201
|
Zhao X, Jin Y, Li L, Xu L, Tang Z, Qi Y, Yin L, Peng J. MicroRNA-128-3p aggravates doxorubicin-induced liver injury by promoting oxidative stress via targeting Sirtuin-1. Pharmacol Res 2019; 146:104276. [PMID: 31112750 DOI: 10.1016/j.phrs.2019.104276] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/31/2022]
Abstract
As one classic anticancer drug, clinical application of Doxorubicin (Dox) is limited due to its side effects. In our previous work, we have investigated the drug targets to treat Dox-induced cardiotoxicity, hepatotoxicity and nephrotoxicity. In this paper, the mechanisms and new drug-target associated with Dox-induced hepatotoxicity were explored. The results showed that Dox markedly inhibited cell viability and cellular respiration, induced cell morphologic change and increased ROS level. Moreover, Dox increased ALT and AST levels, caused pathological damage, increased MDA level and decreased SOD level in mice. Mechanism investigation showed that Dox markedly up-regulated the expression level of miR-128-3p, down-regulated Sirt1 expression level and affected the protein levels of Nrf2, Keap1, Sirt3, NQO1 and HO-1 to cause oxidative stress in liver. Furthermore, double-luciferase reporter assay, and co-transfection test showed that miR-128-3p directly targeted Sirt1. In addition, miR-128-3p mimics in AML-12 cells enhanced Dox-induced oxidative damage via inhibiting cellular respiration, increasing ROS level and mitochondrial superoxide formation. The protein levels of Sirt1, Nrf2, Sirt3, NQO1 and HO-1 in miR-128-3p mimic + Dox group were decreased compared with Dox group. Transfection of miR-128-3p inhibitor weakened Dox-induced oxidative damage via increasing cellular respiration, suppressing cellular ROS level and mitochondrial superoxide formation. The protein levels of Sirt1, Nrf2, Sirt3, NQO1 and HO-1 in miR-128-3p inhibitor + Dox group were increased compared with Dox group. In mice, Dox-induced liver damage was deteriorated by miR-128-3p agomir via increasing the levels of ALT, AST, MDA, and down-regulating the protein levels of Sirt1, Nrf2, Sirt3, NQO1 and HO-1. While, miR-128-3p antagomir alleviated liver injury via decreasing the levels of ALT, AST, MDA, and up-regulating the protein levels of Sirt1, Nrf2, Sirt3, NQO1 and HO-1. Our data showed that miRNA-128-3p aggravated Dox-induced liver injury by promoting oxidative stress via targeting Sirt1, which should be considered as one new drug target to treat Dox-induced liver injury.
Collapse
Affiliation(s)
- Xuerong Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yue Jin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lei Li
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Zeyao Tang
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, China; National-Local Joint Engineering Research Center for Drug Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China.
| |
Collapse
|
202
|
Moghaddam T, Neshati Z. Role of microRNAs in osteogenesis of stem cells. J Cell Biochem 2019; 120:14136-14155. [PMID: 31069839 DOI: 10.1002/jcb.28689] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/21/2022]
Abstract
Osteogenic differentiation is a controlled developmental process in which external and internal factors including cytokines, growth factors, transcription factors (TFs), signaling pathways and microRNAs (miRNAs) play important roles. Various stimulatory and inhibitory TFs contribute to osteogenic differentiation and are responsible for bone development. In addition, cross-talk between several complex signaling pathways regulates the osteogenic differentiation of some stem cells. Although much is known about regulatory genes and signaling pathways in osteogenesis, the role of miRNAs in osteogenic differentiation still needs to be explored. miRNAs are small, approximately 22 nucleotides, single-stranded nonprotein coding RNAs which are abundant in many mammalian cell types. They paly significant regulated roles in various biological processes and serve as promising biomarkers for disease states. Recently, emerging evidence have shown that miRNAs are the key regulators of osteogenesis of stem cells. They may endogenously regulate osteogenic differentiation of stem cells through direct targeting of positive or negative directors of osteogenesis and depending on the target result in the promotion or inhibition of osteogenic differentiation. This review aims to provide a general overview of miRNAs participating in osteogenic differentiation of stem cells and explain their regulatory effect based on the genes targeted with these miRNAs.
Collapse
Affiliation(s)
- Tayebe Moghaddam
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Zeinab Neshati
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
203
|
Xie X, Wu H, Li M, Chen X, Xu X, Ni W, Lu C, Ni R, Bao B, Xiao M. Progress in the application of exosomes as therapeutic vectors in tumor-targeted therapy. Cytotherapy 2019; 21:509-524. [DOI: 10.1016/j.jcyt.2019.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/05/2019] [Accepted: 01/08/2019] [Indexed: 12/13/2022]
|
204
|
Modulation of ADAR mRNA expression in patients with congenital heart defects. PLoS One 2019; 14:e0200968. [PMID: 31039163 PMCID: PMC6490900 DOI: 10.1371/journal.pone.0200968] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 04/05/2019] [Indexed: 12/26/2022] Open
Abstract
Adenosine (A) to inosine (I) RNA editing is a hydrolytic deamination reaction catalyzed by the adenosine deaminase (ADAR) enzyme acting on double-stranded RNA. This posttranscriptional process diversifies a plethora of transcripts, including coding and noncoding RNAs. Interestingly, few studies have been carried out to determine the role of RNA editing in vascular disease. The aim of this study was to determine the potential role of ADARs in congenital heart disease. Strong downregulation of ADAR2 and increase in ADAR1 expression was observed in blood samples from congenital heart disease (CHD) patients. The decrease in expression of ADAR2 was in line with its downregulation in ventricular tissues of dilated cardiomyopathy patients. To further decipher the plausible regulatory pathway of ADAR2 with respect to heart physiology, miRNA profiling of ADAR2 was performed on tissues from ADAR2-/- mouse hearts. Downregulation of miRNAs (miR-29b, miR-405, and miR-19) associated with cardiomyopathy and cardiac fibrosis was observed. Moreover, the upregulation of miR-29b targets COL1A2 and IGF1, indicated that ADAR2 might be involved in cardiac myopathy. The ADAR2 target vascular development associated protein-coding gene filamin B (FLNB) was selected. The editing levels of FLNB were dramatically reduced in ADAR2-/- mice; however, no observable changes in FLNB expression were noted in ADAR2-/- mice compared to wild-type mice. This study proposes that sufficient ADAR2 enzyme activity might play a vital role in preventing cardiovascular defects.
Collapse
|
205
|
Pinchi E, Frati A, Cantatore S, D'Errico S, Russa RL, Maiese A, Palmieri M, Pesce A, Viola RV, Frati P, Fineschi V. Acute Spinal Cord Injury: A Systematic Review Investigating miRNA Families Involved. Int J Mol Sci 2019; 20:E1841. [PMID: 31013946 PMCID: PMC6515063 DOI: 10.3390/ijms20081841] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/06/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
Acute traumatic spinal cord injury (SCI) involves primary and secondary injury mechanisms. The primary mechanism is related to the initial traumatic damage caused by the damaging impact and this damage is irreversible. Secondary mechanisms, which begin as early as a few minutes after the initial trauma, include processes such as spinal cord ischemia, cellular excitotoxicity, ionic dysregulation, and free radical-mediated peroxidation. SCI is featured by different forms of injury, investigating the pathology and degree of clinical diagnosis and treatment strategies, the animal models that have allowed us to better understand this entity and, finally, the role of new diagnostic and prognostic tools such as miRNA could improve our ability to manage this pathological entity. Autopsy could benefit from improvements in miRNA research: the specificity and sensitivity of miRNAs could help physicians in determining the cause of death, besides the time of death.
Collapse
Affiliation(s)
- Enrica Pinchi
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
| | - Alessandro Frati
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
- NESMOS Department ⁻ Neurosurgery Division, "Sapienza" University of Roma, 00189 Rome, Italy.
| | - Santina Cantatore
- Forensic Pathology Institute, University of Foggia, 71122 Foggia, Italy.
| | - Stefano D'Errico
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- Legal Medicine Division, Ospedale Sant'Andrea, 00189 Rome, Italy.
| | - Raffaele La Russa
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
| | - Aniello Maiese
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
| | - Mauro Palmieri
- NESMOS Department ⁻ Neurosurgery Division, "Sapienza" University of Roma, 00189 Rome, Italy.
| | - Alessandro Pesce
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
- NESMOS Department ⁻ Neurosurgery Division, "Sapienza" University of Roma, 00189 Rome, Italy.
| | | | - Paola Frati
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
| | - Vittorio Fineschi
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
| |
Collapse
|
206
|
Smith AM, Dykeman CA, King BL, Yin VP. Modulation of TNFα Activity by the microRNA Let-7 Coordinates Zebrafish Heart Regeneration. iScience 2019; 15:1-15. [PMID: 31026665 PMCID: PMC6482333 DOI: 10.1016/j.isci.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/24/2022] Open
Abstract
The adult zebrafish is capable of regenerating heart muscle, resolving collagen tissue, and fully restoring heart function throughout its life. In this study, we show that the highly upregulated, epicardium-enriched microRNA let-7i functions in wound closure and cardiomyocyte proliferation. RNA sequencing experiments identified upregulated expression of members of the tumor necrosis factor (TNF) signaling pathway in the absence of let-7. Importantly, co-suppression of TNF and let-7 activity rescued epicardium migration and cardiomyocyte proliferation defects induced by depletion of let-7 alone. Sensitizing animals to low levels of TNF activity before injury culminated in repressed cardiomyocyte proliferation and wound closure defects, suggesting that levels of inflammation at the onset of injury are critical for heart regeneration. Our studies indicate that injury-induced reduction in TNF signaling by let-7 in the epicardium creates a pro-regenerative environment for cardiomyocyte proliferation during adult heart regeneration.
Collapse
Affiliation(s)
- Ashley M Smith
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME 04609, USA
| | - Christina A Dykeman
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME 04609, USA
| | - Benjamin L King
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA; Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA
| | - Viravuth P Yin
- Kathryn W. Davis Center for Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME 04609, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
207
|
MicroRNA let-7g acts as tumor suppressor and predictive biomarker for chemoresistance in human epithelial ovarian cancer. Sci Rep 2019; 9:5668. [PMID: 30952937 PMCID: PMC6450929 DOI: 10.1038/s41598-019-42221-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
Remarkable deregulation of microRNAs has been demonstrated in epithelial ovarian cancer (EOC). In particular, some of the let-7 miRNA family members have been proposed as tumor suppressors. Here, we explored the functional roles of let-7g in EOC. The ectopic overexpression of let-7g in OVCAR3 and HEY-A8 EOC cells induced i) a down-regulation of c-Myc and cyclin-D2 thus promoting cell cycle arrest, ii) a reduction of Vimentin, Snail and Slug thus counteracting the progression of epithelial to mesenchymal transition, iii) a chemosensitization to cis-platinum treatment. Next, analysis of human EOC tissues revealed that let-7g expression was significantly reduced in tumor tissue specimens of patients with EOC compared to their non-tumor counterparts (p = 0.0002). Notably, low let-7g tissue levels were significantly associated with acquired chemoresistance of patients with late-stage of EOC (n = 17, p = 0.03194). This finding was further validated in the serum samples collected from the same cohort of patients (n = 17, p = 0.003). To conclude, we demonstrate that let-7g acts as tumor suppressor and might be used to disable EOC tumor progression and chemoresistance to cis-platinum-based chemotherapy. Furthermore, we propose that decreased expression of let-7g could serve as a tissue and serum biomarker able to predict the chemo-resistant features of EOC patients.
Collapse
|
208
|
Exosomal miRNAs as Novel Pharmacodynamic Biomarkers for Cancer Chemopreventive Agent Early Stage Treatments in Chemically Induced Mouse Model of Lung Squamous Cell Carcinoma. Cancers (Basel) 2019; 11:cancers11040477. [PMID: 30987362 PMCID: PMC6520832 DOI: 10.3390/cancers11040477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 01/20/2023] Open
Abstract
Background: Chemopreventive agent (CPA) treatment is one of the main preventive options for lung cancer. However, few studies have been done on pharmacodynamic biomarkers of known CPAs for lung cancer. Materials and methods: In this study, we treated mouse models of lung squamous cell carcinoma with three different CPAs (MEK inhibitor: AZD6244, PI-3K inhibitor: XL-147 and glucocorticoid: Budesonide) and examined circulating exosomal miRNAs in the plasma of each mouse before and after treatment. Results: Compared to baselines, we found differentially expressed exosomal miRNAs after AZD6244 treatment (n = 8, FDR < 0.05; n = 55, raw p-values < 0.05), after XL-147 treatment (n = 4, FDR < 0.05; n = 26, raw p-values < 0.05) and after Budesonide treatment (n = 1, FDR < 0.05; n = 36, raw p-values < 0.05). In co-expression analysis, we found that modules of exosomal miRNAs reacted to CPA treatments differently. By variable selection, we identified 11, 9 and nine exosomal miRNAs as predictors for AZD6244, XL-147 and Budesonide treatment, respectively. Integrating all the results, we highlighted 4 miRNAs (mmu-miR-215-5p, mmu-miR-204-5p, mmu-miR-708-3p and mmu-miR-1298-5p) as the key for AZD6244 treatment, mmu-miR-23a-3p as key for XL-147 treatment, and mmu-miR-125a-5p and mmu-miR-16-5p as key for Budesonide treatment. Conclusions: This is the first study to use circulating exosomal miRNAs as pharmacodynamic biomarkers for CPA treatment in lung cancer.
Collapse
|
209
|
Fan R, Xiao C, Wan X, Cha W, Miao Y, Zhou Y, Qin C, Cui T, Su F, Shan X. Small molecules with big roles in microRNA chemical biology and microRNA-targeted therapeutics. RNA Biol 2019; 16:707-718. [PMID: 30900502 DOI: 10.1080/15476286.2019.1593094] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs that post-transcriptionally regulate gene expression. Aberrant miRNA expression or function have close links with various human diseases. Therefore, therapeutic treatments with disease-associated miRNAs as targets are emerging. However, the intracellular miRNA networks are extremely complicated and poorly understood, which thus hinder the development of miRNA-targeted therapeutics. Small molecules that are able to regulate endogenous miRNAs hold great potential in both elucidation of miRNA networks and treatment of miRNA-related diseases. Herein, we summarize current strategies for discovery of small molecule modifiers of miRNAs, and we highlight aspects of miRNA cellular biology elucidated by using these small molecules and miRNA-targeted therapeutics realized by these small molecules. We envision that this area will expand dramatically in the near future and will ultimately contribute to a better understanding of miRNA-involved cellular processes and development of therapeutic agents for miRNA-associated diseases.
Collapse
Affiliation(s)
- Rengen Fan
- a Department of General Surgery, Yancheng City No. 1 People's Hospital , Yancheng , China
| | - Chaocheng Xiao
- b Department of General Surgery, Yancheng City No. 1 People's Hospital , Yancheng , China
| | - Xinqiang Wan
- c Department of Gynaecology and Obstetrics, Yancheng City No. 1 People's Hospital , Yancheng , China
| | - Wenzhang Cha
- a Department of General Surgery, Yancheng City No. 1 People's Hospital , Yancheng , China
| | - Yufeng Miao
- d Department of Medical Oncology , Wuxi Third People's Hospital , Wuxi , China
| | - Yong Zhou
- a Department of General Surgery, Yancheng City No. 1 People's Hospital , Yancheng , China
| | - Chenglin Qin
- a Department of General Surgery, Yancheng City No. 1 People's Hospital , Yancheng , China
| | - Ting Cui
- e Department of Cardiology, The Third People's Hospital of Yancheng , Yancheng , China
| | - Fenglian Su
- f School of Medical University, Xuzhou , Xuzhou , China
| | - Xiangxiang Shan
- g Department of Geraeology, Yancheng City No.1 People's Hospital , Yancheng , China
| |
Collapse
|
210
|
Yao A, Xiang Y, Si YR, Fan LJ, Li JP, Li H, Guo W, He HX, Liang XJ, Tan Y, Bao LY, Liao XH. PKM2 promotes glucose metabolism through a let-7a-5p/Stat3/hnRNP-A1 regulatory feedback loop in breast cancer cells. J Cell Biochem 2019; 120:6542-6554. [PMID: 30368881 DOI: 10.1002/jcb.27947] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/02/2018] [Indexed: 11/07/2022]
Abstract
Tumor cells metabolize more glucose to lactate in aerobic or hypoxic conditions than normal cells. Pyruvate kinase isoenzyme type M2 (PKM2) is crucial for tumor cell aerobic glycolysis. We established a role for let-7a-5p/Stat3/hnRNP-A1/PKM2 signaling in breast cancer cell glucose metabolism. PKM2 depletion via small interfering RNA (siRNA) inhibits cell proliferation and aerobic glycolysis in breast cancer cells. Signal transducer and activator of transcription 3 (Stat3) promotes upregulation of heterogeneous nuclear ribonucleoprotein (hnRNP)-A1 expression, hnRNP-A1 binding to pyruvate kinase isoenzyme (PKM) pre messenger RNA, and the subsequent formation of PKM2. This pathway is downregulated by the microRNA let-7a-5p, which functionally targets Stat3, whereas hnRNP-A1 blocks the biogenesis of let-7a-5p to counteract its ability to downregulate the Stat3/hnRNP-A1/PKM2 signaling pathway. The downregulation of Stat3/hnRNP-A1/PKM2 by let-7a-5p is verified using a breast cancer. These results suggest that let-7a-5p, Stat3, and hnRNP-A1 form a feedback loop, thereby regulating PKM2 expression to modulate glucose metabolism of breast cancer cells. These findings elucidate a new pathway mediating aerobic glycolysis in breast cancers and provide an attractive potential target for breast cancer therapeutic intervention.
Collapse
Affiliation(s)
- Ao Yao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Yuan Xiang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Yu-Rui Si
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Li-Juan Fan
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Jia-Peng Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Hui Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Wei Guo
- Shenzhen Ritzcon Biological Technology Co, Ltd, Shenzhen, China
| | - Hui-Xin He
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Xing-Jie Liang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Yao Tan
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Le-Yuan Bao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Xing-Hua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| |
Collapse
|
211
|
Jang HS, Shah NM, Du AY, Dailey ZZ, Pehrsson EC, Godoy PM, Zhang D, Li D, Xing X, Kim S, O'Donnell D, Gordon JI, Wang T. Transposable elements drive widespread expression of oncogenes in human cancers. Nat Genet 2019; 51:611-617. [PMID: 30926969 PMCID: PMC6443099 DOI: 10.1038/s41588-019-0373-3] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/12/2019] [Indexed: 11/24/2022]
Abstract
Transposable elements (TEs) are an abundant and rich genetic resource of regulatory sequences1-3. Cryptic regulatory elements within TEs can be epigenetically reactivated in cancer to influence oncogenesis in a process termed onco-exaptation4. However, the prevalence and impact of TE onco-exaptation events across cancer types are poorly characterized. Here, we analyzed 7,769 tumors and 625 normal datasets from 15 cancer types, identifying 129 TE cryptic promoter-activation events involving 106 oncogenes across 3,864 tumors. Furthermore, we interrogated the AluJb-LIN28B candidate: the genetic deletion of the TE eliminated oncogene expression, while dynamic DNA methylation modulated promoter activity, illustrating the necessity and sufficiency of a TE for oncogene activation. Collectively, our results characterize the global profile of TE onco-exaptation and highlight this prevalent phenomenon as an important mechanism for promiscuous oncogene activation and ultimately tumorigenesis.
Collapse
Affiliation(s)
- Hyo Sik Jang
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Nakul M Shah
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Alan Y Du
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Zea Z Dailey
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Erica C Pehrsson
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Paula M Godoy
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - David Zhang
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Daofeng Li
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Sungsu Kim
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- Hope Center for Neurological Disease, Washington University School of Medicine, St Louis, MO, USA
| | - David O'Donnell
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St Louis, MO, USA
| | - Jeffrey I Gordon
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St Louis, MO, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA.
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
212
|
Chen Y, Qiao L, Zhang Z, Hu G, Zhang J, Li H. Let-7a inhibits proliferation and promotes apoptosis of human asthmatic airway smooth muscle cells. Exp Ther Med 2019; 17:3327-3334. [PMID: 30988708 PMCID: PMC6447815 DOI: 10.3892/etm.2019.7363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 12/13/2018] [Indexed: 12/18/2022] Open
Abstract
The present study aimed to examine the changes of let-7a expression in asthmatic airway smooth muscle cells (ASMCs) and to analyze its effect on the proliferation and apoptosis of ASMCs, as well as the potential mechanism of action. Let-7a expression levels in ASMCs from asthmatic and non-asthmatic subjects were detected using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis. Furthermore, let-7a mimics were transfected in vitro into ASMCs isolated from asthmatic patients, and the effect of let-7a on ASMC proliferation was examined using a Cell Counting Kit-8. In addition, the influence of let-7a on ASMC apoptosis was detected using flow cytometry and a caspase-3/7 activity assay. Target genes of let-7a were predicted using bioinformatics software, and the direct regulatory effect of let-7a on the potential target gene signal transducer and activator of transcription 3 (STAT3) was verified through a dual-luciferase reporter gene assay combined with RT-qPCR and western blot analysis. The results demonstrated that let-7a expression was significantly lower in ASMCs of asthmatic subjects compared with that in ASMCs of normal subjects. Furthermore, upregulation of let-7a expression in asthmatic ASMCs markedly inhibited cell proliferation and promoted cell apoptosis. The results of the dual-luciferase reporter gene assay indicated that let-7a selectively binds with the 3′-untranslated region of the STAT3 mRNA. In addition, let-7a mimics evidently reduced the mRNA and protein expression levels of STAT3 in asthmatic ASMCs. In conclusion, the present study demonstrates that let-7a expression is downregulated in ASMCs from asthmatic patients. Furthermore, let-7a suppresses the proliferation and promotes apoptosis of human asthmatic ASMCs, which may, at least partially, be associated with the downregulation of STAT3 expression.
Collapse
Affiliation(s)
- Yan Chen
- Department of Critical Care Medicine, Shengli Oilfield Center Hospital, Dongying, Shandong 257000, P.R. China
| | - Lujun Qiao
- Department of Critical Care Medicine, Shengli Oilfield Center Hospital, Dongying, Shandong 257000, P.R. China
| | - Zewen Zhang
- Department of Respiratory Disease, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Guoxin Hu
- Department of Critical Care Medicine, Shengli Oilfield Center Hospital, Dongying, Shandong 257000, P.R. China
| | - Jian Zhang
- Department of Critical Care Medicine, Shengli Oilfield Center Hospital, Dongying, Shandong 257000, P.R. China
| | - Hongjia Li
- Department of Respiratory Disease, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
213
|
Liu Y, Dong N, Miao J, Li C, Wang X, Ruan J. Lin28 promotes dental pulp cell proliferation via upregulation of cyclin-dependent proteins and interaction with let-7a/IGF2BP2 pathways. Biomed Pharmacother 2019; 113:108742. [PMID: 30851545 DOI: 10.1016/j.biopha.2019.108742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 01/23/2023] Open
Abstract
Caries, pulpitis, and trauma are the main causes of dental pulp damage. The regeneration capacity of dental pulp declines with age. Lin28 is a conserved RNA-binding protein in higher eukaryotes that regulates several important cellular functions associated with development, glucose metabolism, differentiation, and pluripotency. Conditional reactivation of Lin28 gene in adult mice markedly accelerates the wound-healing process in injured digits. However, little is known about its functions and molecular mechanism in human dental pulp. The aim of this study was to investigate the effects and mechanism of overexpression of Lin28 gene on the proliferation of human dental pulp cells (HDPCs). For this purpose, a number of molecular and biochemical analytical techniques, including the ethynyl-2'-deoxyuridine (EdU) incorporation assay, RNA-protein immunoprecipitation (RIP) analysis, and luciferase assays, were used for detailed characterization. In addition, factors regulating HDPCs activation were explored through gain-of-function and loss-of-function analyses. The results demonstrate that Lin28 promotes cell proliferation and the S-G2/M transition of HDPCs and directly binds to a group of cell cycle regulatory mRNAs in HDPCs. Through bioinformatics analysis and luciferase assays, we confirmed that let-7a targets IGF2BP2. Silencing of IGF2BP2 showed similar cellular and molecular effects as let-7a. Similarly, restoration of IGF2BP2 counteracted the effects of let-7a expression. In conclusion, Lin28 promotes cell proliferation by regulation of both mRNA translation (let-7-independent) and miRNA biogenesis (let-7-dependent). Lin28 can promote the expression of pro-proliferative genes by directly enhancing their translation to maintain a tight control over HDPC proliferation.
Collapse
Affiliation(s)
- Yan Liu
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Department of Preventive Dentistry, College of Stomatology, Xi'an Jiaotong University, 98 Xiwu Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Ning Dong
- Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, 98 Xiwu Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Jiyu Miao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Chenxing Li
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Xiaofei Wang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Jianping Ruan
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Department of Preventive Dentistry, College of Stomatology, Xi'an Jiaotong University, 98 Xiwu Road, Xi'an, Shaanxi, 710004, People's Republic of China.
| |
Collapse
|
214
|
Ramezanpour M, Daei P, Tabarzad M, Khanaki K, Elmi A, Barati M. Preliminary study on the effect of nucleolin specific aptamer-miRNA let-7d chimera on Janus kinase-2 expression level and activity in gastric cancer (MKN-45) cells. Mol Biol Rep 2019; 46:207-215. [PMID: 30415442 DOI: 10.1007/s11033-018-4462-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022]
Abstract
Recently, much attention has been focused on the use of miRNAs in cancer treatment. The role of proto-oncogene Janus kinase-2 (JAK-2) in proliferation and survival of gastric cancer has been previously documented. The aim of this study was to evaluate the effect of a chimera consisted of nucleolin specific aptamer (NCL-Apt) and miRNA let-7d on JAK2 expression level and activity in gastric cancer cells. NCL-Apt-miRNA let-7d chimera was prepared by two methods. Gastric cancer (MKN-45) cell line and control cell line of human dermal fibroblast (HDF) were treated with the chimera and the changes in JAK2 expression and activity were determined using real-time PCR and ELISA techniques, respectively. In MKN-45 cells, the chimera caused significant decrease in JAK2 expression level and activity compared to the aptamer alone and miRNA mimic negative control. Nevertheless, transfected miRNA let-7d showed remarkable reduction in the expression level of JAK2 in comparison with control state in both MKN-45 and HDF, confirmed unspecific effect of let-7d on normal and cancerous cells. With regard to the synergic effect of this chimera on JAK2 activity, it might be viewed as a therapeutic candidate in gastric cancer. However, further studies are warranted to prove it.
Collapse
Affiliation(s)
- Mahsa Ramezanpour
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Puyan Daei
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Maryam Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Korosh Khanaki
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Ali Elmi
- Medical Biotechnology Research Center, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahmood Barati
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
215
|
Chen Y, Xie C, Zheng X, Nie X, Wang Z, Liu H, Zhao Y. LIN28/ let-7/PD-L1 Pathway as a Target for Cancer Immunotherapy. Cancer Immunol Res 2019; 7:487-497. [PMID: 30651289 DOI: 10.1158/2326-6066.cir-18-0331] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/15/2018] [Accepted: 01/10/2019] [Indexed: 11/16/2022]
Abstract
The immunocheckpoint protein PD-1/PD-L1 is considered a promising target for cancer immunotherapeutics. However, the objective response rate using antibodies that block the interaction between PD-1 and PD-L1 was less than 40%, and the mechanism underlying regulation of PD-1/PD-L1 expression is poorly understood. In this study, we identified the miRNA let-7 that posttranscriptionally suppresses PD-L1 expression. LIN28, an RNA binding protein upregulated in most cancer cells, inhibits the biogenesis of let-7, thus promoting PD-L1 expression. Therefore, inhibition of LIN28 may be a strategy to prevent immune evasion of cancer cells. We found that treatment with a LIN28 inhibitor, the small compound C1632, increases let-7 and suppresses PD-L1 expression, leading to reactivation of antitumor immunity in vitro and in vivo In addition, C1632 also displayed the capacity to inhibit cancer cell proliferation and tumor growth in mice. Altogether, these findings identified LIN28/let-7 as a target for PD-L1-mediated immunotherapeutics and reveal the potential of C1632 and its derivatives as promising oncotherapeutic agents.
Collapse
Affiliation(s)
- Yanlian Chen
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, P.R. China
| | - Chen Xie
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xiaohui Zheng
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, P.R. China
| | - Xin Nie
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Zining Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, P.R. China
| | - Haiying Liu
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yong Zhao
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China. .,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
216
|
Maria ATJ, Partouche L, Goulabchand R, Rivière S, Rozier P, Bourgier C, Le Quellec A, Morel J, Noël D, Guilpain P. Intriguing Relationships Between Cancer and Systemic Sclerosis: Role of the Immune System and Other Contributors. Front Immunol 2019; 9:3112. [PMID: 30687318 PMCID: PMC6335319 DOI: 10.3389/fimmu.2018.03112] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/17/2018] [Indexed: 01/09/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune connective tissue disorder, characterized by multisystem involvement, vasculopathy, and fibrosis. An increased risk of malignancy is observed in SSc (including breast and lung cancers), and in a subgroup of patients with specific autoantibodies (i.e., anti-RNA polymerase III and related autoantibodies), SSc could be a paraneoplastic syndrome and might be directly related to an immune response against cancer. Herein, we reviewed the literature, focusing on the most recent articles, and shed light onto the potential relationship between cancer and scleroderma regarding temporal and immunological dimensions.
Collapse
Affiliation(s)
- Alexandre Thibault Jacques Maria
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France.,IRMB, INSERM, CHU Montpellier, Montpellier University, Montpellier, France
| | - Léo Partouche
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France
| | - Radjiv Goulabchand
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France
| | - Sophie Rivière
- Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France
| | - Pauline Rozier
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France.,IRMB, INSERM, CHU Montpellier, Montpellier University, Montpellier, France
| | - Céline Bourgier
- Medical School, Montpellier University, Montpellier, France.,Department of Radiation Oncology, INSERM U1194/IRCM, ICM-Val d'Aurelle, Montpellier, France
| | - Alain Le Quellec
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France
| | - Jacques Morel
- Medical School, Montpellier University, Montpellier, France.,Department of Rheumatology, Lapeyronie Hospital, Montpellier, France
| | - Danièle Noël
- IRMB, INSERM, CHU Montpellier, Montpellier University, Montpellier, France
| | - Philippe Guilpain
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France.,IRMB, INSERM, CHU Montpellier, Montpellier University, Montpellier, France
| |
Collapse
|
217
|
Han Z, Li Q, Wang Y, Wang L, Li X, Ge N, Wang Y, Guo C. Niclosamide Induces Cell Cycle Arrest in G1 Phase in Head and Neck Squamous Cell Carcinoma Through Let-7d/CDC34 Axis. Front Pharmacol 2019; 9:1544. [PMID: 30687101 PMCID: PMC6333743 DOI: 10.3389/fphar.2018.01544] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/18/2018] [Indexed: 01/11/2023] Open
Abstract
Niclosamide is a traditional anti-tapeworm drug that exhibits potent anti-cancer activity. Our previous study showed that niclosamide induces cell cycle arrest in G1 phase. Nevertheless, the underlying mechanism remains unknown. The following study investigated the molecular mechanism through which niclosamide induced G1 arrest in head and neck squamous cell carcinoma (HNSCC) cell lines. The effect of niclosamide on human HNSCC cell line WSU-HN6 and CNE-2Z were analyzed using IncuCyte ZOOMTM assay, flow cytometry (FCM), real-time PCR and western blot. Luciferase assay was conducted to demonstrate the interaction between let-7d (a let-7 family member which functions as a tumor suppressor by regulating cell cycle) and 3′UTR of CDC34 mRNA. Xenografts tumor model was established to evaluate the niclosamide treatment efficacy in vivo. Briefly, an exposure to niclosamide treatment led to an increased let-7d expression and a decreased expression of cell cycle regulator CDC34, finally leading to G1 phase arrest. Moreover, an overexpression of let-7d induced G1 phase arrest and downregulated CDC34, while the knockdown of let-7d partially rescued the niclosamide-induced G1 phase arrest. Luciferase assay confirmed the direct inhibition of CDC34 through the targeting of let-7d. Furthermore, niclosamide markedly inhibited the xenografts growth through up-regulation of let-7d and down-regulation of CDC34. To sum up, our findings suggest that niclosamide induces cell cycle arrest in G1 phase in HNSCC through let-7d/CDC34 axis, which enriches the anti-cancer mechanism of niclosamide.
Collapse
Affiliation(s)
- Zewen Han
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Qingxiang Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yifei Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Lin Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiaoxu Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Na Ge
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yixiang Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Chuanbin Guo
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
218
|
Andrés-León E, Rojas AM. miARma-Seq, a comprehensive pipeline for the simultaneous study and integration of miRNA and mRNA expression data. Methods 2019; 152:31-40. [PMID: 30253202 DOI: 10.1016/j.ymeth.2018.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/22/2018] [Accepted: 09/04/2018] [Indexed: 01/06/2023] Open
|
219
|
Swier LJYM, Dzikiewicz‐Krawczyk A, Winkle M, van den Berg A, Kluiver J. Intricate crosstalk between MYC and non-coding RNAs regulates hallmarks of cancer. Mol Oncol 2019; 13:26-45. [PMID: 30451365 PMCID: PMC6322196 DOI: 10.1002/1878-0261.12409] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/10/2018] [Accepted: 10/23/2018] [Indexed: 01/17/2023] Open
Abstract
Myelocytomatosis viral oncogene homolog (MYC) plays an important role in the regulation of many cellular processes, and its expression is tightly regulated at the level of transcription, translation, protein stability, and activity. Despite this tight regulation, MYC is overexpressed in many cancers and contributes to multiple hallmarks of cancer. In recent years, it has become clear that noncoding RNAs add a crucial additional layer to the regulation of MYC and its downstream effects. So far, twenty-five microRNAs and eighteen long noncoding RNAs that regulate MYC have been identified. Thirty-three miRNAs and nineteen lncRNAs are downstream effectors of MYC that contribute to the broad oncogenic role of MYC, including its effects on diverse hallmarks of cancer. In this review, we give an overview of this extensive, multilayered noncoding RNA network that exists around MYC. Current data clearly show explicit roles of crosstalk between MYC and ncRNAs to allow tumorigenesis.
Collapse
Affiliation(s)
- Lotteke J. Y. M. Swier
- Department of Pathology and Medical BiologyUniversity of GroningenUniversity Medical Center GroningenThe Netherlands
| | | | - Melanie Winkle
- Department of Pathology and Medical BiologyUniversity of GroningenUniversity Medical Center GroningenThe Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical BiologyUniversity of GroningenUniversity Medical Center GroningenThe Netherlands
| | - Joost Kluiver
- Department of Pathology and Medical BiologyUniversity of GroningenUniversity Medical Center GroningenThe Netherlands
| |
Collapse
|
220
|
Wan YH, Zhou YJ, Xiao KJ, Nie CP, Zhang J, Liu C, Chen TT, Chu X. Target-assisted self-cleavage DNAzyme probes for multicolor simultaneous imaging of tumor-related microRNAs with signal amplification. Chem Commun (Camb) 2019; 55:3278-3281. [DOI: 10.1039/c9cc00451c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel and highly selective signal amplification strategy was developed based on target-assisted self-cleavage DNAzyme probes for imaging of miRNA-222 and miRNA-223.
Collapse
Affiliation(s)
- Yuan-Hui Wan
- Institute of Chemical Biology and Nanomedicine
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
| | - Yu-Jie Zhou
- Institute of Chemical Biology and Nanomedicine
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
| | - Ke-Jing Xiao
- Institute of Chemical Biology and Nanomedicine
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
| | - Cun-Peng Nie
- Institute of Chemical Biology and Nanomedicine
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
| | - Juan Zhang
- Institute of Chemical Biology and Nanomedicine
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
| | - Chang Liu
- Institute of Chemical Biology and Nanomedicine
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
| | - Ting-Ting Chen
- Institute of Chemical Biology and Nanomedicine
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
| | - Xia Chu
- Institute of Chemical Biology and Nanomedicine
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha 410082
| |
Collapse
|
221
|
Issa F, Milward K, Goto R, Betts G, Wood KJ, Hester J. Transiently Activated Human Regulatory T Cells Upregulate BCL-XL Expression and Acquire a Functional Advantage in vivo. Front Immunol 2019; 10:889. [PMID: 31068951 PMCID: PMC6491764 DOI: 10.3389/fimmu.2019.00889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/05/2019] [Indexed: 01/08/2023] Open
Abstract
Regulatory T cells (Tregs) can control excessive or undesirable immune responses toward autoantigens, alloantigens, and pathogens. In transplantation, host immune responses against the allograft are suppressed through the use of immunosuppressive drugs, however this often results in life-threatening side effects including nephrotoxicity and an increased incidence of cancer and opportunistic infections. Tregs can control graft-vs.-host disease and transplant rejection in experimental models, providing impetus for the use of Tregs as a cellular therapy in clinical transplantation. One of the major barriers to the widespread use of Treg cellular therapy is the requirement to expand cells ex vivo to large numbers in order to alter the overall balance between regulatory and effector cells. Methods that enhance suppressive capacity thereby reducing the need for expansion are therefore of interest. Here, we have compared the function of freshly-isolated and ex vivo-manipulated human Tregs in a pre-clinical humanized mouse model of skin transplantation. Sorted human CD127loCD25+CD4+ Tregs were assessed in three different conditions: freshly-isolated, following transient in vitro activation with antiCD3/antiCD28 beads or after ex vivo-expansion for 2 weeks in the presence of antiCD3/antiCD28 beads and recombinant human IL2. While ex vivo-expansion of human Tregs increased their suppressive function moderately, transient in vitro-activation of freshly isolated Tregs resulted in a powerful enhancement of Treg activity sufficient to promote long-term graft survival of all transplants in vivo. In order to investigate the mechanisms responsible for these effects, we measured the expression of Treg-associated markers and susceptibility to apoptosis in activated Tregs. Transiently activated Tregs displayed enhanced survival and proliferation in vitro and in vivo. On a molecular level, Treg activation resulted in an increased expression of anti-apoptotic BCL2L1 (encoding BCL-XL) which may be at least partially responsible for the observed enhancement in function. Our results suggest that in vitro activation of human Tregs arms them with superior proliferative and survival abilities, enabling them to more effectively control alloresponses. Importantly, this transient activation results in a rapid functional enhancement of freshly-isolated Tregs, thereby providing an opportunity to eliminate the need for in vitro expansion in select circumstances. A protocol employing this technique would therefore benefit from a reduced requirement for large cell numbers for effective therapy.
Collapse
|
222
|
Secretomes from Mesenchymal Stem Cells against Acute Kidney Injury: Possible Heterogeneity. Stem Cells Int 2018; 2018:8693137. [PMID: 30651737 PMCID: PMC6311717 DOI: 10.1155/2018/8693137] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/10/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022] Open
Abstract
A kidney has the ability to regenerate itself after a variety of renal injuries. Mesenchymal stem cells (MSCs) have been shown to ameliorate tissue damages during renal injuries and diseases. The regenerations induced by MSCs are primarily mediated by the paracrine release of soluble factors and extracellular vesicles, including exosomes and microvesicles. Extracellular vesicles contain proteins, microRNAs, and mRNAs that are transferred into recipient cells to induce several repair signaling pathways. Over the past few decades, many studies identified trophic factors from MSCs, which attenuate renal injury in a variety of animal acute kidney injury models, including renal ischemia-reperfusion injury and drug-induced renal injury, using microarray and proteomic analysis. Nevertheless, these studies have revealed the heterogeneity of trophic factors from MSCs that depend on the cell origins and different stimuli including hypoxia, inflammatory stimuli, and aging. In this review article, we summarize the secretomes and regenerative mechanisms induced by MSCs and highlight the possible heterogeneity of trophic factors from different types of MSC and different circumstances for renal regeneration.
Collapse
|
223
|
Cao L, Wan Q, Li F, Tang CE. MiR-363 inhibits cisplatin chemoresistance of epithelial ovarian cancer by regulating snail-induced epithelial-mesenchymal transition. BMB Rep 2018. [PMID: 30037365 PMCID: PMC6177509 DOI: 10.5483/bmbrep.2018.51.9.104] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Chemoresistance is a major barrier to successful cisplatinbased chemotherapy for epithelial ovarian cancer (EOC), and emerging evidences suggest that microRNAs (miRNAs) are involved in the resistance. In this study, it was indicated that miR-363 downregulation was significantly correlated with EOC carcinogenesis and cisplatin resistance. Moreover, miR-363 overexpression could resensitise cisplatin-resistant EOC cells to cisplatin treatment both in vitro and in vivo. In addition, data revealed that EMT inducer Snail was significantly upregulated in cisplatin-resistant EOC cell lines and EOC patients and was a functional target of miR-363 in EOC cells. Furthermore, snail overexpression could significantly attenuate miR-363-suppressed cisplatin resistance of EOC cells, suggesting that miR-363-regulated cisplatin resistance is mediated by snail-induced EMT in EOC cells. Taken together, findings suggest that miR-363 may be a biomarker for predicting responsiveness to cisplatin-based chemotherapy and a potential therapeutic target in EOC. [BMB Reports 2018; 51(9): 456-461].
Collapse
Affiliation(s)
- Lanqin Cao
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qian Wan
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Fengjie Li
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Can-E Tang
- The Institute of Medical Science Research, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P. R., China
| |
Collapse
|
224
|
Swellam M, Mahmoud MS, Hashim M, Hassan NM, Sobeih ME, Nageeb AM. Clinical aspects of circulating miRNA‐335 in breast cancer patients: A prospective study. J Cell Biochem 2018; 120:8975-8982. [DOI: 10.1002/jcb.28168] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/08/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Menha Swellam
- High Throughput Molecular and Genetic Laboratory, Center for Excellences for Advanced Sciences Giza Egypt
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division National Research Centre Dokki Giza Egypt
| | - Magda Sayed Mahmoud
- High Throughput Molecular and Genetic Laboratory, Center for Excellences for Advanced Sciences Giza Egypt
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division National Research Centre Dokki Giza Egypt
| | - Maha Hashim
- High Throughput Molecular and Genetic Laboratory, Center for Excellences for Advanced Sciences Giza Egypt
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division National Research Centre Dokki Giza Egypt
| | - Naglaa M Hassan
- Department of Clinical Pathology National Cancer Institute Cairo Egypt
| | | | - Amira M Nageeb
- High Throughput Molecular and Genetic Laboratory, Center for Excellences for Advanced Sciences Giza Egypt
- Biochemistry Department, Genetic Engineering and Biotechnology Research Division National Research Centre Dokki Giza Egypt
| |
Collapse
|
225
|
Hyperoside and let-7a-5p synergistically inhibits lung cancer cell proliferation via inducing G1/S phase arrest. Gene 2018; 679:232-240. [DOI: 10.1016/j.gene.2018.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/29/2018] [Accepted: 09/06/2018] [Indexed: 01/04/2023]
|
226
|
Saeki N, Saito A, Sugaya Y, Amemiya M, Sasaki H. Indirect Down-regulation of Tumor-suppressive let-7 Family MicroRNAs by LMO1 in Neuroblastoma. Cancer Genomics Proteomics 2018; 15:413-420. [PMID: 30194082 DOI: 10.21873/cgp.20100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/AIM Overall survival for the high-risk group of neuroblastoma (NB) patients still remains at 40-50%, necessitating the establishment of a curable treatment. LIM domain only 1 (LMO1) gene encoding a transcriptional regulator is an NB-susceptibility gene with a tumor-promoting activity. Previously we conducted chromatin immunoprecipitation and DNA sequencing analyses on NB cell lines and identified 3 protein-coding genes regulated by LMO1. In this study, we extended our analyses to capture microRNA genes directly or indirectly regulated by LMO1. MATERIALS AND METHODS Using microarrays, we conducted a comparative gene expression analysis on an NB cell line SK-N-SH; between the cells with and without LMO1 suppression. RESULTS Overall, 18 microRNAs were identified to be indirectly down-regulated by LMO1 including 7 microRNAs of the let-7 family, whose cell proliferation inhibitory activity was observed. CONCLUSION Target genes of the LMO1-regulated microRNAs and their relevant pathways may be a potential therapeutic target.
Collapse
Affiliation(s)
- Norihisa Saeki
- Division of Anatomy and Physiology, Okinawa Prefectural College of Nursing, Okinawa, Japan
| | - Akira Saito
- Statistical Genetics Analysis Division, StaGen Co. Ltd., Tokyo, Japan
| | - Yuki Sugaya
- Statistical Genetics Analysis Division, StaGen Co. Ltd., Tokyo, Japan
| | - Mitsuhiro Amemiya
- Statistical Genetics Analysis Division, StaGen Co. Ltd., Tokyo, Japan
| | - Hiroki Sasaki
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
227
|
Wang S, Zhou H, Wu D, Ni H, Chen Z, Chen C, Xiang Y, Dai K, Chen X, Li X. MicroRNA let-7a regulates angiogenesis by targeting TGFBR3 mRNA. J Cell Mol Med 2018; 23:556-567. [PMID: 30467960 PMCID: PMC6307798 DOI: 10.1111/jcmm.13960] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/16/2018] [Accepted: 09/21/2018] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis has a great impact on human health, owing to its participation in development, wound healing and the pathogenesis of several diseases. It has been reported that let-7a is a tumour suppressor, but whether it plays a role in angiogenesis is unclear. Here we showed that let-7a, a microRNA conserved in vertebrates, regulated angiogenesis by concomitantly down-regulating TGFBR3. Overexpression of let-7a or knockdown of TGFBR3 in cell culture inhibited the tube formation and reduced migration rate. Moreover, xenograft experiments showed that overexpression of let-7a or knockdown of TGFBR3 had smaller tumour size. Downstream genes, such as VEGFC and MMP9, were also down-regulated in let-7a overexpression or TGFBR3 knockdown groups. Therefore, our results revealed a novel mechanism that let-7a regulate angiogenesis through post-transcriptional regulation of TGFBR3.
Collapse
Affiliation(s)
- Shao Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huandong Zhou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dazhou Wu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huajing Ni
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongliang Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youqun Xiang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kezhi Dai
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China.,The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoming Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi Li
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China.,The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
228
|
Thapar R. Regulation of DNA Double-Strand Break Repair by Non-Coding RNAs. Molecules 2018; 23:molecules23112789. [PMID: 30373256 PMCID: PMC6278438 DOI: 10.3390/molecules23112789] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 01/12/2023] Open
Abstract
DNA double-strand breaks (DSBs) are deleterious lesions that are generated in response to ionizing radiation or replication fork collapse that can lead to genomic instability and cancer. Eukaryotes have evolved two major pathways, namely homologous recombination (HR) and non-homologous end joining (NHEJ) to repair DSBs. Whereas the roles of protein-DNA interactions in HR and NHEJ have been fairly well defined, the functions of small and long non-coding RNAs and RNA-DNA hybrids in the DNA damage response is just beginning to be elucidated. This review summarizes recent discoveries on the identification of non-coding RNAs and RNA-mediated regulation of DSB repair.
Collapse
Affiliation(s)
- Roopa Thapar
- Department of Molecular and Cellular Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
229
|
Vahidian F, Mohammadi H, Ali-Hasanzadeh M, Derakhshani A, Mostaan M, Hemmatzadeh M, Baradaran B. MicroRNAs and breast cancer stem cells: Potential role in breast cancer therapy. J Cell Physiol 2018; 234:3294-3306. [PMID: 30362508 DOI: 10.1002/jcp.27246] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) can control cancer and cancer stem cells (CSCs), and this topic has drawn immense attention recently. Stem cells are a tiny population of a bulk of tumor cells that have enormous potential in expansion and metastasis of the tumor. miRNA have a crucial role in the management of the function of stem cells. This role is to either promote or suppress the tumor. In this review, we investigated the function and different characteristics of CSCs and function of the miRNAs that are related to them. We also demonstrated the role and efficacy of these miRNAs in breast cancer and breast cancer stem cells (BCSC). Eventually, we revealed the metastasis, tumor formation, and their role in the apoptosis process. Also, the therapeutic potential of miRNA as an effective method for the treatment of BCSC was described. Extensive research is required to investigate the employment or suppression of these miRNAs for therapeutics approached in different cancers in the future.
Collapse
Affiliation(s)
- Fatemeh Vahidian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Ali-Hasanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Afshin Derakhshani
- Department of Immunology, Birjand University of Medical Sciences, Birjand, Iran.,Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Masoud Mostaan
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Islamic Azad university, Tabriz, Iran
| | - Maryam Hemmatzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
230
|
Gilles ME, Slack FJ. Let-7 microRNA as a potential therapeutic target with implications for immunotherapy. Expert Opin Ther Targets 2018; 22:929-939. [PMID: 30328720 DOI: 10.1080/14728222.2018.1535594] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION MicroRNAs (miRNA) are a class of small non-coding RNA that play a major role in various cellular processes by negatively regulating gene expression. In the past decade, miRNA dysregulation has been reported to be closely linked to inflammatory diseases. The immune response modulates cancer initiation and progression; miRNAs including let-7 family members have been shown to act as key regulators of the immune responses in various diseases and cancers. Notably, the let-7 miRNA has been reported to be closely associated with immunity, specifically with Toll-like receptors that mediate cytokine expression during pathogen infection and with the regulation of various other immune effectors. Areas covered: In this review, the authors describe the discovery of let-7 as the starting point of the RNA revolution and highlight let-7 as an efficient tool for cancer and immune therapy. Expert opinion: let-7 miRNA has emerged as a key player in cancer therapy and immune responses and it has potential role as a new immunotherapeutic target. However, while there are challenges regarding miRNA delivery, the exciting emergence of personalized medicine for cancer and immunotherapy could be beneficial for the development of let-7 therapeutics.
Collapse
Affiliation(s)
- Maud-Emmanuelle Gilles
- a Harvard Medical School initiative for RNA Medicine, Department of Pathology , Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA
| | - Frank J Slack
- a Harvard Medical School initiative for RNA Medicine, Department of Pathology , Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
231
|
Hofbauer SL, de Martino M, Lucca I, Haitel A, Susani M, Shariat SF, Klatte T. A urinary microRNA (miR) signature for diagnosis of bladder cancer. Urol Oncol 2018; 36:531.e1-531.e8. [PMID: 30322728 DOI: 10.1016/j.urolonc.2018.09.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/30/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Bladder cancer (BC) is diagnosed by cystoscopy, which is invasive, costly and causes considerable patient discomfort. MicroRNAs (miR) are dysregulated in BC and may serve as non-invasive urine markers for primary diagnostics and monitoring. The purpose of this study was to identify a urinary miR signature that predicts the presence of BC. METHODS For the detection of potential urinary miR markers, expression of 384 different miRs was analyzed in 16 urine samples from BC patients and controls using a Taqman™ Human MicroRNA Array (training set). The identified candidate gene signature was subsequently validated in an independent cohort of 202 urine samples of patients with BC and controls with microscopic hematuria. The final miR signature was developed from a multivariable logistic regression model. RESULTS Analysis of the training set identified 14 candidate miRs for further analysis within the validation set. Using backward stepwise elimination, we identified a subset of 6 miRs (let-7c, miR-135a, miR-135b, miR-148a, miR-204, miR-345) that distinguished BC from controls with an area under the curve of 88.3%. The signature was most accurate in diagnosing high-grade non-muscle invasive BC (area under the curve = 92.9%), but was capable to identify both low-grade and high-grade disease as well as non-muscle and muscle-invasive BC with high accuracies. CONCLUSIONS We identified a 6-gene miR signature that can accurately predict the presence of BC from urine samples, independent of stage and grade. This signature represents a simple urine assay that may help reducing costs and morbidity associated with invasive diagnostics.
Collapse
Affiliation(s)
- Sebastian L Hofbauer
- Department of Urology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Michela de Martino
- Department of Urology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Ilaria Lucca
- Department of Urology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Andrea Haitel
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Martin Susani
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Shahrokh F Shariat
- Department of Urology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Tobias Klatte
- Department of Urology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| |
Collapse
|
232
|
He Z, Deng W, Jiang B, Liu S, Tang M, Liu Y, Zhang J. Hsa-let-7b inhibits cell proliferation by targeting PLK1 in HCC. Gene 2018; 673:46-55. [PMID: 29913237 DOI: 10.1016/j.gene.2018.06.047] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 06/14/2018] [Indexed: 12/19/2022]
Abstract
Previous studies have shown that high levels of PLK1 are expressed in HCC, and PLK1 inhibitors are being tested in clinical trials. However, the mechanisms, which regulate PLK1 expression in HCC, have not been clarified. Here, we show that induction of let-7b over-expression inhibits the PLK1-regulated luciferase activity in HEK-293T cells, and decreases the levels of PLK1 expression in HCC cells. Furthermore, the levels of let-7b expression were negatively correlated with PLK1 expression in HCC tissues. Let-7b over-expression inhibited the proliferation of HCC cells and promoted their apoptosis, which were partially rescued by increased PLK1 expression. Let-7b over-expression decreased the levels of PLK1, CDC25C and Survivin phosphorylation and CDC2, β-catenin, TCF-4 expression, which were mitigated by increased PLK1 expression in MHCC-97H cells. Let-7b over-expression inhibited the development and growth of implanted HCC tumors in mice by decreasing PLK1 and Survivin expression in the tumors. Together, our data indicated that let-7b targeted PLK1 to inhibit HCC growth and induce their apoptosis by attenuating the PLK1-mediated Survivin phosphorylation. Our findings may provide new insights into the pathogenesis of HCC.
Collapse
Affiliation(s)
- Zili He
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital/the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, People's Republic of China; Key Laboratory of Protein Chemistry, Developmental Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, People's Republic of China; Laboratory of Hepatobiliary Molecular Oncology, Hunan Provincial People's Hospital, Changsha, Hunan 410005, People's Republic of China.
| | - Wen Deng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital/the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, People's Republic of China
| | - Bo Jiang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital/the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, People's Republic of China
| | - Sulai Liu
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital/the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, People's Republic of China
| | - Mingchun Tang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital/the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, People's Republic of China
| | - Yi Liu
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital/the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, People's Republic of China
| | - Jian Zhang
- Key Laboratory of Protein Chemistry, Developmental Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, People's Republic of China.
| |
Collapse
|
233
|
Iqbal MA, Arora S, Prakasam G, Calin GA, Syed MA. MicroRNA in lung cancer: role, mechanisms, pathways and therapeutic relevance. Mol Aspects Med 2018; 70:3-20. [PMID: 30102929 DOI: 10.1016/j.mam.2018.07.003] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/28/2018] [Accepted: 07/30/2018] [Indexed: 12/29/2022]
Abstract
Lung cancer is the cardinal cause of cancer-related deaths with restricted recourse of therapy throughout the world. Clinical success of therapies is not very promising due to - late diagnosis, limited therapeutic tools, relapse and the development of drug resistance. Recently, small ∼20-24 nucleotides molecules called microRNAs (miRNAs) have come into the limelight as they play outstanding role in the process of tumorigenesis by regulating cell cycle, metastasis, angiogenesis, metabolism and apoptosis. miRNAs essentially regulate gene expression via post-transcriptional regulation of mRNA. Nevertheless, few studies have conceded the role of miRNAs in activation of gene expression. A large body of data generated by numerous studies is suggestive of their tumor-suppressing, oncogenic, diagnostic and prognostic biomarker roles in lung cancer. They have also been implicated in regulating cancer cell metabolism and resistance or sensitivity towards chemotherapy and radiotherapy. Further, miRNAs have also been convoluted in regulation of immune checkpoints - Programmed death 1 (PD-1) and its ligand (PD-L1). These molecules play a significant role in tumor immune escape leading to the generation of a microenvironment favouring tumor growth and progression. Therefore, it is imperative to explore the expression of miRNA and understand its relevance in lung cancer and development of anti-cancer strategies (anti - miRs, miR mimics and micro RNA sponges). In view of the above, the role of miRNA in lung cancer has been dissected and the associated mechanisms and pathways are discussed in this review.
Collapse
Affiliation(s)
- Mohammad Askandar Iqbal
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India.
| | - Shweta Arora
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India.
| | - Gopinath Prakasam
- School of Life Sciences, Jawaharlal Nehru University, New Delhi-110067, India.
| | - George A Calin
- Department of Experimental Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX-77030, USA.
| | - Mansoor Ali Syed
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia (A Central University), New Delhi-110025, India.
| |
Collapse
|
234
|
Jiang Y, Seimiya M, Schlumpf TB, Paro R. An intrinsic tumour eviction mechanism in Drosophila mediated by steroid hormone signalling. Nat Commun 2018; 9:3293. [PMID: 30120247 PMCID: PMC6098038 DOI: 10.1038/s41467-018-05794-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 07/11/2018] [Indexed: 12/02/2022] Open
Abstract
Polycomb group proteins are epigenetic regulators maintaining transcriptional memory during cellular proliferation. In Drosophila larvae, malfunction of Polyhomeotic (Ph), a member of the PRC1 silencing complex, results in neoplastic growth. Here, we report an intrinsic tumour suppression mechanism mediated by the steroid hormone ecdysone during metamorphosis. Ecdysone alters neoplastic growth into a nontumorigenic state of the mutant ph cells which then become eliminated during adult stage. We demonstrate that ecdysone exerts this function by inducing a heterochronic network encompassing the activation of the microRNA lethal-7, which suppresses its target gene chronologically inappropriate morphogenesis. This pathway can also promote remission of brain tumours formed in brain tumour mutants, revealing a restraining of neoplastic growth in different tumour types. Given the conserved role of let-7, the identification and molecular characterization of this innate tumour eviction mechanism in flies might provide important clues towards the exploitation of related pathways for human tumour therapy. Drosophila is an excellent model to study both development and tumorigenesis. Here the authors uncover an innate mechanism for a steroid hormone-induced block to tumorigenesis during metamorphosis of Drosophila.
Collapse
Affiliation(s)
- Yanrui Jiang
- D-BSSE, ETH Zürich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Makiko Seimiya
- D-BSSE, ETH Zürich, Mattenstrasse 26, 4058, Basel, Switzerland
| | | | - Renato Paro
- D-BSSE, ETH Zürich, Mattenstrasse 26, 4058, Basel, Switzerland. .,Faculty of Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
235
|
Yin PT, Pongkulapa T, Cho HY, Han J, Pasquale NJ, Rabie H, Kim JH, Choi JW, Lee KB. Overcoming Chemoresistance in Cancer via Combined MicroRNA Therapeutics with Anticancer Drugs Using Multifunctional Magnetic Core-Shell Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2018; 10:26954-26963. [PMID: 30028120 DOI: 10.1021/acsami.8b09086] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this study, we report the use of a multifunctional magnetic core-shell nanoparticle (MCNP), composed of a highly magnetic zinc-doped iron oxide (ZnFe2O4) core nanoparticle and a biocompatible mesoporous silica (mSi) shell, for the simultaneous delivery of let-7a microRNA (miRNA) and anticancer drugs (e.g., doxorubicin) to overcome chemoresistance in breast cancer. Owing to the ability of let-7a to repress DNA repair mechanisms (e.g., BRCA1 and BRCA2) and downregulate drug efflux pumps (e.g., ABCG2), delivery of let-7a could sensitize chemoresistant breast cancer cells (MDA-MB-231) to subsequent doxorubicin chemotherapy both in vitro and in vivo. Moreover, the multifunctionality of our MCNPs allows for the monitoring of in vivo delivery via magnetic resonance imaging. In short, we have developed a multifunctional MCNP-based therapeutic approach to provide an attractive method with which to enhance our ability not only to deliver combined miRNA therapeutics with small-molecule drugs in both selective and effective manner but also to sensitize cancer cells for the enhanced treatment via the combination of miRNA replacement therapy using a single nanoplatform.
Collapse
Affiliation(s)
| | | | - Hyeon-Yeol Cho
- Department of Chemical and Biomolecular Engineering , Sogang University , Seoul 04107 , Republic of Korea
| | - Jiyou Han
- Division of Biotechnology, Laboratory of Stem Cells and Tissue Regeneration, College of Life Sciences and Biotechnology , Korea University , Seoul 02841 , Republic of Korea
- Department of Biological Sciences, Laboratory of Stem Cell Research and Biotechnology , Hyupsung University , Hwaseong-si 18330 , Republic of Korea
| | | | | | - Jong-Hoon Kim
- Division of Biotechnology, Laboratory of Stem Cells and Tissue Regeneration, College of Life Sciences and Biotechnology , Korea University , Seoul 02841 , Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering , Sogang University , Seoul 04107 , Republic of Korea
| | - Ki-Bum Lee
- College of Pharmacy , Kyung Hee University , 26 Kyungheedae-ro , Dongdaemun-gu, Seoul 02447 , Republic of Korea
| |
Collapse
|
236
|
Mortazavi D, Sharifi M. Antiproliferative effect of upregulation of hsa-let-7c-5p in human acute erythroleukemia cells. Cytotechnology 2018; 70:1509-1518. [PMID: 30073438 DOI: 10.1007/s10616-018-0241-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022] Open
Abstract
New achievements in the field of cancer treatment are results of recent advances in molecular medicine and gene therapy. Usage of microRNAs (miRNAs) which are small noncoding RNAs is one of the molecular research lines for the diagnosis and treatment of cancers. miRNAs have an important role in post-transcriptional regulation of the gene expression and are involved in cellular activities such as growth, differentiation, cell death and cancer development. One of the miRNAs that showed downregulation in human acute erythroleukemia is hsa-let-7c-5p. Down-regulation of hsa-let-7c-5p has been reported in in vitro studies of different cancers. In the present study, upregulation of hsa-let-7c-5p is performed in human acute erythroleukemia cell line (KG-1) using miRNA mimic. qRT-PCR, MTT assay, Annexin-V, and propidium iodide staining at different time points after miRNA mimic transfection were accomplished to assess the expression level of hsa-let-7c-5p, cell viability, apoptosis and late apoptosis. In addition, the expression level of PBX2 oncogene, a validated target gene of hsa-let-7c-5p, is evaluated by RT-qPCR to show the effectivity of this approach on erythroleukemia cancer cells. Our results can be used in translational medicine for future investigation in acute erythroleukemia and to approach treatment based on miRNA mimic therapy.
Collapse
Affiliation(s)
- Deniz Mortazavi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, 81744-176, Iran
| | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, 81744-176, Iran.
| |
Collapse
|
237
|
Frankel D, Delecourt V, Harhouri K, De Sandre-Giovannoli A, Lévy N, Kaspi E, Roll P. MicroRNAs in hereditary and sporadic premature aging syndromes and other laminopathies. Aging Cell 2018; 17:e12766. [PMID: 29696758 PMCID: PMC6052405 DOI: 10.1111/acel.12766] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
Hereditary and sporadic laminopathies are caused by mutations in genes encoding lamins, their partners, or the metalloprotease ZMPSTE24/FACE1. Depending on the clinical phenotype, they are classified as tissue‐specific or systemic diseases. The latter mostly manifest with several accelerated aging features, as in Hutchinson–Gilford progeria syndrome (HGPS) and other progeroid syndromes. MicroRNAs are small noncoding RNAs described as powerful regulators of gene expression, mainly by degrading target mRNAs or by inhibiting their translation. In recent years, the role of these small RNAs has become an object of study in laminopathies using in vitro or in vivo murine models as well as cells/tissues of patients. To date, few miRNAs have been reported to exert protective effects in laminopathies, including miR‐9, which prevents progerin accumulation in HGPS neurons. The recent literature has described the potential implication of several other miRNAs in the pathophysiology of laminopathies, mostly by exerting deleterious effects. This review provides an overview of the current knowledge of the functional relevance and molecular insights of miRNAs in laminopathies. Furthermore, we discuss how these discoveries could help to better understand these diseases at the molecular level and could pave the way toward identifying new potential therapeutic targets and strategies based on miRNA modulation.
Collapse
Affiliation(s)
- Diane Frankel
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Service de Biologie Cellulaire; Marseille France
| | | | | | - Annachiara De Sandre-Giovannoli
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Département de Génétique Médicale; Marseille France
| | - Nicolas Lévy
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Département de Génétique Médicale; Marseille France
| | - Elise Kaspi
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Service de Biologie Cellulaire; Marseille France
| | - Patrice Roll
- Aix Marseille Univ; INSERM; MMG; Marseille France
- APHM, Hôpital la Timone; Service de Biologie Cellulaire; Marseille France
| |
Collapse
|
238
|
Silver J, Wadley G, Lamon S. Mitochondrial regulation in skeletal muscle: A role for non‐coding RNAs? Exp Physiol 2018; 103:1132-1144. [PMID: 29885080 DOI: 10.1113/ep086846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Jessica Silver
- Institute for Physical Activity and Nutrition (IPAN) Deakin University Geelong Victoria Australia
| | - Glenn Wadley
- Institute for Physical Activity and Nutrition (IPAN) Deakin University Geelong Victoria Australia
| | - Séverine Lamon
- Institute for Physical Activity and Nutrition (IPAN) Deakin University Geelong Victoria Australia
| |
Collapse
|
239
|
Yao Q, Wang Y, Wang J, Chen S, Liu H, Jiang Z, Zhang X, Liu S, Yuan Q, Zhou X. An Ultrasensitive Diagnostic Biochip Based on Biomimetic Periodic Nanostructure-Assisted Rolling Circle Amplification. ACS NANO 2018; 12:6777-6783. [PMID: 29924598 DOI: 10.1021/acsnano.8b01950] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Developing portable and sensitive devices for point of care detection of low abundance bioactive molecules is highly valuable in early diagnosis of disease. Herein, an ultrasensitive photonic crystals-assisted rolling circle amplification (PCs-RCA) biochip was constructed and further applied to circulating microRNAs (miRNAs) detection in serum. The biochip integrated the optical signal enhancement capability of biomimetic PCs surface with the thousand-fold signal amplification feature of RCA. The biomimetic PCs displayed periodic dielectric nanostructure and significantly enhanced the signal intensity of RCA reaction, leading to efficient improvement of detection sensitivity. A limit of detection (LOD) as low as 0.7 aM was obtained on the PCs-RCA biochip, and the LOD was 7 orders of magnitude lower than that of standard RCA. Moreover, the PCs-RCA biochip could discriminate a single base variation in miRNAs. Accurate quantification of ultralow-abundance circulating miRNAs in clinical serum samples was further achieved with the PCs-RCA biochip, and patients with the nonsmall cell lung carcinoma were successfully distinguished from healthy donors. The PCs-RCA biochip can detect bioactive molecules with ultrahigh sensitivity and good specificity, making it valuable in clinical disease diagnosis and health assessment.
Collapse
Affiliation(s)
- Qian Yao
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education) , Wuhan University , Wuhan 430072 , China
| | - Yingqian Wang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education) , Wuhan University , Wuhan 430072 , China
| | - Jie Wang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education) , Wuhan University , Wuhan 430072 , China
| | - Shaomin Chen
- Center for Gene Diagnosis , Zhongnan Hospital of Wuhan University, Wuhan University , Wuhan 430072 , China
| | - Haoyang Liu
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education) , Wuhan University , Wuhan 430072 , China
| | - Zhuoran Jiang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education) , Wuhan University , Wuhan 430072 , China
| | - Xiaoe Zhang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education) , Wuhan University , Wuhan 430072 , China
| | - Songmei Liu
- Center for Gene Diagnosis , Zhongnan Hospital of Wuhan University, Wuhan University , Wuhan 430072 , China
| | - Quan Yuan
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education) , Wuhan University , Wuhan 430072 , China
| | - Xiang Zhou
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education) , Wuhan University , Wuhan 430072 , China
| |
Collapse
|
240
|
Li Z, Pan W, Shen Y, Chen Z, Zhang L, Zhang Y, Luo Q, Ying X. IGF1/IGF1R and microRNA let-7e down-regulate each other and modulate proliferation and migration of colorectal cancer cells. Cell Cycle 2018; 17:1212-1219. [PMID: 29886785 DOI: 10.1080/15384101.2018.1469873] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
MicroRNA let-7 has been reported to be down-regulated in several human cancers and is now characterized as a tumor suppressor. IGF1R is over-expressed in many cancers and IGF1/IGF1R pathway is attractive target for anticancer therapy. However, the crosstalk between let-7 and IGF1/IGF1R are largely unknown. The present study showed IGF1R were significantly over-expressed in colorectal cancer tissues compared with adjacent normal tissues through immunohistochemical analysis. qRT-PCR results showed that let-7a, let-7b and let-7e were down-regulated in colorectal cancer tissues. Bioinformatics analysis revealed that both IGF1 and IGF1R mRNA are potential targets for let-7 miRNA family. Ectopic transfection of let-7e led to a significant reduction in IGF1R at protein level and their downstream Akt inhibition, as well as a reduction in cell proliferation, migration and invasion in colorectal cancer cells, while inhibition of let-7e enhanced the expression of IGF1R. On the other hand, IGF1 stimulation can significantly down-regulate the expression of let-7e in colorectal cancer cells. Taken together, our findings identify a negative feedback regulation between let-7e and IGF1/IGF1R, and suggest that let-7e could be used in IGF1R-targeted therapeutics in anticancer therapy. ABBREVIATIONS IGF1: insulin-like growth factor 1; IGF1R: IGF1 receptor; miRNA: microRNA; CRC: colorectal cancer; EGFR: epidermal growth factor receptor; HRP: horseradish peroxidase; MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide; p-Akt: phospho-Akt; PI3K: phosphoinositide 3-kinase; qRT-PCR: quantitative reverse transcription-PCR; IHC: immunohistochemical; siRNA: small interfering RNA; 3'-UTR: 3'-untranslated region.
Collapse
Affiliation(s)
- Zhenjun Li
- a Department of Colorectal Surgery , Shaoxing People's Hospital , Shaoxing , China
| | - Weihuo Pan
- a Department of Colorectal Surgery , Shaoxing People's Hospital , Shaoxing , China
| | - Yi Shen
- a Department of Colorectal Surgery , Shaoxing People's Hospital , Shaoxing , China
| | - Zhiliang Chen
- a Department of Colorectal Surgery , Shaoxing People's Hospital , Shaoxing , China
| | - Lihua Zhang
- a Department of Colorectal Surgery , Shaoxing People's Hospital , Shaoxing , China
| | - Yuping Zhang
- a Department of Colorectal Surgery , Shaoxing People's Hospital , Shaoxing , China
| | - Quan Luo
- b Digestive Endoscopy Center , Shaoxing People's Hospital , Shaoxing , China
| | - Xiaojiang Ying
- a Department of Colorectal Surgery , Shaoxing People's Hospital , Shaoxing , China
| |
Collapse
|
241
|
Xu L, Li T, Ding W, Cao Y, Ge X, Wang Y. Combined seven miRNAs for early hepatocellular carcinoma detection with chronic low-dose exposure to microcystin-LR in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 628-629:271-281. [PMID: 29438936 DOI: 10.1016/j.scitotenv.2018.02.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/31/2018] [Accepted: 02/02/2018] [Indexed: 06/08/2023]
Abstract
Aberrant miRNA expression has been detected in various tumor tissues, which may be considered as a marker for early cancer diagnosis. One miRNA has multiple downstream target genes, which can be regulated by multiple upstream other miRNAs. Hence, this dynamic regulation is likely characterized by volatility, and thus, finding the appropriate time point for tests becomes essential for the use of miRNAs as an early marker of tumor diagnosis. In this study, we established a chronic liver cancer progression model in mice by using low doses of the harmful substance microcystin-LR (MC-LR). On the basis of miRNAs microarray assay, we further tested seven miRNAs that showed characteristic expression changes in pre-hepatocarcinogenesis. Our results showed that the levels of four miRNAs (miR-122-5p, miR-125-5p, miR-199a-5p, and miR-503-5p) decreased dramatically, whereas those of two miRNAs (miR-222-5p and miR-590-5p) increased significantly in the early stages, which were all accompanied by an increase in atypia of hepatocytes. MiR-490-5p was a sensitive molecular, suitable only for evaluation of pathological changes in young mice. Therefore the combination the seven of miRNAs for a set may prove to be an effective method in healthy assessment of environmental toxicants for detection of hepatocarcinogenesis caused by hazardous materials.
Collapse
Affiliation(s)
- Lizhi Xu
- Basic Medical Education Center, Nanjing University School of Medicine, Nanjing, Jiangsu 210093, People's Republic of China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing, Jiangsu 210093, People's Republic of China.
| | - Tianfeng Li
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, Jiangsu 210093, People's Republic of China; Center for Reproductive Medicine, The Affiliated Shenzhen City Maternity and Child Healthcare Hospital of Southern Medical University, Shenzhen, Guangdong 518017, People's Republic of China
| | - Weidong Ding
- Basic Medical Education Center, Nanjing University School of Medicine, Nanjing, Jiangsu 210093, People's Republic of China
| | - Yu Cao
- Basic Medical Education Center, Nanjing University School of Medicine, Nanjing, Jiangsu 210093, People's Republic of China
| | - Xiaolong Ge
- Basic Medical Education Center, Nanjing University School of Medicine, Nanjing, Jiangsu 210093, People's Republic of China
| | - Yaping Wang
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, Jiangsu 210093, People's Republic of China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing, Jiangsu 210093, People's Republic of China
| |
Collapse
|
242
|
MiRAR-miRNA Activity Reporter for Living Cells. Genes (Basel) 2018; 9:genes9060305. [PMID: 29921790 PMCID: PMC6027049 DOI: 10.3390/genes9060305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/08/2018] [Accepted: 06/15/2018] [Indexed: 12/18/2022] Open
Abstract
microRNA (miRNA) activity and regulation are of increasing interest as new therapeutic targets. Traditional approaches to assess miRNA levels in cells rely on RNA sequencing or quantitative PCR. While useful, these approaches are based on RNA extraction and cannot be applied in real-time to observe miRNA activity with single-cell resolution. We developed a green fluorescence protein (GFP)-based reporter system that allows for a direct, real-time readout of changes in miRNA activity in live cells. The miRNA activity reporter (MiRAR) consists of GFP fused to a 3′ untranslated region containing specific miRNA binding sites, resulting in miRNA activity-dependent GFP expression. Using qPCR, we verified the inverse relationship of GFP fluorescence and miRNA levels. We demonstrated that this novel optogenetic reporter system quantifies cellular levels of the tumor suppressor miRNA let-7 in real-time in single Human embryonic kidney 293 (HEK 293) cells. Our data shows that the MiRAR can be applied to detect changes in miRNA levels upon disruption of miRNA degradation pathways. We further show that the reporter could be adapted to monitor another disease-relevant miRNA, miR-122. With trivial modifications, this approach could be applied across the miRNome for quantification of many specific miRNA in cell cultures, tissues, or transgenic animal models.
Collapse
|
243
|
Xie L, Xu J. Role of MiR-98 and Its Underlying Mechanisms in Systemic Lupus Erythematosus. J Rheumatol 2018; 45:1397-1405. [DOI: 10.3899/jrheum.171290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2018] [Indexed: 12/12/2022]
Abstract
Objective.T-lymphocyte apoptosis plays a critical role in the pathogenesis of systemic lupus erythematosus (SLE). However, the underlying regulatory mechanisms of apoptosis in SLE remain unclear. The aim of this study was to explore the role of miR-98 in SLE and its underlying mechanisms.Methods.Western blotting and quantitative reverse transcription PCR (qRT-PCR) were used to analyze miR-98 and Fas expression. Luciferase reporter assays were performed to identify miR-98 targets. To modify miRNA levels, miR-98 mimics and inhibitor were transfected into cells. A lentiviral construct was used to overexpress the level of Fas in SLE CD4+ T cells. Gene and protein expression were determined by qRT-PCR and Western blotting. Apoptosis levels were evaluated by annexin V staining and flow cytometry.Results.Compared to those of healthy donors, miR-98 was downregulated in SLE CD4+ T cells, whereas Fas mRNA and protein expression were upregulated. Upregulation of miR-98 by mimic transfection protected Jurkat cells against Fas-mediated apoptosis at both mRNA and protein levels, while miR-98 inhibitor induced the completely opposite effect. Luciferase reporter assays demonstrated that miR-98 directly targeted Fas mRNA. Further, miR-98 inhibitor induced apoptosis in primary healthy CD4+ T cells through the Fas-caspase axis, while upregulation of miR-98 in SLE CD4+ T cells led to the opposite effect.Conclusion.The current study revealed that downregulation of miR-98 induces apoptosis by modulating the Fas-mediated apoptotic signaling pathway in SLE CD4+ T cells. These results suggest that miR-98 might serve as a potential target for SLE treatment.
Collapse
|
244
|
Macedo F, Ladeira K, Longatto-Filho A, Martins SF. Editor’s Pick: Pyruvate Kinase and Gastric Cancer: A Potential Marker. EUROPEAN MEDICAL JOURNAL 2018. [DOI: 10.33590/emj/10313567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Gastric cancer is the second most common cause of cancer-related deaths worldwide, and the 5-year overall survival rate for advanced gastric cancer is ≤25%. Metabolism is a critical process for maintaining growth and other functions in cancer cells; in these cells, the metabolic process shifts from oxidative phosphorylation to aerobic glycolysis and the expression of pyruvate kinase (PK) splice isoform M2 (PKM2) is upregulated. A PubMed search focussing on PK in gastric cancer was conducted and 32 articles were initially collected; 12 articles were subsequently excluded from this review. PKM2 is responsible for tumour growth and invasion and correlates with short survival times and cancer differentiation. Pyruvate dehydrogenase kinase 1 is associated with cell proliferation, lymph node metastasis, and invasion. Measurement of PKM2 or pyruvate dehydrogenase kinase 1 in the blood or stools could be a good marker for gastric cancer in combination with the glycoprotein CA72-4. The review arose from the need for new biomarkers in the management of gastric cancer and had the primary objective of determining whether PK could be used as a marker to diagnose and monitor gastric cancer.
Collapse
Affiliation(s)
- Filipa Macedo
- Portuguese Oncology Institute – Coimbra, Coimbra, Portugal
| | - Kátia Ladeira
- Portuguese Oncology Institute – Lisbon, Lisbon, Portugal; Life and Health Science Research Institute, School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B’s-PT Government Associate Laboratory, Braga, Portugal
| | - Adhemar Longatto-Filho
- Life and Health Science Research Institute, School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B’s-PT Government Associate Laboratory, Braga, Portugal 5. Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo, Brazil; Laboratory of Medical Investigation 14, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Sandra F. Martins
- Life and Health Science Research Institute, School of Health Sciences, University of Minho, Braga, Portugal; ICVS/3B’s-PT Government Associate Laboratory, Braga, Portugal; Surgery Department, Coloproctology Unit, Braga Hospital, Braga, Portugal
| |
Collapse
|
245
|
Yang Y, Pan JJ, Zhou XG, Zhou XY, Cheng R. Differentially expressed miRNAs in premature infants with retinopathy-a bioinformatics analysis. Int J Ophthalmol 2018; 11:773-779. [PMID: 29862174 DOI: 10.18240/ijo.2018.05.09] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 01/29/2018] [Indexed: 12/16/2022] Open
Abstract
AIM To reveal the role of miRNAs in retinopathy of prematurity (ROP) by bioinformatics analysis. METHODS The raw data of this study came from the researches of Wang et al and Zhao et al who analyzed the microRNA (miRNA) expression profile between ROP and controls. Based on the identified differentially expressed miRNAs, the related target genes, lncRNA and circRNA were predicted. Then we performed functional enrichment analysis to further analyze the functions of target genes. RESULTS Hsa-miRNA-128-3p and hsa-miRNA-9-5p showed significantly different expression in both studies. LncRNA of POLDIP2, GAS5, NEFL and UHRF1, circRNA of ZNF280C_hsa_circ_001211 and SIAE_hsa_circ_002083, tar-get gene of QKI showed meaningful differential expression in ROP. Enrichment analysis showed that TGF-β signaling pathway, PI3K-Akt signaling pathway and MAPK signaling pathway might play important roles in the prog-ress of ROP. CONCLUSION This research may provide a comprehensive bioinformatics analysis of differentially expressed miRNAs which are possibly involved in ROP.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neonatology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Jing-Jing Pan
- Department of Neonatology, Jiangsu Provincial People's Hospital Affiliated to Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Xiao-Guang Zhou
- Department of Neonatology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Xiao-Yu Zhou
- Department of Neonatology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Rui Cheng
- Department of Neonatology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
246
|
Bulgakova O, Zhabayeva D, Kussainova A, Pulliero A, Izzotti A, Bersimbaev R. miR-19 in blood plasma reflects lung cancer occurrence but is not specifically associated with radon exposure. Oncol Lett 2018; 15:8816-8824. [PMID: 29805621 PMCID: PMC5950512 DOI: 10.3892/ol.2018.8392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022] Open
Abstract
Radon is one of the most powerful carcinogens, particularly in terms of lung cancer onset and development. miRNAs may be considered not only as markers of the ongoing tumorigenesis but also as a hallmark of exposure to radiation, including radon and its progeny. Therefore, the purpose of the present study was to estimate the value of plasma miR-19b-3p level as the prospective marker of the response to radon exposure in lung cancer pathogenesis. A total of 136 subjects were examined, including 49 radon-exposed patients with lung cancer, 37 patients with lung cancer without radon exposure and 50 age/sex matched healthy controls. Total RNA from blood samples was extracted and used to detect miR-19b-3p expression via reverse transcription quantitative-polymerase chain reaction. The 2-ΔΔCq method was used to quantify the amount of relative miRNA. The plasma level of p53 protein was determined using a Human p53 ELISA kit. Plasma miR-19b-3p level was significantly higher in the patients with lung cancer groups, compared with the healthy control group (P<0.0001). No other statistically significant differences were determined in the expression level of plasma miR-19b-3p between patients diagnosed with lung cancer exposed to radon and not exposed to radon. The expression level of free circulating miR-19b-3p was higher in the group of non-smoking patients with lung cancer, compared with smokers with lung cancer. The miR-19b-3p was 1.4-fold higher in non-smokers than in smokers (P<0.05). No association between plasma levels of p53 protein and miR-19b-3p freely circulating in patients with lung cancer was observed. No other statistically significant differences were determined in the plasma p53 protein level between patients diagnosed with lung cancer exposed and not exposed to radon. These results indicated that detection of miR-19b-3p levels in plasma potentially could be exploited as a noninvasive method for the lung cancer diagnostics. However, this miRNA is not suitable as the precise marker for radon impact.
Collapse
Affiliation(s)
- Olga Bulgakova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Akmola 010008, Kazakhstan
| | - Dinara Zhabayeva
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Akmola 010008, Kazakhstan
| | - Assiya Kussainova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Akmola 010008, Kazakhstan
| | - Alessandra Pulliero
- Department of Health Sciences, University of Genoa, Genoa, I-16132 Liguria, Italy
| | - Alberto Izzotti
- Department of Health Sciences, University of Genoa, Genoa, I-16132 Liguria, Italy
| | - Rakhmetkazhi Bersimbaev
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Akmola 010008, Kazakhstan
| |
Collapse
|
247
|
Fang C, Li XP, Chen YX, Wu NY, Yin JY, Zhang W, Zhou HH, Liu ZQ. Functional miRNA variants affect lung cancer susceptibility and platinum-based chemotherapy response. J Thorac Dis 2018; 10:3329-3340. [PMID: 30069329 DOI: 10.21037/jtd.2018.05.145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Platinum-based chemotherapy is widely used as the first-line treatment of lung cancer. MicroRNAs have an important role in lung carcinogenesis and progression. Single-nucleotide polymorphisms (SNPs) in miRNA involved in miRNA biogenesis and structural alteration may affect miRNA expression. In this study, we aimed to investigate the association of functional miRNA variants with the lung cancer susceptibility and platinum-based chemotherapy response. Methods Nine genetic polymorphisms in miR-605, 146a, 149, 196a-2, 27a, 499, 30c-1, 5197 and let-7a-2 were selected with comprehensive collection strategy and genotyped by MALDI-TOF mass spectrometry in a total of 215 health control and 507 lung cancer patients (386 patients received at least two consecutive cycles of platinum-based chemotherapy). Results We found that an allele carriers of miR-146a rs2910164 (P=0.022, OR=1.315) and C allele carriers of miR-149 rs71428439 (P=0.042, OR=1.372) performance a high risk of lung cancer. Mir-30c-1 rs928508 (P=0.005, in recessive model) and let-7a-2 rs629367 (P=0.030 and P=0.021, in additive and dominant models, respectively) showed strong relationship with lung cancer risk in age under 57 years. The rs11614913 (miR-196a-2) C allele or rs9280508 (miR-30c-1) G allele carriers shown more sensitive to platinum both in additive (P=0.010, P=0.022, respectively) and dominant models (P=0.001, P=0.018, respectively). Conclusions These findings suggested that SNPs rs71428439 (miR-149), rs2910164 (miR-146a), rs928508 (mir-30c-1) and rs629367 (let-7a-2) were associated with the lung cancer prevalence, polymorphisms of rs11614913 (miR-196a-2) and rs9280508 (miR-30c-1) significantly influenced the patients' response to platinum-based chemotherapy, which may serve as potential clinical biomarkers to predict lung cancer risk and platinum-based chemotherapy response.
Collapse
Affiliation(s)
- Chao Fang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha 410013, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Xiang-Ping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yi-Xin Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Na-Yiyuan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.,Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.,Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, China
| |
Collapse
|
248
|
Liu G, Niu X, Meng X, Zhang Z. Sensitive miRNA markers for the detection and management of NSTEMI acute myocardial infarction patients. J Thorac Dis 2018; 10:3206-3215. [PMID: 30069316 DOI: 10.21037/jtd.2018.05.141] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background NSTEMI patients will benefit greatly with better biomarker screening to detect and prognose the disease. Using miRNAs, we evaluated the clinical utility in acute myocardial infarction (AMI) patients during disease onset and therapy. Methods A total of 145 NSTEMI patients and 30 healthy volunteers with no history of cardiovascular disease (CVD) were recruited. miRNA levels in plasma were measured during disease manifestation and serially during treatment phase. Levels of multiple candidates (miR-1, miR-133, miR-208, miR-499) were analysed. The miRNA levels were directly compared between NSTEMI and healthy volunteers. Results Cardiac related miRNAs levels demonstrated significant increase compared with healthy controls. miR-499 exhibited the highest elevation with more than 6.03-fold change compared with healthy participants. Conventional cTnT measurements were in good agreement to miRNA relative expressions. In serial measurements, miR-499 demonstrated large fluctuations and could be linked to the secondary complications. In contrast, miR-133 showed insignificant variations in mean levels during serial sampling. Conclusions miRNA is a potentially sensitive biomarker for NSTEMI AMI patients for disease detection and treatment monitoring. The sensitivities were comparable to cTnT for diagnostic accuracy and patients with sustained or higher levels were correlated to secondary complications.
Collapse
Affiliation(s)
- Guoyong Liu
- Heart Center, The First Affiliated Hospital of Lanzhou University, Lanzhou 730000, China.,The Cardiovascular Department, Qinghai Provincial People's Hospital, Xining 810007, China
| | - Xiaowei Niu
- Heart Center, The First Affiliated Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiaoxue Meng
- Heart Center, The First Affiliated Hospital of Lanzhou University, Lanzhou 730000, China
| | - Zheng Zhang
- Heart Center, The First Affiliated Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
249
|
Soliman B, Salem A, Ghazy M, Abu-Shahba N, El Hefnawi M. Bioinformatics functional analysis of let-7a, miR-34a, and miR-199a/b reveals novel insights into immune system pathways and cancer hallmarks for hepatocellular carcinoma. Tumour Biol 2018; 40:1010428318773675. [PMID: 29775159 DOI: 10.1177/1010428318773675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Let-7a, miR-34a, and miR-199 a/b have gained a great attention as master regulators for cellular processes. In particular, these three micro-RNAs act as potential onco-suppressors for hepatocellular carcinoma. Bioinformatics can reveal the functionality of these micro-RNAs through target prediction and functional annotation analysis. In the current study, in silico analysis using innovative servers (miRror Suite, DAVID, miRGator V3.0, GeneTrail) has demonstrated the combinatorial and the individual target genes of these micro-RNAs and further explored their roles in hepatocellular carcinoma progression. There were 87 common target messenger RNAs (p ≤ 0.05) that were predicted to be regulated by the three micro-RNAs using miRror 2.0 target prediction tool. In addition, the functional enrichment analysis of these targets that was performed by DAVID functional annotation and REACTOME tools revealed two major immune-related pathways, eight hepatocellular carcinoma hallmarks-linked pathways, and two pathways that mediate interconnected processes between immune system and hepatocellular carcinoma hallmarks. Moreover, protein-protein interaction network for the predicted common targets was obtained by using STRING database. The individual analysis of target genes and pathways for the three micro-RNAs of interest using miRGator V3.0 and GeneTrail servers revealed some novel predicted target oncogenes such as SOX4, which we validated experimentally, in addition to some regulated pathways of immune system and hepatocarcinogenesis such as insulin signaling pathway and adipocytokine signaling pathway. In general, our results demonstrate that let-7a, miR-34a, and miR-199 a/b have novel interactions in different immune system pathways and major hepatocellular carcinoma hallmarks. Thus, our findings shed more light on the roles of these miRNAs as cancer silencers.
Collapse
Affiliation(s)
- Bangly Soliman
- 1 Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt.,2 Informatics and Systems Department, Biomedical Informatics and Chemo-Informatics Group, Centre of Excellence for Advanced Sciences (CEAS), Division of Engineering Research, National Research Centre, Cairo, Egypt
| | - Ahmed Salem
- 1 Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohamed Ghazy
- 1 Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Nourhan Abu-Shahba
- 3 Stem Cells Research Group, Medical Centre of Excellence, Medical Molecular Genetics Department, National Research Centre, Cairo, Egypt
| | - Mahmoud El Hefnawi
- 2 Informatics and Systems Department, Biomedical Informatics and Chemo-Informatics Group, Centre of Excellence for Advanced Sciences (CEAS), Division of Engineering Research, National Research Centre, Cairo, Egypt.,4 Centre for Informatics, Nile University, Sheikh Zayed City, Egypt
| |
Collapse
|
250
|
Tetik Vardarlı A, Düzgün Z, Erdem C, Kaymaz BT, Eroglu Z, Çetintas VB. Matrine induced G0/G1 arrest and apoptosis in human acute T-cell lymphoblastic leukemia (T-ALL) cells. Bosn J Basic Med Sci 2018; 18:141-149. [PMID: 29045804 DOI: 10.17305/bjbms.2017.2457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/13/2022] Open
Abstract
Matrine, a natural product extracted from the root of Sophora flavescens, is a promising alternative drug in different types of cancer. Here, we aimed to investigate the therapeutic effects and underlying molecular mechanisms of matrine on human acute lymphoblastic leukemia (ALL) cell line, CCRF-CEM. Cell viability and IC50 values were determined by WST-1 cell cytotoxicity assay. Cell cycle distribution and apoptosis rates were analyzed by flow cytometry. Expression patterns of 44 selected miRNAs and 44 RNAs were analyzed by quantitative reverse transcription polymerase chain reaction (qRT-PCR) using the Applied Biosystems 7500 Fast Real-Time PCR System. Matrine inhibited cell viability and induced apoptosis of CCRF-CEM cells in a dose-dependent manner. Cell cycle analysis demonstrated that matrine-treated CCRF-CEM cells significantly accumulated in the G0/G1 phase compared with the untreated control cells. hsa-miR-376b-3p (-37.09 fold, p = 0.008) and hsa-miR-106b-3p (-16.67 fold, p = 0.028) expressions were decreased, whereas IL6 (95.47 fold, p = 0.000011) and CDKN1A (140.03 fold, p = 0.000159) expressions were increased after matrine treatment. Our results suggest that the downregulation of hsa-miR-106b-3p leads to the upregulation of target p21 gene, CDKN1A, and plays a critical role in the cell cycle progression by arresting matrine-treated cells in the G0/G1 phase.
Collapse
|