51
|
Kornmueller K, Amri EZ, Scheideler M, Prassl R. Delivery of miRNAs to the adipose organ for metabolic health. Adv Drug Deliv Rev 2022; 181:114110. [PMID: 34995679 DOI: 10.1016/j.addr.2021.114110] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/14/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022]
Abstract
Despite the increasing prevalence of obesity and diabetes, there is no efficient treatment to combat these epidemics. The adipose organ is the main site for energy storage and plays a pivotal role in whole body lipid metabolism and energy homeostasis, including remodeling and dysfunction of adipocytes and adipose tissues in obesity and diabetes. Thus, restoring and balancing metabolic functions in the adipose organ is in demand. MiRNAs represent a novel class of drugs and drug targets, as they are heavily involved in the regulation of many cellular and metabolic processes and diseases, likewise in adipocytes. In this review, we summarize key regulatory activities of miRNAs in the adipose organ, discuss various miRNA replacement and inhibition strategies, promising delivery systems for miRNAs and reflect the future of novel miRNA-based therapeutics to target adipose tissues with the ultimate goal to combat metabolic disorders.
Collapse
Affiliation(s)
- Karin Kornmueller
- Department of Biophysics, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | | | - Marcel Scheideler
- Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ruth Prassl
- Department of Biophysics, Gottfried Schatz Research Center, Medical University of Graz, Austria.
| |
Collapse
|
52
|
Havasi A, Sur D, Cainap SS, Lungulescu CV, Gavrilas LI, Cainap C, Vlad C, Balacescu O. Current and New Challenges in the Management of Pancreatic Neuroendocrine Tumors: The Role of miRNA-Based Approaches as New Reliable Biomarkers. Int J Mol Sci 2022; 23:1109. [PMID: 35163032 PMCID: PMC8834851 DOI: 10.3390/ijms23031109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 12/17/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PanNETs) are rare tumors; however, their incidence greatly increases with age, and they occur more frequently among the elderly. They represent 5% of all pancreatic tumors, and despite the fact that low-grade tumors often have an indolent evolution, they portend a poor prognosis in an advanced stages and undifferentiated tumors. Additionally, functional pancreatic neuroendocrine tumors greatly impact quality of life due to the various clinical syndromes that result from abnormal hormonal secretion. With limited therapeutic and diagnostic options, patient stratification and selection of optimal therapeutic strategies should be the main focus. Modest improvements in the management of pancreatic neuroendocrine tumors have been achieved in the last years. Therefore, it is imperative to find new biomarkers and therapeutic strategies to improve patient survival and quality of life, limiting the disease burden. MicroRNAs (miRNAs) are small endogenous molecules that modulate the expression of thousands of genes and control numerous critical processes involved in tumor development and progression. New data also suggest the implication of miRNAs in treatment resistance and their potential as prognostic or diagnostic biomarkers and therapeutic targets. In this review, we discusses the current and new challenges in the management of PanNETs, including genetic and epigenetic approaches. Furthermore, we summarize the available data on miRNAs as potential prognostic, predictive, or diagnostic biomarkers and discuss their function as future therapeutic targets.
Collapse
Affiliation(s)
- Andrei Havasi
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.C.)
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
- MedEuropa Radiotherapy Center, 410191 Oradea, Romania
| | - Daniel Sur
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.C.)
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
| | - Simona Sorana Cainap
- Department of Mother and Child, Pediatric Cardiology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
| | | | - Laura-Ioana Gavrilas
- Department of Bromatology, Hygiene, Nutrition, University of Medicine and Pharmacy “Iuliu Hatieganu”, 23 Marinescu Street, 400337 Cluj-Napoca, Romania;
| | - Calin Cainap
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania; (A.H.); (C.C.)
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
| | - Catalin Vlad
- Department of Surgery, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34–36, Republicii Street, 400015 Cluj-Napoca, Romania;
- Department of Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8, Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Ovidiu Balacescu
- 11th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400015 Cluj-Napoca, Romania;
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta’’, 400015 Cluj-Napoca, Romania
| |
Collapse
|
53
|
Park JW, Kim Y, Lee SB, Oh CW, Lee EJ, Ko JY, Park JH. Autophagy inhibits cancer stemness in triple-negative breast cancer via miR-181a-mediated regulation of ATG5 and/or ATG2B. Mol Oncol 2022; 16:1857-1875. [PMID: 35029026 PMCID: PMC9067148 DOI: 10.1002/1878-0261.13180] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022] Open
Abstract
Autophagy has a dual role in the maintenance of cancer stem cells (CSCs), but the precise relationship between autophagy and cancer stemness requires further investigation. In this study, it was found that luminal and triple‐negative breast cancers require distinct therapeutic approaches because of their different amounts of autophagy flux. We identified that autophagy flux was inhibited in triple‐negative breast cancer (TNBC) CSCs. Moreover, miRNA‐181a (miR‐181a) expression is upregulated in both TNBC CSCs and patient tissues. Autophagy‐related 5 (ATG5) and autophagy‐related 2B (ATG2B) participate in the early formation of autophagosomes and were revealed as targets of miR‐181a. Inhibition of miR‐181a expression led to attenuation of TNBC stemness and an increase in autophagy flux. Furthermore, treatment with curcumin led to attenuation of cancer stemness in TNBC CSCs; the expression of ATG5 and ATG2B was enhanced and there was an increase of autophagy flux. These results indicated that ATG5 and ATG2B are involved in the suppression of cancer stemness in TNBC. In summary, autophagy inhibits cancer stemness through the miR‐181a‐regulated mechanism in TNBC. Promoting tumor‐suppressive autophagy using curcumin may be a potential method for the treatment of TNBC.
Collapse
Affiliation(s)
- Jee Won Park
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Yesol Kim
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Soo-Been Lee
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Chae Won Oh
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Eun Ji Lee
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Je Yeong Ko
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jong Hoon Park
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| |
Collapse
|
54
|
Teerapakpinyo C, Areeruk W, Tantbirojn P, Phupong V, Shuangshoti S, Lertkhachonsuk R. MicroRNA Expression Profiling in Hydatidiform Mole for the Prediction of Postmolar GTN : MicroRNA Profile in Postmolar GTN. Technol Cancer Res Treat 2022; 21:15330338211067309. [PMID: 35023789 PMCID: PMC8785350 DOI: 10.1177/15330338211067309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Objectives: The primary aim of the study was to identify miRNAs that were differentially expressed between complete hydatidiform moles (CHMs) that turned out to be gestational trophoblastic neoplasia (GTN) [GTN moles] and CHMs that regressed spontaneously after evacuation [remission moles]. The secondary aim was to study the profiles of miRNA expressions in CHMs. Methods: A case-control study was conducted on GTN moles and remission moles. We quantitatively assessed the expression of 800 human miRNAs from molar tissues using Nanostring nCounter. Results: From a pilot study, 21 miRNAs were significantly downregulated in GTN moles compared to the remission moles. Five of them (miR-566, miR-608, miR-1226-3p, miR-548ar-3p and miR-514a-3p) were downregulated for >4 folds. MiR-608 was selected as a candidate for further analysis on 18 CHMs (9 remission moles and 9 GTN moles) due to its striking association with malignant formation. MiR-608 expression was slightly lower in GTN moles compared to the remission moles, that is, 2.22 folds change [p = 0.063]. Conclusion: We identified 21 miRNAs that were differentially expressed between GTN moles and remission moles suggesting that miRNA profiles can distinguish between the two groups. Although not reaching statistically significant, miR-608 expression was slightly lower in GTN moles compared to remission moles.
Collapse
Affiliation(s)
| | - Wilasinee Areeruk
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Patou Tantbirojn
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Vorapong Phupong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Shanop Shuangshoti
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ruangsak Lertkhachonsuk
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
55
|
Tu C, Wei L, Wang L, Tang Y. Eight Differential miRNAs in DN Identified by Microarray Analysis as Novel Biomarkers. Diabetes Metab Syndr Obes 2022; 15:907-920. [PMID: 35359345 PMCID: PMC8961165 DOI: 10.2147/dmso.s355783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/11/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is the common cause of renal diseases such as end-stage renal disease (ESRD) and chronic kidney disease (CKD). Various diagnostic applications and treatment methods are used for clinical but remain some prognosis issues. To avoid morbidity and mortality related to DN, early detection of disease complications as well as targeted therapeutic strategies is essential. Considerable evidence indicates that non-coding RNA plays a vital role in the biological processes of various diseases, used as biomarkers and therapeutic targets. And the most known ncRNAs are the microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs). MATERIALS AND METHODS Our study aimed to identify potential prognostic ncRNAs involved in DN by bioinformatics analysis and validated expression levels through quantitative polymerase chain reaction (qPCR) and GEO database. Our research focuses on differential expression miRNAs (DEmiRNAs) in DN and their interactions with critical genes. RESULTS We identified 8 up-regulated DEmiRNAs, including miR-103a-2-5p, miR-297, miR-548x-3p, miR-604, miR-644a, miR-1256, miR-3911 and miR-5047 finally. We further validated these miRNAs in a murine model. CONCLUSION Identifying these up-regulated genes and elucidating these miRNAs regulatory network will contribute to a better understanding of the molecular mechanism of DN and how they can be used as new biomarkers and potential therapeutic targets for DN.
Collapse
Affiliation(s)
- Chao Tu
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, People’s Republic of China
| | - Lan Wei
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, People’s Republic of China
| | - Liangzhi Wang
- Department of Internal Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, People’s Republic of China
| | - Ying Tang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213000, People’s Republic of China
- Correspondence: Ying Tang, Department of Rehabilitation Medicine, The Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, Jiangsu, 213000, People’s Republic of China, Tel +86 0519 68872146, Email
| |
Collapse
|
56
|
Alemohammad H, Najafzadeh B, Asadzadeh Z, Baghbanzadeh A, Ghorbaninezhad F, Najafzadeh A, Safarpour H, Bernardini R, Brunetti O, Sonnessa M, Fasano R, Silvestris N, Baradaran B. The importance of immune checkpoints in immune monitoring: A future paradigm shift in the treatment of cancer. Biomed Pharmacother 2021; 146:112516. [PMID: 34906767 DOI: 10.1016/j.biopha.2021.112516] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
The growth and development of cancer are directly correlated to the suppression of the immune system. A major breakthrough in cancer immunotherapy depends on various mechanisms to detect immunosuppressive factors that inhibit anti-tumor immune responses. Immune checkpoints are expressed on many immune cells such as T-cells, regulatory B cells (Bregs), dendritic cells (DCs), natural killer cells (NKs), regulatory T (Tregs), M2-type macrophages, and myeloid-derived suppressor cells (MDSCs). Immune inhibitory molecules, including CTLA-4, TIM-3, TIGIT, PD-1, and LAG-3, normally inhibit immune responses via negatively regulating immune cell signaling pathways to prevent immune injury. However, the up-regulation of inhibitory immune checkpoints during tumor progression on immune cells suppresses anti-tumor immune responses and promotes immune escape in cancer. It has recently been indicated that cancer cells can up-regulate various pathways of the immune checkpoints. Therefore, targeting immune inhibitory molecules through antibodies or miRNAs is a promising therapeutic strategy and shows favorable results. Immune checkpoint inhibitors (ICIs) are introduced as a new immunotherapy strategy that enhance immune cell-induced antitumor responses in many patients. In this review, we highlighted the function of each immune checkpoint on different immune cells and therapeutic strategies aimed at using monoclonal antibodies and miRNAs against inhibitory receptors. We also discussed current challenges and future strategies for maximizing these FDA-approved immunosuppressants' effectiveness and clinical success in cancer treatment.
Collapse
Affiliation(s)
- Hajar Alemohammad
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Basira Najafzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Arezoo Najafzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, Catania, Italy
| | - Oronzo Brunetti
- Medical Oncological Unite, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy
| | - Margherita Sonnessa
- Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Rossella Fasano
- Medical Oncological Unite, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy
| | - Nicola Silvestris
- Medical Oncological Unite, IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
57
|
Rencelj A, Gvozdenovic N, Cemazar M. MitomiRs: their roles in mitochondria and importance in cancer cell metabolism. Radiol Oncol 2021; 55:379-392. [PMID: 34821131 PMCID: PMC8647792 DOI: 10.2478/raon-2021-0042] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/28/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are short non-coding RNAs that play important roles in almost all biological pathways. They regulate post-transcriptional gene expression by binding to the 3'untranslated region (3'UTR) of messenger RNAs (mRNAs). MitomiRs are miRNAs of nuclear or mitochondrial origin that are localized in mitochondria and have a crucial role in regulation of mitochondrial function and metabolism. In eukaryotes, mitochondria are the major sites of oxidative metabolism of sugars, lipids, amino acids, and other bio-macromolecules. They are also the main sites of adenosine triphosphate (ATP) production. CONCLUSIONS In the review, we discuss the role of mitomiRs in mitochondria and introduce currently well studied mitomiRs, their target genes and functions. We also discuss their role in cancer initiation and progression through the regulation of mRNA expression in mitochondria. MitomiRs directly target key molecules such as transporters or enzymes in cell metabolism and regulate several oncogenic signaling pathways. They also play an important role in the Warburg effect, which is vital for cancer cells to maintain their proliferative potential. In addition, we discuss how they indirectly upregulate hexokinase 2 (HK2), an enzyme involved in glucose phosphorylation, and thus may affect energy metabolism in breast cancer cells. In tumor tissues such as breast cancer and head and neck tumors, the expression of one of the mitomiRs (miR-210) correlates with hypoxia gene signatures, suggesting a direct link between mitomiR expression and hypoxia in cancer. The miR-17/92 cluster has been shown to act as a key factor in metabolic reprogramming of tumors by regulating glycolytic and mitochondrial metabolism. This cluster is deregulated in B-cell lymphomas, B-cell chronic lymphocytic leukemia, acute myeloid leukemia, and T-cell lymphomas, and is particularly overexpressed in several other cancers. Based on the current knowledge, we can conclude that there is a large number of miRNAs present in mitochondria, termed mitomiR, and that they are important regulators of mitochondrial function. Therefore, mitomiRs are important players in the metabolism of cancer cells, which need to be further investigated in order to develop a potential new therapies for cancer.
Collapse
Affiliation(s)
- Andrej Rencelj
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Nada Gvozdenovic
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Ljubljana, Slovenia
| | - Maja Cemazar
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| |
Collapse
|
58
|
Gubu A, Su W, Zhao X, Zhang X, Fan X, Wang J, Wang Q, Tang X. Circular Antisense Oligonucleotides for Specific RNase-H-Mediated microRNA Inhibition with Reduced Off-Target Effects and Nonspecific Immunostimulation. J Med Chem 2021; 64:16046-16055. [PMID: 34672619 DOI: 10.1021/acs.jmedchem.1c01421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Antisense microRNA oligodeoxynucleotides (AMOs) are powerful tools to regulate microRNA functions. Unfortunately, severe off-target effects are sometimes observed. Due to the special topological and enzymatic properties of circular oligodeoxynucleotides (c-ODNs), we rationally designed and developed circular AMOs, which effectively inhibited microRNA functions with high target specificity and low off-target effects. Binding and enzymatic assays indicated that small circular AMOs could selectively bind to and further digest the target mature miR 21, which suggested that the topological properties of circular c-ODNs significantly decreased their off-target effects as microRNA inhibitors. Compared with their linear corresponding phosphorothioated AMOs, circular phosphorothioated AMOs could more effectively reduce the amount of carcinogenic miR 21 and miR 222 and upregulate the expression levels of downstream antitumor proteins of PTEN and PDCD4. In addition, c-PS-antimiRs induced much less nonspecific immunostimulatory effects compared with their linear partner PS-ODNs, further indicating the advantages of circular ODNs in nonspecific immunostimulation.
Collapse
Affiliation(s)
- Amu Gubu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China.,Chemical Biology Center, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
| | - Wenbo Su
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China.,Chemical Biology Center, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
| | - Xiaoran Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China.,Chemical Biology Center, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
| | - Xueli Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China.,Chemical Biology Center, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
| | - Xinli Fan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China.,Chemical Biology Center, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
| | - Xinjing Tang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, No. 38 Xueyuan Road, Beijing 100191, China.,Chemical Biology Center, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
| |
Collapse
|
59
|
Peng ZT, Gu P. Sulforaphane suppresses autophagy during the malignant progression of gastric carcinoma via activating miR-4521/PIK3R3 pathway. Hum Exp Toxicol 2021; 40:S711-S720. [PMID: 34749521 DOI: 10.1177/09603271211054437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Sulforaphane, which exerts an effective anti-cancer ability, is a phytochemical converted from cruciferous plants. Here, we aimed to identify whether sulforaphane could suppress autophagy during the malignant progression of gastric carcinoma and to explore the underlying mechanisms. METHODS SGC7901 cells were transfected with miR-4521 mimics, inhibitor, and pcDNA3.1-PIK3R3, and treated with sulforaphane or autophagy inhibitor. Cell proliferation, apoptosis, and miR-4521 or PIK3R3 expression were detected. RESULTS MiR-4521 over-expression suppressed LC3-II/I ratio and Beclin-1 expression but induced p62 expression in SGC7901 cells. MiR-4521 also reduced gastric carcinoma cell proliferation and promoted apoptosis in vitro. In the mechanical observation, we identified that miR-4521 directly targeted PIK3R3 to repress its expression, and PIK3R3 up-regulation partly antagonized miR-4521-mediated autophagy, proliferation, and apoptosis in gastric carcinoma cells. In addition, sulforaphane exerted effective anti-cancer functions by repressing autophagy and growth in tumor cells at a concentration-dependent way. MiR-4521 inhibition or PIK3R3 over-expression weakened the anti-cancer functions of sulforaphane in gastric carcinoma cells. CONCLUSION Consequently, miR-4521 suppressed autophagy during the malignant progression of gastric carcinoma by targeting PIK3R3. Thus, miR-4521 may be applied as a therapeutic target for sulforaphane in gastric carcinoma.
Collapse
Affiliation(s)
- Zi-Tan Peng
- Edong Healthcare Group,Department of Clinical Laboratory, Huangshi Central Hospital, 162732Affiliated Hospital of Hubei Polytechnic University, Huangshi, China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, China
| | - Pei Gu
- Edong Healthcare Group,Department of Clinical Laboratory, Huangshi Central Hospital, 162732Affiliated Hospital of Hubei Polytechnic University, Huangshi, China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, China
| |
Collapse
|
60
|
da Silva Gomes PR, Candido P, Ghazarian V, Camargo JA, Guimarães VR, Gonçalves GL, Romão P, Silva IA, Srougi M, Nahas WC, Leite KR, Reis ST, Pimenta R, Viana NI. Can increased expression of miR-Let-7c reduce the transition potential of high-grade urothelial carcinoma? Mol Biol Rep 2021; 48:7947-7952. [PMID: 34708341 DOI: 10.1007/s11033-021-06825-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/08/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Bladder cancer is the leading transitional cell carcinoma affecting men and women with high morbidity and mortality rates, justifying the need to develop new molecular target therapies using microRNAs. This study aimed to evaluate the behavior of the T24 cell line after transfection with miR-Let-7c precursor mimic through invasion, migration, apoptosis, and cell cycle assays. METHODS AND RESULTS: T24 cell was transfected with the Let-7c mimic and its respective control and evaluated after 24 h. The expression levels of miR-Let-7c were analyzed by qPCR. We performed wound healing, Matrigel and flow cytometry, apoptosis, and cell cycle assays to determine its effect on cellular processes. Cells transfected with miR-Let-7c showed increased apoptosis rates (p = 0.019), decreased migration 24 h (p = 0.031) and 48 h (p = 0.0006), invasion potential (p = 0.0007), and cell proliferation (p = 0.002). CONCLUSIONS Our results demonstrate that miR-Let-7c can act in different pathways of the carcinogenic cellular processes of muscle-invasive urothelial carcinoma cells, inhibiting cell proliferation and increasing apoptosis levels, consequently limiting their invasion potential. However, further studies should be carried out better to elucidate this microRNA's role in high-grade urothelial carcinomas and unveil which targets this microRNA may present, which are intrinsically related to the cancer survival pathways.
Collapse
Affiliation(s)
- Paulo Ricardo da Silva Gomes
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
- Faculdade de Medicina, Universidade Federal do Pará, Belém, PA, Brazil
| | - Patricia Candido
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Vitória Ghazarian
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Juliana A Camargo
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Vanessa R Guimarães
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Guilherme L Gonçalves
- Laboratory of Renal Physiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Poliana Romão
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Iran A Silva
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Miguel Srougi
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil
| | - William C Nahas
- Uro-Oncology Group, Urology Department, University of Sao Paulo Medical School and Institute of Cancer Estate of Sao Paulo (ICESP), São Paulo, Brazil
| | - Kátia R Leite
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Sabrina T Reis
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Ruan Pimenta
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil
| | - Nayara Izabel Viana
- Laboratorio de Investigação Médica 55 (LIM55), Faculdade de Medicina, Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil.
- Universidade do Estado de Minas Gerais - UEMG, Passos, MG, Brazil.
| |
Collapse
|
61
|
Magnetic ionic liquids as microRNA extraction solvents and additives for the exponential amplification reaction. Anal Chim Acta 2021; 1181:338900. [PMID: 34556230 DOI: 10.1016/j.aca.2021.338900] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022]
Abstract
The detection of microRNAs (miRNAs) from highly complex matrices has become an area of immense interest as their characterization in biological samples has been utilized for disease diagnosis and body fluid identification. However, conventional northern blotting miRNA detection lacks the sensitivity required to detect circulating miRNAs. Additionally, polymerase chain reaction-based methods for miRNA detection require modified oligonucleotides that are difficult to design. Exponential amplification reaction (EXPAR) is an isothermal amplification method used for miRNA detection that is simple to design but suffers from non-specific amplification that masks low concentration miRNAs. Previous studies have shown that magnetic ionic liquids (MILs) are a promising alternative to traditional nucleic acid extraction methods capable of preconcentrating DNA from complex matrices. In this study, three hydrophobic magnetic ionic liquids (MILs) were investigated as EXPAR additives and miRNA extraction solvents. The addition of MIL to the EXPAR buffer decreased the background signal from non-specific amplification and increased the reaction rate. Reactions containing MIL could detect miRNA at concentration levels down to 10 aM. In comparison, reactions that did not contain MIL could not discriminate 10 fM lethal-7a (let-7a) standards from the no trigger control (NTC). All three MILs extracted miRNA from 2-fold diluted plasma, artificial urine, and artificial saliva with only a 1 min dispersion step. By integrating the miRNA-enriched MIL into the EXPAR buffer, the extraction and detection of femtomolar concentrations of miRNA required only 10 min. In contrast, conventional spin column kits require at least 20 min to isolate miRNA, indicating that a dispersive MIL-based extraction is ideal for high throughput analysis of miRNA.
Collapse
|
62
|
Yamada Y. Nucleic Acid Drugs-Current Status, Issues, and Expectations for Exosomes. Cancers (Basel) 2021; 13:cancers13195002. [PMID: 34638486 PMCID: PMC8508492 DOI: 10.3390/cancers13195002] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Nucleic acid drugs provide novel therapeutic modalities with characteristics that differ from those of small molecules and antibodies. In this review, I focus on the various mechanisms through which nucleic acid drugs act on, the status of their clinical development, and discuss several hurdles that need to be surmounted. In addition, by listing examples of how the progress in exosome biology can lead to the solution of problems in nucleic acid drug therapy, I hope that many more nucleic acid drugs including anticancer drugs will be developed in the future. Abstract Nucleic acid drugs are being developed as novel therapeutic modalities. They have great potential to treat human diseases such as cancers, viral infections, and genetic disorders due to unique characteristics that make it possible to approach undruggable targets using classical small molecule or protein/antibody-based biologics. In this review, I describe the advantages, classification, and clinical status of nucleic acid therapeutics. To date, more than 10 products have been launched, and many products have been tested in clinics. To promote the use of nucleic acid therapeutics such as antibodies, several hurdles need to be surmounted. The most important issue is the delivery of nucleic acids and several other challenges have been reported. Recent advanced delivery platforms are lipid nanoparticles and ligand conjugation approaches. With the progress of exosome biology, exosomes are expected to contribute to the solution of various problems associated with nucleic acid drugs.
Collapse
Affiliation(s)
- Yoji Yamada
- Research Management Office, Research Unit, R&D Division, Kyowa Kirin Co. Ltd., 1-9-2, Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
63
|
Bazrgar M, Khodabakhsh P, Prudencio M, Mohagheghi F, Ahmadiani A. The role of microRNA-34 family in Alzheimer's disease: A potential molecular link between neurodegeneration and metabolic disorders. Pharmacol Res 2021; 172:105805. [PMID: 34371173 DOI: 10.1016/j.phrs.2021.105805] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/27/2021] [Accepted: 08/05/2021] [Indexed: 02/09/2023]
Abstract
Growing evidence indicates that overexpression of the microRNA-34 (miR-34) family in the brain may play a crucial role in Alzheimer's disease (AD) pathogenesis by targeting and downregulating genes associated with neuronal survival, synapse formation and plasticity, Aβ clearance, mitochondrial function, antioxidant defense system, and energy metabolism. Additionally, elevated levels of the miR-34 family in the liver and pancreas promote the development of metabolic syndromes (MetS), such as diabetes and obesity. Importantly, MetS represent a well-documented risk factor for sporadic AD. This review focuses on the recent findings regarding the role of the miR-34 family in the pathogenesis of AD and MetS, and proposes miR-34 as a potential molecular link between both disorders. A comprehensive understanding of the functional roles of miR-34 family in the molecular and cellular pathogenesis of AD brains may lead to the discovery of a breakthrough treatment strategy for this disease.
Collapse
Affiliation(s)
- Maryam Bazrgar
- Neuroscience Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Pariya Khodabakhsh
- Department of Pharmacology, Shahid Beheshti University of Medical Science, Tehran, Iran
| | | | - Fatemeh Mohagheghi
- Institute of Experimental Hematology, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran.
| |
Collapse
|
64
|
Interplay between Epigenetics and Cellular Metabolism in Colorectal Cancer. Biomolecules 2021; 11:biom11101406. [PMID: 34680038 PMCID: PMC8533383 DOI: 10.3390/biom11101406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 01/30/2023] Open
Abstract
Cellular metabolism alterations have been recognized as one of the most predominant hallmarks of colorectal cancers (CRCs). It is precisely regulated by many oncogenic signaling pathways in all kinds of regulatory levels, including transcriptional, post-transcriptional, translational and post-translational levels. Among these regulatory factors, epigenetics play an essential role in the modulation of cellular metabolism. On the one hand, epigenetics can regulate cellular metabolism via directly controlling the transcription of genes encoding metabolic enzymes of transporters. On the other hand, epigenetics can regulate major transcriptional factors and signaling pathways that control the transcription of genes encoding metabolic enzymes or transporters, or affecting the translation, activation, stabilization, or translocation of metabolic enzymes or transporters. Interestingly, epigenetics can also be controlled by cellular metabolism. Metabolites not only directly influence epigenetic processes, but also affect the activity of epigenetic enzymes. Actually, both cellular metabolism pathways and epigenetic processes are controlled by enzymes. They are highly intertwined and are essential for oncogenesis and tumor development of CRCs. Therefore, they are potential therapeutic targets for the treatment of CRCs. In recent years, both epigenetic and metabolism inhibitors are studied for clinical use to treat CRCs. In this review, we depict the interplay between epigenetics and cellular metabolism in CRCs and summarize the underlying molecular mechanisms and their potential applications for clinical therapy.
Collapse
|
65
|
Wu H, Luo YX, Hu W, Zhao ML, Bie J, Yang M, Pan R, Huang NX, Feng G, Liu K, Song G. MicroRNA-382-5p inhibits osteosarcoma development and progression by negatively regulating VEZF1 expression. Oncol Lett 2021; 22:752. [PMID: 34539856 PMCID: PMC8436354 DOI: 10.3892/ol.2021.13013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
Human osteosarcoma is the most frequent malignant primary bone tumor that mainly occurs in young adults and children. MicroRNAs (miRNAs/miRs) are abnormally expressed in human osteosarcoma and contribute to osteosarcoma initiation and development. The present study aimed to investigate the role of miR-382-5p in the nosogenesis of osteosarcoma and to identify a novel target for osteosarcoma treatment. miR-382-5p expression was detected in human osteosarcoma clinical tissues and cell lines, including 143B, U2OS and MG63, via reverse transcription-quantitative PCR analysis. Multiple bioinformatic prediction toowe used to identify the potential target genes of miR-382-5p and vascular endothelial zinc finger 1 (VEZF1), which were validated via the dual-luciferase reporter assay. MG63 and U2OS cells were transfected with miR-382-5p mimics. The Cell Counting Kit-8 assay was performed to assess cell proliferation, while the Transwell assay was performed to assess migration and invasion. Cell colony formation was measured via crystal violet staining, and apoptosis was assessed via Annexin V/propidium iodide staining. The wound healing assay was performed to assess the migratory ability of U2OS and MG63 cells. Antitumor effects of miR-382-5p were evaluated in nude mice xenografts using U2OS cells. The results demonstrated that miR-382-5p expression was markedly downregulated in human osteosarcoma tissues and cell lines compared with adjacent normal tissues. Transfection of miR-382-5p mimics into MG63 and U2OS cells significantly inhibited the malignant behaviors of cells, including decreased proliferation, migration, diminished colony formation and invasion, and promoted osteosarcoma cell apoptosis. Bioinformatics prediction indicated that VEZF1 is a direct target gene of miR-382-5p. Overexpression of VEZF1 restored osteosarcoma tumor development inhibited by miR-382-5p in vivo. In addition, overexpression of miR-382-5p restrained the growth of xenograft osteosarcoma in nude mice following co-transfection, and overexpression of VEZF1 attenuated the inhibitory effect of miR-382-5p in nude mice. miR-382-5p acted as a tumor suppressor gene and inhibited the malignant biological behaviors of human osteosarcoma cells and functions associated with directly targeting VEZF1. Taken together, these results suggest that the miR-382-5p/VEZF1 interaction has an important role in osteosarcoma development and progression, and thus may be used as a diagnostic and therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Hui Wu
- Department of Orthopedics, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yu-Xi Luo
- The First Clinical College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wen Hu
- School of Medical Imaging, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Mao-Lin Zhao
- School of Medical Imaging, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Jun Bie
- Oncology Department, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Mi Yang
- Oncology Department, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Rongqiang Pan
- Oncology Department, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Nan-Xiang Huang
- Department of Pediatric Surgery, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Gang Feng
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Institute of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guiqin Song
- School of Basic Medicine, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
66
|
Ma Y, Shen N, Wicha MS, Luo M. The Roles of the Let-7 Family of MicroRNAs in the Regulation of Cancer Stemness. Cells 2021; 10:cells10092415. [PMID: 34572067 PMCID: PMC8469079 DOI: 10.3390/cells10092415] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/01/2021] [Accepted: 09/08/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer has long been viewed as a disease of normal development gone awry. Cancer stem-like cells (CSCs), also termed as tumor-initiating cells (TICs), are increasingly recognized as a critical tumor cell population that drives not only tumorigenesis but also cancer progression, treatment resistance and metastatic relapse. The let-7 family of microRNAs (miRNAs), first identified in C. elegans but functionally conserved from worms to human, constitutes an important class of regulators for diverse cellular functions ranging from cell proliferation, differentiation and pluripotency to cancer development and progression. Here, we review the current state of knowledge regarding the roles of let-7 miRNAs in regulating cancer stemness. We outline several key RNA-binding proteins, long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) involved in the regulation of let-7 biogenesis, maturation and function. We then highlight key gene targets and signaling pathways that are regulated or mutually regulated by the let-7 family of miRNAs to modulate CSC characteristics in various types of cancer. We also summarize the existing evidence indicating distinct metabolic pathways regulated by the let-7 miRNAs to impact CSC self-renewal, differentiation and treatment resistance. Lastly, we review current preclinical studies and discuss the clinical implications for developing let-7-based replacement strategies as potential cancer therapeutics that can be delivered through different platforms to target CSCs and reduce/overcome treatment resistance when applied alone or in combination with current chemo/radiation or molecularly targeted therapies. By specifically targeting CSCs, these strategies have the potential to significantly improve the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Yuxi Ma
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (Y.M.); (N.S.)
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Na Shen
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (Y.M.); (N.S.)
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Max S. Wicha
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (Y.M.); (N.S.)
- Correspondence: (M.S.W.); (M.L.)
| | - Ming Luo
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (Y.M.); (N.S.)
- Correspondence: (M.S.W.); (M.L.)
| |
Collapse
|
67
|
Sriram V, Lee JY. Calcium phosphate-polymeric nanoparticle system for co-delivery of microRNA-21 inhibitor and doxorubicin. Colloids Surf B Biointerfaces 2021; 208:112061. [PMID: 34492599 DOI: 10.1016/j.colsurfb.2021.112061] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 08/13/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022]
Abstract
Targeted combination therapy has shown promise to achieve maximum therapeutic efficacy by overcoming drug resistance. MicroRNA-21 (miR-21) is frequently overexpressed in various cancer types including breast and non-small cell lung cancer and its functions can be inhibited by miR inhibitor (miR-21i). A combination of miR-21i and a chemo drug, doxorubicin (Dox), can provide synergistic effects. Here, we developed a calcium phosphate (CaP)-coated nanoparticle (NP) formulation to co-deliver miR-21i along with Dox. This NP design can be used to deliver the two agents with different physiochemical properties. The NP formulation was optimized for particle size, polydispersity, Dox loading, and miR-21i loading. The NP formulation was confirmed to downregulate miR-21 levels and upregulate tumor suppressor gene levels. The cytotoxic efficacy of the combined miR-21i and Dox-containing NPs was found to be higher than that of Dox. Therefore, the CaP-coated hybrid lipid-polymeric NPs hold potential for the delivery of miR-21i and Dox.
Collapse
Affiliation(s)
- Vishnu Sriram
- Chemical Engineering Program, Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH, 45221-0012, United States
| | - Joo-Youp Lee
- Chemical Engineering Program, Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH, 45221-0012, United States.
| |
Collapse
|
68
|
Zhang X, Lin ZI, Yang J, Liu GL, Hu Z, Huang H, Li X, Liu Q, Ma M, Xu Z, Xu G, Yong KT, Tsai WC, Tsai TH, Ko BT, Chen CK, Yang C. Carbon Dioxide-Derived Biodegradable and Cationic Polycarbonates as a New siRNA Carrier for Gene Therapy in Pancreatic Cancer. NANOMATERIALS 2021; 11:nano11092312. [PMID: 34578632 PMCID: PMC8472555 DOI: 10.3390/nano11092312] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/18/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is an aggressive malignancy associated with poor prognosis and a high tendency in developing infiltration and metastasis. K-ras mutation is a major genetic disorder in pancreatic cancer patient. RNAi-based therapies can be employed for combating pancreatic cancer by silencing K-ras gene expression. However, the clinical application of RNAi technology is appreciably limited by the lack of a proper siRNA delivery system. To tackle this hurdle, cationic poly (cyclohexene carbonate) s (CPCHCs) using widely sourced CO2 as the monomer are subtly synthesized via ring-opening copolymerization (ROCOP) and thiol-ene functionalization. The developed CPCHCs could effectively encapsulate therapeutic siRNA to form CPCHC/siRNA nanoplexes (NPs). Serving as a siRNA carrier, CPCHC possesses biodegradability, negligible cytotoxicity, and high transfection efficiency. In vitro study shows that CPCHCs are capable of effectively protecting siRNA from being degraded by RNase and promoting a sustained endosomal escape of siRNA. After treatment with CPCHC/siRNA NPs, the K-ras gene expression in both pancreatic cancer cell line (PANC-1 and MiaPaCa-2) are significantly down-regulated. Subsequently, the cell growth and migration are considerably inhibited, and the treated cells are induced into cell apoptotic program. These results demonstrate the promising potential of CPCHC-mediated siRNA therapies in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Xinmeng Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
| | - Zheng-Ian Lin
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan;
| | - Jingyu Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
| | - Guan-Lin Liu
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan;
| | - Zulu Hu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
| | - Haoqiang Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
| | - Xiang Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
| | - Qiqi Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
| | - Mingze Ma
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Wei-Chung Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (W.-C.T.); (T.-H.T.)
| | - Tzu-Hsien Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan; (W.-C.T.); (T.-H.T.)
| | - Bao-Tsan Ko
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan;
- Correspondence: (B.-T.K.); (C.-K.C.); (C.Y.); Tel.: +886-4-2284-0411 (ext. 715) (B.-T.K.); +886-7-525-2000 (ext. 4060) (C.-K.C.); +86-0755-2693-2683 (C.Y.)
| | - Chih-Kuang Chen
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan;
- Correspondence: (B.-T.K.); (C.-K.C.); (C.Y.); Tel.: +886-4-2284-0411 (ext. 715) (B.-T.K.); +886-7-525-2000 (ext. 4060) (C.-K.C.); +86-0755-2693-2683 (C.Y.)
| | - Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China; (X.Z.); (J.Y.); (Z.H.); (H.H.); (X.L.); (Q.L.); (M.M.); (Z.X.); (G.X.)
- Correspondence: (B.-T.K.); (C.-K.C.); (C.Y.); Tel.: +886-4-2284-0411 (ext. 715) (B.-T.K.); +886-7-525-2000 (ext. 4060) (C.-K.C.); +86-0755-2693-2683 (C.Y.)
| |
Collapse
|
69
|
Sindhu KJ, Venkatesan N, Karunagaran D. MicroRNA Interactome Multiomics Characterization for Cancer Research and Personalized Medicine: An Expert Review. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:545-566. [PMID: 34448651 DOI: 10.1089/omi.2021.0087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) that are mutually modulated by their interacting partners (interactome) are being increasingly noted for their significant role in pathogenesis and treatment of various human cancers. Recently, miRNA interactome dissected with multiomics approaches has been the subject of focus since individual tools or methods failed to provide the necessary comprehensive clues on the complete interactome. Even though single-omics technologies such as proteomics can uncover part of the interactome, the biological and clinical understanding still remain incomplete. In this study, we present an expert review of studies involving multiomics approaches to identification of miRNA interactome and its application in mechanistic characterization, classification, and therapeutic target identification in a variety of cancers, and with a focus on proteomics. We also discuss individual or multiple miRNA-based interactome identification in various pathological conditions of relevance to clinical medicine. Various new single-omics methods that can be integrated into multiomics cancer research and the computational approaches to analyze and predict miRNA interactome are also highlighted in this review. In all, we contextulize the power of multiomics approaches and the importance of the miRNA interactome to achieve the vision and practice of predictive, preventive, and personalized medicine in cancer research and clinical oncology.
Collapse
Affiliation(s)
- K J Sindhu
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Nalini Venkatesan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
70
|
Huang B, Cui DJ, Yan F, Yang LC, Zhang MM, Zhao X. Circ_0087862 promotes the progression of colorectal cancer by sponging miR-142-3p and up-regulating BACH1 expression. Kaohsiung J Med Sci 2021; 37:1048-1057. [PMID: 34390174 DOI: 10.1002/kjm2.12437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 06/22/2021] [Accepted: 07/08/2021] [Indexed: 12/15/2022] Open
Abstract
Circular RNAs (circRNAs) feature prominently in regulating the malignant biological behaviors of colorectal cancer (CRC), including cell viability, cell cycle progression, apoptosis, migration, invasion, and so on. This study is performed to probe into the biological function and molecular mechanism of circ_0087862 in CRC. The expression profile of GSE138589 was available from Gene Expression Omnibus (GEO), and the differentially expressed circRNAs were analyzed by GEO2R. The expression of circ_0087862, miR-142-3p, and BACH1 mRNA in CRC tissues and cells was measured by qRT-PCR. CCK-8 assay was employed to determine the proliferation of CRC cells. Scratch wound healing and transwell assays were used to examine the migration and invasion of CRC cells. The targeting relationships between circ_0087862 and miR-142-3p, and between miR-142-3p and BACH1 3'UTR were verified by dual-luciferase reporter gene assay and RIP assay. BACH1 protein expression was probed by western blot. Circ_0087862 was highly expressed in CRC tissues and cell lines. Knocking down circ_0087862 significantly restrained the multiplication, migration and invasion of CRC cells. miR-142-3p inhibition weakened the impact of circ_0087862 knockdown on CRC cells. Circ_0087862 regulated BACH1 expressions by targeting miR-142-3p. Circ_0087862 regulates BACH1 expressions through sponging miR-142-3p, and promotes the proliferation, migration, and invasion of CRC cells.
Collapse
Affiliation(s)
- Bo Huang
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| | - De-Jun Cui
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| | - Fang Yan
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| | - Liu-Chan Yang
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| | - Man-Man Zhang
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| | - Xun Zhao
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| |
Collapse
|
71
|
Yan G, Yan S, Wang J, Lei S, Tian W, Yue X, Zhang Y. MicroRNA-296-5p inhibits cell proliferation by targeting HMGA1 in colorectal cancer. Exp Ther Med 2021; 22:793. [PMID: 34093749 PMCID: PMC8170657 DOI: 10.3892/etm.2021.10225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 11/08/2019] [Indexed: 01/10/2023] Open
Abstract
An increasing body of evidence indicates the involvement of microRNAs (miRNAs/miRs) in the initiation and progression of colorectal cancer (CRC). miR-296-5p was recently identified as a tumor suppressor in a variety of human cancer types; however, its function in CRC remains largely unknown. The present study demonstrated that the expression of miR-296-5p was significantly downregulated in CRC tissues and cell lines. The overexpression of miR-296-5p markedly inhibited proliferation, and induced cell cycle arrest and apoptosis in CRC cells. Bioinformatics analysis suggested that high mobility group AT-hook 1 (HMGA1) may be a target of miR-296-5p in CRC cells. Further experiments showed that miR-296-5p bound the 3'-untranslated region of HMGA1 and decreased its expression in CRC cells. HMGA1 was overexpressed in CRC tissues and was inversely correlated with the expression of miR-296-5p. The restoration of HMGA1 significantly reversed the inhibitory effect of miR-296-5p on the proliferation of CRC cells. Overall, the findings of the present study indicate that miR-296-5p suppressed the progression of CRC, at least partially via targeting HMGA1. Thus, miR-296-5p is a potential target for novel therapies in CRC.
Collapse
Affiliation(s)
- Guohui Yan
- The Medical Department of the Xiamen University, Xiamen, Fujian 361000, P.R. China
- The Medical Department of the Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
- Department of Ultrasound, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Shuidi Yan
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Jiajia Wang
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Shen Lei
- The Medical Department of the Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
| | - Weimin Tian
- Department of Paediatrics, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Xin Yue
- Department of Imaging, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Yang Zhang
- The Medical Department of the Xiamen University, Xiamen, Fujian 361000, P.R. China
- The Medical Department of the Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian 361004, P.R. China
| |
Collapse
|
72
|
Dai R, Jiang Q, Zhou Y, Lin R, Lin H, Zhang Y, Zhang J, Gao X. Lnc-STYK1-2 regulates bladder cancer cell proliferation, migration, and invasion by targeting miR-146b-5p expression and AKT/STAT3/NF-kB signaling. Cancer Cell Int 2021; 21:408. [PMID: 34332611 PMCID: PMC8325849 DOI: 10.1186/s12935-021-02114-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/24/2021] [Indexed: 02/07/2023] Open
Abstract
Background Epigenetic modulation by noncoding RNAs substantially contributes to human cancer development, but noncoding RNAs involvement in bladder cancer remains poorly understood. This study investigated the role of long noncoding RNA (lncRNA) lnc-STYK1-2 in tumorigenesis in cancerous bladder cells. Methods Differential lncRNA and mRNA profiles were characterized by high-throughput RNA sequencing combined with validation via quantitative PCR. Bladder cancer cell proliferation was assessed through MTS, and bladder cancer cell migration and invasion were assessed through a Transwell system. The in vivo tumorigenesis of bladder cancer cells was evaluated using the cancer cell line-based xenograft model. The dual-luciferase reporter assay verified the association of miR-146b-5p with lnc-STYK1-2 and the target gene. Protein abundances and phosphorylation were detected by Western blotting. Results Alterations in lncRNA profiles, including decreased lnc-STYK1-2 expression, were detected in bladder cancer tissues compared with adjacent noncancerous tissues. lnc-STYK1-2 silencing effectively promoted proliferation, migration, and invasion in two bladder cancer cell lines, 5637 and T24, and their tumorigenesis in nude mice. lnc-STYK1-2 siRNA promoted miR-146b-5p and reduced ITGA2 expression in bladder cancer cells. Moreover, miR-146b-5p suppressed ITGA2 expression in bladder cancer cells through direct association. Also, lnc-STYK1-2 directly associated with miR-146b-5p. Finally, miR-146b-5p inhibitors abrogated the alterations in bladder cell functions, ITGA2 expression, and phosphorylation of AKT, STAT3, and P65 proteins in 5637 and T24 cells induced by lnc-STYK1-2 silencing. Conclusion lnc-STYK1-2 inhibited bladder cancer cell proliferation, migration, and tumorigenesis by targeting miR-146b-5p to regulate ITGA2 expression and AKT/STAT3/NF-kB signaling.
Collapse
Affiliation(s)
- Ranran Dai
- Guangdong Key Laboratory of Urology, Guangzhou Medical University, Guangzhou, China
| | - Qingping Jiang
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - You Zhou
- Guangdong Key Laboratory of Urology, Guangzhou Medical University, Guangzhou, China
| | - Ruifeng Lin
- Guangdong Key Laboratory of Urology, Guangzhou Medical University, Guangzhou, China
| | - Hai Lin
- Guangdong Key Laboratory of Urology, Guangzhou Medical University, Guangzhou, China
| | - Yumin Zhang
- Department of Children's Stomatology, Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinhu Zhang
- Guangdong Key Laboratory of Urology, Guangzhou Medical University, Guangzhou, China
| | - Xingcheng Gao
- Guangdong Key Laboratory of Urology, Guangzhou Medical University, Guangzhou, China. .,Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang road, Yuexiu district, Guangzhou, 510120, China.
| |
Collapse
|
73
|
Naidoo M, Levine F, Gillot T, Orunmuyi AT, Olapade-Olaopa EO, Ali T, Krampis K, Pan C, Dorsaint P, Sboner A, Ogunwobi OO. MicroRNA-1205 Regulation of FRYL in Prostate Cancer. Front Cell Dev Biol 2021; 9:647485. [PMID: 34386489 PMCID: PMC8354587 DOI: 10.3389/fcell.2021.647485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 07/06/2021] [Indexed: 01/01/2023] Open
Abstract
High mortality rates of prostate cancer (PCa) are associated with metastatic castration-resistant prostate cancer (CRPC) due to the maintenance of androgen receptor (AR) signaling despite androgen deprivation therapies (ADTs). The 8q24 chromosomal locus is a region of very high PCa susceptibility that carries genetic variants associated with high risk of PCa incidence. This region also carries frequent amplifications of the PVT1 gene, a non-protein coding gene that encodes a cluster of microRNAs including, microRNA-1205 (miR-1205), which are largely understudied. Herein, we demonstrate that miR-1205 is underexpressed in PCa cells and tissues and suppresses CRPC tumors in vivo. To characterize the molecular pathway, we identified and validated fry-like (FRYL) as a direct molecular target of miR-1205 and observed its overexpression in PCa cells and tissues. FRYL is predicted to regulate dendritic branching, which led to the investigation of FRYL in neuroendocrine PCa (NEPC). Resistance toward ADT leads to the progression of treatment related NEPC often characterized by PCa neuroendocrine differentiation (NED), however, this mechanism is poorly understood. Underexpression of miR-1205 is observed when NED is induced in vitro and inhibition of miR-1205 leads to increased expression of NED markers. However, while FRYL is overexpressed during NED, FRYL knockdown did not reduce NED, therefore revealing that miR-1205 induces NED independently of FRYL.
Collapse
Affiliation(s)
- Michelle Naidoo
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States.,Department of Biology and Biochemistry, The Graduate Center of the City University of New York, New York, NY, United States
| | - Fayola Levine
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States
| | - Tamara Gillot
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States
| | - Akintunde T Orunmuyi
- Department of Radiation Oncology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Thahmina Ali
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States
| | - Konstantinos Krampis
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States
| | - Chun Pan
- Department of Mathematics and Statistics, Hunter College of the City University of New York, New York, NY, United States
| | - Princesca Dorsaint
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Andrea Sboner
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Olorunseun O Ogunwobi
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States.,Department of Biology and Biochemistry, The Graduate Center of the City University of New York, New York, NY, United States.,Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
74
|
Taefehshokr S, Taefehshokr N, Derakhshani A, Baghbanzadeh A, Astamal RV, Safaei S, Abbasi S, Hajazimian S, Maroufi NF, Isazadeh A, Hajiasgharzadeh K, Baradaran B. The regulatory role of pivotal microRNAs in the AKT signaling pathway in breast cancer. Curr Mol Med 2021; 22:263-273. [PMID: 34238182 DOI: 10.2174/1566524021666210708095051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/29/2021] [Accepted: 05/06/2021] [Indexed: 11/22/2022]
Abstract
Breast cancer is the most prevalent type of cancer among women, and it remains the main challenge despite improved treatments. MicroRNAs (miRNAs) are a small non-coding family of RNAs that play an indispensable role in regulating major physiological processes, including differentiation, proliferation, invasion, migration, cell cycle regulation, stem cell maintenance, apoptosis, and organ development. The dysregulation of these tiny molecules is associated with various human malignancies. More than 50% of these non-coding RNA sequences estimated have been placed on genomic regions or fragile sites linked to cancer. Following the discovery of the first signatures of specific miRNA in breast cancer, numerous researches focused on involving these tiny RNAs in breast cancer physiopathology as a new therapeutic approach or as reliable prognostic biomarkers. In the current review, we focus on recent findings related to the involvement of miRNAs in breast cancer via the AKT signaling pathway and the related clinical implications.
Collapse
Affiliation(s)
- Sina Taefehshokr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Taefehshokr
- Division of Biosciences, Department of Life Sciences, Brunel University London, Kingston Lane, UB8 3PH, United Kingdom
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Vaezi Astamal
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samane Abbasi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Saba Hajazimian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Fathi Maroufi
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
75
|
Y-Box Binding Protein 1 Regulates Angiogenesis in Bladder Cancer via miR-29b-3p-VEGFA Pathway. JOURNAL OF ONCOLOGY 2021; 2021:9913015. [PMID: 34306080 PMCID: PMC8270724 DOI: 10.1155/2021/9913015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/23/2021] [Indexed: 12/26/2022]
Abstract
Angiogenesis plays a vital role in the development of bladder cancer (BC). The Y-box-binding protein 1 (YB-1) is a well-known oncoprotein which is closely related to angiogenesis of tumors, but the relationship and mechanism of YB-1 and angiogenesis in BC remain unclear. Based on 56 clinical BC specimens, this study found that high expression of YB-1 samples demonstrated a higher expression of vascular endothelial growth factor A (VEGFA) than those of YB-1 low expression. Subsequently, the expression of YB-1 and miR-29b-3p was regulated in the BC cell lines where we noted that YB-1 promoted VEGFA expression by downregulating the expression of miR- 29b-3p. The ability of BC cells to induce angiogenesis decreased after YB-1 was knocked down. Moreover, the in vivo study further confirmed that YB-1 promotes angiogenesis in BC. Our findings enhance the understanding of how YB-1 promotes angiogenesis in BC and provide evidence for YB-1 as a therapeutic target of BC. Moreover, this may provide new inspiration for miRNAs replacement therapies.
Collapse
|
76
|
PD-L1 regulation revisited: impact on immunotherapeutic strategies. Trends Mol Med 2021; 27:868-881. [PMID: 34187739 DOI: 10.1016/j.molmed.2021.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/29/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
A particularly promising cancer treatment is the use of monoclonal antibodies (mAbs) against immune checkpoints (i.e., immune checkpoint inhibitors; ICIs). However, many patients experience relapse and severe adverse events. To overcome these negative issues and improve efficiency, current approaches rely on combinatorial treatments, including some modulating the expression of programmed cell death receptor 1 (PD-1)/programmed death ligand 1 (PD-L1) immune checkpoints directly. In this review, we examine the recently discovered pathways involved in PD-L1 expression and highlight the relevant druggable strategies that are being developed to both improve the response rate and avoid the onset of resistance. Altogether, these new strategies will pave the way for effective treatment combinations in future oncology clinical trials.
Collapse
|
77
|
Li G, Wang Q, Li Z, Shen Y. Serum miR-21 and miR-210 as promising non-invasive biomarkers for the diagnosis and prognosis of colorectal cancer. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2021; 112:832-837. [PMID: 33054296 DOI: 10.17235/reed.2020.6801/2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE this study aimed to investigate the expression and clinical significance of miR-21 and miR-210 in serum of patients with colorectal cancer (CRC). METHODS the expression levels of serum miR-21 and miR-210 in 40 CRC patients (CRC group) and 20 healthy patients (control group) were measured by qRT-PCR. Correlation analysis was performed of the relationship between serum miR-21 and miR-210 levels with clinical characteristics, including gender, age, tumor location, tumor size, tumor stage, local invasion and TNM staging. The expression levels of miR-21 and miR-210 in the CRC group were separately measured before and after surgery. ROC analysis was performed to evaluate the diagnostic value of miR-21 and miR-210. RESULTS serum miR-21 and miR-210 in the CRC group were much higher than those in the control group. Meanwhile, the levels of serum miR-21 and miR-210 were closely related to tumor size (p = 0.028, p = 0.047), lymphatic metastasis (p = 0.038, p = 0.028), TNM staging (p = 0.014, p = 0.047) and tumor stage (p = 0.014, p = 0.017), but independent of gender, age and tumor location. In addition, serum miR-21 and miR-210 in the CRC group (n = 18) after surgery were lower than those before surgery (p < 0.001). ROC curves showed that miR-21 (AUC = 0.863) and miR-210 (AUC = 0.818) both had diagnostic efficacy in CRC patients. CONCLUSION miR-21 and miR-210 can be used as novel non-invasive biomarkers for CRC diagnosis and prognosis.
Collapse
Affiliation(s)
- Gang Li
- Colorectal Surgery, Shaoxing People's Hospital
| | - Qi Wang
- Colorectal Surgery, Shaoxing People's Hospital
| | - Zhenjun Li
- Colorectal Surgery, Shaoxing People's Hospital
| | - Yi Shen
- Colorectal Surgery, Shaoxing People's Hospital,
| |
Collapse
|
78
|
Cai Y, Wang B, Li B, Huang X, Guo H, Liu Y, Chen B, Zhao S, Wu S, Li W, Wang L, Jia K, Wang H, Chen P, Jiang M, Tang X, Qi H, Dai C, Ye J, He Y. Collection on reports of molecules linked to epithelial-mesenchymal transition in the process of treating metastasizing cancer: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:946. [PMID: 34350261 PMCID: PMC8263858 DOI: 10.21037/atm-20-7002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/12/2021] [Indexed: 12/26/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a morphological process in which epithelial cells transform into mesenchymal cells via a specific procedure. EMT plays an important role in the cancer invasion-metastasis cascade and the current treatment of metastatic cancer, influences the migration, polarity, and adhesion of tumor cells, promotes their migration, invasiveness, anti-apoptotic ability. It contributes to the changes of the tumor microenvironment and suppresses the sensitivity of tumor cells to chemotherapy, causing cancer metastasis and worse, hindering the control and therapy of it. This paper reviews the mechanisms, detection, and treatments of cancer metastasis that have been identified and applied to date, summarizes the EMT-related biological molecules, providing a reference for EMT-targeted research and therapy. As EMT is significant in the progress of tumor metastasis, it is meaningful for the therapy and control of metastatic cancer to understand the mechanism of EMT at the molecular level. We summarized the mechanisms, detection and therapeutic implications of EMT, listed the research progress of molecules like genes, miRNAs, signaling pathways in EMT. We also discussed the prospects of EMT-targeted treatment in cancer metastasis interventions and the challenges the treatment and researches are facing. The summary is conducive to the treatment and further research of EMT and metastatic cancer.
Collapse
Affiliation(s)
- Yiyi Cai
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Boyuan Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Bingying Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Xintong Huang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Haoyue Guo
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Yu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Bin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Sha Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shengyu Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Keyi Jia
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Xuzhen Tang
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Hui Qi
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Chunlei Dai
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Junyan Ye
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
79
|
Yang X, Chen C, Li L, Xiao T, Zou YD, Zheng D. Current research advances in microRNA-mediated regulation of Krüppel-like factor 4 in cancer: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:948. [PMID: 34350263 PMCID: PMC8263881 DOI: 10.21037/atm-21-2347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/28/2021] [Indexed: 11/06/2022]
Abstract
Objective The purpose of this study was to investigate the miRNAs and related mechanisms that regulates KLF4 in different cancers. Furthermore, we summarized the potential targets of miRNAs regulating the KLF4 pathway in cancer research. Background MiRNAs are single-stranded, endogenous non-coding small RNAs, some of which are related to human cancers. miRNAs carry out post-transcriptional gene regulation through translation inhibition and degradation of target messenger RNAs (mRNAs) via complementarily pairing with their 3' untranslated regions. KLF4 is an important transcription factor with complex involvement in cancer. Increasing evidence shows that miRNAs are dysregulated in cancer and can regulate cancer-related signaling pathways, thereby affecting tumor progression. Methods Systematic scientific literature searches were undertaken on PubMed using the following terms: "miRNAs and KLF4", "KLF4 and cancer", "miRNAs and cancer", and "miRNAs, KLF4 and cancer". Relevant papers were retrieved and further results were found by reviewing related papers and the references of the retrieved papers. We then conducted a narrative overview of the literature to summarize the results of the papers. Conclusions The role of KLF4 in cancer varies in a context-dependent manner. KLF4-regulating miRNAs in different tumors include miR-124, miR-9-5p, miR-10b, miR-18a, miR-25-3p, miR-10b, miR-92a, miR-103, miR-155, miR-135b-5p, miR-32-5p, miR-148-3p, miR-152-3p, miR-10b, miR-25, miR-3120-5p, miR-7, miR-1233-3p, miR-10b, miR-145, miR-139-5p, miR-16, miR-152, miR-375, and miR-145.
Collapse
Affiliation(s)
- Xi Yang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China.,Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Cheng Chen
- Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Li Li
- Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Tian Xiao
- Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Yong-Dong Zou
- Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Duo Zheng
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China.,Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| |
Collapse
|
80
|
Rizkita LD, Astuti I. The potential of miRNA-based therapeutics in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection: A review. J Pharm Anal 2021; 11:265-271. [PMID: 33782640 PMCID: PMC7989072 DOI: 10.1016/j.jpha.2021.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Since the World Health Organization (WHO) declared COVID-19, the disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as a pandemic in March 2020, and more than 117 million people worldwide have been confirmed to have been infected. Scientists, medical professionals, and other stakeholders are racing against time to find and develop effective medicines for COVID-19. However, no drug with high efficacy to treat SARS-CoV-2 infection has been approved. With the increasing popularity of gene therapy, scientists have explored the utilization of small RNAs such as microRNAs (miRNAs) as therapeutics. miRNAs are non-coding RNAs with high affinity for the 3'-UTRs of targeted messenger RNAs (mRNAs). Interactions between host cells and viral genomes may induce the upregulation or downregulation of various miRNAs. Therefore, understanding the expression patterns of these miRNAs and their functions will provide insights into potential miRNA-based therapies. This review systematically summarizes the potential targets of miRNA-based therapies for SARS-CoV-2 infection and examines the viability of possible transfection methods.
Collapse
Affiliation(s)
- Leonny Dwi Rizkita
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Indwiani Astuti
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| |
Collapse
|
81
|
Karkhane M, Lashgarian HE, Hormozi M, Fallahi S, Cheraghipour K, Marzban A. Oncogenesis and Tumor Inhibition by MicroRNAs and its Potential Therapeutic Applications: A Systematic Review. Microrna 2021; 9:198-215. [PMID: 31686643 DOI: 10.2174/2211536608666191104103834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/01/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022]
Abstract
MicroRNAs appear as small molecule modifiers, which improve many new findings and mechanical illustrations for critically important biological phenomena and pathologic events. The best-characterized non-coding RNA family consists of about 2600 human microRNAs. Rich evidence has revealed their crucial importance in maintaining normal development, differentiation, growth control, aging, modulation of cell survival or apoptosis, as well as migration and metastasis as microRNAs dysregulation leads to cancer incidence and progression. By far, microRNAs have recently emerged as attractive targets for therapeutic intervention. The rationale for developing microRNA therapeutics is based on the premise that aberrantly expressed microRNAs play a significant role in the emergence of a variety of human diseases ranging from cardiovascular defects to cancer, and that repairing these microRNA deficiencies by either antagonizing or restoring microRNA function may yield a therapeutic benefit. Although microRNA antagonists are conceptually similar to other inhibitory therapies, improving the performance of microRNAs by microRNA replacement or inhibition that is a less well- described attitude. In this assay, we have condensed the last global knowledge and concepts regarding the involvement of microRNAs in cancer emergence, which has been achieved from the previous studies, consisting of the regulation of key cancer-related pathways, such as cell cycle control and the DNA damage response and the disruption of profile expression in human cancer. Here, we have reviewed the special characteristics of microRNA replacement and inhibition therapies and discussed explorations linked with the delivery of microRNA mimics in turmeric cells. Besides, the achievement of biomarkers based on microRNAs in clinics is considered as novel non-invasive biomarkers in diagnostic and prognostic assessments.
Collapse
Affiliation(s)
- Maryam Karkhane
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hamed Esmaeil Lashgarian
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Maryam Hormozi
- Department of Biochemistry, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Shirzad Fallahi
- Department of Medical Parasitology and Mycology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Kourosh Cheraghipour
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Abdolrazagh Marzban
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
82
|
Mohamad SFS, Elias MH. Potential treatment for chronic myeloid leukemia using microRNA: in silico comparison between plants and human microRNAs in targeting BCR-ABL1 gene. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00156-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Chronic myeloid leukemia (CML) is a myeloproliferative disorder characterized by the expression of the BCR-ABL1 fusion gene. Tyrosine kinase inhibitors (TKI) are used to treat CML, but mutations in the tyrosine kinase domain contribute to CML chemo-resistance. Therefore, finding alternative molecular-targeted therapy is important for the comprehensive treatment of CML. MicroRNAs (miRNA) are small non-coding regulatory RNAs which suppress the expression of their target genes by binding to the 3′ untranslated region (3′UTR) of the target mRNA. Hypothetically, the miRNA-mRNA interaction would suppress BCR-ABL1 expression and consequently reduce and inhibit CML cell proliferation. Thus, our objective was to determine the target interaction of human and plant miRNAs targeting the 3′UTR region of BCR-ABL1 in terms of miRNA binding conformity, protein interaction network, and pathways using in silico analysis. The 3′UTR sequence of BCR-ABL1 is obtained from Ensembl Genome Browser while the binding conformity was determined using the PsRNATarget Analysis Server, RNA22, Target Rank Server, and DIANA TOOLS. Protein-protein interaction network and pathway analysis are determined using STRING, Cytoscape, and KEGG pathway analysis.
Results
Five plants and five human miRNAs show strong binding conformity with 3′UTR of BCR-ABL1. The strongest binding conformity was shown by Oryza sativa’s Osa-miR1858a and osa-miR1858b with −24.4 kcal/mol folding energy and a p value of 0.0077. Meanwhile, in human miRNA, the hsa-miR-891a-3p shows the highest miTG score of 0.99 with −12 kcal/mol folding energy and a p value of 0.037. Apart from ABL1, osa-miR1858a/osa-miR1858b and hsa-miR891a-3p also target other 720 and 645 genes, respectively. The interaction network of Osa-miR1858a/osa-miR1858b and hsa-miR891a-3p identifies nineteen and twelve ABL1’s immediate neighboring proteins, respectively. The pathways analysis focuses on the RAS, MAPK, CML, and hematopoietic cell lineage pathway.
Conclusion
Both plant and human miRNAs tested in this study could be a potential therapeutic prospect in CML treatment, but thermodynamically, osa-miR1858a/osa-miR1858b binding to ABL1 is more favorable. However, it is important to carry out more research in vitro and in vivo and clinical studies to assess its efficacy as a targeted therapy for CML.
Graphical abstract
Collapse
|
83
|
Sartorius K, An P, Winkler C, Chuturgoon A, Li X, Makarova J, Kramvis A. The Epigenetic Modulation of Cancer and Immune Pathways in Hepatitis B Virus-Associated Hepatocellular Carcinoma: The Influence of HBx and miRNA Dysregulation. Front Immunol 2021; 12:661204. [PMID: 33995383 PMCID: PMC8117219 DOI: 10.3389/fimmu.2021.661204] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/15/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatitis B virus (HBV)-associated hepatocellular carcinoma (HBV-HCC) pathogenesis is fueled by persistent HBV infection that stealthily maintains a delicate balance between viral replication and evasion of the host immune system. HBV is remarkably adept at using a combination of both its own, as well as host machinery to ensure its own replication and survival. A key tool in its arsenal, is the HBx protein which can manipulate the epigenetic landscape to decrease its own viral load and enhance persistence, as well as manage host genome epigenetic responses to the presence of viral infection. The HBx protein can initiate epigenetic modifications to dysregulate miRNA expression which, in turn, can regulate downstream epigenetic changes in HBV-HCC pathogenesis. We attempt to link the HBx and miRNA induced epigenetic modulations that influence both the HBV and host genome expression in HBV-HCC pathogenesis. In particular, the review investigates the interplay between CHB infection, the silencing role of miRNA, epigenetic change, immune system expression and HBV-HCC pathogenesis. The review demonstrates exactly how HBx-dysregulated miRNA in HBV-HCC pathogenesis influence and are influenced by epigenetic changes to modulate both viral and host genome expression. In particular, the review identifies a specific subset of HBx induced epigenetic miRNA pathways in HBV-HCC pathogenesis demonstrating the complex interplay between HBV infection, epigenetic change, disease and immune response. The wide-ranging influence of epigenetic change and miRNA modulation offers considerable potential as a therapeutic option in HBV-HCC.
Collapse
Affiliation(s)
- Kurt Sartorius
- Hepatitis Virus Diversity Research Unit, School of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa.,Department of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa.,Department of Surgery, University of KwaZulu-Natal Gastrointestinal Cancer Research Centre, Durban, South Africa
| | - Ping An
- Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Cheryl Winkler
- Basic Research Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, South Africa
| | - Xiaodong Li
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Julia Makarova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia.,Higher School of Economics University, Moscow, Russia
| | - Anna Kramvis
- Hepatitis Virus Diversity Research Unit, School of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
84
|
Trovato M, Sciacchitano S, Facciolà A, Valenti A, Visalli G, Di Pietro A. Interleukin‑6 signalling as a valuable cornerstone for molecular medicine (Review). Int J Mol Med 2021; 47:107. [PMID: 33907833 PMCID: PMC8057292 DOI: 10.3892/ijmm.2021.4940] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
The biological abilities of interleukin-6 (IL-6) have been under investigation for nearly 40 years. IL-6 works through an interaction with the complex peptide IL-6 receptor (IL-6R). IL-6 is built with four α-chain nanostructures, while two different chains, IL-6Rα (gp80) and gp130/IL6β (gp130), are included in IL-6R. The three-dimensional shapes of the six chains composing the IL-6/IL-6R complex are the basis for the nanomolecular roles of IL-6 signalling. Genes, pseudogenes and competitive endogenous RNAs of IL-6 have been identified. In the present review, the roles played by miRNA in the post-transcriptional regulation of IL-6 expression are evaluated. mRNAs are absorbed via the 'sponge' effect to dynamically balance mRNA levels and this has been assessed with regard to IL-6 transcription efficiency. According to current knowledge on molecular and nanomolecular structures involved in active IL-6 signalling, two different IL-6 models have been proposed. IL-6 mainly has functions in inflammatory processes, as well as in cognitive activities. Furthermore, the abnormal production of IL-6 has been found in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2; also known as COVID-19). In the present review, both inflammatory and cognitive IL-6 models were analysed by evaluating the cytological and histological locations of IL-6 signalling. The goal of this review was to illustrate the roles of the classic and trans-signalling IL-6 pathways in endocrine glands such as the thyroid and in the central nervous system. Specifically, autoimmune thyroid diseases, disorders of cognitive processes and SARS-CoV-2 virus infection have been examined to determine the contribution of IL-6 to these disease states.
Collapse
Affiliation(s)
- Maria Trovato
- Department of Clinical and Experimental Medicine, University Hospital, I‑98125 Messina, Italy
| | | | - Alessio Facciolà
- Department of Clinical and Experimental Medicine, University Hospital, I‑98125 Messina, Italy
| | - Andrea Valenti
- Department of Clinical and Experimental Medicine, University Hospital, I‑98125 Messina, Italy
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Policlinico Universitario, I‑98125 Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Policlinico Universitario, I‑98125 Messina, Italy
| |
Collapse
|
85
|
Therapeutic strategies for miRNA delivery to reduce hepatocellular carcinoma. Semin Cell Dev Biol 2021; 124:134-144. [PMID: 33926792 DOI: 10.1016/j.semcdb.2021.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/21/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022]
Abstract
Malignancies of hepatocellular carcinoma (HCC) are rapidly spreading and commonly fatal. Like most cancers, the gene expression patterns in HCC vary significantly from patient to patient. Moreover, the expression networks during HCC progression are largely controlled by microRNAs (miRNAs) regulating multiple oncogenes and tumor supressors. Therefore, miRNA-based therapeutic strategies altering these networks may significantly influence the cellular behavior enough for them to cure HCC. However, the most substantial challenges in developing such therapies are the stability of the oligos themselves and that of their delivery systems. Here we provide a comprehensive update describing various miRNA delivery systems, including virus-based delivery and non-viral delivery. The latter may be achieved using inorganic nanoparticles, polymer based nano-carriers, lipid-based vesicles, exosomes, and liposomes. Leaky vasculature in HCC-afflicted livers helps untargeted nanocarriers to accumulate in the tumor tissue but may result in side effects during higher dose of treatment. On the other hand, the strategies for actively targeting miRNA therepeutics to cancerous cells through nano-conjugates or vesicles by decorating their surface with antibodies against or ligands for HCC-specific antigens or receptors are more efficient in preventing damage to healthy tissue and cancer recurrence.
Collapse
|
86
|
Zhang T, Wu Y, Yang D, Wu C, Li H. Preparation, characterization, and in vitro tumor-suppressive effect of anti-miR-21-equipped RNA nanoparticles. Biochem Biophys Res Commun 2021; 558:107-113. [PMID: 33906109 DOI: 10.1016/j.bbrc.2021.04.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
MicroRNAs play an irreplaceable role in gene expression regulation. Upregulation of several miRNAs increases the risk of invasion and metastasis of breast cancer cells. An oncogenic miRNA, miR-21, is highly expressed in triple-negative breast cancer (TNBC) and is associated with tumor proliferation, invasion, carcinogenesis, prognosis, and therapeutic resistance. However, targeted delivery of therapeutic anti-miRNAs into cancer cells remains challenging, especially for TNBC. In this study, we report the application of an RNA nanotechnology-based platform for the targeted delivery of anti-miR-21 by epidermal growth factor receptor (EGFR) aptamer in vitro to TNBC and chemical-resistant breast cancer cells. RNA nanoparticles reduced cell viability and sensitized breast cancer cells to doxorubicin (DOX) treatment in vitro. Inhibition of miR-21 by RNA nanoparticles suppressed TNBC cell invasion, migration, and colony formation. The results indicate the potential application of nanotechnology-based delivery platforms in clinical anti-cancer therapeutics.
Collapse
Affiliation(s)
- Tinghong Zhang
- Department of Urology, Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, PR China; Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325011, PR China
| | - Yunlong Wu
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325011, PR China; School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian Province, 361102, PR China
| | - Dejun Yang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, PR China.
| | - Cunzao Wu
- Department of Urology, Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, PR China.
| | - Huaqiong Li
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325011, PR China
| |
Collapse
|
87
|
microRNA-1298 inhibits the malignant behaviors of breast cancer cells via targeting ADAM9. Biosci Rep 2021; 40:226894. [PMID: 33146718 PMCID: PMC7729294 DOI: 10.1042/bsr20201215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs (miRNAs) regulate the progression of human malignancy by targeting oncogenes or tumor suppressors, which are 12 promising targets for cancer treatment. Increasing evidence has suggested the aberrant expression and tumor-suppressive function of miR-1298 in cancers, however, the regulatory mechanism of miR-1298 in breast cancer (BC) remains unclear. Here, our findings showed that miR-1298 was down-regulated in BC tissues and cell lines. Lower level of miR-1298 was significantly correlated with the advanced progression of BC patients. Experimental study showed that overexpression of miR-1298 inhibited the proliferation, induced apoptosis and cell cycle arrest in BC cells. The in vivo xenograft mice model showed that highly expressed miR-1298 significantly reduced the tumor growth and metastasis. Further mechanism analysis revealed that miR-1298 bound the 3′-untranslated region (UTR) of a disintegrin and metalloproteinase 9 domain (ADAM9) and suppressed the expression of ADAM9 in BC cells. ADAM9 was overexpressed in BC tissues and inversely correlated with miR-1298. Down-regulation of ADAM9 induced apoptosis and cell cycle arrest of BC cells. Moreover, ectopic expression of ADAM9 by transiently transfecting with vector encoding the full coding sequence of ADAM9 attenuated the inhibitory effects of miR-1298 on the proliferation and cell cycle progression of BC cells. Collectively, our results illustrated that miR-1298 played a suppressive role in regulating the phenotype of BC cells through directly repressing ADAM9, suggesting the potential application of miR-1298 in the therapy of BC.
Collapse
|
88
|
Molecular Landscape of the Epithelial-Mesenchymal Transition in Endometrioid Endometrial Cancer. J Clin Med 2021; 10:jcm10071520. [PMID: 33917330 PMCID: PMC8038735 DOI: 10.3390/jcm10071520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/02/2021] [Indexed: 12/25/2022] Open
Abstract
Modern diagnostics are based on molecular analysis and have been focused on searching for new molecular markers to use in diagnostics. Included in this has been the search for the correlation between gene expression in tissue samples and liquid biological materials. The aim of this study was to evaluate the differences in the expression profile of messenger RNA (mRNA) and micro-RNA (miRNA) related to the epithelial-mesenchymal transition (EMT) in different grades of endometrial cancer (G1-G3), in order to select the most promising molecular markers. The study material consisted of tissue samples and whole blood collected from 30 patients with endometrial cancer (study group; G1 = 15; G2 = 8; G3 = 7) and 30 without neoplastic changes (control group). The molecular analysis included the use of the microarray technique and RTqPCR. Microarray analysis indicated the following number of mRNA differentiating the endometrial cancer samples from the control (tissue/blood): G1 vs. C = 21/18 mRNAs, G2 vs. C = 19/14 mRNAs, and G3 vs. C = 10/9 mRNAs. The common genes for the tissue and blood samples (Fold Change; FC > 3.0) were G1 vs. C: TGFB1, WNT5A, TGFB2, and NOTCH1; G2 vs. C: BCL2L, SOX9, BAMBI, and SMAD4; G3 vs. C STAT1 and TGFB1. In addition, mRNA TGFB1, NOTCH1, and BCL2L are common for all grades of endometrial cancer. The analysis showed that miR-144, miR-106a, and miR-30d are most strongly associated with EMT, making them potential diagnostic markers.
Collapse
|
89
|
Zhuo J, Zheng Y, Hu W, Yin G. Sufentanil Inhibits Proliferation, Migration, and Invasion of Hepatocellular Carcinoma Cells by Upregulating miRNA-204. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Sufentanil is a powerful analgesic that acts on μ-receptors, but there are few studies on sufentanil in cancer. The biological function and underlying mechanisms of sufentanil on the hepatocellular carcinoma (HCC) cells were explored in the present study. HCC cells were first treated
with different concentrations of sufentanil and the most optimum concentration of sufentanil was determined. The expression of miR-204 in HCC cells was changed by transfected with miR-204 inhibitor and the transfection efficiency was assessed by qRT-PCR. CCK-8, wound-healing and Transwell
assays were performed to evaluate the proliferation, migration and invasion of HCC cells, respectively. The level of AKT and PI3K phosphorylation (p-AKT and p-PI3K) were assessed by western blot analysis. Our results demonstrated that sufentanil effectively inhibited cell proliferation,migration
and invasion in both Huh7 and Hep3B cells, and significantly decreased the expression of p-AKT and p-PI3K. In addition, miR-204 was upregulated in Huh7 and Hep3B cells treated with sufentanil, and low expression of miR-204 attenuated the damage of sufentanil on the viability of Huh7 and Hep3B
cells. Taken together, sufentanil suppressed the proliferation, migration and invasion of HCC cells via inhibiting AKT/PI3K signaling pathway by targeting miR-204.
Collapse
Affiliation(s)
- Jiuwu Zhuo
- Department of Intensive Care Unit, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| | - Yishan Zheng
- Department of Intensive Care Unit, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| | - Wanying Hu
- Operating Room, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| | - Guoping Yin
- Department of Anesthesiology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210003, China
| |
Collapse
|
90
|
Arrighetti N, Beretta GL. miRNAs as Therapeutic Tools and Biomarkers for Prostate Cancer. Pharmaceutics 2021; 13:380. [PMID: 33805590 PMCID: PMC7999286 DOI: 10.3390/pharmaceutics13030380] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
Prostate cancer (PCa) is the fifth cause of tumor-related deaths in man worldwide. Despite the considerable improvement in the clinical management of PCa, several limitations emerged both in the screening for early diagnosis and in the medical treatment. The use of prostate-specific antigen (PSA)-based screening resulted in patients' overtreatment and the standard therapy of patients suffering from locally advanced/metastatic tumors (e.g., radical prostatectomy, radiotherapy, and androgen deprivation therapy) showed time-limited efficacy with patients undergoing progression toward the lethal metastatic castration-resistant PCa (mCRPC). Although valuable alternative therapeutic options have been recently proposed (e.g., docetaxel, cabazitaxel, abiraterone, enzalutamide, and sipuleucel-T), mCRPC remains incurable. Based on this background, there is an urgent need to identify new and more accurate prostate-specific biomarkers for PCa diagnosis and prognosis and to develop innovative medical approaches to counteract mCRPC. In this context, microRNA (miRNAs) emerged as potential biomarkers in prostate tissues and biological fluids and appeared to be promising therapeutic targets/tools for cancer therapy. Here we overview the recent literature and summarize the achievements of using miRNAs as biomarkers and therapeutic targets/tools for fighting PCa.
Collapse
Affiliation(s)
| | - Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| |
Collapse
|
91
|
|
92
|
Lotfinejad P, Kazemi T, Safaei S, Amini M, Roshani Asl E, Baghbani E, Sandoghchian Shotorbani S, Jadidi Niaragh F, Derakhshani A, Abdoli Shadbad M, Silvestris N, Baradaran B. PD-L1 silencing inhibits triple-negative breast cancer development and upregulates T-cell-induced pro-inflammatory cytokines. Biomed Pharmacother 2021; 138:111436. [PMID: 33667790 DOI: 10.1016/j.biopha.2021.111436] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/22/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an invasive tumor with a high incidence of distant metastasis and poor prognosis. In TNBC cells, high PD-L1 expression can induce an immunosuppressive tumor microenvironment, repressing the anti-tumoral immune responses. Although FDA-approved agents targeting the PD-1/PD-L1 axis are potent to eliminate tumoral cells, their immune-related adverse events have become worrisome. As the regulator of gene expression, siRNAs can directly target PD-L1 in breast cancer cells. The gene modification of tumoral PD-L1 can reduce our reliance on the current method of targeting the PD-L1/PD-1 axis. We initiated the study with bioinformatics analysis; the results indicated that TNBC and the MDA-MB-231 cells significantly overexpressed PD-L1 compared to other breast cancer subtypes and cell lines. Our results demonstrated that PD-L1 silencing substantially reduced PD-L1 expression at mRNA and protein levels in MDA-MB-231 cells. Moreover, our results demonstrated that PD-L1 knockdown reduced cancer cell proliferation and induced apoptosis via intrinsic and extrinsic apoptosis pathways. We observed that PD-L1 silencing effectively inhibited the migration of TNBC cells. Further investigation also displayed that silencing of PD-L1 in breast cancer cells induced T-cell cytotoxic function by upregulating the gene expression of pro-inflammatory cytokines, i.e., IL-2, IFN-γ, and TNF-α, and downregulating the gene expression of anti-inflammatory cytokines, i.e., IL-10, and TGF-β, in a co-culture system.
Collapse
Affiliation(s)
- Parisa Lotfinejad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Roshani Asl
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Italy
| | - Mahdi Abdoli Shadbad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nicola Silvestris
- IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO-University of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
93
|
Smith DM, Keller A. DNA Nanostructures in the Fight Against Infectious Diseases. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000049. [PMID: 33615315 PMCID: PMC7883073 DOI: 10.1002/anbr.202000049] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Throughout history, humanity has been threatened by countless epidemic and pandemic outbreaks of infectious diseases, from the Justinianic Plague to the Spanish flu to COVID-19. While numerous antimicrobial and antiviral drugs have been developed over the last 200 years to face these threats, the globalized and highly connected world of the 21st century demands for an ever-increasing efficiency in the detection and treatment of infectious diseases. Consequently, the rapidly evolving field of nanomedicine has taken up the challenge and developed a plethora of strategies to fight infectious diseases with the help of various nanomaterials such as noble metal nanoparticles, liposomes, nanogels, and virus capsids. DNA nanotechnology represents a comparatively recent addition to the nanomedicine arsenal, which, over the past decade, has made great progress in the area of cancer diagnostics and therapy. However, the past few years have seen also an increasing number of DNA nanotechnology-related studies that particularly focus on the detection and inhibition of microbial and viral pathogens. Herein, a brief overview of this rather young research field is provided, successful concepts as well as potential challenges are identified, and promising directions for future research are highlighted.
Collapse
Affiliation(s)
- David M. Smith
- DNA Nanodevices UnitDepartment DiagnosticsFraunhofer Institute for Cell Therapy and Immunology IZI04103LeipzigGermany
- Peter Debye Institute for Soft Matter PhysicsFaculty of Physics and Earth SciencesUniversity of Leipzig04103LeipzigGermany
- Institute of Clinical ImmunologyUniversity of Leipzig Medical School04103LeipzigGermany
- Dhirubhai Ambani Institute of Information and Communication TechnologyGandhinagar382 007India
| | - Adrian Keller
- Technical and Macromolecular ChemistryPaderborn UniversityWarburger Str. 10033098PaderbornGermany
| |
Collapse
|
94
|
Takafuji Y, Tatsumi K, Kawao N, Okada K, Muratani M, Kaji H. MicroRNA-196a-5p in Extracellular Vesicles Secreted from Myoblasts Suppresses Osteoclast-like Cell Formation in Mouse Cells. Calcif Tissue Int 2021; 108:364-376. [PMID: 33090325 DOI: 10.1007/s00223-020-00772-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
Muscle/bone interaction has been recently noted. Extracellular vesicles (EVs) play a vital role in physiological and pathophysiological processes by transferring microRNA (miRNA) to distant tissues. We previously reported that EVs secreted from C2C12 myoblasts (Myo-EVs) suppress osteoclast differentiation. In the present study, we identified 4 miRNAs in Myo-EVs that suppressed osteoclast-like cell formation in Raw264.7 cells using small RNA sequencing analysis. Among them, miR-196a-5p expression was higher in C2C12 cells compared to mouse osteoblasts and bone marrow cells. Transfection of miR-196a-5p mimic suppressed the mRNA levels of osteoclast-related genes and mitochondrial energy metabolism induced by receptor activator of nuclear factor-κB ligand in Raw264.7 cells. In contrast, miR-196a-5p mimic enhanced osteoblastic differentiation in ST-2 cells and MC3T3-E1 cells. In conclusion, we demonstrated that miR-196-5p suppresses osteoclast-like cell formation and mitochondrial energy metabolism in mouse cells, suggesting that it might be a crucial factor for muscle/bone interaction via EVs.
Collapse
Affiliation(s)
- Yoshimasa Takafuji
- Faculty of Medicine, Department of Physiology and Regenerative Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, 589-8511, Japan
| | - Kohei Tatsumi
- Faculty of Medicine, Department of Physiology and Regenerative Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, 589-8511, Japan
| | - Naoyuki Kawao
- Faculty of Medicine, Department of Physiology and Regenerative Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, 589-8511, Japan
| | - Kiyotaka Okada
- Faculty of Medicine, Department of Physiology and Regenerative Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, 589-8511, Japan
| | - Masafumi Muratani
- Faculty of Medicine, Department of Genome Biology, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Kaji
- Faculty of Medicine, Department of Physiology and Regenerative Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, 589-8511, Japan.
| |
Collapse
|
95
|
He F, Zheng WW, Chen BB, Zeng YM. MiR-139-5p inhibits invasion and migration of gastric cancer cells by targeting PAK5 gene to block Wnt/β-catenin signaling pathway. Shijie Huaren Xiaohua Zazhi 2021; 29:174-181. [DOI: 10.11569/wcjd.v29.i4.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the most common type of cancer of the digestive system. Local or systemic metastasis is the main cause of poor prognosis. MicroRNAs (miRNAs) are an important regulatory factor in the development of gastric cancer. However, the effect and mechanism of miR-139-5p on the invasion and metastasis of gastric cancer cells are still unclear.
AIM To explore the effect of miR-139-5p on the invasion and migration of gastric cancer cells and the underlying mechanism.
METHODS Real-time fluorescent quantitative PCR (qRT-PCR) and Western blot were used to detect the expression of miR-139-5p and PAK5, respectively, in immortalized gastric mucosal cell line GES1 and gastric cancer cell lines SGC-7901, AGS, and BGC-823. MiR-139-5p mimic was transfected into gastric cancer SGC-7901 cells, and qRT-PCR was used detect the transfection efficiency. Transwell invasion and scratch assays were used to detect the effect of overexpression of miR-139-5p on the invasion and migration of SGC-7901 cells. Dual luciferase reporter gene assay and Western blot were used to detect the targeted regulation of miR-139-5p on PAK5. Western blot was used to detect the effect of overexpression of miR-139-5p on the activation of Wnt/β-catenin signaling pathway.
RESULTS The expression level of miR-139-5p in gastric cancer cells was significantly lower than that of normal gastric mucosal cells (P < 0.05), and the expression of PAK5 mRNA and protein was significantly higher than that of normal gastric mucosal cells (P < 0.05). Transfection of miR-139-5p mimic up-regulated the expression of miR-139-5p in SGC-7901 cells. Overexpression of miR-139-5p significantly inhibited the invasion and migration of SGC-7901 cells. The results of dual luciferase reporter gene assay and Western blot showed that miR-139-5p can target and negatively regulate the expression of PAK5. After overexpression of miR-139-5p, the expression of Wnt3a, β-catenin, and Cyclin D1 proteins in SGC-7901 cells was significantly down-regulated.
CONCLUSION MiR-139-5p inhibits the invasion and migration of gastric cancer cells by targeting the PAK5 gene to block the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Fan He
- Department of Gastroenterology, Wenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Wenzhou 325000, Zhejiang Province, China
| | - Wei-Wei Zheng
- Department of Gastroenterology, Wenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Wenzhou 325000, Zhejiang Province, China
| | - Bing-Bing Chen
- Department of Gastroenterology, Wenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Wenzhou 325000, Zhejiang Province, China
| | - Yao-Ming Zeng
- Department of Gastroenterology, Wenzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Wenzhou 325000, Zhejiang Province, China
| |
Collapse
|
96
|
Dong X, Li Y, Cao R, Xu H. MicroRNA-363-3p Inhibits the Expression of Renal Fibrosis Markers in TGF-β1-Treated HK-2 Cells by Targeting TGF-β2. Biochem Genet 2021; 59:1033-1048. [PMID: 33630202 DOI: 10.1007/s10528-021-10044-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/29/2021] [Indexed: 11/30/2022]
Abstract
This study aimed to explore the role of miR-363-3p in renal fibrosis (RF) in vitro. HK-2 cells were treated with transforming growth factor (TGF)-β1 for 72 h to establish an in vitro model of RF. Subsequently, western blot analysis and reverse transcription-quantitative PCR were used to detect the protein and mRNA expression levels of RF markers in TGF-β1-treated HK-2 cells, respectively. The results showed that the protein and mRNA expression levels of TGF-β2, α-smooth muscle actin (SMA), fibronectin, vimentin, collagen II and N-cadherin were increased, while the protein and mRNA expression levels of E-cadherin were decreased in TGF-β1-treated HK-2 cells. The level of miR-363-3p was significantly decreased in TGF-β1-treated HK-2 cells. TargetScan indicated that TGF-β2 was a direct target gene for miR-363-3p, which was further verified using dual luciferase reporter gene assays. Further analyses revealed that the increased protein and mRNA expression levels of TGF-β2, α-SMA, fibronectin, vimentin, collagen II, N-cadherin, increased phosphorylated-Smad3 protein level, and decreased E-cadherin protein and mRNA expression in TGF-β1-treated HK-2 cells were significantly reversed by miR-363-3p mimics. However, all the effects were suppressed by a TGF-β2-plasmid. The results suggested that miR-363-3p plays a protective role in RF by regulating the TGF-β2/Smad3 signaling pathway.
Collapse
Affiliation(s)
- Xiangnan Dong
- Department of Urinary Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Yang Li
- Department of Nephrology, Qingdao Municipal Hospital, 1 Jiaozhou Road, Shibei, Qingdao, 266000, Shandong, China
| | - Rui Cao
- Department of Blood Purification Center, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Honglan Xu
- Department of Nephrology, Qingdao Municipal Hospital, 1 Jiaozhou Road, Shibei, Qingdao, 266000, Shandong, China.
| |
Collapse
|
97
|
Liang XL, Wang YL, Wang PR. MiR-200a with CDC7 as a direct target declines cell viability and promotes cell apoptosis in Wilm's tumor via Wnt/β-catenin signaling pathway. Mol Cell Biochem 2021; 476:2409-2420. [PMID: 33599894 DOI: 10.1007/s11010-021-04090-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/29/2021] [Indexed: 12/25/2022]
Abstract
MiR-200a acts as a key role in tumor malignant progression. This work purposed to assess the function of miR-200a in Wilm's tumor. Based on bioinformatics analysis, the expression, prognostic value and related pathways of miR-200a and CDC7 (a potential downstream molecule of miR-200a) in Wilm's tumor were analyzed. qRT-PCR was conducted to confirm the miR-200a level in Wilm's tumor cells. The luciferase reporter assay was carried out to verify the binding of miR-200a to 3'-UTR of CDC7. Then, the impacts of miR-200a and CDC7 on cell viability and apoptosis were measured using CCK-8 and flow cytometry assays. Also, western blot was applied to measure the expression of CDC7 as well as Wnt/β-catenin signaling pathway-related proteins and apoptosis proteins. Herein, we revealed that miR-200a was lowly expressed in Wilm's tumor tissues and cells and the low miR-200a expression is closely bound up with death and poor outcomes. Moreover, miR-200a directly targeted and inhibited CDC7 in Wilm's tumor cells. Biological function experiments illustrated that overexpression of miR-200a reduced the viability and elevated the apoptosis of Wilm's tumor cells, while overexpression of CDC7 reversed the inhibitory impact of miR-200a on cell viability and the promoting impact of miR-200a on cell apoptosis. Besides, we revealed that miR-200a/CDC7 axis can decrease the expression of β-Catenin, Cyclin D1 and C-Myc as well as the phosphorylation of GSK-3β, thus inhibiting the Wnt/β-catenin signaling pathway. Furthermore, blocking the Wnt/β-catenin signaling pathway caused an increase on cell apoptosis, while overexpression of CDC7 can reverse these impacts. Collectively, miR-200a/CDC7 axis involved in regulating the malignant phenotype of Wilm's tumor through Wnt/β-catenin signaling pathway, which provides a theoretical basis for targeted molecular therapy of Wilm's tumor.
Collapse
Affiliation(s)
- Xiu-Ling Liang
- Department of Pediatrics, Second Hospital Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Street, Jinan, People's Republic of China.,Department of Pediatric Internal Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, People's Republic of China
| | - Yu-Long Wang
- Department of Pediatrics, Second Hospital Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Street, Jinan, People's Republic of China
| | - Pei-Rong Wang
- Department of Pediatrics, Second Hospital Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Street, Jinan, People's Republic of China.
| |
Collapse
|
98
|
Esfandyari YB, Doustvandi MA, Amini M, Baradaran B, Zaer SJ, Mozammel N, Mohammadzadeh M, Mokhtarzadeh A. MicroRNA-143 Sensitizes Cervical Cancer Cells to Cisplatin: a Promising Anticancer Combination Therapy. Reprod Sci 2021; 28:2036-2049. [PMID: 33569751 DOI: 10.1007/s43032-021-00479-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Cisplatin-based chemotherapy is commonly used for cervical cancer treatment. However, the development of chemoresistance is considered the main obstacle to the effectiveness of this therapeutic agent. MicroRNAs are illustrated to play a major role in the regulation of cancer cell chemosensitivity. Therefore, this study was aimed to investigate the potential therapeutic role of miRNA-143 in combination with cisplatin on cervical cancer cells. Then, CaSki cell line with low expression levels of miRNA-143 was selected for functional experiments. The cells were treated with miRNA-143 and cisplatin individually or in combination. The cell viability and apoptosis induction were evaluated by MTT, Annexin V-FITC/PI, and DAPI staining tests. Cell migration was further evaluated by wound healing assay. The effect of miRNA-143 and cisplatin combination on gene expression was quantified by real-time PCR. Furthermore, the combination therapy effect on cell cycle progression and autophagy induction was also evaluated by flow cytometry. Our results showed that miRNA-143 overexpression could increase cisplatin-induced apoptosis and increase the sensitivity of CaSki cells to low doses of this chemotherapeutic agent via modulating the expression of apoptosis-related genes including Bcl-2, Bax, and caspase-9. Besides, miRNA-143 and cisplatin were demonstrated to cooperatively increase the cell cycle arrest at the sub-G1 and G2-M phases, induce autophagy activation, and via downregulation of vimentin inhibit CaSki cell migration. Moreover, c-Myc as an important regulator of cell growth was downregulated in treatment groups compared to the control. In conclusion, regarding that miRNA-143 could sensitize cervical cancer cells to cisplatin, it may be considered a promising therapeutic strategy for the treatment of this malignancy.
Collapse
Affiliation(s)
- Yalda Baghay Esfandyari
- Department of Biology, Faculty of Sciences, Urmia University, Urmia, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sheyda Jodeiry Zaer
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Mozammel
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
99
|
Zhang G, Xia M, Guo J, Huang Y, Huang J, Wei K, Zhang X, Zeng J, Liang W. microRNA-1296 Inhibits Glioma Cell Growth by Targeting ABL2. Technol Cancer Res Treat 2021; 20:1533033821990009. [PMID: 33550941 PMCID: PMC7876570 DOI: 10.1177/1533033821990009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Aberrant expression of microRNAs (miRNAs) has been reported to play a role in tumorigenesis. Dysfunction of miR-1296 was found in a variety of cancers, however, the function of miR-1296 in the progression of glioma remains largely understood. Here, our results showed that miR-1296 was significantly down-regulated in glioma tissues and cell lines. Decreased expression of miR-1296 was associated with the tumor size, WHO grade and karnofsky performance scale (KPS) of glioma patients. Low expression of miR-1296 was significantly correlated with the shorter 5-year overall survival of glioma patients. Overexpression of miR-1296 inhibited the proliferation, colony formation, migration and induced apoptosis of glioma cells. MiR-1296 was found to bind the 3’-untranslated region (UTR) of ABL proto-oncogene 2 (ABL2) and subsequently repressed both the mRNA and protein expression of ABL2. ABL2 was overexpressed in glioma tissues and inversely correlated with that of miR-1296. Ectopic expressed ABL2 could reverse the inhibitory effects of miR-1296 on glioma cell proliferation. Our results illustrated the novel tumor-suppressive function of miR-1296 in glioma via repressing ABL2, suggesting a potential application of miR-1296 in the treatment of glioma.
Collapse
Affiliation(s)
- Gaolian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Meng Xia
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Jianhui Guo
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Yi Huang
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Jianrong Huang
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Kecong Wei
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Xiaoning Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Jing Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| | - Weibin Liang
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People's Republic of China
| |
Collapse
|
100
|
Tian J, Zhao Y, Li L, Cui Y, Wu Y. MicroRNA-936 Targets JAG1 and Inhibits the Proliferation of Hepatocellular Carcinoma Cells. Technol Cancer Res Treat 2021; 20:1533033820985785. [PMID: 33550933 PMCID: PMC7876578 DOI: 10.1177/1533033820985785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors. Investigating the underlying molecular mechanism is essential for the treatment and prognosis of HCC. Emerging evidence suggests that microRNAs (miRNAs) play pivotal roles in cancer progression. Down-regulation of miR-936 has been found in several cancers, which serves as a tumor suppressor to inhibit the development of cancers. However, the clinical significance and functional roles of miR-936 in HCC have not been determined. To explore this, the expression of miR-936 in HCC tissues and cells was detected by RT-qPCR. Cell Counting Kit-8 (CCK-8) assay, cell migration and cell cycle analysis were performed to evaluate the effects of miR-936 on the growth of HCC cells. The targets of miR-936 were predicted using the miRDB database and confirmed by luciferase reporter experiments. The protein expression of targets was determined by western blot. The results showed that miR-936 was significantly decreased in HCC tissues and cell lines. Low expression of miR-936 was associated with the advance progression and poor survival of HCC patients (P = 0.0036). Functional study revealed that overexpression of miR-936 inhibited the proliferation, migration (decreased to ∼0.26 fold) and induced cell cycle arrested in G1 phase (from 35.3% to 44.7%) of HCC cells. Additionally, miR-936 targeted the 3′-untranslated region (UTR) of jagged-1 (JAG1) and reduced the expression of JAG1 (decreased to ∼0.35 fold). JAG1 was found to be up-regulated in HCC tissues and was inversely correlated with the expression of miR-936 (Pearson r = −0.4633; P = 0.0007). The anti-cancer effects of miR-936 on the proliferation of HCC cells were partially reversed by the rescue of JAG1. Therefore, these results suggested that miR-936 might be a potential target for HCC treatment.
Collapse
Affiliation(s)
- Junmei Tian
- Nutritional Department, Luoyang Central Hospital Affiliated Zhengzhou University, Luoyang, Henan, People's Republic of China
| | - Yongfei Zhao
- Nutritional Department, Luoyang Central Hospital Affiliated Zhengzhou University, Luoyang, Henan, People's Republic of China
| | - Li Li
- Nutritional Department, Luoyang Central Hospital Affiliated Zhengzhou University, Luoyang, Henan, People's Republic of China
| | - Yanling Cui
- Nutritional Department, Kaifeng Central Hospital, Kaifeng, Henan, People's Republic of China
| | - Yang Wu
- Oncology Department, Luoyang Central Hospital Affiliated Zhengzhou University, Luoyang, Henan, People's Republic of China
| |
Collapse
|