1
|
Shoari A, Ashja Ardalan A, Dimesa AM, Coban MA. Targeting Invasion: The Role of MMP-2 and MMP-9 Inhibition in Colorectal Cancer Therapy. Biomolecules 2024; 15:35. [PMID: 39858430 PMCID: PMC11762759 DOI: 10.3390/biom15010035] [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: 12/09/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the most prevalent and lethal cancers worldwide, prompting ongoing research into innovative therapeutic strategies. This review aims to systematically evaluate the role of gelatinases, specifically MMP-2 and MMP-9, as therapeutic targets in CRC, providing a critical analysis of their potential to improve patient outcomes. Gelatinases, specifically MMP-2 and MMP-9, play critical roles in the processes of tumor growth, invasion, and metastasis. Their expression and activity are significantly elevated in CRC, correlating with poor prognosis and lower survival rates. This review provides a comprehensive overview of the pathophysiological roles of gelatinases in CRC, highlighting their contribution to tumor microenvironment modulation, angiogenesis, and the metastatic cascade. We also critically evaluate recent advancements in the development of gelatinase inhibitors, including small molecule inhibitors, natural compounds, and novel therapeutic approaches like gene silencing techniques. Challenges such as nonspecificity, adverse side effects, and resistance mechanisms are discussed. We explore the potential of gelatinase inhibition in combination therapies, particularly with conventional chemotherapy and emerging targeted treatments, to enhance therapeutic efficacy and overcome resistance. The novelty of this review lies in its integration of recent findings on diverse inhibition strategies with insights into their clinical relevance, offering a roadmap for future research. By addressing the limitations of current approaches and proposing novel strategies, this review underscores the potential of gelatinase inhibitors in CRC prevention and therapy, inspiring further exploration in this promising area of oncological treatment.
Collapse
Affiliation(s)
- Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Arghavan Ashja Ardalan
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | | | - Mathew A. Coban
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA;
| |
Collapse
|
2
|
Liu X, Wang CX, Feng Q, Zhang T. lncRNA TINCR promotes the development of cervical cancer via the miRNA‑7/mTOR axis in vitro. Exp Ther Med 2023; 26:487. [PMID: 37745037 PMCID: PMC10515118 DOI: 10.3892/etm.2023.12186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/09/2023] [Indexed: 09/26/2023] Open
Abstract
The present study aimed to examine the effects of the long non-coding (lnc)RNA expressed by tissue differentiation-inducing non-protein coding RNA (TINCR) on cervical cancer development. For this purpose, adjacent normal and cancer tissues were obtained from patients with cervical cancer and the lncRNA TINCR level was examined using reverse transcription-quantitative PCR (RT-qPCR) and in situ hybridization. The association between lncRNA TINCR and the clinicopathological characteristics and prognosis of patients with cervical cancer was also analyzed. Furthermore, the expression levels of lncRNA TINCR, miRNA-7, mTOR, hypoxia-inducible factor 1 subunit α and VEGF were measured using RT-qPCR and western blot analysis. Cell proliferation, apoptosis, and invasion and migration were examined using MTT assay, 5-ethynyl-2'-deoxyuridine staining, flow cytometry, TUNEL assay, and Transwell and wound healing assays. The association between lncRNA TINCR, miRNA-7 and mTOR was also examined using a luciferase assay. The results revealed that the lncRNA TINCR level was significantly increased in cervical cancer tissues and was associated with the overall survival of patients (low vs. high expression group; P=0.0391). LncRNA TINCR was also associated with the clinicopathological characteristics of patients with cervical cancer. Following the knockdown of lncRNA TINCR using small interfering (si)RNA, cell proliferation was significantly decreased and cell apoptosis was significantly increased (P<0.001 for both); cell invasion and migration were also significantly decreased (P<0.001 for both) following transfection with mimics miRNA-7. Transfection with miRNA-7 antisense oligonucleotide decreased the antitumor effects of si-TINCR in Siha and HeLa cell lines. As shown using the dual-luciferase assay, lncRNA TINCR could target miRNA-7 and miRNA-7 could directly regulate mTOR in HeLa and SiHa cell lines. In conclusion, the present study demonstrated that lncRNA TINCR could promote cervical cancer development via regulation of the miRNA-7/mTOR axis in vitro.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Gynecology, Women and Children's Hospital, Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Cui Xia Wang
- Department of Pediatrics, Eighth People's Hospital of Qingdao Shandong, Qingdao, Shandong 266000, P.R. China
| | - Qin Feng
- Department of Imaging, Women and Children's Hospital, Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Tao Zhang
- Department of General Internal Medicine, Women and Children's Hospital, Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
3
|
Ignjatović Jocić V, Janković Miljuš J, Išić Denčić T, Živaljević V, Tatić S, Đorić I, Šelemetjev S. Expression of pY397-FAK and Its miR Regulators Drive Dedifferentiation in the Thyroid Neoplasia Spectrum. Cells 2023; 12:1721. [PMID: 37443754 PMCID: PMC10340340 DOI: 10.3390/cells12131721] [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: 05/17/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 07/15/2023] Open
Abstract
Thyroid carcinomas are growing malignancies worldwide. They encompass several diagnostic categories with varying degrees of dedifferentiation. Focal adhesion kinase is involved in cellular communication and locomotion. It is regulated on a posttranscriptional level by miR-7, miR-135a, and miR-138 and on a posttranslational level by autophosphorylation at Y397 (pY397-FAK). We related regulators of FAK with histologic dedifferentiation, clinicopathological factors, and differential diagnosis in the thyroid neoplasia spectrum. We classified 82 cases into 5 groups with increasing aggressiveness: healthy tissue, follicular and classical variants of papillary thyroid carcinoma (PTC), dedifferentiated PTC, and anaplastic carcinoma. MiRs were analyzed by RT-qPCR. Protein expression of pY397-FAK was analyzed by immunohistochemistry (separately in the membrane, cytoplasm, and nuclear compartment) and Western blot. All three miRs were upregulated in healthy tissue compared to malignant, while pY397-FAK was downregulated. MiRs and pY397-FAK were not mutually correlated. MiR-135a-5p was decreasing while membranous and cytoplasmic pY397-FAK increased with dedifferentiation. Neither miR correlated with clinicopathological factors. MiR-135a-5p, miR-138-5p, and membranous and cytoplasmic pY397-FAK discriminated the follicular from the classical variant of PTC. Disturbances of FAK regulation on different levels contribute to neoplastic dedifferentiation. pY397-FAK exerts its oncogenic role in the membrane and cytoplasm. Diagnostically, miRs-135a-5p, miR-138-5p, and membranous and cytoplasmic pY397-FAK differentiated between classical and follicular PTC.
Collapse
Affiliation(s)
- Valentina Ignjatović Jocić
- Department of Endocrinology and Radioimmunology, Institute for the Application of Nuclear Energy—INEP, University of Belgrade, Banatska 31b, 11000 Belgrade, Serbia
| | - Jelena Janković Miljuš
- Department of Endocrinology and Radioimmunology, Institute for the Application of Nuclear Energy—INEP, University of Belgrade, Banatska 31b, 11000 Belgrade, Serbia
| | - Tijana Išić Denčić
- Department of Endocrinology and Radioimmunology, Institute for the Application of Nuclear Energy—INEP, University of Belgrade, Banatska 31b, 11000 Belgrade, Serbia
| | - Vladan Živaljević
- Center for Endocrine Surgery, University Clinical Center of Serbia, Doktora Subotića 13, 11000 Belgrade, Serbia
| | - Svetislav Tatić
- Institute for Pathology, Faculty of Medicine, University of Belgrade, Doktora Subotića Starijeg 1, 11000 Belgrade, Serbia
| | - Ilona Đorić
- Department of Endocrinology and Radioimmunology, Institute for the Application of Nuclear Energy—INEP, University of Belgrade, Banatska 31b, 11000 Belgrade, Serbia
| | - Sonja Šelemetjev
- Department of Endocrinology and Radioimmunology, Institute for the Application of Nuclear Energy—INEP, University of Belgrade, Banatska 31b, 11000 Belgrade, Serbia
| |
Collapse
|
4
|
Sell MC, Ramlogan-Steel CA, Steel JC, Dhungel BP. MicroRNAs in cancer metastasis: biological and therapeutic implications. Expert Rev Mol Med 2023; 25:e14. [PMID: 36927814 PMCID: PMC10407223 DOI: 10.1017/erm.2023.7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 01/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Cancer metastasis is the primary cause of cancer-related deaths. The seeding of primary tumours at a secondary site is a highly inefficient process requiring substantial alterations in the genetic architecture of cancer cells. These alterations include significant changes in global gene expression patterns. MicroRNAs are small, non-protein coding RNAs which play a central role in regulating gene expression. Here, we focus on microRNA determinants of cancer metastasis and examine microRNA dysregulation in metastatic cancer cells. We dissect the metastatic process in a step-wise manner and summarise the involvement of microRNAs at each step. We also discuss the advantages and limitations of different microRNA-based strategies that have been used to target metastasis in pre-clinical models. Finally, we highlight current clinical trials that use microRNA-based therapies to target advanced or metastatic tumours.
Collapse
Affiliation(s)
- Marie C. Sell
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia
| | - Charmaine A. Ramlogan-Steel
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia
| | - Jason C. Steel
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, QLD 4701, Australia
| | - Bijay P. Dhungel
- Gene & Stem Cell Therapy Program Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
- Faculty of Medicine & Health, The University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
5
|
Antonio LGL, Meola J, Rosa-e-Silva ACJDS, Nogueira AA, Candido dos Reis FJ, Poli-Neto OB, Rosa-e-Silva JC. Altered Differential Expression of Genes and microRNAs Related to Adhesion and Apoptosis Pathways in Patients with Different Phenotypes of Endometriosis. Int J Mol Sci 2023; 24:ijms24054434. [PMID: 36901866 PMCID: PMC10002379 DOI: 10.3390/ijms24054434] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 03/12/2023] Open
Abstract
We aim to investigate the expression of genes (MAPK1 and CAPN2) and microRNAs (miR-30a-5p, miR-7-5p, miR-143-3p, and miR-93-5p) involved in adhesion and apoptosis pathways in superficial peritoneal endometriosis (SE), deep infiltrating endometriosis (DE), and ovarian endometrioma (OE), and to evaluate whether these lesions share the same pathophysiological mechanisms. We used samples of SE (n = 10), DE (n = 10), and OE (n = 10), and endometrial biopsies of these respective patients affected with endometriosis under treatment at a tertiary University Hospital. Endometrial biopsies collected in the tubal ligation procedure from women without endometriosis comprised the control group (n = 10). Quantitative real-time polymerase chain reaction was performed. The expression of MAPK1 (p < 0.0001), miR-93-5p (p = 0.0168), and miR-7-5p (p = 0.0006) was significantly lower in the SE group than in the DE and OE groups. The expression of miR-30a (p = 0.0018) and miR-93 (p = 0.0052) was significantly upregulated in the eutopic endometrium of women with endometriosis compared to the controls. MiR-143 (p = 0.0225) expression also showed a statistical difference between the eutopic endometrium of women with endometriosis and the control group. In summary, SE showed lower pro-survival gene expression and miRNAs involved in this pathway, indicating that this phenotype has a different pathophysiological mechanism compared to DE and OE.
Collapse
|
6
|
Qiao D, Xing J, Duan Y, Wang S, Yao G, Zhang S, Jin J, Lin Z, Chen L, Piao Y. The molecular mechanism of baicalein repressing progression of gastric cancer mediating miR-7/FAK/AKT signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154046. [PMID: 35306368 DOI: 10.1016/j.phymed.2022.154046] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/20/2022] [Accepted: 03/11/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Baicalein (BAI) has a significant anti-cancerous function in the treatment of gastric cancer (GC). Focal adhesion kinase (FAK) is a key regulatory molecule in integrin and growth factor receptor mediated signaling. MicroRNA-7 (miR-7), has been considered as a potential tumor suppressor in a variety of cancers. However, the possible mechanisms by which BAI inhibiting progression of gastric cancer mediating miR-7/FAK/AKT signaling pathway remain unclear. PURPOSE To investigate the molecular mechanism and effects of BAI inhibiting progression of gastric cancer mediating miR-7/FAK/AKT signaling pathway. METHODS Gastric cancer cell lines with FAK knockdown and overexpression were constructed by lentivirus transfection. After BAI treatment, the effects of FAK protein on proliferation, metastasis and angiogenesis of gastric cancer cells were detected by MTT, EdU, colony formation, wound healing, transwell and Matrigel tube formation assays. In vivo experiment was performed by xenograft model. Immunofluorescence and western blot assay were used to detect the effects of FAK protein on the expression levels of EMT markers and PI3K/AKT signaling pathway related proteins. qRT-PCR and luciferase reporter assay were used to clarify the targeting relationship between miR-7 and FAK. RESULTS BAI can regulate FAK to affect proliferation, metastasis and angiogenesis of gastric cancer cells through PI3K/AKT signaling pathway. qRT-PCR showed BAI can upregulated the expression of miR-7 and luciferase reporter assay showed the targeting relationship between miR-7 and FAK. Additionally, miR-7 mediates cell proliferation, metastasis and angiogenesis by directly targeting FAK 3'UTR to inhibit FAK expression. CONCLUSION BAI repressing progression of gastric cancer mediating miR-7/FAK/AKT signaling pathway.
Collapse
Affiliation(s)
- Dan Qiao
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China
| | - Jian Xing
- Department of Image, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, P.R. China
| | - Yunxiao Duan
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China
| | - Shiyu Wang
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China
| | - Guangyuan Yao
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China
| | - Shengjun Zhang
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China
| | - Jingchun Jin
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China; Department of Internal Medicine of Yanbian University Hospital, Yanji 133000, P.R. China
| | - Zhenhua Lin
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China; Department of Internal Medicine of Yanbian University Hospital, Yanji 133000, P.R. China
| | - Liyan Chen
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China
| | - Yingshi Piao
- Cancer Research Center, Yanbian University Medical College, Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Research and Innovation Group of Yanbian University, Yanji, P.R. China.
| |
Collapse
|
7
|
Emerging roles of circular RNAs in cancer: a narrative review. JOURNAL OF PANCREATOLOGY 2022. [DOI: 10.1097/jp9.0000000000000087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
8
|
Rauschner M, Hüsing T, Lange L, Jarosik K, Reime S, Riemann A, Thews O. Role of acidosis-sensitive microRNAs in gene expression and functional parameters of tumors in vitro and in vivo. Neoplasia 2021; 23:1275-1288. [PMID: 34781085 PMCID: PMC8605108 DOI: 10.1016/j.neo.2021.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/04/2021] [Indexed: 11/09/2022] Open
Abstract
Background: The acidic extracellular environment of tumors has been shown to affect the malignant progression of tumor cells by modulating proliferation, cell death or metastatic potential. The aim of the study was to analyze whether acidosis-dependent miRNAs play a role in the signaling cascade from low pH through changes in gene expression to functional properties of tumors in vitro and in vivo. Methods: In two experimental tumor lines the expression of 13 genes was tested under acidic conditions in combination with overexpression or downregulation of 4 pH-sensitive miRNAs (miR-7, 183, 203, 215). Additionally, the impact on proliferation, cell cycle distribution, apoptosis, necrosis, migration and cell adhesion were measured. Results: Most of the genes showed a pH-dependent expression, but only a few of them were additionally regulated by miRNAs in vitro (Brip1, Clspn, Rif1) or in vivo (Fstl, Tlr5, Txnip). Especially miR-215 overexpression was able to counteract the acidosis effect in some genes. The impact on proliferation was cell line-dependent and most pronounced with overexpression of miR-183 and miR-203, whereas apoptosis and necrosis were pH-dependent but not influenced by miRNAs. The tumor growth was markedly regulated by miR-183 and miR-7. In addition, acidosis had a strong effect on cell adhesion, which could be modulated by miR-7, miR-203 and miR-215. Conclusions: The results indicate that the acidosis effect on gene expression and functional properties of tumor cells could be mediated by pH-dependent miRNAs. Many effects were cell line dependent and therefore do not reflect universal intracellular signaling cascades. However, the role of miRNAs in the adaptation to an acidic environment may open new therapeutic strategies.
Collapse
Affiliation(s)
- Mandy Rauschner
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6, Halle (Saale) 06112, Germany
| | - Thea Hüsing
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6, Halle (Saale) 06112, Germany
| | - Luisa Lange
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6, Halle (Saale) 06112, Germany
| | - Kristin Jarosik
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6, Halle (Saale) 06112, Germany
| | - Sarah Reime
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6, Halle (Saale) 06112, Germany
| | - Anne Riemann
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6, Halle (Saale) 06112, Germany
| | - Oliver Thews
- Julius Bernstein Institute of Physiology, University of Halle-Wittenberg, Magdeburger Str. 6, Halle (Saale) 06112, Germany.
| |
Collapse
|
9
|
Khorsandi K, Esfahani H, Abrahamse H. Characteristics of circRNA and its approach as diagnostic tool in melanoma. Expert Rev Mol Diagn 2021; 21:1079-1094. [PMID: 34380368 DOI: 10.1080/14737159.2021.1967749] [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: 12/08/2022]
Abstract
One of the most common types of cancer in the world is skin cancer, which has been divided into two groups: non-melanoma and melanoma skin cancer. Different external and internal agents are considered as risk factors for melanoma skin cancer pathogenesis but the exact mechanisms are not yet confirmed. Genetic and epigenetic changes, UV exposure, arsenic compounds, and chemical substances are contributory factors to the development of melanoma. A correlation has emerged between new therapies and the discovery of a basic molecular pattern for skin cancer patients. Circular RNAs (circRNAs) are described as a unique group of extensively expressed endogenous regulatory RNAs with closed-loop structure bonds connecting the 5' and 3' ends, which are commonly expressed in mammalian cells. In this review, we describe the biogenesis of circular RNAs and its function in cancerous conditions focusing on the crosstalk between different circRNAs and melanoma. Increasing evidence suggests that circRNAs appears to be relative to the origin and development of skin-related diseases like malignant melanoma. Different circular RNAs like hsa_circ_0025039, hsa_circRNA006612, circRNA005537, and circANRIL, by targeting different cellular and molecular targets (e.g., CDK4, DAB2IP, ZEB1, miR-889, and let-7 c-3p), can participate in melanoma cancer progression.
Collapse
Affiliation(s)
- Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - HomaSadat Esfahani
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Heidi Abrahamse
- Laser Research Centre, Nrf SARChI Chair: Laser Applications in Health, Faculty of Health Sciences, University of Johannesburg, Auckland Park, South Africa
| |
Collapse
|
10
|
Korać P, Antica M, Matulić M. MiR-7 in Cancer Development. Biomedicines 2021; 9:325. [PMID: 33806891 PMCID: PMC8004586 DOI: 10.3390/biomedicines9030325] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNA involved in the regulation of specific mRNA translation. They participate in cellular signaling circuits and can act as oncogenes in tumor development, so-called oncomirs, as well as tumor suppressors. miR-7 is an ancient miRNA involved in the fine-tuning of several signaling pathways, acting mainly as tumor suppressor. Through downregulation of PI3K and MAPK pathways, its dominant role is the suppression of proliferation and survival, stimulation of apoptosis and inhibition of migration. Besides these functions, it has numerous additional roles in the differentiation process of different cell types, protection from stress and chromatin remodulation. One of the most investigated tissues is the brain, where its downregulation is linked with glioblastoma cell proliferation. Its deregulation is found also in other tumor types, such as in liver, lung and pancreas. In some types of lung and oral carcinoma, it can act as oncomir. miR-7 roles in cell fate determination and maintenance of cell homeostasis are still to be discovered, as well as the possibilities of its use as a specific biotherapeutic.
Collapse
Affiliation(s)
- Petra Korać
- Department of Biology, Division of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102, 10000 Zagreb, Croatia;
| | - Mariastefania Antica
- Division of Molecular Biology, Rudjer Bosković Institute, Bijenička 54, 10000 Zagreb, Croatia;
| | - Maja Matulić
- Department of Biology, Division of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102, 10000 Zagreb, Croatia;
| |
Collapse
|
11
|
Chang L, Zhou D, Luo S. Novel lncRNA LINC00941 Promotes Proliferation and Invasion of Colon Cancer Through Activation of MYC. Onco Targets Ther 2021; 14:1173-1186. [PMID: 33654409 PMCID: PMC7910107 DOI: 10.2147/ott.s293519] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/22/2021] [Indexed: 01/05/2023] Open
Abstract
Purpose We conducted the study to elucidate how LncRNA LINC00941 affects colon cancer progression and its possible regulatory mechanism. Methods The expression level of LINC00941 in colon cancer tissues and cells was detected by qRT-PCR. The function of LINC00941 on colon cancer cell proliferation, migration, and invasion was detected by CCK-8 and Transwell assay respectively. The target interactions among LINC00941, miR-205-5p, and MYC were further confirmed by dual-luciferase reporter gene assays and RNA pull-down experiments. Meanwhile, in vivo experiments were carried out to study the role of LINC00941 in the xenotransplantation model. Results LINC00941 expression level was elevated in colon cancer tissues and cells. LINC00941 overexpression accelerated proliferation, migration, and invasion of colon cancer cells, while the LINC00941 knockdown showed the opposite results. In addition, LINC00941 regulated the expression of MYC by sponging miR-205-5p as a competitive endogenous RNA, and miR-205-5p knockdown reversed the tumor inhibition of LINC00941 knockdown on colon cancer cells. Xenograft model assay confirmed that LINC00941 silencing could inhibit colon cancer cell growth and metastasis. Conclusion LINC00941 may markedly promote colon cancer progression by acting on the miR-205-5p/MYC axis as a ceRNA, which offers novel clues for lncRNA to guide the treatment and prognosis of colon cancer.
Collapse
Affiliation(s)
- Lin Chang
- Intensive Care Unit, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Dongmin Zhou
- Intensive Care Unit, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Suxia Luo
- Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| |
Collapse
|
12
|
Ku GW, Kang Y, Yu SL, Park J, Park S, Jeong IB, Kang MW, Son JW, Kang J. LncRNA LINC00240 suppresses invasion and migration in non-small cell lung cancer by sponging miR-7-5p. BMC Cancer 2021; 21:44. [PMID: 33422052 PMCID: PMC7796488 DOI: 10.1186/s12885-020-07755-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background lncRNAs have important roles in regulating cancer biology. Accumulating evidence has established a link between the dysregulation of lncRNAs and microRNA in cancer progression. In previous studies, miR-7-5p has been found to be significantly down-regulated in mesenchymal-like lung cancer cell lines and directly regulated EGFR. In this work, we investigated the lncRNA partner of miR-7-5p in the progression of lung cancer. Methods We investigated the expression of miR-7-5p and the lncRNA after transfection with an miR-7-5p mimics using a microarray. The microarray results were validated using quantitative real time-polymerase Chain Reaction (qRT-PCR). The regulatory effects of lncRNA on miR-7-5p and its target were evaluated by changes in the expression of miR-7-5p after transfection with siRNAs for lncRNA and the synthesis of full-length lncRNA. The effect of miR-7-5p on lncRNA and the miRNA target was evaluated after transfection with miRNA mimic and inhibitor. The role of lncRNA in cancer progression was determined using invasion and migration assays. The level of lncRNA and EGFR in lung cancer and normal lung tissue was analyzed using TCGA data. Results We found that LINC00240 was downregulated in lung cancer cell line after miR-7-5p transfection with an miR-7-5p mimic. Further investigations revealed that the knockdown of LINC00240 induced the overexpression of miR-7-5p. The overexpression of miR-7-5p diminished cancer invasion and migration. The EGFR expression was down regulated after siRNA treatment for LINC00240. Silencing LINC00240 suppressed the invasion and migration of lung cancer cells, whereas LINC00240 overexpression exerted the opposite effect. The lower expression of LINC00240 in squamous lung cancer was analyzed using TCGA data. Conclusions Taken together, LINC00240 acted as a sponge for miR-7-5p and induced the overexpression of EGFR. LINC00240 may represent a potential target for the treatment of lung cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-020-07755-8.
Collapse
Affiliation(s)
- Gwan Woo Ku
- Department of Thoracic Surgery, Konyang University Hospital, Daejeon, 35365, Republic of Korea
| | - Yujin Kang
- Priority Research Center, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Seong-Lan Yu
- Priority Research Center, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Joonghoon Park
- Graduate School of International Agricultural Technology and Institute of GreenBio Science and Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Sejin Park
- Department of Internal Medicine, Konyang University Hospital, Daejeon, Republic of Korea
| | - In Beom Jeong
- Department of Internal Medicine, Konyang University Hospital, Daejeon, Republic of Korea
| | - Min Woong Kang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
| | - Ji Woong Son
- Department of Internal Medicine, Konyang University Hospital, Daejeon, Republic of Korea.
| | - Jaeku Kang
- Priority Research Center, Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea. .,Department of Pharmacology, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea.
| |
Collapse
|
13
|
Grzywa TM, Klicka K, Włodarski PK. Regulators at Every Step-How microRNAs Drive Tumor Cell Invasiveness and Metastasis. Cancers (Basel) 2020; 12:E3709. [PMID: 33321819 PMCID: PMC7763175 DOI: 10.3390/cancers12123709] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor cell invasiveness and metastasis are the main causes of mortality in cancer. Tumor progression is composed of many steps, including primary tumor growth, local invasion, intravasation, survival in the circulation, pre-metastatic niche formation, and metastasis. All these steps are strictly controlled by microRNAs (miRNAs), small non-coding RNA that regulate gene expression at the post-transcriptional level. miRNAs can act as oncomiRs that promote tumor cell invasion and metastasis or as tumor suppressor miRNAs that inhibit tumor progression. These miRNAs regulate the actin cytoskeleton, the expression of extracellular matrix (ECM) receptors including integrins and ECM-remodeling enzymes comprising matrix metalloproteinases (MMPs), and regulate epithelial-mesenchymal transition (EMT), hence modulating cell migration and invasiveness. Moreover, miRNAs regulate angiogenesis, the formation of a pre-metastatic niche, and metastasis. Thus, miRNAs are biomarkers of metastases as well as promising targets of therapy. In this review, we comprehensively describe the role of various miRNAs in tumor cell migration, invasion, and metastasis.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Klaudia Klicka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (T.M.G.); (K.K.)
| |
Collapse
|
14
|
Zhang Y, Chen XF, Li J, He F, Li X, Guo Y. lncRNA Neat1 Stimulates Osteoclastogenesis Via Sponging miR-7. J Bone Miner Res 2020; 35:1772-1781. [PMID: 32353178 DOI: 10.1002/jbmr.4039] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/10/2020] [Accepted: 04/22/2020] [Indexed: 12/25/2022]
Abstract
Increasing evidence uncover the essential role of long noncoding RNA (lncRNAs) in bone metabolism and the association of lncRNA with genetic risk of osteoporosis. However, whether lncRNA nuclear paraspeckle assembly transcript 1 (Neat1) is involved remains largely unknown. In the present study, we found that Neat1 is induced by osteoclastic differentiation stimuli. Knockdown of Neat1 attenuates osteoclast formation whereas overexpression of Neat1 accelerates osteoclast formation. In vivo evidence showed that enhanced Neat1 expression stimulates osteoclastogenesis and reduces bone mass in mice. Mechanically, Neat1 competitively binds with microRNA 7 (miR-7) and blocks its function for regulating protein tyrosine kinase 2 (PTK2). Intergenic SNP rs12789028 acts as allele-specific long-range enhancer for NEAT1 via chromatin interactions. We establish for the first time that Neat1 plays an essential role in osteoclast differentiation, and provide genetic mechanism underlying the association of NEAT1 locus with osteoporosis risk. These results enrich the current knowledge of NEAT1 function, and uncover the potential of NEAT1 as a therapeutic target for osteoporosis. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yan Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiao-Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Jing Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Fang He
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xu Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, and Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
15
|
Luo Q, Zeng L, Tang C, Zhang Z, Chen Y, Zeng C. TLR9 induces colitis-associated colorectal carcinogenesis by regulating NF-κB expression levels. Oncol Lett 2020; 20:110. [PMID: 32863923 PMCID: PMC7448563 DOI: 10.3892/ol.2020.11971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
Abstract
Chronic colorectal inflammation has been associated with colorectal cancer (CRC); however, its exact molecular mechanisms remain unclear. The present study aimed to investigate the effect of Toll-like receptor 9 (TLR9) on the development of colitis-associated CRC (CAC) through its regulation of the NF-κB signaling pathway. By using a CAC mouse model and immunohistochemistry, the present study discovered that the protein expression levels of TLR9 were gradually upregulated during the development of CRC. In addition, the expression levels of TLR9 were revealed to be positively correlated with NF-κB and Ki67 expression levels. In vitro, inhibiting TLR9 expression levels using chloroquine decreased the cell viability, proliferation and migration of the CRC cell line HT29, and further experiments indicated that this may occur through downregulating the expression levels of NF-κB, proliferating cell nuclear antigen and Bcl-xl. In conclusion, the findings of the present study suggested that TLR9 may serve an important role in the development of CAC by regulating NF-κB signaling.
Collapse
Affiliation(s)
- Qingtian Luo
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China.,Department of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi 341000, P.R. China
| | - Ling Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chaotao Tang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhendong Zhang
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Youxiang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chunyan Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
16
|
Hallajzadeh J, Amirani E, Mirzaei H, Shafabakhsh R, Mirhashemi SM, Sharifi M, Yousefi B, Mansournia MA, Asemi Z. Circular RNAs: new genetic tools in melanoma. Biomark Med 2020; 14:563-571. [PMID: 32462914 DOI: 10.2217/bmm-2019-0567] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Melanoma is the most lethal form of skin cancer. New technologies have resulted in major advances in the diagnosis and treatment of melanoma and other cancer types. Recently, some studies have investigated the role of circular RNAs (circRNAs) in different cancers. CircRNAs are a member of long noncoding RNA family mainly formed through back-splicing and have a closed-loop structure. These molecules affect several biological and oncogenic cascades in diverse ways via acting as microRNA sponge, interacting with RNA-binding proteins and acting as a transcription regulator. In this review, we made an insight into the impact of circRNA dysregulation in the melanoma tumorigenesis based on the presented evidences.
Collapse
Affiliation(s)
- Jamal Hallajzadeh
- Department of Biochemistry & Nutrition, Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Elaheh Amirani
- Research Center for Biochemistry & Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry & Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Rana Shafabakhsh
- Research Center for Biochemistry & Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyyed M Mirhashemi
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mehran Sharifi
- Department of Hematology & Oncology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahman Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad A Mansournia
- Department of Epidemiology & Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry & Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
17
|
Luan C, Li Y, Liu Z, Zhao C. Long Noncoding RNA MALAT1 Promotes the Development of Colon Cancer by Regulating miR-101-3p/STC1 Axis. Onco Targets Ther 2020; 13:3653-3665. [PMID: 32431516 PMCID: PMC7200234 DOI: 10.2147/ott.s242300] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/08/2020] [Indexed: 12/24/2022] Open
Abstract
Purpose Colon cancer (CC) is a leading cause of cancer-related deaths worldwide. This study aimed to clarify the effect of long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) on CC progression and the potential mechanism. Methods CC cell lines HCT116 and HT29 were selected for functional analysis. The expression of MALAT1, microRNA (miR)-101-3p, and stanniocalcin 1 (STC1) in CC tissues and cells were measured by quantitative reverse transcription PCR (qRT-PCR). Cell proliferation, apoptosis, migration and invasion were measured by Cell Counting Kit-8 (CCK-8), flow cytometry, wound scratch and transwell assay, respectively. The target relationships (MALAT1 and miR-101-3p, miR-101-3p and STC1) were validated by dual-luciferase reporter and RNA pull-down assay. Results The expression of MALAT1 was elevated in CC tissues compared with adjacent normal tissues and was associated with lymph node metastasis, depth of invasion and tumor-node-metastasis (TNM) stage. Up-regulation of MALAT1 promoted the proliferation, migration, and invasion and inhibited the apoptosis of CC cells; while MALAT1 knockdown exhibited opposite results. MiR-101-3p was a target of MALAT1, which was negatively regulated by MALAT1. Silencing of miR-101-3p reverses the anti-tumor effect of MALAT1 knockdown on CC cells. STC1 was a target of miR-101-3p, which was negatively regulated by miR-101-3p. Silencing of STC1 reverses the tumor promoting effects of MALAT1 up-regulation and miR-101-3p down-regulation on CC cells. Conclusion MALAT1 may function as an oncogene in CC progression by affecting the miR-101-3p/STC1 axis, providing a hopeful therapeutic option for CC.
Collapse
Affiliation(s)
- Chunyan Luan
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, People's Republic of China
| | - Yongzhu Li
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, People's Republic of China
| | - Zhigang Liu
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, People's Republic of China
| | - Cunxin Zhao
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, People's Republic of China
| |
Collapse
|
18
|
He J, Yang H, Xu Z, Li J, Chen G, Jiang L, Wu L, Zhou X. A functional polymorphism in the paired basic amino acid-cleaving enzyme 4 gene confers osteoarthritis risk in a population of Eastern China. Genet Mol Biol 2020; 43:e20190115. [PMID: 32167127 PMCID: PMC7197988 DOI: 10.1590/1678-4685-gmb-2019-0115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/19/2019] [Indexed: 01/01/2023] Open
Abstract
Paired basic amino acid-cleaving enzyme 4 (PACE4), a proprotein convertase, is
involved in the activation of aggrecanases (ADAMTS-4 and ADAMTS-5) in
osteoarthritic and cytokine-stimulated cartilage. Activated aggrecanases cause
aggrecan degradation and thus, contribute to osteoarthritis (OA). In this study,
we investigated the association between PACE4 gene
polymorphisms and OA risk. One single-nucleotide polymorphism (rs4965833) in the
PACE4 gene was genotyped in 432 OA patients and 523 healthy
controls using matrix-assisted laser desorption/ionization time-of-flight mass
spectrometry. Quantitative reverse transcription PCR (qRT-PCR) was used to
determine the relative expression of PACE4 in blood samples
from 90 OA patients (30 for each genotype). The relative expression level of
PACE4 mRNA was higher in the GG genotype as compared to the
AA/AG group. Moreover, the PACE4 rs4965833 polymorphism was
associated with increased risk of OA, especially among individuals aged ≥55
years and with a body mass index ≥25. There was no significant association
between the PACE4 rs4965833 polymorphism and clinical
parameters of OA patients, such as erythrocyte sedimentation rate, C-reactive
protein, Visual Analog Scale for pain and Lequesne’s index. In conclusion, the
rs4965833 polymorphism in the 3’-UTR of PACE4 is associated
with OA susceptibility.
Collapse
Affiliation(s)
- Jin He
- Department of Orthopedics, Jintan Hospital Affiliated to Jiangsu University, Changzhou, China
| | - Haoyu Yang
- Department of Orthopedics, Wuxi No.9 People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, China
| | - Zhonghua Xu
- Department of Orthopedics, Jintan Hospital Affiliated to Jiangsu University, Changzhou, China
| | - Jin Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Gang Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lifeng Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lidong Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
19
|
Kyei B, Li L, Yang L, Zhan S, Zhang H. CDR1as/miRNAs-related regulatory mechanisms in muscle development and diseases. Gene 2020; 730:144315. [PMID: 31904497 DOI: 10.1016/j.gene.2019.144315] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
Muscles are critical tissues for mammals due to their close association with movement and physiology. Myogenesis involves proliferation, differentiation, and fusion of myoblast, in which many well-known protein-coding genes, as well as linear non-coding RNAs such as microRNAs (miRNAs), are involved. Recently, circular RNAs (circRNAs) have attracted much attention since several circRNAs are known to play significant roles in muscle development and diseases through limited mechanisms, particularly through sponging miRNAs. Through advanced researches, increasing evidence suggests that Cerebellar Degeneration-Related protein 1 antisense (CDR1as) is an important circRNA that regulates the levels of mRNAs expression via competitively sponged miRNAs. Here, we reviewed the robust expression and base pairing relationships of CDR1as and several myogenic miRNAs, as well as these miRNAs and their targeted genes in muscles or some muscle-related diseases. These potential CDR1as/miRNAs/mRNA pathways will provide the basis for further research on the function of CDR1as in muscle development, and eventually extend the versatile roles of CDR1as in mammals.
Collapse
Affiliation(s)
- Bismark Kyei
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Liu Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Siyuan Zhan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Hongping Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
20
|
Shih JC, Lin HH, Hsiao AC, Su YT, Tsai S, Chien CL, Kung HN. Unveiling the role of microRNA-7 in linking TGF-β-Smad-mediated epithelial-mesenchymal transition with negative regulation of trophoblast invasion. FASEB J 2019; 33:6281-6295. [PMID: 30789794 DOI: 10.1096/fj.201801898rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Several pregnancy complications result from abnormal trophoblast invasion. The dichotomous effect of TGF-β on epithelial-mesenchymal transition (EMT) between trophoblast invasion and cancer progression remains unknown and a critical concern. We attenuated the expression of TGF-β type 1 receptor (coding by TGFBR1) with RNA interference in trophoblastic cells and significantly enhanced the trophoblastic invasion. Analysis of microRNA profiles in trophoblasts indicated microRNA-7 as a key molecule linking TGF-β with the negative regulation of trophoblast invasion. We then attenuated TGFBR1 and miR-7 transcription by transducing either short hairpin RNA targeting TGFBR1 or anti-miR-7-locked nucleonic acid, and we observed an up-regulation of EMT-related transcription factors (TFs) and their downstream effectors, causing a mesenchymal transition of trophoblasts. Conversely, overexpression of TGFBR1 or miR-7 led to the epithelial transition of trophoblasts. Our results showed that TGF-β-induced miR-7 expression negatively modulated the TGF-β-SMAD family member 2-mediated EMT pathway via targeting EMT-related TFs and down-regulating their mesenchymal markers. These findings possibly explain, at least in part, why TGF-β exerts an opposite effect on EMT during trophoblast invasion and cancer progression.-Shih, J.-C., Lin, H.-H., Hsiao, A.-C., Su, Y.-T., Tsai, S., Chien, C.-L., Kung, H.-N. Unveiling the role of microRNA-7 in linking TGF-β-Smad-mediated epithelial-mesenchymal transition with negative regulation of trophoblast invasion.
Collapse
Affiliation(s)
- Jin-Chung Shih
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hua-Heng Lin
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - An-Che Hsiao
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Ting Su
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shawn Tsai
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-Liang Chien
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsiu-Ni Kung
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
21
|
Yin CY, Kong W, Jiang J, Xu H, Zhao W. miR-7-5p inhibits cell migration and invasion in glioblastoma through targeting SATB1. Oncol Lett 2018; 17:1819-1825. [PMID: 30675243 PMCID: PMC6341908 DOI: 10.3892/ol.2018.9777] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 11/16/2018] [Indexed: 02/05/2023] Open
Abstract
MicroRNAs (miRNAs/miRs) have been revealed to influence the development and progression of glioblastoma. Although a number of miRNAs are abnormally expressed in glioblastoma it is not clear whether they are a factor associated with glioblastoma pathogenesis. In the present study, miR-7-5p was identified as being aberrantly downregulated in glioblastoma tissues and cell lines. miR-7-5p overexpression significantly decreased the migratory and invasive capacity of the cells, while miR-7-5p silencing had the opposite effect. In addition, a luciferase assay confirmed that special AT rich sequence binding protein 1 (SATB1) was a direct target gene of miR-7-5p in glioblastoma. The overexpression of SATB1 in glioblastoma was revealed to promote cell migration and invasion. In addition, SATB1 overexpression may weaken the inhibitory effect of miR-7-5p on cell migration and invasion. miR-7-5p overexpression reversed the effects of SATB1 on cell migration and invasion in glioblastoma cells. In conclusion, miR-7-5p may be a useful therapeutic target for the diagnosis and treatment of patients with glioblastoma.
Collapse
Affiliation(s)
- Chang-You Yin
- Department of Neurosurgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Wei Kong
- Department of Neurosurgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Jing Jiang
- Department of Emergency, Yantaishan Hospital, Yantai, Shandong 264001, P.R. China
| | - Hao Xu
- Department of Neurosurgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Wei Zhao
- Department of Neurosurgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
22
|
Neve B, Jonckheere N, Vincent A, Van Seuningen I. Epigenetic Regulation by lncRNAs: An Overview Focused on UCA1 in Colorectal Cancer. Cancers (Basel) 2018; 10:E440. [PMID: 30441811 PMCID: PMC6266399 DOI: 10.3390/cancers10110440] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/06/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancers have become the second leading cause of cancer-related deaths. In particular, acquired chemoresistance and metastatic lesions occurring in colorectal cancer are a major challenge for chemotherapy treatment. Accumulating evidence shows that long non-coding (lncRNAs) are involved in the initiation, progression, and metastasis of cancer. We here discuss the epigenetic mechanisms through which lncRNAs regulate gene expression in cancer cells. In the second part of this review, we focus on the role of lncRNA Urothelial Cancer Associated 1 (UCA1) to integrate research in different types of cancer in order to decipher its putative function and mechanism of regulation in colorectal cancer cells. UCA1 is highly expressed in cancer cells and mediates transcriptional regulation on an epigenetic level through the interaction with chromatin modifiers, by direct regulation via chromatin looping and/or by sponging the action of a diversity of miRNAs. Furthermore, we discuss the role of UCA1 in the regulation of cell cycle progression and its relation to chemoresistance in colorectal cancer cells.
Collapse
Affiliation(s)
- Bernadette Neve
- Inserm UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer (JPArc), Team "Mucins, Epithelial Differentiation and Carcinogenesis"; University Lille; CHU Lille,59045, Lille CEDEX, France.
| | - Nicolas Jonckheere
- Inserm UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer (JPArc), Team "Mucins, Epithelial Differentiation and Carcinogenesis"; University Lille; CHU Lille,59045, Lille CEDEX, France.
| | - Audrey Vincent
- Inserm UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer (JPArc), Team "Mucins, Epithelial Differentiation and Carcinogenesis"; University Lille; CHU Lille,59045, Lille CEDEX, France.
| | - Isabelle Van Seuningen
- Inserm UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer (JPArc), Team "Mucins, Epithelial Differentiation and Carcinogenesis"; University Lille; CHU Lille,59045, Lille CEDEX, France.
| |
Collapse
|
23
|
He H, Dai J, Zhuo R, Zhao J, Wang H, Sun F, Zhu Y, Xu D. Study on the mechanism behind lncRNA MEG3 affecting clear cell renal cell carcinoma by regulating miR-7/RASL11B signaling. J Cell Physiol 2018; 233:9503-9515. [PMID: 29968912 DOI: 10.1002/jcp.26849] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/10/2018] [Indexed: 12/17/2022]
Abstract
The goal of this research was to study the relationships between maternally expressed gene 3 (MEG3), microRNA-7 (miR-7), and RASL11B, and explore their influence on the progression of clear cell renal cell carcinoma (CCRCC). Microarray analysis was conducted using the data provided by The Cancer Genome Atlas. The expression levels of MEG3 and miR-7 in CCRCC and adjacent tissue samples were ascertained by quantitative real-time polymerase chain reaction (qRT-PCR). The cell proliferation activity was unmasked by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and cell apoptosis and cell cycle were investigated by flow cytometry. A dual luciferase reporter assay was used to verify target relationships. Wound healing assay and transwell assay were used to detect cell migration and invasion ability. Decreased MEG3 expression was observed in CCRCC tissues and cells. Overexpression of MEG3 accelerated apoptosis; inhibited cell proliferation, migration and invasion; and induced G0/G1 phase cell cycle arrest in CCRCC. MiR-7, directly binding to MEG3, was overexpressed in the CCRCC tissues and could inhibit the apoptosis and promote the migration and invasion of CCRCC cells. RASL11B, lowly expressed in CCRCC, was a target of miR-7. After the overexpression of RASL11B, G0/G1 phase cell cycle arrest was induced; cell apoptosis was promoted; and the proliferation, invasion, and migration of CCRCC cells were inhibited. MEG3 could up-regulate RASL11B to inhibit the cell proliferation, invasion, and migration; induce G0/G1 cell cycle arrest; and promote cell apoptosis by suppressing miR-7 in CCRCC.
Collapse
Affiliation(s)
- Hongchao He
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jun Dai
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ran Zhuo
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Juping Zhao
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haofei Wang
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fukang Sun
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Zhu
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Danfeng Xu
- Department of Urology, Shanghai Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Naser R, Aldehaiman A, Díaz-Galicia E, Arold ST. Endogenous Control Mechanisms of FAK and PYK2 and Their Relevance to Cancer Development. Cancers (Basel) 2018; 10:E196. [PMID: 29891810 PMCID: PMC6025627 DOI: 10.3390/cancers10060196] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/31/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023] Open
Abstract
Focal adhesion kinase (FAK) and its close paralogue, proline-rich tyrosine kinase 2 (PYK2), are key regulators of aggressive spreading and metastasis of cancer cells. While targeted small-molecule inhibitors of FAK and PYK2 have been found to have promising antitumor activity, their clinical long-term efficacy may be undermined by the strong capacity of cancer cells to evade anti-kinase drugs. In healthy cells, the expression and/or function of FAK and PYK2 is tightly controlled via modulation of gene expression, competing alternatively spliced forms, non-coding RNAs, and proteins that directly or indirectly affect kinase activation or protein stability. The molecular factors involved in this control are frequently deregulated in cancer cells. Here, we review the endogenous mechanisms controlling FAK and PYK2, and with particular focus on how these mechanisms could inspire or improve anticancer therapies.
Collapse
Affiliation(s)
- Rayan Naser
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Abdullah Aldehaiman
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Escarlet Díaz-Galicia
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| | - Stefan T Arold
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia.
| |
Collapse
|
25
|
Chen C, Yue D, Lei L, Wang H, Lu J, Zhou Y, Liu S, Ding T, Guo M, Xu L. Promoter-Operating Targeted Expression of Gene Therapy in Cancer: Current Stage and Prospect. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:508-514. [PMID: 29858085 PMCID: PMC5992480 DOI: 10.1016/j.omtn.2018.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/24/2018] [Accepted: 04/09/2018] [Indexed: 02/07/2023]
Abstract
The technique of targeted expression of interesting genes, including distinct delivery systems and specific gene promoter-operating expression, is an important strategy for gene therapy against cancers. Up to now, extensive literature documented the efficacy of distinct delivery systems, such as the liposome system, nano-particle system, polyetherimide (PEI) system, and so on, in cancer gene therapy. However, a related document on the potential value of using a specific gene promoter, such as a tumor suppressor, in cancer gene therapy was still scary. The main obstacle might be that the selection of an ideal gene promoter to operate interesting gene expression in cancer gene therapy is still not fully understood. Therefore, many efforts need to be done in order to make it a real power tool for the human clinical treatment of cancer patients. The purpose of this review is to clarify the current state and some problematics in development of promoter-operating targeted expression of interesting genes and highlight its potential in cancer gene therapy.
Collapse
Affiliation(s)
- Chao Chen
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Dongxu Yue
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Liangyu Lei
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Hairong Wang
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Jia Lu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Ya Zhou
- Department of Medical Physics, Zunyi Medical University, Guizhou 563000, China
| | - Shiming Liu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Tao Ding
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Lin Xu
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China.
| |
Collapse
|
26
|
Liu X, He B, Xu T, Pan Y, Hu X, Chen X, Wang S. MiR-490-3p Functions As a Tumor Suppressor by Inhibiting Oncogene VDAC1 Expression in Colorectal Cancer. J Cancer 2018; 9:1218-1230. [PMID: 29675103 PMCID: PMC5907670 DOI: 10.7150/jca.23662] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/28/2018] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide, usually with poor prognosis because many CRC patients are diagnosed at an advanced stage. Therefore, novel potential diagnostic and prognostic biomarkers are urgently needed. MicroRNAs have been reported to regulate a variety of biological processes, such as cell proliferation, differentiation and apoptosis. Accumulating studies have demonstrated that miR-490-3p could regulate the development and progression of multiple cancers, but its clinical significance and molecular mechanism in CRC are still elusive. Here, we try to further elucidate the regulatory mechanism of miR-490-3p in CRC. In the present study, miR-490-3p expression level observably down-regulated in CRC tissues and cell lines, and miR-490-3p expression in CRC tissues was significantly associated with TNM stage, histological grade, tumor size and overall survival (OS). In addition, we observed miR-490-3p expression was also decreased in CRC plasmas and could act as a promising diagnostic biomarker for screening CRC. Further studies in vitro demonstrated Voltage Dependent Anion Channel 1 (VDAC1) which highly expressed in CRC tissues and cell lines is a direct target of miR-490-3p, and miR-490-3p could markedly inhibit CRC cells proliferation, metastasis, invasion and anti-apoptosis through suppressing VDAC1/AMPK/mTOR pathway. These results indicated that miR-490-3p functions as a tumor suppressor in CRC, and may be a novel potential diagnostic and prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Xiangxiang Liu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Bangshun He
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Yuqin Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Xiuxiu Hu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Xiaoxiang Chen
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Shukui Wang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210000, China
| |
Collapse
|
27
|
Feng S, Wang Y, Zhang R, Yang G, Liang Z, Wang Z, Zhang G. Curcumin exerts its antitumor activity through regulation of miR-7/Skp2/p21 in nasopharyngeal carcinoma cells. Onco Targets Ther 2017; 10:2377-2388. [PMID: 28496336 PMCID: PMC5422505 DOI: 10.2147/ott.s130055] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Curcumin, a natural polyphenol compound, exhibits tumor suppressive activity in a wide spectrum of cancers, including nasopharyngeal carcinoma cells. However, the exact molecular mechanisms governing this tumor suppressive activity remain elusive. Multiple studies have revealed that miRNAs are critically involved in tumorigenesis, indicating that targeting miRNAs could be a therapeutic strategy for treating human cancer. In the current study, we set out to determine whether curcumin regulates miR-7 expression in nasopharyngeal carcinoma cells. We found that curcumin inhibited cell growth, induced apoptosis, retarded cell migration and invasion, and triggered cell cycle arrest in the human nasopharyngeal carcinoma cell lines CNE1 and CNE2. Importantly, we observed that curcumin upregulated the expression of miR-7 and subsequently inhibited Skp2, a direct miR-7 target. Our results identified that upregulation of miR-7 by curcumin could benefit nasopharyngeal carcinoma patients.
Collapse
Affiliation(s)
- Shaoyan Feng
- Department of Otorhinolaryngology, Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou.,Department of Otolaryngology, Head and Neck Surgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai
| | - Yu Wang
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou
| | | | - Guangwei Yang
- Department of Radiation Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, People's Republic of China
| | - Zibin Liang
- Department of Radiation Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, People's Republic of China
| | - Zhiwei Wang
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou
| | - Gehua Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou
| |
Collapse
|
28
|
Lei L, Chen C, Zhao J, Wang H, Guo M, Zhou Y, Luo J, Zhang J, Xu L. Targeted Expression of miR-7 Operated by TTF-1 Promoter Inhibited the Growth of Human Lung Cancer through the NDUFA4 Pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 6:183-197. [PMID: 28325285 PMCID: PMC5363496 DOI: 10.1016/j.omtn.2016.12.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/04/2016] [Accepted: 12/05/2016] [Indexed: 01/05/2023]
Abstract
Targeted expression of gene technique is an important therapeutic strategy for lung cancer. MicroRNA-7 has been well documented as a promising tumor suppressor but never been test in specific gene-promoter-targeted expression in cancer gene therapy. Here, we first evaluated the efficacy of miR-7 expression operated by the promoter of TTF-1, a lineage-specific oncogene in lung cancer, in vitro using an eukaryotic vector of TTF-1-promoter-operated expression of miR-7 (termed as p-T-miR-7). Interestingly, using a nude mice model, the growth and metastasis of human lung cancer cells in vivo were significantly reduced in remote hypodermic injection of the p-T-miR-7 group, accompanied by increased expression of miR-7 and reduced transduction of the Akt and Erk pathway in situ. Mechanism aspect, global gene expression analysis showed that downregulation of NDUFA4, a novel target of miR-7, contributed to the effects of miR-7 expression operated by TTF-1 promoter on the growth and metastasis of human lung cancer cells, as well as altered transduction of the Akt and Erk pathway. Finally, there was no significant difference in weight or histopathology of other organs. These data provided a basis for development of novel modality of miRNA-based targeted expression therapy against clinical lung cancer.
Collapse
Affiliation(s)
- Liangyu Lei
- Department of Immunology, Zunyi Medical College, Guizhou 563000, China
| | - Chao Chen
- Department of Immunology, Zunyi Medical College, Guizhou 563000, China
| | - Juanjuan Zhao
- Department of Immunology, Zunyi Medical College, Guizhou 563000, China
| | - HaiRong Wang
- Department of Immunology, Zunyi Medical College, Guizhou 563000, China
| | - Mengmeng Guo
- Department of Immunology, Zunyi Medical College, Guizhou 563000, China
| | - Ya Zhou
- Department of Medical Physics, Zunyi Medical College, Guizhou 563000, China
| | - Junming Luo
- Department of Immunology, Zunyi Medical College, Guizhou 563000, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical College, Guizhou 563000, China
| | - Lin Xu
- Department of Immunology, Zunyi Medical College, Guizhou 563000, China.
| |
Collapse
|
29
|
MicroRNA Regulation of Endothelial Junction Proteins and Clinical Consequence. Mediators Inflamm 2016; 2016:5078627. [PMID: 27999452 PMCID: PMC5143735 DOI: 10.1155/2016/5078627] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/03/2016] [Indexed: 12/18/2022] Open
Abstract
Cellular junctions play a critical role in structural connection and signal communication between cells in various tissues. Although there are structural and functional varieties, cellular junctions include tight junctions, adherens junctions, focal adhesion junctions, and tissue specific junctions such as PECAM-1 junctions in endothelial cells (EC), desmosomes in epithelial cells, and hemidesmosomes in EC. Cellular junction dysfunction and deterioration are indicative of clinical diseases. MicroRNAs (miRNA) are ~20 nucleotide, noncoding RNAs that play an important role in posttranscriptional regulation for almost all genes. Unsurprisingly, miRNAs regulate junction protein gene expression and control junction structure integrity. In contrast, abnormal miRNA regulation of junction protein gene expression results in abnormal junction structure, causing related diseases. The major components of tight junctions include zonula occluden-1 (ZO-1), claudin-1, claudin-5, and occludin. The miRNA regulation of ZO-1 has been intensively investigated. ZO-1 and other tight junction proteins such as claudin-5 and occludin were positively regulated by miR-126, miR-107, and miR21 in different models. In contrast, ZO-1, claudin-5, and occludin were negatively regulated by miR-181a, miR-98, and miR150. Abnormal tight junction miRNA regulation accompanies cerebral middle artery ischemia, brain trauma, glioma metastasis, and so forth. The major components of adherens junctions include VE-cadherin, β-catenin, plakoglobin, P120, and vinculin. VE-cadherin and β-catenin were regulated by miR-9, miR-99b, miR-181a, and so forth. These regulations directly affect VE-cadherin-β-catenin complex stability and further affect embryo and tumor angiogenesis, vascular development, and so forth. miR-155 and miR-126 have been shown to regulate PECAM-1 and affect neutrophil rolling and EC junction integrity. In focal adhesion junctions, the major components are integrin β4, paxillin, and focal adhesion kinase (FAK). Integrin β4 has been regulated by miR-184, miR-205, and miR-9. Paxillin has been regulated by miR-137, miR-145, and miR-218 in different models. FAK has been regulated by miR-7, miR-138, and miR-135. Deregulation of miRNAs is caused by viral infections, tumorigenesis, and so forth. By regulation of posttranscription, miRNAs manipulate junction protein expression in all cellular processes and further determine cellular fate and development. Elucidation of these regulatory mechanisms will become a new alternative therapy for many diseases, such as cancers and inflammatory diseases.
Collapse
|