Research Division, Kurume University Research Center for Innovative Cancer Therapy, Kurume, Japan
- Center for Multidisciplinary Treatment of Cancer, Kurume University Hospital, Kurume, Japan
| | - Hiroyuki Suzuki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Liver Cancer Research Division, Kurume University Research Center for Innovative Cancer Therapy, Kurume, Japan
| | - Hideya Suga
- Department of Gastroenterology and Hepatology, Yanagawa Hospital, Yanagawa, Japan
| | - Shingo Hirai
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toru Hisaka
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Yoshiki Naito
- Department of Clinical Laboratory Medicine, Kurume University Hospital, Kurume, Japan
| | - Keisuke Ohta
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, Japan
| | - Kei-Ichiro Nakamura
- Division of Microscopic and Developmental Anatomy, Department of Anatomy, Kurume University School of Medicine, Kurume, Japan
| | - Karuppaiyah Selvendiran
- Division of Gynecologic Oncology, Department of Obstetrics/Gynecology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Yoshinobu Okabe
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takuji Torimura
- Department of Gastroenterology, Omuta City Hospital, Omuta, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
7
|
Becker AP, Becker V, McElroy J, Webb A, Han C, Guo Y, Bell EH, Fleming J, Popp I, Staszewski O, Prinz M, Otero JJ, Haque SJ, Grosu AL, Chakravarti A. Proteomic Analysis of Spatial Heterogeneity Identifies HMGB2 as Putative Biomarker of Tumor Progression in Adult-Type Diffuse Astrocytomas. Cancers (Basel) 2024; 16:1516. [PMID: 38672598 PMCID: PMC11049315 DOI: 10.3390/cancers16081516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Although grading is defined by the highest histological grade observed in a glioma, most high-grade gliomas retain areas with histology reminiscent of their low-grade counterparts. We sought to achieve the following: (i) identify proteins and molecular pathways involved in glioma evolution; and (ii) validate the high mobility group protein B2 (HMGB2) as a key player in tumor progression and as a prognostic/predictive biomarker for diffuse astrocytomas. We performed liquid chromatography tandem mass spectrometry (LC-MS/MS) in multiple areas of adult-type astrocytomas and validated our finding in multiplatform-omics studies and high-throughput IHC analysis. LC-MS/MSdetected proteomic signatures characterizing glioma evolution towards higher grades associated with, but not completely dependent, on IDH status. Spatial heterogeneity of diffuse astrocytomas was associated with dysregulation of specific molecular pathways, and HMGB2 was identified as a putative driver of tumor progression, and an early marker of worse overall survival in grades 2 and 3 diffuse gliomas, at least in part regulated by DNA methylation. In grade 4 astrocytomas, HMGB2 expression was strongly associated with proliferative activity and microvascular proliferation. Grounded in proteomic findings, our results showed that HMGB2 expression assessed by IHC detected early signs of tumor progression in grades 2 and 3 astrocytomas, as well as identified GBMs that had a better response to the standard chemoradiation with temozolomide.
Collapse
Affiliation(s)
- Aline P. Becker
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA; (A.P.B.); (V.B.); (C.H.); (Y.G.); (J.F.); (S.J.H.)
| | - Valesio Becker
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA; (A.P.B.); (V.B.); (C.H.); (Y.G.); (J.F.); (S.J.H.)
| | - Joseph McElroy
- Center for Biostatistics, The Ohio State University, Columbus, OH 43210, USA;
| | - Amy Webb
- School of Biomedical Science-Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA;
| | - Chunhua Han
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA; (A.P.B.); (V.B.); (C.H.); (Y.G.); (J.F.); (S.J.H.)
| | - Yingshi Guo
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA; (A.P.B.); (V.B.); (C.H.); (Y.G.); (J.F.); (S.J.H.)
| | - Erica H. Bell
- Department of Neurology, The Ohio State University, Columbus, OH 43210, USA;
| | - Jessica Fleming
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA; (A.P.B.); (V.B.); (C.H.); (Y.G.); (J.F.); (S.J.H.)
| | - Ilinca Popp
- Department of Radiation Oncology, University of Freiburg, 79110 Freiburg, Germany; (I.P.); (A.-L.G.)
| | - Ori Staszewski
- Institute of Neuropathology, Medical Faculty of the Saarland University, 66421 Homburg, Germany;
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
- Signalling Research Centres BIOSS & CIBSS, University of Freiburg, 79098 Freiburg, Germany
| | - Jose J. Otero
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA;
| | - Saikh Jaharul Haque
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA; (A.P.B.); (V.B.); (C.H.); (Y.G.); (J.F.); (S.J.H.)
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, University of Freiburg, 79110 Freiburg, Germany; (I.P.); (A.-L.G.)
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA; (A.P.B.); (V.B.); (C.H.); (Y.G.); (J.F.); (S.J.H.)
| |
Collapse
|
8
|
Hosseinzadeh A, Pourhanifeh MH, Amiri S, Sheibani M, Irilouzadian R, Reiter RJ, Mehrzadi S. Therapeutic potential of melatonin in targeting molecular pathways of organ fibrosis. Pharmacol Rep 2024; 76:25-50. [PMID: 37995089 DOI: 10.1007/s43440-023-00554-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/24/2023]
Abstract
Fibrosis, the excessive deposition of fibrous connective tissue in an organ in response to injury, is a pathological condition affecting many individuals worldwide. Fibrosis causes the failure of tissue function and is largely irreversible as the disease progresses. Pharmacologic treatment options for organ fibrosis are limited, but studies suggest that antioxidants, particularly melatonin, can aid in preventing and controlling fibrotic damage to the organs. Melatonin, an indole nocturnally released from the pineal gland, is commonly used to regulate circadian and seasonal biological rhythms and is indicated for treating sleep disorders. While it is often effective in treating sleep disorders, melatonin's anti-inflammatory and antioxidant properties also make it a promising molecule for treating other disorders such as organ fibrosis. Melatonin ameliorates the necrotic and apoptotic changes that lead to fibrosis in various organs including the heart, liver, lung, and kidney. Moreover, melatonin reduces the infiltration of inflammatory cells during fibrosis development. This article outlines the protective effects of melatonin against fibrosis, including its safety and potential therapeutic effects. The goal of this article is to provide a summary of data accumulated to date and to encourage further experimentation with melatonin and increase its use as an anti-fibrotic agent in clinical settings.
Collapse
Affiliation(s)
- Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Shiva Amiri
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Sheibani
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rana Irilouzadian
- Clinical Research Development Unit of Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Ma M, Zeng G, Tan B, Zhao G, Su Q, Zhang W, Song Y, Liang J, Xu B, Wang Z, Chen J, Hou M, Yang C, Yun J, Huang Y, Lin Y, Chen D, Han Y, DeMorrow S, Liang L, Lai J, Huang L. DAGLβ is the principal synthesizing enzyme of 2-AG and promotes aggressive phenotype of intrahepatic cholangiocarcinoma via AP-1/DAGLβ/miR4516 feedforward circuitry. Am J Physiol Gastrointest Liver Physiol 2023; 325:G213-G229. [PMID: 37366545 PMCID: PMC10435072 DOI: 10.1152/ajpgi.00243.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023]
Abstract
The endocannabinoid system (ECS) is dysregulated in various liver diseases. Previously, we had shown that the major endocannabinoid 2-arachidonoyl glycerol (2-AG) promoted tumorigenesis of intrahepatic cholangiocarcinoma (ICC). However, biosynthesis regulation and clinical significance of 2-AG remain elusive. In the present study, we quantified 2-AG by gas chromatography/mass spectrometry (GC/MS) and showed that 2-AG was enriched in patients with ICC samples as well as in thioacetamide-induced orthotopic rat ICC model. Moreover, we found that diacylglycerol lipase β (DAGLβ) was the principal synthesizing enzyme of 2-AG that significantly upregulated in ICC. DAGLβ promoted tumorigenesis and metastasis of ICC in vitro and in vivo and positively correlated with clinical stage and poor survival in patients with ICC. Functional studies showed that activator protein-1 (AP-1; heterodimers of c-Jun and FRA1) directly bound to the promoter and regulated transcription of DAGLβ, which can be enhanced by lipopolysaccharide (LPS). miR-4516 was identified as the tumor-suppressing miRNA of ICC that can be significantly suppressed by LPS, 2-AG, or ectopic DAGLβ overexpression. FRA1 and STAT3 were targets of miR-4516 and overexpression of miRNA-4516 significantly suppressed expression of FRA1, SATA3, and DAGLβ. Expression of miRNA-4516 was negatively correlated with FRA1, SATA3, and DAGLβ in patients with ICC samples. Our findings identify DAGLβ as the principal synthesizing enzyme of 2-AG in ICC. DAGLβ promotes oncogenesis and metastasis of ICC and is transcriptionally regulated by a novel AP-1/DAGLβ/miR4516 feedforward circuitry.NEW & NOTEWORTHY Dysregulated endocannabinoid system (ECS) had been confirmed in various liver diseases. However, regulation and function of 2-arachidonoyl glycerol (2-AG) and diacylglycerol lipase β (DAGLβ) in intrahepatic cholangiocarcinoma (ICC) remain to be elucidated. Here, we demonstrated that 2-AG was enriched in ICC, and DAGLβ was the principal synthesizing enzyme of 2-AG in ICC. DAGLβ promotes tumorigenesis and metastasis in ICC via a novel activator protein-1 (AP-1)/DAGLβ/miR4516 feedforward circuitry.
Collapse
Affiliation(s)
- Mingjian Ma
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Guangyan Zeng
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Bingyan Tan
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Guangyin Zhao
- Laboratory Animal Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qiao Su
- Laboratory Animal Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wenhui Zhang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Song
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiahua Liang
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Borui Xu
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zicheng Wang
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiancong Chen
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Mengjun Hou
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chuntao Yang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jingping Yun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yuhua Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yansong Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Demeng Chen
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yuyan Han
- School of Biological Sciences, University of Northern Colorado, Greeley, Colorado, United States
| | - Sharon DeMorrow
- Research Division, Central Texas Veterans Health Care System, Temple, Texas, United States
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, United States
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas, United States
| | - Lijian Liang
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiaming Lai
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Huang
- Department of Pancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Center of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
10
|
Nafe R, Hattingen E. The Spectrum of Molecular Pathways in Gliomas-An Up-to-Date Review. Biomedicines 2023; 11:2281. [PMID: 37626776 PMCID: PMC10452344 DOI: 10.3390/biomedicines11082281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
During the last 20 years, molecular alterations have gained increasing significance in the diagnosis and biological assessment of tumors. Gliomas represent the largest group of tumors of the central nervous system, and the main aim of this review is to present the current knowledge on molecular pathways and their alterations in gliomas. A wide range of new insights has been gained, including evidence for the involvement of the WNT pathway or the hippo pathway in the pathobiology of gliomas, indicating a broad involvement of different pathways formerly not considered to play a central role in gliomas. Even new aspects of angiogenic, apoptotic, and metabolic pathways are presented, as well as the rapidly growing field of epigenetic processes, including non-coding RNAs. The two major conclusions drawn from the present review are the distinct interconnectivity of the whole spectrum of molecular pathways and the prominent role of non-coding RNAs, especially circular RNAs, in the regulation of specific targets. All these new insights are discussed, even considering the topic of the resistance to therapy of gliomas, along with aspects that are still incompletely understood, like the role of hydroxymethylation, or even ferroptosis, in the pathobiology of gliomas.
Collapse
Affiliation(s)
- Reinhold Nafe
- Department of Neuroradiology, Clinics of Johann Wolfgang Goethe-University, Schleusenweg 2-16, D-60528 Frankfurt am Main, Germany;
| | | |
Collapse
|
11
|
Gray A, Cui T, Bell EH, McElroy J, Sebastian E, Li F, Geurts M, Liu K, Robe P, Haque SJ, Chakravarti A. MicroRNA-575 acts as a novel oncogene via targeting multiple signaling pathways in glioblastoma. Exp Mol Pathol 2022; 128:104813. [PMID: 35901926 DOI: 10.1016/j.yexmp.2022.104813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 05/09/2022] [Accepted: 07/16/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE Glioblastoma (GBM) patients currently face poor survival outcomes with an average survival period of <15 months, while only 3-5% of patients survive longer than 36 months. Although the mechanisms of tumorigenesis are still being elucidated, miRNAs are promising candidates to explore as novel and prognostic biomarkers in GBM. In this study, we identified the association between miR-575 expression and overall survival (OS) of primary GBM patients and undertook functional studies to discern the contribution of miR-575 to GBM tumorigenesis. METHODS Total RNAs were isolated from 254 FFPE GBM tumor samples and miR expression was assayed (simultaneously) using NanoString Technologies. To determine the association between miR-575 and patients' prognosis, Kaplan-Meier, univariable and multivariable Cox regression analyses were performed. Cell proliferation, colony formation, migration assays were conducted to investigate the function of miR-575 in vitro and in vivo. In silico target gene network analysis was performed to identify the putative targets of miR-575 in GBM, which were further verified by luciferase reporter assay, as well as qPCR and immunoblotting. RESULTS Our clinical data (n = 254) show that miR-575 is associated with worse GBM OS by univariable analysis (UVA, HR = 1.27, p-value<0.001) and multivariable (MVA, HR = 1.23, p = 0.007) analysis incorporating critical clinical variables. Functional studies indicated that overexpression of miR-575 significantly increased cell proliferation and migration of GBM cells in vitro, as well as tumor growth in vivo. Subsequent in silico target gene network and mechanistic studies identified CDKN1B/p27 and PTEN, as potential targets of miR-575 in GBM. MicroRNA-575 can also regulate the activity of AKT and ERK pathways in GBM. CONCLUSION miR-575 has prognostic value in GBM, with higher expression associating with worse OS of patients, and contributes to GBM tumorigenesis by regulating multiple signaling pathways in GBM.
Collapse
Affiliation(s)
- Ashley Gray
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Tiantian Cui
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, OH, USA
| | - Erica Hlavin Bell
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, OH, USA
| | - Joseph McElroy
- The Ohio State University Center for Biostatistics, Department of Biomedical Informatics, Columbus, OH, USA
| | - Ebin Sebastian
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, OH, USA
| | - Fuhai Li
- Institute for Informatics, Washington University School of Medicine, St. Louis, MO, USA
| | - Marjolein Geurts
- Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Kevin Liu
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Pierre Robe
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S Jaharul Haque
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, OH, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
12
|
Bibby G, Krasniqi B, Reddy I, Sekar D, Ross K. Capturing the RNA castle: Exploiting MicroRNA inhibition for wound healing. FEBS J 2022; 289:5137-5151. [PMID: 34403569 DOI: 10.1111/febs.16160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/14/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023]
Abstract
The growing pipelines of RNA-based therapies herald new opportunities to deliver better patient outcomes for complex disorders such as chronic nonhealing wounds associated with diabetes. Members of the microRNA (miRNA) family of small noncoding RNAs have emerged as targets for diverse elements of cutaneous wound repair, and both miRNA enhancement with mimics or inhibition with antisense oligonucleotides represent tractable approaches for miRNA-directed wound healing. In this review, we focus on miRNA inhibition strategies to stimulate skin repair given advances in chemical modifications to enhance the performance of antisense miRNA (anti-miRs). We first explore miRNAs whose inhibition in keratinocytes promotes keratinocyte migration, an essential part of re-epithelialisation during wound repair. We then focus on miRNAs that can be targeted for inhibition in endothelial cells to promote neovascularisation for wound healing in the context of diabetic mouse models. The picture that emerges is that direct comparisons of different anti-miRNAs modifications are required to establish the most translationally viable options in the chronic wound environment, that direct comparisons of the impact of inhibition of different miRNAs are needed to quantify and rank their relative efficacies in promoting wound repair, and that a standardised human ex vivo model of the diabetic wound is needed to reduce reliance on mouse models that do not necessarily enhance mechanistic understanding of miRNA-targeted wound healing.
Collapse
Affiliation(s)
- George Bibby
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, UK
| | - Blerta Krasniqi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, UK
| | - Izaak Reddy
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, UK
| | - Durairaj Sekar
- Dental Research Cell and Biomedical Research Unit (DRC-BRULAC), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science (SIMATS), Saveetha University, Chennai, India
| | - Kehinde Ross
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, UK
| |
Collapse
|
13
|
Tang X, Qi C, Zhou H, Liu Y. Critical roles of PTPN family members regulated by non-coding RNAs in tumorigenesis and immunotherapy. Front Oncol 2022; 12:972906. [PMID: 35957898 PMCID: PMC9360549 DOI: 10.3389/fonc.2022.972906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/04/2022] [Indexed: 12/22/2022] Open
Abstract
Since tyrosine phosphorylation is reversible and dynamic in vivo, the phosphorylation state of proteins is controlled by the opposing roles of protein tyrosine kinases (PTKs) and protein tyrosine phosphatase (PTPs), both of which perform critical roles in signal transduction. Of these, intracellular non-receptor PTPs (PTPNs), which belong to the largest class I cysteine PTP family, are essential for the regulation of a variety of biological processes, including but not limited to hematopoiesis, inflammatory response, immune system, and glucose homeostasis. Additionally, a substantial amount of PTPNs have been identified to hold crucial roles in tumorigenesis, progression, metastasis, and drug resistance, and inhibitors of PTPNs have promising applications due to striking efficacy in antitumor therapy. Hence, the aim of this review is to summarize the role played by PTPNs, including PTPN1/PTP1B, PTPN2/TC-PTP, PTPN3/PTP-H1, PTPN4/PTPMEG, PTPN6/SHP-1, PTPN9/PTPMEG2, PTPN11/SHP-2, PTPN12/PTP-PEST, PTPN13/PTPL1, PTPN14/PEZ, PTPN18/PTP-HSCF, PTPN22/LYP, and PTPN23/HD-PTP, in human cancer and immunotherapy and to comprehensively describe the molecular pathways in which they are implicated. Given the specific roles of PTPNs, identifying potential regulators of PTPNs is significant for understanding the mechanisms of antitumor therapy. Consequently, this work also provides a review on the role of non-coding RNAs (ncRNAs) in regulating PTPNs in tumorigenesis and progression, which may help us to find effective therapeutic agents for tumor therapy.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of Clinical Laboratory Diagnostics, Binzhou Medical University, Binzhou, China
| | - Chumei Qi
- Department of Clinical Laboratory, Dazhou Women and Children’s Hospital, Dazhou, China
| | - Honghong Zhou
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Honghong Zhou, ; Yongshuo Liu,
| | - Yongshuo Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- *Correspondence: Honghong Zhou, ; Yongshuo Liu,
| |
Collapse
|
14
|
Pao SI, Lin LT, Chen YH, Chen CL, Chen JT. MicroRNA-4516 suppresses proliferative vitreoretinopathy development via negatively regulating OTX1. PLoS One 2022; 17:e0270526. [PMID: 35771766 PMCID: PMC9246108 DOI: 10.1371/journal.pone.0270526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/11/2022] [Indexed: 11/18/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) progression is associated with TGF-β2-induced epithelial–mesenchymal transition (EMT) in retinal pigment epithelial (RPE) cells. In cancer cells, miR-4516 downregulates orthodenticle homeobox 1 (OTX1)-mediated cell invasion. Moreover, OTX1 is shown to be involved in invasion and EMT. The purpose of this study was to assess whether microRNA (miR-4516) suppresses EMT in RPE cells. EMT features were assessed using Western blotting, immunocytochemical staining, scratch-wound healing, modified Boyden chamber assay, and collagen gel contraction assay. For in vivo testing, a rabbit model was used, which involved induction of PVR by injection of transfected spontaneously arising RPE (ARPE) cells into the vitreous chamber. The putative target of miR-4516 was identified by luciferase reporter assay. Results showed that TGF-β2-induced transdifferentiation and migration of RPE cells was inhibited by miR-4516 delivery. Overexpression of miR-4516 led to upregulation of zonula occludens-1, downregulation of α-smooth muscle actin and vimentin, and cell contractility—all EMT features—in the TGF-β2-treated ARPE-19 cells. MiR-4516 regulated OTX1 expression negatively by binding to its 3’-UTR. TGF-β2-induced phosphorylated ERK was inhibited in miR-4516-overexpressing ARPE-19 cells. MiR-4516 suppressed experimental PVR in vitro and in vivo. In conclusion, the overexpression of miR-4516 suppresses TGF-β2-induced EMT in a PVR model, and its role in PVR depends on OTX1/ERK. Further research is needed to develop a feasible treatment method to prevent and treat PVR.
Collapse
Affiliation(s)
- Shu-I Pao
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Le-Tien Lin
- Department of Ophthalmology, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Hao Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Long Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Jiann-Torng Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
15
|
Xu Q, Xu JL, Chen WQ, Xu WX, Song YX, Tang WJ, Xu D, Jiang MP, Tang J. Roles and mechanisms of miR-195-5p in human solid cancers. Biomed Pharmacother 2022; 150:112885. [PMID: 35453003 DOI: 10.1016/j.biopha.2022.112885] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 11/02/2022] Open
Abstract
Cancer persists as a worldwide disease that contributes to high morbidity and mortality rates. As a class of non-coding RNA, microRNAs (miRNAs) are one kind of important regulators in cancer and frequently implicated in tumor development and progression. Emerging experiments have suggested that miRNA-195-5p (miR-195-5p) can regulate neoplastic processes in many pathways. For instance, miR-195-5p can not only regulate proliferation, migration and invasion of tumor cells but also promote tumor cell apoptosis. Furthermore, low expression of miR-195-5p could induce drug resistance. Our review focuses on the expression of miR-195-5p in various tumors and elucidates the related mechanisms of which miR-195-5p participates in tumor biology, as well as summarizes the roles of miR-195-5p in tumor progression. We believe that miR-195-5p might have potential utility as a novel diagnostic biomarker and therapeutic target for cancer.
Collapse
Affiliation(s)
- Qi Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jia-Lin Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wen-Quan Chen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wen-Xiu Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Yu-Xin Song
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wen-Juan Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Di Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Meng-Ping Jiang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jinhai Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China.
| |
Collapse
|
16
|
Enge AM, Sprenger H, Braeuning A, Hessel-Pras S. Identification of microRNAs Implicated in Modulating Senecionine-Induced Liver Toxicity in HepaRG Cells. Foods 2022; 11:foods11040532. [PMID: 35206009 PMCID: PMC8871147 DOI: 10.3390/foods11040532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
Abstract
1,2-unsaturated Pyrrolizidine Alkaloids (PAs) are secondary plant metabolites that occur as food contaminants. Upon consumption, they can cause severe liver damage. PAs have been shown to induce apoptosis, to have cytotoxic and genotoxic effects, and to impair bile acid homeostasis in the human hepatoma cell line HepaRG. The major mode of action of PAs is DNA- and protein-adduct formation. Beyond that, nuclear receptor activation has only been observed for one receptor and two PAs, yielding the possibility that other cellular mediators are involved in PA-mediated toxicity. Here, the mode of action of Senecionine (Sc), a prominent and ubiquitous representative of hepatotoxic PAs, was investigated by analyzing 7 hepatic microRNAs (miRNAs) in HepaRG cells. Ultimately, 11 target genes that were predicted with Ingenuity Pathway Analysis software (IPA) were found to be significantly downregulated, while their assigned miRNAs showed significant upregulation of gene expression. According to IPA, these targets are positively correlated with apoptosis and cellular death and are involved in diseases such as hepatocellular carcinoma. Subsequent antagomiR-inhibition analysis revealed a significant correlation between PA-induced miRNA-4434 induction and P21-Activated Kinase-1 (PAK1) downregulation. PAK1 downregulation is usually associated with cell cycle arrest, suggesting a new function of Sc-mediated toxicity in human liver cells.
Collapse
|
17
|
Wang Y, Bao G, Zhang M, Xiang J, Zhou H, Wahafu A, Wu W, Ma X, Huo L, Bai X, Xie W, Liu P, Wang M. CRB2 enhances malignancy of glioblastoma via activation of the NF-κB pathway. Exp Cell Res 2022; 414:113077. [DOI: 10.1016/j.yexcr.2022.113077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/04/2022] [Accepted: 02/13/2022] [Indexed: 11/27/2022]
|
18
|
Li Q, Wang W, Yang T, Li D, Huang Y, Bai G, Li Q. LINC00520 up-regulates SOX5 to promote cell proliferation and invasion by miR-4516 in human hepatocellular carcinoma. Biol Chem 2022; 403:665-678. [PMID: 35089659 DOI: 10.1515/hsz-2021-0316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 01/05/2022] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common human cancers. Long non-coding RNA (lncRNA) has been demonstrated to play an important role in regulating tumor development. The current study aims to explore the specific role of LINC00520 during HCC progression. The present study identified that LINC00520 was upregulated in HCC tissues and indicated poor patient survival. Overexpression of LINC00520 promoted HCC cell proliferation, migration and invasion, while LINC00520 downregulation led to the opposite effects. Besides, LINC00520 knockdown was found to inhibit tumor growth in vivo. Furthermore, LINC00520 acted as a sponge of miR-4516 to regulate SRY-related high mobility group box 5 (SOX5). In addition, the inhibition of miR-4516 partly reversed the inhibitory effect of LINC00520 silencing on HCC cell proliferation, migration and invasion. In conclusion, the inhibition of LINC00520 suppressed HCC cell proliferation, migration and invasion through mediating miR-4516/SOX5 axis. Therefore, our study provides a basis for the development of treatment strategies for HCC.
Collapse
Affiliation(s)
- Qing Li
- Department of Internal Medicine, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Wei Wang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Tao Yang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Dongsheng Li
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Yinpeng Huang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Guang Bai
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Qiang Li
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| |
Collapse
|
19
|
Li J, Wang H. H3K27ac-activated EGFR-AS1 promotes cell growth in cervical cancer through ACTN4-mediated WNT pathway. Biol Direct 2022; 17:3. [PMID: 34998421 PMCID: PMC8742952 DOI: 10.1186/s13062-021-00315-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/29/2021] [Indexed: 01/09/2023] Open
Abstract
Background Recently, extensive studies unveiled that lncRNAs exert critical function in the development and progression of cervical cancer (CC). EGFR-AS1 is a novel lncRNA which has not been well-explored in CC. Aims Our study aimed to research the function and molecular mechanism of EGFR-AS1 in CC cells. qRT-PCR analysis was performed to detect gene expression. Colony formation, EdU, flow cytometry, TUNEL, western blot and transwell assays were performed to assess the effect of EGFR-AS1 on CC cell growth. The regulatory mechanism of EGFR-AS1 was dug out through mechanism experiments. Results EGFR-AS1 was notably overexpressed in CC cell lines. Loss-of-functional experiments revealed that EGFR-AS1 promoted CC cell proliferation, migration and invasion, and suppressed cell apoptosis. Mechanistically, up-regulation of EGFR-AS1 was attributed to the activation of H3K27 acetylation (H3K27ac). Further, EGFR-AS1 was revealed to function as miR-2355-5p sponge. Additionally, miR-2355-5p was down-regulated in CC cells and ACTN4 was identified as a target gene of miR-2355-5p. Ultimately, overexpressed ACTN4 could reserve the suppressive role of EGFR-AS1 silencing in CC cell growth. Last but not least, EGFR-AS1 facilitated CC cell growth via ACTN4-mediated WNT pathway. Conclusions H3K27ac-activated EGFR-AS1 sponged miR-2355-5p and promoted CC cell growth through ACTN4-mediated WNT pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s13062-021-00315-5.
Collapse
Affiliation(s)
- Jingyan Li
- Zibo Maternal and Child Health Hospital of Shandong Province, Zibo, 255000, Shandong, China
| | - Hongbing Wang
- Department of Gynecology and Oncology, Hubei Cancer Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Hongshan District, No. 116 Zhuodaoquan South Road, Wuhan, 430079, Hubei, China.
| |
Collapse
|
20
|
Zhang Y, Zhang Y, Wang S, Li Q, Cao B, Huang B, Wang T, Guo R, Liu N. SP1-induced lncRNA ZFPM2 antisense RNA 1 (ZFPM2-AS1) aggravates glioma progression via the miR-515-5p/Superoxide dismutase 2 (SOD2) axis. Bioengineered 2021; 12:2299-2310. [PMID: 34077295 PMCID: PMC8806534 DOI: 10.1080/21655979.2021.1934241] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022] Open
Abstract
Glioma is a common life-threatening tumor with high malignancy and high invasiveness. LncRNA ZFPM2 antisense RNA 1 (ZFPM2-AS1) was confirmed to be implicated in numerous tumors, while its biological function and mechanism have not been thoroughly understood in glioma. The gene expression was measured by RT-qPCR. Cell proliferation, cell cycle, and cell apoptosis of glioma cells were validated by CCK-8, colony formation, flow cytometry and TUNEL assays. The effect of ZFPM2-AS1 on tumor growth was verified by in vivo assay. The exploration on ZFPM2-AS1-mediated mechanism was carried out via ChIP, luciferase reporter, and RIP assays. In the present study, ZFPM2-AS1 was demonstrated as a highly-expressed lncRNA in glioma tissues and cells. ZFPM2-AS1 silencing suppressed cell proliferation and cell cycle, but facilitated cell apoptosis. In addition, the inhibitive effect of silenced ZFPM2-AS1 was also observed in tumor growth. Furthermore, we found that SP1 interacted with ZFPM2-AS1 promoter to transcriptionally activate ZFPM2-AS1 expression. Moreover, ZFPM2-AS1 was identified as a competing endogenous RNA (ceRNA) for miR-515-5p to target SOD2. Rescue assays verified that SOD2 overexpression partially abolished the suppressive impact of ZFPM2-AS1 silencing on glioma cell growth. In conclusion, this study corroborated the regulatory mechanism of SP1/ZFPM2-AS1/miR-515-5p/SOD2 axis in glioma, indicating that targeting ZFPM2-AS1 might be an effective way to treat glioma.
Collapse
Affiliation(s)
- Yaxuan Zhang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, China
| | - Yin Zhang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, China
| | - Sen Wang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, China
| | - Qingquan Li
- Department of Neurosurgery, The Second Affiliated Hospital of Nanjing Medical University
| | - Boqiang Cao
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, China
| | - Baosheng Huang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, China
| | - Tianlu Wang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, China
| | - Ruijuan Guo
- Department of ICU, The Affiliated Sir Run Run Hospital of Nanjing Medical University
| | - Ning Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University
| |
Collapse
|
21
|
Linck-Paulus L, Lämmerhirt L, Völler D, Meyer K, Engelmann JC, Spang R, Eichner N, Meister G, Kuphal S, Bosserhoff AK. Learning from Embryogenesis-A Comparative Expression Analysis in Melanoblast Differentiation and Tumorigenesis Reveals miRNAs Driving Melanoma Development. J Clin Med 2021; 10:2259. [PMID: 34073664 PMCID: PMC8197100 DOI: 10.3390/jcm10112259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022] Open
Abstract
Malignant melanoma is one of the most dangerous tumor types due to its high metastasis rates and a steadily increasing incidence. During tumorigenesis, the molecular processes of embryonic development, exemplified by epithelial-mesenchymal transition (EMT), are often reactivated. For melanoma development, the exact molecular differences between melanoblasts, melanocytes, and melanoma cells are not completely understood. In this study, we aimed to identify microRNAs (miRNAs) that promote melanoma tumorigenesis and progression, based on an in vitro model of normal human epidermal melanocyte (NHEM) de-differentiation into melanoblast-like cells (MBrCs). Using miRNA-sequencing and differential expression analysis, we demonstrated in this study that a majority of miRNAs have an almost equal expression level in NHEMs and MBrCs but are significantly differentially regulated in primary tumor- and metastasis-derived melanoma cell lines. Further, a target gene analysis of strongly regulated but functionally unknown miRNAs yielded the implication of those miRNAs in many important cellular pathways driving malignancy. We hypothesize that many of the miRNAs discovered in our study are key drivers of melanoma development as they account for the tumorigenic potential that differentiates melanoma cells from proliferating or migrating embryonic cells.
Collapse
Affiliation(s)
- Lisa Linck-Paulus
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.L.-P.); (L.L.); (D.V.); (S.K.)
| | - Lisa Lämmerhirt
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.L.-P.); (L.L.); (D.V.); (S.K.)
| | - Daniel Völler
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.L.-P.); (L.L.); (D.V.); (S.K.)
| | - Katharina Meyer
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany; (K.M.); (R.S.)
| | - Julia C. Engelmann
- Department of Marine Microbiology and Biogeochemistry, NIOZ Royal Netherlands Institute for Sea Research, 1790 AB Den Burg, The Netherlands;
| | - Rainer Spang
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany; (K.M.); (R.S.)
| | - Norbert Eichner
- Department of Biochemistry I, University of Regensburg, 93053 Regensburg, Germany; (N.E.); (G.M.)
| | - Gunter Meister
- Department of Biochemistry I, University of Regensburg, 93053 Regensburg, Germany; (N.E.); (G.M.)
| | - Silke Kuphal
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.L.-P.); (L.L.); (D.V.); (S.K.)
| | - Anja Katrin Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.L.-P.); (L.L.); (D.V.); (S.K.)
| |
Collapse
|
22
|
Fan X, Sun Y, Guo X, He C, Han B, Sun X. Long non-coding RNA LINC01116 regulated miR-744-5p/SCN1B axis to exacerbate lung squamous cell carcinoma. Cancer Biomark 2021; 28:473-482. [PMID: 32538822 DOI: 10.3233/cbm-190945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Lung squamous cell carcinoma (LUSC) is a kind of lung cancer which possesses high morbidity and mortality. Long non-coding RNAs (lncRNAs) have been abundantly reported to participate in regulating cellular activities of various diseases, including cancers. LINC01116 was reported as a tumor promoter in some cancers, whereas its function has not been clarified in LUSC. OBJECTIVE This exploration aimed to study the modulatory role of LINC01116 in LUSC. METHODS The expressions of LINC01116, miR-744-5p and SCN1B were determined by RT-qPCR. CCK-8, EdU and transwell assays were conducted to evaluate the proliferative, migratory and invasive abilities of A549 and H1299 cells. The protein expression of SCN1B or EMT-associated proteins was examined through western blot assay. The interaction between miR-744-5p and LINC01116 (or SCN1B) was confirmed by RNA pull down and luciferase reporter assays. RESULTS LINC01116 was up-regulated in LUSC tissues and cells, and LINC01116 repression limited the proliferative, migratory, invasive capabilities and EMT process in LUSC cells. In mechanism, LINC01116 directly interacted with miR-744-5p, and its expression was negatively correlated with miR-744-5p expression. SCN1B, overexpressed in LUSC tissues and cells, was proved to be targeted by miR-744-5p. Furthermore, SCN1B expression was in a negative association with miR-744-5p expression. At last, SCN1B amplification recovered the inhibitive effect of LINC01116 knockdown on cell proliferation, migration, invasion and EMT process in LUSC. CONCLUSION LINC01116 regulated miR-744-5p/SCN1B axis to exacerbate LUSC, providing a helpful theoretic basis for the exploration of LUSC treatment.
Collapse
Affiliation(s)
- Xiangyu Fan
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Haerbin, Heilongjiang, China
| | - Yingying Sun
- TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Haerbin, Heilongjiang, China
| | - Xu Guo
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Haerbin, Heilongjiang, China
| | - Chunbo He
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Haerbin, Heilongjiang, China
| | - Beiqiu Han
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Haerbin, Heilongjiang, China
| | - Xilin Sun
- TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Haerbin, Heilongjiang, China
| |
Collapse
|
23
|
MicroRNA-183-5p contributes to malignant progression through targeting PDCD4 in human hepatocellular carcinoma. Biosci Rep 2021; 40:226717. [PMID: 33078826 PMCID: PMC7601345 DOI: 10.1042/bsr20201761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/21/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common malignant tumors worldwide. The present study aimed to investigate the biological role of microRNA-183-5p (miR-183-5p), a novel tumor-related microRNA (miRNA), in HCC and illuminate the possible molecular mechanisms. The expression patterns of miR-183-5p in clinical samples were characterized using qPCR analysis. Kaplan–Meier survival curve was applied to evaluate the correlation between miR-183-5p expression and overall survival of HCC patients. Effects of miR-183-5p knockdown on HCC cell proliferation, apoptosis, migration and invasion capabilities were determined via Cell Counting Kit-8 (CCK8) assays, flow cytometry, scratch wound healing assays and Transwell invasion assays, respectively. Mouse neoplasm transplantation models were established to assess the effects of miR-183-5p knockdown on tumor growth in vivo. Bioinformatics analysis, dual-luciferase reporter assays and rescue assays were performed for mechanistic researches. Results showed that miR-183-5p was highly expressed in tumorous tissues compared with adjacent normal tissues. Elevated miR-183-5p expression correlated with shorter overall survival of HCC patients. Moreover, miR-183-5p knockdown significantly suppressed proliferation, survival, migration and invasion of HCC cells compared with negative control treatment. Consistently, miR-183-5p knockdown restrained tumor growth in vivo. Furthermore, programmed cell death factor 4 (PDCD4) was identified as a direct target of miR-183-5p. Additionally, PDCD4 down-regulation was observed to abrogate the inhibitory effects of miR-183-5p knockdown on malignant phenotypes of HCC cells. Collectively, our data suggest that miR-183-5p may exert an oncogenic role in HCC through directly targeting PDCD4. The current study may offer some new insights into understanding the role of miR-183-5p in HCC.
Collapse
|
24
|
Cai X, Yu L, Chen Z, Ye F, Ren Z, Jin P. Arsenic trioxide-induced upregulation of miR-1294 suppresses tumor growth in hepatocellular carcinoma by targeting TEAD1 and PIM1. Cancer Biomark 2021; 28:221-230. [PMID: 32280078 DOI: 10.3233/cbm-190490] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recently, Arsenic trioxide (ATO) has been reported as an efficient drug for suppression of cancer cell growth. Existing studies revealed the extensive involvement of microRNAs (miRNAs) in initiation and development of hepatocellular carcinoma (HCC). However, the potential correlation between ATO and miRNAs in HCC progression remains to be explored. To conduct our research, we applied a qRT-PCR analysis to find miRNAs that were upregulated in HCC cells treated with ATO. In our present study, miR-1294 was found to be significantly upregulated in ATO-treated HCC cells. To confirm the function of ATO and miR-1294 in HCC progression, gain-of function assays were designed and conducted. As expected, proliferative ability of ATO-treated HCC cells was markedly weakened compared to DMSO-treated HCC cells. More importantly, proliferation was further suppressed in ATO-induced HCC cells after overexpression of miR-1294. Through bioinformatics analysis, some potential targets of miR-1294 were predicted. Further investigation revealed that Pim-1 proto-oncogene (PIM1) and TEA domain transcription factor 1 (TEAD1) were two downstream targets of miR-1294 and could be negatively regulated by ATO. Functionally, we determined that cell proliferation and apoptosis resistance suppressed by miR-1294 and ATO were recovered by introduction of TEAD1 and PIM1. Collectively, this study revealed that a novel ATO-miR-1294-TEAD1/PIM1 axis regulated HCC cell growth, offering a potential insight into the HCC therapy.
Collapse
Affiliation(s)
- Xiaoniao Cai
- Department of Gastroenterology, Pingyang People's Hospital, Pingyang, Wenzhou, Zhejiang, China
| | - Leilei Yu
- Department of Gastroenterology, Ruian People's Hospital, Ruian, Wenzhou, Zhejiang, China
| | - Zhen Chen
- Department of General Surgery, Ruian People's Hospital, Ruian, Wenzhou, Zhejiang, China
| | - Fangpeng Ye
- Department of Gastroenterology, Ruian People's Hospital, Ruian, Wenzhou, Zhejiang, China
| | - Zonghai Ren
- Department of Gastroenterology, Ruian People's Hospital, Ruian, Wenzhou, Zhejiang, China
| | - Peisheng Jin
- Department of Gastroenterology, Ruian People's Hospital, Ruian, Wenzhou, Zhejiang, China
| |
Collapse
|
25
|
MicroRNA-522-3p plays an oncogenic role in glioblastoma through activating Wnt/β-catenin signaling pathway via targeting SFRP2. Neuroreport 2021; 32:88-98. [PMID: 33395185 DOI: 10.1097/wnr.0000000000001565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Increasing studies have suggested that microRNAs (miRNAs) contribute to the occurrence and development of glioblastoma. MiR-522-3p is a novel miRNA, which has been found to modulate tumorigenesis and tumor progression. However, its pathological role and functional mechanism in glioblastoma remain elusive at present. METHOD The miR-522-3p expression in glioblastoma and adjacent normal tissues, human fetal astrocyte HA1800, and glioblastoma cell lines was detected by reverse transcription-PCR. The proliferation, migration, and invasion were detected through Cell Counting Kit-8 (CCK8) and Transwell assay, and apoptosis was calculated through flow cytometry. The downstream target of miR-522-3p was analyzed through bioinformatics, and the correlation between miR-522-3p and secreted frizzled-related protein 2 (SFRP2) was verified through dual-luciferase reporter assay and RNA immunoprecipitation (RIP) experiment. Besides, western blot was conducted to test the level of SFRP2 and the Wnt/β-catenin pathway. RESULTS MiR-522-3p was overexpressed in glioblastoma tissues compared with that in normal tissues, and the inhibition of miR-522-3p reduced cell proliferation, migration, and invasion and promoted apoptosis in glioblastoma. Bioinformatics revealed that SFRP2 was an essential downstream target of miR-522-3p, and it inhibited the malignant biological behaviors induced by miR-522-3p and inactivated the Wnt/β-catenin pathway. CONCLUSION MiR-522-3p is an oncogene in glioblastoma by targeting SFRP2 through the Wnt/β-catenin pathway.
Collapse
|
26
|
Zhou T, Wu L, Ma N, Tang F, Yu Z, Jiang Z, Li Y, Zong Z, Hu K. SOX9-activated FARSA-AS1 predetermines cell growth, stemness, and metastasis in colorectal cancer through upregulating FARSA and SOX9. Cell Death Dis 2020; 11:1071. [PMID: 33318478 PMCID: PMC7736271 DOI: 10.1038/s41419-020-03273-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
SRY-box transcription factors (SOXs) are effective inducers for the formation of stem-like phenotypes. As a member of SOX family, SOX9 (SRY-box transcription factor 9) has been reported to be highly expressed and exert oncogenic functions in multiple human cancers. In this study, we hypothesized that SOX9 could regulate the function of cancer stem/initiating cells (CSCs) to further facilitate the progression of colorectal cancer (CRC). Then, stable transfection of shRNAs was used to silence indicated genes. Loss-of-function experiments were conducted to demonstrate the in vitro function of CRC cells. In vivo study was conducted to determine the changes in tumorigenesis and metastasis in vivo. Bioinformatics analyses and mechanistic experiments were employed to explore the downstream molecules. Presently, GEPIA data indicated that SOX9 was upregulated in 275 COAD (colon adenocarcinoma) samples relative to 349 normal tissues. Besides, we also proved the upregulation of SOX9 in CRC cell lines (HCT15, SW480, SW1116, and HT-29) compared to normal NCM-460 cells. Silencing of SOX9 suppressed cell growth, stemness, migration, and invasion. Mechanistically, SOX9 activated the transcription of lncRNA phenylalanyl-tRNA synthetase subunit alpha antisense RNA 1 (FARSA-AS1), while FARSA-AS1 elevated SOX9 in turn by absorbing miR-18b-5p and augmented FARSA via sequestering miR-28-5p. Furthermore, loss of FARSA-AS1 hindered malignant phenotypes in vitro and blocked tumor growth and metastasis in vivo. Notably, we testified that FARSA-AS1 aggravated the malignancy in CRC by enhancing SOX9 and FARSA. Our study unveiled a mechanism of SOX9-FARSA-AS1-SOX9/FARSA loop in CRC, which provides some clews of promising targets for CRC.
Collapse
Affiliation(s)
- Taicheng Zhou
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, 510655, Guangzhou, Guangdong, China
| | - Lili Wu
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-sen University, Guangdong Key Laboratory of Liver Disease Research, 510630, Guangzhou, Guangdong, China
| | - Ning Ma
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, 510655, Guangzhou, Guangdong, China
| | - Fuxin Tang
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, 510655, Guangzhou, Guangdong, China
| | - Zhuomin Yu
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, 510655, Guangzhou, Guangdong, China
| | - Zhipeng Jiang
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, 510655, Guangzhou, Guangdong, China
| | - Yingru Li
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, 510655, Guangzhou, Guangdong, China
| | - Zhen Zong
- Department of Gastroenterological Surgery, The Second Affiliated Hospital of Nanchang University, No.1 Mingde Road, 330006, Nanchang, Jiangxi, China.
| | - Kunpeng Hu
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No.2693 Kaichuang Road, Huangpu, 510000, Guangzhou, Guangdong, China.
| |
Collapse
|
27
|
Caponnetto F, Dalla E, Mangoni D, Piazza S, Radovic S, Ius T, Skrap M, Di Loreto C, Beltrami AP, Manini I, Cesselli D. The miRNA Content of Exosomes Released from the Glioma Microenvironment Can Affect Malignant Progression. Biomedicines 2020; 8:biomedicines8120564. [PMID: 33287106 PMCID: PMC7761654 DOI: 10.3390/biomedicines8120564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Low-grade gliomas (LGG) are infiltrative primary brain tumors that in 70% of the cases undergo anaplastic transformation, deeply affecting prognosis. However, the timing of progression is heterogeneous. Recently, the tumor microenvironment (TME) has gained much attention either as prognostic factor or therapeutic target. Through the release of extracellular vesicles, the TME contributes to tumor progression by transferring bioactive molecules such as microRNA. The aim of the study was to take advantage of glioma-associated stem cells (GASC), an in vitro model of the glioma microenvironment endowed with a prognostic significance, and their released exosomes, to investigate the possible role of exosome miRNAs in favoring the anaplastic transformation of LGG. Therefore, by deep sequencing, we analyzed and compared the miRNA profile of GASC and exosomes obtained from LGG patients characterized by different prognosis. Results showed that exosomes presented a different signature, when compared to their cellular counterpart and that, although sharing several miRNAs, exosomes of patients with a bad prognosis, selectively expressed some miRNAs possibly responsible for the more aggressive phenotype. These findings get insights into the value of TME and exosomes as potential biomarkers for precision medicine approaches aimed at improving LGG prognostic stratification and therapeutic strategies.
Collapse
Affiliation(s)
- Federica Caponnetto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
- Correspondence: (F.C.); (I.M.); Tel.: +39-0432-559-412 (F.C. & I.M.)
| | - Emiliano Dalla
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
| | - Damiano Mangoni
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy;
| | - Silvano Piazza
- International Center for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy;
| | | | - Tamara Ius
- Neurosurgery Unit, Department of Neurosciences, University Hospital of Udine, 33100 Udine, Italy; (T.I.); (M.S.)
| | - Miran Skrap
- Neurosurgery Unit, Department of Neurosciences, University Hospital of Udine, 33100 Udine, Italy; (T.I.); (M.S.)
| | - Carla Di Loreto
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy
| | - Antonio Paolo Beltrami
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
| | - Ivana Manini
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy
- Correspondence: (F.C.); (I.M.); Tel.: +39-0432-559-412 (F.C. & I.M.)
| | - Daniela Cesselli
- Department of Medicine, University of Udine, 33100 Udine, Italy; (E.D.); (C.D.L.); (A.P.B.); (D.C.)
- Institute of Pathology, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
28
|
Rezaei O, Honarmand K, Nateghinia S, Taheri M, Ghafouri-Fard S. miRNA signature in glioblastoma: Potential biomarkers and therapeutic targets. Exp Mol Pathol 2020; 117:104550. [PMID: 33010295 DOI: 10.1016/j.yexmp.2020.104550] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/19/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are transcripts with sizes of about 22 nucleotides, which are produced through a multistep process in the nucleus and cytoplasm. These transcripts modulate the expression of their target genes through binding with certain target regions, particularly 3' suntranslated regions. They are involved in the pathogenesis of several kinds of cancers, such as glioblastoma. Several miRNAs, including miR-10b, miR-21, miR-17-92-cluster, and miR-93, have been up-regulated in glioblastoma cell lines and clinical samples. On the other hand, expression of miR-7, miR-29b, miR-32, miR-34, miR-181 family members, and a number of other miRNAs have been decreased in this type of cancer. In the current review, we explain the role of miRNAs in the pathogenesis of glioblastoma through providing a summary of studies that reported dysregulation of these epigenetic effectors in this kind of brain cancer.
Collapse
Affiliation(s)
- Omidvar Rezaei
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kasra Honarmand
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeedeh Nateghinia
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Cui T, Bell EH, McElroy J, Liu K, Sebastian E, Johnson B, Gulati PM, Becker AP, Gray A, Geurts M, Subedi D, Yang L, Fleming JL, Meng W, Barnholtz-Sloan JS, Venere M, Wang QE, Robe PA, Haque SJ, Chakravarti A. A Novel miR-146a-POU3F2/SMARCA5 Pathway Regulates Stemness and Therapeutic Response in Glioblastoma. Mol Cancer Res 2020; 19:48-60. [PMID: 32973101 DOI: 10.1158/1541-7786.mcr-20-0353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/24/2020] [Accepted: 09/21/2020] [Indexed: 11/16/2022]
Abstract
Rapid tumor growth, widespread brain-invasion, and therapeutic resistance critically contribute to glioblastoma (GBM) recurrence and dismal patient outcomes. Although GBM stem cells (GSC) are shown to play key roles in these processes, the molecular pathways governing the GSC phenotype (GBM-stemness) remain poorly defined. Here, we show that epigenetic silencing of miR-146a significantly correlated with worse patient outcome and importantly, miR-146a level was significantly lower in recurrent tumors compared with primary ones. Further, miR-146a overexpression significantly inhibited the proliferation and invasion of GBM patient-derived primary cells and increased their response to temozolomide (TMZ), both in vitro and in vivo. Mechanistically, miR-146a directly silenced POU3F2 and SMARCA5, two transcription factors that mutually regulated each other, significantly compromising GBM-stemness and increasing TMZ response. Collectively, our data show that miR-146a-POU3F2/SMARCA5 pathway plays a critical role in suppressing GBM-stemness and increasing TMZ-response, suggesting that POU3F2 and SMARCA5 may serve as novel therapeutic targets in GBM. IMPLICATIONS: miR-146a predicts favorable prognosis and the miR-146a-POU3F2/SMARCA5 pathway is important for the suppression of stemness in GBM.
Collapse
Affiliation(s)
- Tiantian Cui
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Erica H Bell
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Joseph McElroy
- The Ohio State University Center for Biostatistics, Department of Biomedical Informatics, Columbus, Ohio
| | - Kevin Liu
- The Ohio State University College of Medicine, Columbus, Ohio
| | - Ebin Sebastian
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Benjamin Johnson
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Pooja Manchanda Gulati
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Aline Paixao Becker
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Ashley Gray
- The Ohio State University College of Medicine, Columbus, Ohio
| | - Marjolein Geurts
- Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | | | - Linlin Yang
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Jessica L Fleming
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Wei Meng
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Jill S Barnholtz-Sloan
- Department of Population and Quantitative Health Sciences and Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Monica Venere
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Qi-En Wang
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Pierre A Robe
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S Jaharul Haque
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio
| | - Arnab Chakravarti
- Department of Radiation Oncology, Arthur G. James Hospital/Ohio State Comprehensive Cancer Center, Columbus, Ohio.
| |
Collapse
|
30
|
Hao B, Shi A, Li X, Li J, Liu Z, Yuan H. miR-4516 inhibits the apoptosis of RB tumor cells by targeting the PTEN/AKT signaling pathway. Exp Eye Res 2020; 200:108224. [PMID: 32919989 DOI: 10.1016/j.exer.2020.108224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/15/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Bing Hao
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Anjie Shi
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiuhong Li
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jia Li
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhibin Liu
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hongfeng Yuan
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
31
|
Wang Z, Xu R. lncRNA PART1 Promotes Breast Cancer Cell Progression by Directly Targeting miR-4516. Cancer Manag Res 2020; 12:7753-7760. [PMID: 32922076 PMCID: PMC7457826 DOI: 10.2147/cmar.s249296] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Breast cancer is a serious threat to human health. It is meaningful to study the pathogenesis of breast cancer. lncRNAs have been found to play vital roles in numerous biological processes including development, immunology and cancer. Methods qRT-PCR was performed to examine the expressions of PART1 and miR-4516. CCK-8 assay, colony formation assay and transwell assay were used to examine the progression of breast cancer cells. Results In this study, we showed that lncRNA PART1 was highly expressed in breast cancer cells. Knockdown of PART1 induced decreased proliferation, invasion and migration of breast cancer cells. Moreover, we found that PART1 can bind to miR-4516 directly. We also found that inhibition of miR-4516 could rescue the decreased proliferation, migration and invasion of breast cancer cells induced by knockdown of PART1. Discussion lncRNA PART1 and miR-4516 were proven to be involved in the progression of many cancers. However, the roles of lncRNA PART1 and miR-4516 in the regulation of breast cancer remain unknown. Here, we demonstrated that PART1 can bind to miR-4516 to decrease the expression of miR-4516 and promote the development of breast cancer.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Breast Surgery, The First People's Hospital of Jingzhou, Jingzhou 434000, Hubei, People's Republic of China
| | - Ruqing Xu
- Department of Breast Surgery, The First People's Hospital of Jingzhou, Jingzhou 434000, Hubei, People's Republic of China
| |
Collapse
|
32
|
Liu C, Wu Y, Ma J. Interaction of non-coding RNAs and Hippo signaling: Implications for tumorigenesis. Cancer Lett 2020; 493:207-216. [PMID: 32822816 DOI: 10.1016/j.canlet.2020.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Hippo signaling is an evolutionarily conserved pathway that controls organ size by regulating cell proliferation, apoptosis, and stem cell self-renewal by "turning off" or "turning on" the kinase cascade chain reaction to manipulate the expression of downstream genes. Dysregulation of the Hippo pathway contributes to cancer development and metastasis. Emerging evidence has revealed new insights into tumorigenesis through the interplay between the Hippo pathway and non-coding RNAs (ncRNAs), especially microRNA, long non-coding RNA and circular RNA. Here, we reviewed the interactions between the Hippo pathway and ncRNAs and their implication for a variety of tumor-promoting or tumor-repressing effects. These interactions have the potential to serve as cancer biomarkers and therapeutic targets in clinical applications.
Collapse
Affiliation(s)
- Can Liu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yangge Wu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Cancer Research Institute, School of Basic Medical Science, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
33
|
Li Y, Chen X, Xue W, Liang J, Wang L. MiR-874 Inhibits Cell Proliferation, Migration, and Invasion of Glioma Cells and Correlates with Prognosis of Glioma Patients. Neuromolecular Med 2020; 23:247-255. [PMID: 32803522 DOI: 10.1007/s12017-020-08608-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/04/2020] [Indexed: 01/28/2023]
Abstract
Previous studies showed that miR-874 expression was abnormally expressed in many tumors. However, the potential role of miR-874 in glioma remains a mystery. This study aimed to investigate its expression, clinical significance, and cellular function in glioma. A total of 105 glioma patients were enrolled in the present study. The RT-qPCR analysis was used to detect the expression of miR-874 in glioma tissues and cells. The χ2 test was used to analyze the association between miR-874 expression and clinical characteristics of patients. Kaplan-Meier method and multivariate Cox regression assays were used to analyze the prognostic value of miR-874 in glioma. Cell counting kit-8 and Transwell assays were used to explore the alterations in a series of cancer-related phenotypes affected by miR-874, including cell proliferation, migration, and invasion capacities. The expression of miR-874 was significantly downregulated in human glioma tissue specimens and cell lines. Furthermore, the expression of miR-874 was associated with tumor size, KPS, and WHO grade. The decreased expression of miR-874 was associated with shorter overall survival. Then functional assays indicated that upregulation of miR-874 suppressed proliferation, migration, and invasion of glioma cells in vitro. The present study indicated that miR-874 inhibited cell proliferation, migration, and invasion of glioma cells and might be a novel prognostic biomarker and potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Yongjuan Li
- Department of Gastroanorectal Surgery, Anqiu People's Hospital, Weifang, 262100, China
| | - Xiaoyan Chen
- Department of Cardiothoracic Surgery, Anqiu People's Hospital, Weifang, 262100, China
| | - Wei Xue
- Department of Neurosurgery, Anqiu People's Hospital, No. 159 Xuefu Street, Weifang, 262100, China
| | - Junjun Liang
- Department of Neurosurgery, Anqiu People's Hospital, No. 159 Xuefu Street, Weifang, 262100, China
| | - Liang Wang
- Department of Neurosurgery, Anqiu People's Hospital, No. 159 Xuefu Street, Weifang, 262100, China.
| |
Collapse
|
34
|
Yoon YM, Go G, Yun CW, Lim JH, Lee JH, Lee SH. Melatonin Suppresses Renal Cortical Fibrosis by Inhibiting Cytoskeleton Reorganization and Mitochondrial Dysfunction through Regulation of miR-4516. Int J Mol Sci 2020; 21:ijms21155323. [PMID: 32727098 PMCID: PMC7432329 DOI: 10.3390/ijms21155323] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/11/2022] Open
Abstract
Renal fibrosis, a major risk factor for kidney failure, can lead to chronic kidney disease (CKD) and is caused by cytoskeleton reorganization and mitochondrial dysfunction. In this study, we investigated the potential of melatonin treatment to reduce renal fibrosis by recovering the cytoskeleton reorganization and mitochondrial dysfunction. We found that miR-4516 expression was downregulated in the renal cortex of CKD mice and P-cresol-treated TH1 cells. Decreased miR-4516 expression stimulated cytoskeleton reorganization and mitochondrial dysfunction, and induced renal fibrosis. Melatonin treatment suppressed fibrosis by inhibiting cytoskeleton reorganization and restoring mitochondrial function via increased miR-4516 expression. More specifically, melatonin treatment increased miR-4516 expression while decreasing ITGA9 expression, thereby inhibiting cytoskeleton reorganization. In addition, increased expression of miR-4516 by melatonin treatment reduced ROS formation and restored mitochondrial function. These findings suggest that melatonin may be a promising treatment for patients with CKD having renal fibrosis. Moreover, regulation of miR-4516 expression may be a novel strategy for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Yeo Min Yoon
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea; (Y.M.Y.); (C.W.Y.); (J.H.L.); (J.H.L.)
| | - Gyeongyun Go
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea;
| | - Chul Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea; (Y.M.Y.); (C.W.Y.); (J.H.L.); (J.H.L.)
| | - Ji Ho Lim
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea; (Y.M.Y.); (C.W.Y.); (J.H.L.); (J.H.L.)
| | - Jun Hee Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea; (Y.M.Y.); (C.W.Y.); (J.H.L.); (J.H.L.)
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea;
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea; (Y.M.Y.); (C.W.Y.); (J.H.L.); (J.H.L.)
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea;
- Correspondence: ; Tel.: +82-2-709-9029
| |
Collapse
|
35
|
Wang R, Du Y, Shang J, Dang X, Niu G. PTPN14 acts as a candidate tumor suppressor in prostate cancer and inhibits cell proliferation and invasion through modulating LATS1/YAP signaling. Mol Cell Probes 2020; 53:101642. [PMID: 32645410 DOI: 10.1016/j.mcp.2020.101642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/02/2020] [Accepted: 07/05/2020] [Indexed: 12/22/2022]
Abstract
Protein tyrosine phosphatase, non-receptor type 14 (PTPN14) exerts a profound effect in the progression of multiple malignant tumors. However, whether PTPN14 plays a role in prostate cancer has not been well investigated. Herein, we evaluated the function and potential underlying mechanism of PTPN14 in prostate cancer. Decreased PTPN14 expression was detected in prostate cancer, and restoration of PTPN14 expression in prostate cancer cells inhibited the proliferative and invasive potential. Mechanistically, PTPN14 increased the phosphorylation of Yes-associated protein (YAP) by activation of large tumor suppressor 1 (LATS1), an action that resulted in a significant reduction in YAP-mediated transcriptional activity. Inactivation of YAP by its inhibitor markedly abrogated the PTPN14-knockdown-induced promotion effect on prostate cancer cell proliferation and invasion. Notably, PTPN14 up-regulation also exerted a remarkable suppressive impact on tumorigenesis of prostate cancer in vivo. Taken together, the study reveals a tumor-inhibition role of PTPN14 that represses the proliferation and invasion of prostate cancer by down-regulating YAP activation.
Collapse
Affiliation(s)
- Rong Wang
- The Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Yonghao Du
- The Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Jin Shang
- The Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Xiaomin Dang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Gang Niu
- The Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
36
|
Liang G, Duan C, He J, Ma W, Dai X. PTPN14, a target gene of miR-4295, restricts the growth and invasion of osteosarcoma cells through inactivation of YAP1 signalling. Clin Exp Pharmacol Physiol 2020; 47:1301-1310. [PMID: 32141101 DOI: 10.1111/1440-1681.13296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 11/26/2022]
Abstract
Non-receptor tyrosine phosphatase 14 (PTPN14) has emerged as a novel tumour-suppressor in a wide range of human cancer types. However, the role of PTPN14 in osteosarcoma remains undetermined. In the present study, we aimed to explore the expression pattern, biological function, and regulation of PTPN14 in osteosarcoma. Low PTPN14 expression levels were detected in osteosarcoma cells, and PTPN14 overexpression markedly decreased the proliferation, colony formation, and invasive potential of osteosarcoma cells. Bioinformatics analysis predicted PTPN14 as a potential target gene of microRNA-4295 (miR-4295), and this prediction was validated by a dual-luciferase reporter assay. PTPN14 expression was negatively modulated by miR-4295 in osteosarcoma cells. Moreover, PTPN14 expression was inversely correlated with miR-4295 expression in osteosarcoma tissues. Notably, miR-4295 inhibition significantly restricted the proliferation and invasion of osteosarcoma cells. PTPN14 overexpression or miR-4295 inhibition increased the phosphorylation of Yes-associated protein 1 (YAP1) and impeded YAP1 nuclear translocation, leading to inhibition of YAP1-mediated transcriptional activity. However, the miR-4925-inhibition-mediated antitumour effect was partially reversed by PTPN14 knockdown. Overall, these results demonstrate that PTPN14 is a miR-4295 target gene and it exerts a tumour-suppressive function in osteosarcoma cells via inactivation of YAP1. Our study uncovers a miR-4295-PTPN14-YAP1 signalling pathway that may play a crucial role in the progression of osteosarcoma.
Collapse
Affiliation(s)
- Gaofeng Liang
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Orthopaedics, 521 Hospital of Norinco Group, Xi'an, China
| | - Chaopeng Duan
- Department of Orthopaedics, 521 Hospital of Norinco Group, Xi'an, China
| | - June He
- 521 Hospital of Norinco Group, Xi'an, China
| | - Wei Ma
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xing Dai
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
37
|
Lu F, Li C, Sun Y, Jia T, Li N, Li H. Upregulation of miR-1825 inhibits the progression of glioblastoma by suppressing CDK14 though Wnt/β-catenin signaling pathway. World J Surg Oncol 2020; 18:147. [PMID: 32605563 PMCID: PMC7325653 DOI: 10.1186/s12957-020-01927-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/18/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mounting evidences displayed that miRNAs play crucial roles in tumor initiation and development. However, the regulation and relevant mechanism of miR-1825 in glioblastoma (GBM) remain unclear. METHODS qRT-PCR was used to detect miR-1825 and CDK14 mRNA expression. Western blot was applied for testing protein levels (VEGF, E-cadherin, N-cadherin, vimentin, β-catenin, c-myc, p-c-Jun). MTT and transwell assays were used for detecting GBM cell progression, including cell viability, migration, and invasion. RESULTS The results showed that miR-1825 was decreased in GBM tissue specimens by qRT-PCR and it was confirmed as a prognostic marker of GBM by Kaplan-Meier survival analysis. Moreover, we also found that miR-1825 upregulation suppressed GBM cell viability, tumor growth, invasion, and migration. Furthermore, CDK14 was first identified as the direct target of miR-1825 by Luciferase reporter assay. CDK14 acted as an oncogene in GBM development by immunohistochemistry. In addition, Western blot analysis demonstrated that miR-1825 regulated Wnt/β-catenin signaling pathway in GBM development. CONCLUSION In conclusion, miR-1825 upregulation suppressed GBM progression by targeting CDK14 through Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Fengqin Lu
- Department of Geriatrics, Jinan Zhangqiu District Hospital of TCM, Jinan, 250200, China
| | - Chunhong Li
- Department of Public Health, Jinan Zhangqiu District Hospital of TCM, Jinan, 250200, China
| | - Yuping Sun
- Department of Neurology, the People's Hospital of Zhangqiu Area, Jinan, 250200, China
| | - Ting Jia
- Department of Gynaecology, the People's Hospital of Zhangqiu Area, Jinan, 250200, China
| | - Na Li
- Department of Radiology, the People's Hospital of Zhangqiu Area, Jinan, 250200, China
| | - Haiyan Li
- Department of Neurology, Qingdao Central Hospital, Qingdao University, No.127 Siliunan Road, Qingdao, 266042, China.
| |
Collapse
|
38
|
Li W, Pan T, Jiang W, Zhao H. HCG18/miR-34a-5p/HMMR axis accelerates the progression of lung adenocarcinoma. Biomed Pharmacother 2020; 129:110217. [PMID: 32559619 DOI: 10.1016/j.biopha.2020.110217] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
As the most common subtype of lung cancer, lung adenocarcinoma (LUAD) is the frequently occurred cancers in human. Therefore, thorough investigation is necessary for understanding the progression of LUAD. HMMR has functioned as a regulator in some cancers, whereas its biological role still needs to be investigated in LUAD. By bioinformatics analysis, we found that HMMR was highly expressed in LUAD tissues and associated with patients' poor prognosis. Further, qRT-PCR demonstrated that HMMR was up-regulated in LUAD tissues and cells. Loss-of-function assays manifested that HMMR knockdown refrained cell proliferation, migration and invasion and enhanced cell apoptosis in LUAD. Later, HMMR was identified as a target gene of miR-34a-5p, which expressed at a low level in LUAD cell and played an anti-oncogenic role in LUAD. Simultaneously, we discovered that miR-34a-5p could directly bind to HCG18. Subsequent assays revealed that HCG18 mediated HMMR expression by sequestering miR-34a-5p. At last, rescue assays proved the carcinogenic role of HCG18/miR-34a-5p/HMMR axis in LUAD cells growth. Importantly, HCG18 was found to facilitate tumor growth in LUAD. Conclusively, HCG18 acted an oncogene in LUAD and enhanced LUAD progression by targeting miR-34a-5p/HMMR axis.
Collapse
Affiliation(s)
- Wei Li
- Department of Thoracic Surgery, The People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, PR China
| | - Tinghong Pan
- The Cardiothoracic Surgery of Weifang Yidu Central Hospital, Weifang, 262500, Shandong, PR China
| | - Wei Jiang
- The Pediatrics of Weifang Yidu Central Hospital, Weifang, 262500, Shandong, PR China
| | - Hongying Zhao
- Department of Medical Oncology, Xuzhou Cancer Hospital, Xuzhou Hospital Affiliated to Jiangsu University, Xuzhou, 221000, Jiangsu, PR China.
| |
Collapse
|
39
|
Chen S, Xu M, Zhao J, Shen J, Li J, Liu Y, Cao G, Ma J, He W, Chen X, Shan T. MicroRNA-4516 suppresses pancreatic cancer development via negatively regulating orthodenticle homeobox 1. Int J Biol Sci 2020; 16:2159-2169. [PMID: 32549762 PMCID: PMC7294951 DOI: 10.7150/ijbs.45933] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer remains one of the most lethal human cancers without efficient therapeutic strategy. MicoRNAs (miRNAs) are a group of small non-coding RNAs involved in multiple biological processes including tumor development and progression. In this study, we investigated the expression and function of miR-4516 in pancreatic cancer. MiR-4516 was low-expressed in pancreatic cancer tissues and cell lines. Overexpression of miR-4516 inhibited pancreatic cancer cell proliferation, migration and invasion, while promoted cell apoptosis in vitro. Further, overexpression of miR-4516 suppressed xenograft pancreatic tumor growth in vivo. Bioinformatics analysis was performed and miR-4516 was predicted to negatively regulate orthodenticle homeobox 1 (OTX1) expression by binding to its 3'-UTR. Consistently, OTX1 was highly expressed in pancreatic cancer tissues and cell lines. Knockdown of OTX1 expression suppressed pancreatic cancer cell migration and invasion, with down-regulated MMP2 and MMP9 expression. Moreover, we demonstrated that miR-4516 regulated pancreatic cancer cell growth, migration, invasion and apoptosis via targeting OTX1. Overexpression of OTX1 could partially abrogate the inhibitory effect of miR-4516. Taken together, we conclude that miR-4516 could function as a tumor suppressor via targeting OTX1. These findings suggest that miR-4516/OTX1 axis might be a novel therapeutic target for miRNA-based therapy for pancreatic cancer patients.
Collapse
Affiliation(s)
- Shuo Chen
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| | - Meng Xu
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| | - Jing Zhao
- School of Science, Xi'an Jiaotong University, PR China
| | - Jiaqi Shen
- School of Life Science, Xiamen University, PR China
| | - Junhui Li
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| | - Yang Liu
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| | - Gang Cao
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| | - Jiancang Ma
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| | - Weizhou He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| | - Xi Chen
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| | - Tao Shan
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, PR China
| |
Collapse
|
40
|
Yoon YM, Lee JH, Song KH, Noh H, Lee SH. Melatonin-stimulated exosomes enhance the regenerative potential of chronic kidney disease-derived mesenchymal stem/stromal cells via cellular prion proteins. J Pineal Res 2020; 68:e12632. [PMID: 31989677 DOI: 10.1111/jpi.12632] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/07/2020] [Accepted: 01/22/2020] [Indexed: 12/21/2022]
Abstract
Chronic kidney disease (CKD) is caused by dysfunctional kidneys, which result in complications like cardiovascular diseases. Chronic kidney disease-induced pathophysiological conditions decrease efficacy of autologous mesenchymal stem/stromal cell (MSC)-based therapy by reducing MSC functionality. To enhance therapeutic potential in patients with CKD, we isolated exosomes derived from melatonin-treated healthy MSCs (MT exosomes) and assessed the biological functions of MT exosome-treated MSCs isolated from patients with CKD (CKD-MSCs). Treatment with melatonin increased the expression of cellular prion protein (PrPC ) in exosomes isolated from MSCs through the upregulation of miR-4516. Treatment with MT exosomes protected mitochondrial function, cellular senescence, and proliferative potential of CKD-MSCs. MT exosomes significantly increased the level of angiogenesis-associated proteins in CKD-MSCs. In a murine hindlimb ischemia model with CKD, MT exosome-treated CKD-MSCs improved functional recovery and vessel repair. These findings elucidate the regenerative potential of MT exosome-treated CKD-MSCs via the miR-4516-PrPC signaling axis. This study suggests that the treatment of CKD-MSCs with MT exosomes might be a powerful strategy for developing autologous MSC-based therapeutics for patients with CKD. Furthermore, miR-4516 and PrPC could be key molecules for enhancing the regenerative potential of MSCs in ischemic diseases.
Collapse
Affiliation(s)
- Yeo Min Yoon
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Jun Hee Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
- Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Keon-Hyoung Song
- Department of Pharmaceutical Engineering, College of Medical Science, Soonchunhyang University, Asan, Korea
| | - Hyunjin Noh
- Department of Internal Medicine, Soonchunhyang University, Seoul, Korea
- Hyonam Kidney Laboratory, Soonchunhyang University, Seoul, Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
- Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
41
|
Jin XH, Lu S, Wang AF. Expression and clinical significance of miR-4516 and miR-21-5p in serum of patients with colorectal cancer. BMC Cancer 2020; 20:241. [PMID: 32293319 PMCID: PMC7087363 DOI: 10.1186/s12885-020-06715-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 03/05/2020] [Indexed: 12/24/2022] Open
Abstract
Background This study sought to detect the expression and clinical significance of miR-4516 and miR-21-5p in serum of patients with colorectal cancer. Methods Bioinformatics methods were used to analyze the expression patterns of miR-4516 and miR-21-5p in colorectal cancer. A total of 80 patients with colorectal cancer, 65 patients with benign colorectal tumors and 50 healthy persons were selected. qRT-PCR was performed to detect the expression levels of serum miR-4516 and miR-21-5p before and after operation or postoperative recurrence. The correlation of miR-4516 and miR-21-5p expression levels with the clinical characteristics and prognosis of colorectal cancer was analyzed, and that with the patient’s survival was further examined by Kaplan-Meier analysis. Results MiR-4516 was poorly expressed in colorectal cancer in the preoperative group, and miR-21-5p was highly expressed. While in the postoperative group, miR-4516 was up-regulated, and miR-21-5p was down-regulated. The low expression of miR-4516 was shown to be related to TNM staging, invasion degree, lymph node metastasis and distant metastasis of the patients. Whereas the high expression of miR-21-5p was proved to be correlated with TNM staging and lymph node metastasis. Kaplan-Meier survival analysis showed that high expression of miR-4516 or low expression of miR-21-5p could contribute to better overall survival. Conclusion Low miR-4516 or high miR-21-5p could be used as an independent risk factor for prognosis of colorectal cancer.
Collapse
Affiliation(s)
- Xi-Han Jin
- Department of Colorectal Surgery, Jinhua Hospital of Zhejiang University, Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Sen Lu
- Department of Colorectal Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Ai-Fen Wang
- Department of Oncology, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, No. 1882 South Zhonghuan Road, Nanhu District, Jiaxing, 314001, China.
| |
Collapse
|
42
|
Wu B, Liu G, Jin Y, Yang T, Zhang D, Ding L, Zhou F, Pan Y, Wei Y. miR-15b-5p Promotes Growth and Metastasis in Breast Cancer by Targeting HPSE2. Front Oncol 2020; 10:108. [PMID: 32175269 PMCID: PMC7054484 DOI: 10.3389/fonc.2020.00108] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/21/2020] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) can participate in many behaviors of various tumors. Prior studies have reported that miR-15b-5p in different tumors can either promote or inhibit tumor progression. In breast cancer, the role of miR-15b-5p is unclear. The main objective of this paper is to explore miR-15b-5p effects and their mechanisms in breast cancer using both in vitro and in vivo experiments. This study showed that miR-15b-5p expression was upregulated in breast cancer compared with normal breast tissue and was positively correlated with poor overall survival in patients. Knockdown of miR-15b-5p in MCF-7 and MD-MBA-231 breast cancer cells restrained cell growth and invasiveness and induced apoptosis, whereas overexpression of miR-15b-5p achieved the opposite effects. We next revealed a negative correlation between miR-15b-5p and heparanase-2 (HPSE2) expression in breast cancer. Knockdown of miR-15b-5p significantly increased HPSE2 expression at both mRNA and protein levels in breast cancer cells in vitro. The underlying mechanisms of miR-15-5p in breast cancer were investigated using luciferase activity reporter assay and rescue experiments. In addition, miR-15b-5p knockdown significantly inhibited tumor growth in a xenograft model in mice. In summary, we showed that miR-15b-5p promotes breast cancer cell proliferation, migration, and invasion by directly targeting HPSE2. Accordingly, miR-15b-5p may serve both as a tool for prognosis and as a target for therapy of breast cancer patients.
Collapse
Affiliation(s)
- Balu Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yanxia Jin
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Tian Yang
- Department of Clinical Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Dongdong Zhang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Lu Ding
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yongchang Wei
- Hubei Key Laboratory of Tumor Biological Behaviors, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
43
|
Chen X, Zhang X, Sun S, Zhu M. MicroRNA‑432 inhibits the aggressiveness of glioblastoma multiforme by directly targeting IGF‑1R. Int J Mol Med 2019; 45:597-606. [PMID: 31894251 DOI: 10.3892/ijmm.2019.4429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 11/04/2019] [Indexed: 11/05/2022] Open
Abstract
MicroRNA‑432 (miR‑432) has been studied in multiple tumors, but the expression status, biological functions and the mechanism of action of miR‑432 in glioblastoma multiforme (GBM) are yet to be elucidated. In the present study, miR‑432 expression in GBM was determined and its clinical significance was evaluated among patients with GBM. The effects on the malignancy of GBM in vitro and in vivo were examined in detail and the interactions between miR‑432 and insulin‑like growth factor 1 receptor (IGF‑1R) mRNA were then explored. miR‑432 expression in GBM tissue samples and cell lines was measured by reverse transcription‑quantitative (RT‑q)PCR. GBM cell proliferation, apoptosis, migration and invasion in vitro and tumor growth in vivo were evaluated by a Cell Counting Kit‑8 assay, flow‑cytometric analysis, Transwell migration and invasion assays, and a tumor xenograft experiment, respectively. Bioinformatic analysis followed by a luciferase reporter assay, RT‑qPCR and western blotting was applied to demonstrate that IGF‑1R is a direct target gene of miR‑432 in GBM cells. It was found that miR‑432 is downregulated in GBM tumors and cell lines. miR‑432 under expression obviously correlated with the Karnofsky Performance Status score and shorter overall survival among patients with GBM. Exogenous miR‑432 expression significantly reduced proliferation and induced apoptosis of GBM cells. In addition, miR‑432 overexpression impaired the migratory and invasive abilities of GBM cells in vitro and decreased their tumor growth in vivo. Furthermore, IGF‑1R was validated as a direct target gene of miR‑432 in GBM cells. IGF‑1R knockdown imitated the tumor‑suppressive actions of miR‑432 overexpression in GBM cells. Rescue experiments proved IGF‑1R downregulation to be essential for the effects of miR‑432 on GBM cells. The results of the present study revealed a tumor‑suppressive role of the miR‑432‑IGF‑1R axis in GBM cells and this axis may have implications for GBM therapy.
Collapse
Affiliation(s)
- Xudong Chen
- Department of Neurosurgery, Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, P.R. China
| | - Xufei Zhang
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Shunjin Sun
- Department of Neurosurgery, Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, P.R. China
| | - Meixiao Zhu
- Department of TCM Pharmacy, Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, P.R. China
| |
Collapse
|
44
|
Po'uha ST, Le Grand M, Brandl MB, Gifford AJ, Goodall GJ, Khew-Goodall Y, Kavallaris M. Stathmin levels alter PTPN14 expression and impact neuroblastoma cell migration. Br J Cancer 2019; 122:434-444. [PMID: 31806880 PMCID: PMC7000740 DOI: 10.1038/s41416-019-0669-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/15/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Stathmin mediates cell migration and invasion in vitro, and metastasis in vivo. To investigate stathmin's role on the metastatic process, we performed integrated mRNA-miRNA expression analysis to identify pathways regulated by stathmin. METHODS MiRNA and gene arrays followed by miRNA-target-gene integration were performed on stathmin-depleted neuroblastoma cells (CtrlshRNA vs. Stmn Seq2shRNA). The expression of the predicted target PTPN14 was evaluated by RT-qPCR, western blot and immunohistochemistry. Gene-silencing technology was used to assess the role of PTPN14 on proliferation, migration, invasion and signalling pathway. RESULTS Stathmin levels modulated the expression of genes and miRNA in neuroblastoma cells, leading to a deregulation of migration and invasion pathways. Consistent with gene array data, PTPN14 mRNA and protein expression were downregulated in stathmin- depleted neuroblastoma cells and xenografts. In two independent neuroblastoma cells, suppression of PTPN14 expression led to an increase in cell migration and invasion. PTPN14 and stathmin expression did not act in a feedback regulatory loop in PTPN14- depleted cells, suggesting a complex interplay of signalling pathways. The effect of PTPN14 on YAP pathway activation was cell-type dependent. CONCLUSIONS Our findings demonstrate that stathmin levels can regulate PTPN14 expression, which can modulate neuroblastoma cell migration and invasion.
Collapse
Affiliation(s)
- Sela T Po'uha
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensignton, NSW, 2052, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia.,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, NSW, 2052, Australia
| | - Marion Le Grand
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensignton, NSW, 2052, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia.,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, NSW, 2052, Australia
| | - Miriam B Brandl
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensignton, NSW, 2052, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia.,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, NSW, 2052, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensignton, NSW, 2052, Australia.,Department of Anatomical Pathology (SEALS), Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | - Gregory J Goodall
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Discipline of Medicine and Dept of Molecular and Biomedical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Discipline of Medicine and Dept of Molecular and Biomedical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensignton, NSW, 2052, Australia. .,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia. .,School of Women's and Children's Health, Faculty of Medicine, UNSW Sydney, NSW, 2052, Australia.
| |
Collapse
|
45
|
Liu C, Ma T, Jiang T, Jia G, Yang C, Peng Y, Qian Y, Wang R, Wang S. Abnormal increase of miR-4262 promotes cell proliferation and migration by targeting large tumor suppressor 1 in gliomas. Pathol Res Pract 2019; 216:152778. [PMID: 31831299 DOI: 10.1016/j.prp.2019.152778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/20/2019] [Accepted: 12/01/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND miRNA was recently detected as tumor suppressor or inducer in various cancers including gliomas. Due to the abnormal expression of miR-4262 in glioma cancer, we supposed that miR-4262 made efforts in proliferation and migration in glioma cancer. METHODS CCK-8, Transwell migration Assay and Wound-healing assay were appraisal assays for cell proliferation and migration. qRT-PCR and western blot were performed to test the expression of miR-4262, MMP2, MMP13 and LATS1 in glioma cancers tissues and cancer cells. The targeting detection between miR-4262 and LATS1 was detected by luciferase reporter assay. RESULTS miR-4262 expression was dramatically higher in glioma tumor tissues than in para-tumor control. Inhibition of miR-4262 in glioma cancer cells prominently inhibited cell proliferation and migration. Mechanically, downregulation of miR-4262 inhibited expression of matrix metalloproteinase (MMP) -2, -13. In addition, miR-4262 directly and negatively modulated expression of large tumor suppressor 1 (LATS1). Moreover, we discovered that overexpression of LATS1 could reverse the effects of miR-4262 on cell proliferation and migration, as well as the production of MMP-2, -13. CONCLUSIONS In glioma cancer, miR-4262 regulated cell proliferation and migration mediated by LATS1. This indicated that miR-4262 is a tumor inducer in glioma cancer and may be a feasible target for glioma therapy.
Collapse
Affiliation(s)
- Chunbo Liu
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China
| | - Tao Ma
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China
| | - Tianwei Jiang
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China
| | - Geng Jia
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China
| | - Changchun Yang
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China
| | - Ya Peng
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China
| | - Yitao Qian
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China
| | - Rong Wang
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China
| | - Suinuan Wang
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou First Peoples' Hospital, Changzhou City, 213003, Jiangsu Province, China.
| |
Collapse
|
46
|
The stromal loss of miR-4516 promotes the FOSL1-dependent proliferation and malignancy of triple negative breast cancer. Cancer Lett 2019; 469:256-265. [PMID: 31672492 DOI: 10.1016/j.canlet.2019.10.039] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/24/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022]
Abstract
Stroma-derived exosomal microRNA (exomiR) contributes to tumor progression, however, which remains poorly understood. In our study, we analyzed exomiRs from the cancer-associated fibroblast (CAF) and normal fibroblast (NF) isolated from an invasive ductal carcinoma (IDC) patient and found that the level of microRNA (miR)-4516 was approximately 5-fold lower in CAF-derived exosomes than NF-derived ones. In gene annotation analysis, miR-4516 target genes were mainly associated with the regulation of proliferation. miR-4516 overexpression or mimic treatment suppressed the proliferation of breast cancer cells, especially triple negative breast cancer (TNBC) cells. Among miR-4516 targets, FOSL1 was overexpressed in TNBC cells compared to non-TNBC cells and promoted tumor proliferation. The expression of miR-4516 and FOSL1 was reversely correlated in breast cancer patient tissues. Particularly, TNBC patients with high FOSL1 expression showed a significant poorer survival than those with low FOSL1 expression. Our results show that the loss of miR-4516 from CAF-derived exosomes is associated with FOSL1-dependent TNBC progression and suggest that miR-4516 can be used as an anti-cancer drug for TNBC.
Collapse
|
47
|
Liu Y, Li X, Zhang Y, Wang H, Rong X, Peng J, He L, Peng Y. An miR-340-5p-macrophage feedback loop modulates the progression and tumor microenvironment of glioblastoma multiforme. Oncogene 2019; 38:7399-7415. [PMID: 31427735 DOI: 10.1038/s41388-019-0952-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 06/14/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) have been shown to be involved in the progression and tumor microenvironment of glioblastoma multiforme (GBM). Our previous research has indicated that miR-340-5p has an antitumor effect in vitro. However, the role of miR-340-5p in GBM has not been fully elucidated. Here, we show that downregulation of miR-340-5p in GBM is correlated with tumor size, recurrence, and poor survival. Moreover, we found that miR-340-5p levels are correlated with the density of tumor-associated macrophages (TAMs) and M2-polarized TAMs in GBM. Biofunctional investigations revealed that downregulation of miR-340-5p promoted TAM recruitment and M2-TAMs polarization in vitro and in vivo. In addition, we found that upregulation of miR-340-5p inhibited tumor growth and was associated with good prognosis in vivo. Through gene expression profiles and bioinformatics analysis, we showed that miR-340-5p directly targets POSTN, which recruited TAMs through integrin αvβ3. Downregulation of miR-340-5p in GBM did not induce the differentiation of TAMs into polarized M2 cells but was able to promote the M2 polarization of TAMs through directly targeting LTBP-1. Furthermore, we found that M2-TAMs promoted tumorigenesis and were associated with a poor prognosis in vivo. In an in vitro study, we demonstrated that M2-TAMs inhibited miR-340-5p expression in GBM cells by upregulation of TGFβ-1, which increased HMGA-2 expression in GBM. A ChIP assay confirmed that HMGA-2 transcriptionally suppressed miR-340-5p expression. Patients with low-miR-340-5p expression, high CD163, high POSTN, high LIBP1 levels, and high HMGA-2 had a poor prognosis with shorter overall survival, confirming data from the TCGA database. These findings suggest that an miR-340-5p-macrophage feedback loop modulates the progression and tumor microenvironment of GBM and may represent a prognostic biomarker and therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Yunyun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Xiaoyu Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Yuanpei Zhang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Hongxuan Wang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Xiongming Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Jialing Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Lei He
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Ying Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China.
| |
Collapse
|
48
|
Zhang C, Wang L, Yang J, Fu Y, Li H, Xie L, Cui Y. MicroRNA-33a-5p suppresses esophageal squamous cell carcinoma progression via regulation of lncRNA DANCR and ZEB1. Eur J Pharmacol 2019; 861:172590. [PMID: 31401160 DOI: 10.1016/j.ejphar.2019.172590] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/11/2019] [Accepted: 08/07/2019] [Indexed: 12/28/2022]
Abstract
Increasing evidence displays that microRNAs (miRNAs) participate in the development of various human malignancies, including esophageal squamous cell carcinoma (ESCC). MicroRNA-33a-5p (miR-33a-5p) has recently been reported to function as a tumor suppressor in many human cancers. However, the expression and role of miR-33a-5p in ESCC remains largely unknown. Herein, we discovered that miR-33a-5p was down-regulated in both ESCC tissues and cell lines, compared with their normal counterparts, and decreased expression of miR-33a-5p was found to be closely associated with poor patient prognosis of ESCC. Moreover, functional experiments revealed that up-regulation of miR-33a-5p inhibited cell proliferation and metastasis of ESCC cells. In addition, the expression level of miR-33a-5p was found to be negatively correlated with the long non-coding RNA (lncRNA) differentiation antagonizing non-protein coding RNA (DANCR), in both ESCC tissues and cell lines. Furthermore, zinc-finger-enhancer binding protein 1 (ZEB1) was predicted and confirmed to be a direct target of miR-33a-5p in ESCC. Further mechanistic investigation indicated that DANCR may function as a competing endogenous RNA (ceRNA) to sponge miR-33a-5p, and thereby up-regulate ZEB1 expression in ESCC. This study highlights the functions of miR-33a-5p in the progression of ESCC and suggests that the DANCR/miR-33a-5p/ZEB1 axis may be a potential prognostic and therapeutic target.
Collapse
Affiliation(s)
- Chunyan Zhang
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Lingxu Wang
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Juanjuan Yang
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Yin Fu
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Hongzhi Li
- Department of Infedtious Diseases, The Sixth People's Hospital of Zhengzhou, Zhengzhou, 450007, China
| | - Linsen Xie
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China.
| | - Yuanbo Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
49
|
Zhang L, Wang H, Xu M, Chen F, Li W, Hu H, Yuan Q, Su Y, Liu X, Wuri J, Yan T. Long noncoding RNA HAS2-AS1 promotes tumor progression in glioblastoma via functioning as a competing endogenous RNA. J Cell Biochem 2019; 121:661-671. [PMID: 31385362 DOI: 10.1002/jcb.29313] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/15/2019] [Indexed: 12/25/2022]
Abstract
Glioblastoma multiforme (GBM) is a refractory tumor with poor prognosis and requires more effective treatment regimens. It has been confirmed that long noncoding RNAs (lncRNAs) substantially regulate various human disease including GBM. However, the biological roles and its underlying molecular mechanisms still need to be further investigated. In this study, the biological function and potential molecular mechanism of lncHAS2-AS1 in GBM were explored. It was discovered that HAS2-AS1 was elevated in glioma tissues and correlated with the prognosis of patients with glioma. Reduction of HAS2-AS1 suppressed the migration and invasion in vitro and in vivo. The transcription factor STAT1 could raise HAS2-AS1 by binding to its promoter region. Besides, HAS2-AS1 could adjust PRPS1 via sponging miR-608 in a direct manner. On the whole, the results of this study evidence that HAS2-AS1 is an oncogene and a potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Liqun Zhang
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Hong Wang
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Meijie Xu
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurology, Xiqing Hospital, Tianjin, China
| | - Fangyu Chen
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurology, Langfang Hospital of Traditional Chinese Medicine, Langfang, Hebei Province, China
| | - Wenkui Li
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Haotian Hu
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Quan Yuan
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Yue Su
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Xiaoxuan Liu
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Jimusi Wuri
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Tao Yan
- Department of Neurology, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| |
Collapse
|