1
|
Xu D, Wang W, Wang D, Ding J, Zhou Y, Zhang W. Long noncoding RNA MALAT-1: A versatile regulator in cancer progression, metastasis, immunity, and therapeutic resistance. Noncoding RNA Res 2024; 9:388-406. [PMID: 38511067 PMCID: PMC10950606 DOI: 10.1016/j.ncrna.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 03/22/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) are RNA transcripts longer than 200 nucleotides that do not code for proteins but have been linked to cancer development and metastasis. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT-1) influences crucial cancer hallmarks through intricate molecular mechanisms, including proliferation, invasion, angiogenesis, apoptosis, and the epithelial-mesenchymal transition (EMT). The current article highlights the involvement of MALAT-1 in drug resistance, making it a potential target to overcome chemotherapy refractoriness. It discusses the impact of MALAT-1 on immunomodulatory molecules, such as major histocompatibility complex (MHC) proteins and PD-L1, leading to immune evasion and hindering anti-tumor immune responses. MALAT-1 also plays a significant role in cancer immunology by regulating diverse immune cell populations. In summary, MALAT-1 is a versatile cancer regulator, influencing tumorigenesis, chemoresistance, and immunotherapy responses. Understanding its precise molecular mechanisms is crucial for developing targeted therapies, and therapeutic strategies targeting MALAT-1 show promise for improving cancer treatment outcomes. However, further research is needed to fully uncover the role of MALAT-1 in cancer biology and translate these findings into clinical applications.
Collapse
Affiliation(s)
- Dexin Xu
- Department of Orthopedics, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Wenhai Wang
- Department of Cardiology, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Duo Wang
- Department of Geriatrics, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Jian Ding
- Department of Electrodiagnosis, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Yunan Zhou
- Department of Orthopedics, Jilin Province FAW General Hospital, Changchun, 130000, China
| | - Wenbin Zhang
- Department of Cardiology, Jilin Province FAW General Hospital, Changchun, 130000, China
| |
Collapse
|
2
|
Adu-Gyamfi EA, Cheeran EA, Salamah J, Lee BK. Mechanistic actions of long non-coding RNA MALAT1 within the ovary and at the feto-maternal interface. Mol Biol Rep 2024; 51:301. [PMID: 38353828 DOI: 10.1007/s11033-024-09220-2] [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: 12/05/2023] [Accepted: 01/06/2024] [Indexed: 02/16/2024]
Abstract
Long non-coding RNAs (LncRNAs) are being unveiled as crucial regulators of several biological processes and pathways. Among the lncRNAs is metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), which is also known as nuclear enriched abundant transcript 2 (NEAT2). MALAT1 is highly conserved in mammals, and controls cellular processes such as proliferation, migration, invasion, angiogenesis, and apoptosis in both physiological and pathological conditions. Roles of MALAT1 in the female reproductive system are gradually getting explored. Within the ovarian micro-environment, the physiological expression of MALAT1 potentially modulates folliculogenesis while its upregulation promotes the metastasis of epithelial ovarian cancers. Interestingly, women with polycystic ovary syndrome have been shown to exhibit aberrant ovarian expression of MALAT1 and this is believed to contribute to the development of the disease. At the feto-maternal interface, MALAT1 potentially promotes trophoblast development. While its placental downregulation is linked to the pathogenesis of preeclampsia, its placental upregulation is associated with placenta increta and placenta percreta. Hence, abnormal expression of MALAT1 is a candidate molecular biomarker and therapeutic target for the treatment of these obstetric and gynecologic anomalies. To enhance a quick uncovering and detailed characterization of the mechanistic actions of MALAT1 in the female reproductive system, we have highlighted some knowledge deficits and have recommended ideal experimental models to be employed in prospective investigations.
Collapse
Affiliation(s)
- Enoch Appiah Adu-Gyamfi
- Department of Biomedical Sciences, University at Albany - State University of New York, Rensselaer, NY, 12144, USA.
- Cancer Research Center, University at Albany - State University of New York, Rensselaer, NY, 12144, USA.
| | - Elisha Ann Cheeran
- Department of Biomedical Sciences, University at Albany - State University of New York, Rensselaer, NY, 12144, USA
- Cancer Research Center, University at Albany - State University of New York, Rensselaer, NY, 12144, USA
| | - Joudi Salamah
- Department of Biomedical Sciences, University at Albany - State University of New York, Rensselaer, NY, 12144, USA
- Cancer Research Center, University at Albany - State University of New York, Rensselaer, NY, 12144, USA
| | - Bum-Kyu Lee
- Department of Biomedical Sciences, University at Albany - State University of New York, Rensselaer, NY, 12144, USA.
- Cancer Research Center, University at Albany - State University of New York, Rensselaer, NY, 12144, USA.
| |
Collapse
|
3
|
Malgundkar SH, Tamimi Y. The pivotal role of long non-coding RNAs as potential biomarkers and modulators of chemoresistance in ovarian cancer (OC). Hum Genet 2024; 143:107-124. [PMID: 38276976 DOI: 10.1007/s00439-023-02635-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024]
Abstract
Ovarian cancer (OC) is a fatal gynecological disease that is often diagnosed at later stages due to its asymptomatic nature and the absence of efficient early-stage biomarkers. Previous studies have identified genes with abnormal expression in OC that couldn't be explained by methylation or mutation, indicating alternative mechanisms of gene regulation. Recent advances in human transcriptome studies have led to research on non-coding RNAs (ncRNAs) as regulators of cancer gene expression. Long non-coding RNAs (lncRNAs), a class of ncRNAs with a length greater than 200 nucleotides, have been identified as crucial regulators of physiological processes and human diseases, including cancer. Dysregulated lncRNA expression has also been found to play a crucial role in ovarian carcinogenesis, indicating their potential as novel and non-invasive biomarkers for improving OC management. However, despite the discovery of several thousand lncRNAs, only one has been approved for clinical use as a biomarker in cancer, highlighting the importance of further research in this field. In addition to their potential as biomarkers, lncRNAs have been implicated in modulating chemoresistance, a major problem in OC. Several studies have identified altered lncRNA expression upon drug treatment, further emphasizing their potential to modulate chemoresistance. In this review, we highlight the characteristics of lncRNAs, their function, and their potential to serve as tumor markers in OC. We also discuss a few databases providing detailed information on lncRNAs in various cancer types. Despite the promising potential of lncRNAs, further research is necessary to fully understand their role in cancer and develop effective strategies to combat this devastating disease.
Collapse
Affiliation(s)
- Shika Hanif Malgundkar
- Biochemistry Department, College of Medicine and Health Sciences, Sultan Qaboos University, PC 123, PO Box 35, Muscat, Sultanate of Oman
| | - Yahya Tamimi
- Biochemistry Department, College of Medicine and Health Sciences, Sultan Qaboos University, PC 123, PO Box 35, Muscat, Sultanate of Oman.
| |
Collapse
|
4
|
Marques IS, Tavares V, Savva-Bordalo J, Rei M, Liz-Pimenta J, de Melo IG, Assis J, Pereira D, Medeiros R. Long Non-Coding RNAs: Bridging Cancer-Associated Thrombosis and Clinical Outcome of Ovarian Cancer Patients. Int J Mol Sci 2023; 25:140. [PMID: 38203310 PMCID: PMC10778953 DOI: 10.3390/ijms25010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Ovarian cancer (OC) and venous thromboembolism (VTE) have a close relationship, in which tumour cells surpass the haemostatic system to drive cancer progression. Long non-coding RNAs (lncRNAs) have been implicated in VTE pathogenesis, yet their roles in cancer-associated thrombosis (CAT) and their prognostic value are unexplored. Understanding how these lncRNAs influence venous thrombogenesis and ovarian tumorigenesis may lead to the identification of valuable biomarkers for VTE and OC management. Thus, this study evaluated the impact of five lncRNAs, namely MALAT1, TUG1, NEAT1, XIST and MEG8, on a cohort of 40 OC patients. Patients who developed VTE after OC diagnosis had worse overall survival compared to their counterparts (log-rank test, p = 0.028). Elevated pre-chemotherapy MEG8 levels in peripheral blood cells (PBCs) predicted VTE after OC diagnosis (Mann-Whitney U test, p = 0.037; Χ2 test, p = 0.033). In opposition, its low levels were linked to a higher risk of OC progression (adjusted hazard ratio (aHR) = 3.00; p = 0.039). Furthermore, low pre-chemotherapy NEAT1 levels in PBCs were associated with a higher risk of death (aHR = 6.25; p = 0.008). As for the remaining lncRNAs, no significant association with VTE incidence, OC progression or related mortality was observed. Future investigation with external validation in larger cohorts is needed to dissect the implications of the evaluated lncRNAs in OC patients.
Collapse
Affiliation(s)
- Inês Soares Marques
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (I.G.d.M.)
- Faculty of Sciences of the University of Porto (FCUP), 4169-007 Porto, Portugal
| | - Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (I.G.d.M.)
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Joana Savva-Bordalo
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Mariana Rei
- Department of Gynaecology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal;
| | - Joana Liz-Pimenta
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
- Department of Medical Oncology, Centro Hospitalar de Trás-os-Montes e Alto Douro (CHTMAD), 5000-508 Vila Real, Portugal
| | - Inês Guerra de Melo
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (I.G.d.M.)
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
| | - Joana Assis
- Clinical Research Unit, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal;
| | - Deolinda Pereira
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal; (I.S.M.); (V.T.); (I.G.d.M.)
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
- Abel Salazar Institute for the Biomedical Sciences (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
5
|
Mazarei M, Shahabi Rabori V, Ghasemi N, Salehi M, Rayatpisheh N, Jahangiri N, Saberiyan M. LncRNA MALAT1 signaling pathway and clinical applications in overcome on cancers metastasis. Clin Exp Med 2023; 23:4457-4472. [PMID: 37695391 DOI: 10.1007/s10238-023-01179-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/27/2023] [Indexed: 09/12/2023]
Abstract
In spite of its high mortality rate and difficulty in finding a cure, scientific advancements have contributed to a reduction in cancer-related fatalities. Aberrant gene expression during carcinogenesis emphasizes the importance of targeting the signaling networks that control gene expression in cancer treatment. Long noncoding RNAs (lncRNAs), which are transcribed RNA molecules that play a role in gene expression regulation, are a recent innovative therapeutic approach for diagnosing and treating malignancies. MALAT1, a well-known lncRNA, functions in gene expression, RNA processing, and epigenetic control. High expression levels of MALAT1 are associated with several human disorders, including metastasis, invasion, autophagy, and proliferation of cancer cells. MALAT1 affects various signaling pathways and microRNAs (miRNAs), and this study aims to outline its functional roles in cancer metastasis and its interactions with cellular signaling pathways. Moreover, MALAT1 and its interactions with signaling pathways can be promising target for cancer treatment.
Collapse
Affiliation(s)
- Madineh Mazarei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Nazila Ghasemi
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | - Mehrnaz Salehi
- School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Najmeh Rayatpisheh
- School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Negin Jahangiri
- Department of Biology, Faculty of Basic Sciences and Engineering, Gonbad Kavous University, Gonbad-e Kavus, Iran
| | - Mohammadreza Saberiyan
- Department of Medical Genetics, Faculty of Medicine, School of Medical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
6
|
Sharma T, Nisar S, Masoodi T, Macha MA, Uddin S, Akil AAS, Pandita TK, Singh M, Bhat AA. Current and emerging biomarkers in ovarian cancer diagnosis; CA125 and beyond. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:85-114. [PMID: 36707207 DOI: 10.1016/bs.apcsb.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Ovarian cancer (OC) is one of the most common causes of cancer-related death in women worldwide. Its five-year survival rates are worse than the two most common gynecological cancers, cervical and endometrial. This is because it is asymptomatic in the early stages and usually detected in the advanced metastasized stage. Thus, survival is increasingly dependent on timely diagnosis. The delay in detection is contributed partly by the occurrence of non-specific clinical symptoms in the early stages and the lack of effective biomarkers and detection approaches. This underlines the need for biomarker identification and clinical validation, enabling earlier diagnosis, effective prognosis, and response to therapy. Apart from the traditional diagnostic biomarkers for OC, several new biomarkers have been delineated using advanced high-throughput molecular approaches in recent years. They are currently being clinically evaluated for their true diagnostic potential. In this chapter, we document the commonly utilized traditional screening markers and recently identified emerging biomarkers in OC diagnosis, focusing on secretory and protein biomarkers. We also briefly reviewed the recent advances and prospects in OC diagnosis.
Collapse
Affiliation(s)
- Tarang Sharma
- Department of Medical Oncology, Dr. B.R Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sabah Nisar
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Tariq Masoodi
- Laboratory of Cancer immunology and genetics, Sidra Medicine, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Jammu and Kashmir, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Ammira Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, United States
| | - Mayank Singh
- Department of Medical Oncology, Dr. B.R Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
7
|
Functional roles of long noncoding RNA MALAT1 in gynecologic cancers. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:48-65. [PMID: 36042115 DOI: 10.1007/s12094-022-02914-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
Abstract
Gynecologic cancers are reproductive disorders characterized by pelvic pain and infertility. The identification of new predictive markers and therapeutic targets for the treatment of gynecologic cancers is urgently necessary. One of the recent successes in gynecologic cancers research is identifying the role of signaling pathways in the pathogenesis of the disease. Recent experiments showed long noncoding RNAs (lncRNA) can be novel therapeutic approaches for the diagnosis and treatment of gynecologic cancers. LncRNA are transcribed RNA molecules that play pivotal roles in multiple biological processes by regulating the different steps of gene expression. Metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) is a well-known lncRNA that plays functional roles in gene expression, RNA processing, and epigenetic regulation. High expression of MALAT1 is closely related to numerous human diseases. It is generally believed that MALAT1 expression is associated with cancer cell growth, autophagy, invasion, and metastasis. MALAT1 by targeting multiple signaling pathways and microRNAs (miRNAs) could contribute to the pathogenesis of gynecologic cancers. In this review, we will summarize functional roles of MALAT1 in the most common gynecologic cancers, including endometrium, breast, ovary, and cervix.
Collapse
|
8
|
Keyvani V, Mahmoudian RA, Mollazadeh S, Kheradmand N, Ghorbani E, Khazaei M, Saeed Al-Hayawi I, Hassanian SM, Ferns GA, Avan A, Anvari K. Insight into RNA-based Therapies for Ovarian Cancer. Curr Pharm Des 2023; 29:2692-2701. [PMID: 37916491 DOI: 10.2174/0113816128270476231023052228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/14/2023] [Indexed: 11/03/2023]
Abstract
Ovarian cancer (OC) is one of the most common malignancies in women and is associated with poor outcomes. The treatment for OC is often associated with resistance to therapies and hence this has stimulated the search for alternative therapeutic approaches, including RNA-based therapeutics. However, this approach has some challenges that include RNA degradation. To solve this critical issue, some novel delivery systems have been proposed. In current years, there has been growing interest in the improvement of RNAbased therapeutics as a promising approach to target ovarian cancer and improve patient outcomes. This paper provides a practical insight into the use of RNA-based therapeutics in ovarian cancers, highlighting their potential benefits, challenges, and current research progress. RNA-based therapeutics offer a novel and targeted approach to treat ovarian cancer by exploiting the unique characteristics of RNA molecules. By targeting key oncogenes or genes responsible for drug resistance, siRNAs can effectively inhibit tumor growth and sensitize cancer cells to conventional therapies. Furthermore, messenger RNA (mRNA) vaccines have emerged as a revolutionary tool in cancer immunotherapy. MRNA vaccines can be designed to encode tumor-specific antigens, stimulating the immune system to distinguish and eliminate ovarian cancer cells. A nano-based delivery platform improves the release of loaded RNAs to the target location and reduces the off-target effects. Additionally, off-target effects and immune responses triggered by RNA molecules necessitate careful design and optimization of these therapeutics. Several preclinical and clinical researches have shown promising results in the field of RNA-based therapeutics for ovarian cancer. In a preclinical study, siRNA-mediated silencing of the poly (ADP-ribose) polymerase 1 (PARP1) gene, involved in DNA repair, sensitized ovarian cancer cells to PARP inhibitors, leading to enhanced therapeutic efficacy. In clinical trials, mRNA-based vaccines targeting tumor-associated antigens have demonstrated safety and efficacy in stimulating immune responses in ovarian cancer patients. In aggregate, RNA-based therapeutics represent a promising avenue for the therapy of ovarian cancers. The ability to specifically target oncogenes or stimulate immune responses against tumor cells holds great potential for improving patient outcomes. However, further research is needed to address challenges related to delivery, permanence, and off-target effects. Clinical trials assessing the care and effectiveness of RNAbased therapeutics in larger patient cohorts are warranted. With continued advancements in the field, RNAbased therapeutics have the potential to develop the management of ovarian cancer and provide new hope for patients.
Collapse
Affiliation(s)
- Vahideh Keyvani
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reihaneh Alsadat Mahmoudian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Nahid Kheradmand
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Seyed Mahdi Hassanian
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane 4059, Australia
| | - Kazem Anvari
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Rakheja I, Ansari AH, Ray A, Chandra Joshi D, Maiti S. Small molecule quercetin binds MALAT1 triplex and modulates its cellular function. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:241-256. [PMID: 36284512 PMCID: PMC9576543 DOI: 10.1016/j.omtn.2022.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 09/20/2022] [Indexed: 11/25/2022]
Abstract
The triple-helix structure at the 3' end of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long non-coding RNA, has been considered to be a target for modulating the oncogenic functions of MALAT1. This study examines the binding of quercetin-a known triplex binding molecule-to the MALAT1 triplex. By employing UV-visible spectroscopy, circular dichroism spectroscopy, and isothermal titration calorimetry, we observed that quercetin binds to the MALAT1 triplex with a stoichiometry of 1:1 and K d of 495 ± 61 nM, along with a negative change in free energy, indicating a spontaneous interaction. Employing real-time PCR measurements, we observed around 50% downregulation of MALAT1 transcript levels in MCF7 cells, and fluorescence in situ hybridization (FISH) experiments showed concomitantly reduced levels of MALAT1 in nuclear speckles. This interaction is likely a result of a direct interaction between the molecule and the RNA, as indicated by a transcription-stop experiment. Further, transcriptome-wide analysis of alternative splicing changes induced by quercetin revealed modulation of MALAT1 downstream genes. Collectively, our study shows that quercetin strongly binds to the MALAT1 triplex and modulates its functions. It can thus be used as a scaffold for further development of therapeutics or as a chemical tool to understand MALAT1 functions.
Collapse
Affiliation(s)
- Isha Rakheja
- Chemical and Systems Biology Unit, Council of Scientific and Industrial Research-Institute of Genomics & Integrative Biology, New Delhi 110025, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-Human Resource Development Centre, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201002, India
| | - Asgar Hussain Ansari
- Chemical and Systems Biology Unit, Council of Scientific and Industrial Research-Institute of Genomics & Integrative Biology, New Delhi 110025, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-Human Resource Development Centre, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201002, India
| | - Arjun Ray
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-Delhi), Okhla Industrial Estate, Phase III, New Delhi 110020, India
| | - Dheeraj Chandra Joshi
- Chemical and Systems Biology Unit, Council of Scientific and Industrial Research-Institute of Genomics & Integrative Biology, New Delhi 110025, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-Human Resource Development Centre, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201002, India
| | - Souvik Maiti
- Chemical and Systems Biology Unit, Council of Scientific and Industrial Research-Institute of Genomics & Integrative Biology, New Delhi 110025, India
- Institute of Genomics and Integrative Biology (IGIB)-National Chemical Laboratory (NCL) Joint Center, Council of Scientific and Industrial Research-NCL, Pune 411008, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-Human Resource Development Centre, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
10
|
Malakoti F, Targhazeh N, Karimzadeh H, Mohammadi E, Asadi M, Asemi Z, Alemi F. The Multiple Function of lncRNA MALAT1 in Cancer Occurrence and Progression. Chem Biol Drug Des 2021; 101:1113-1137. [PMID: 34918470 DOI: 10.1111/cbdd.14006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 11/28/2022]
Abstract
Long non-coding RNAs (lncRNAs) have received particular attention in the last decade due to its engaging in carcinogenesis and tumorigenesis. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a lncRNA that plays physiological and pathological roles in many aspects of genome function as well as biological processes involved in cell development, differentiation, proliferation, invasion, and migration. In this article, we will review the effects of lncRNA MALAT1 on the progression of six prevalent human cancers by focusing on MALAT1 ability to regulate post-transcriptional modification and signaling pathways.
Collapse
Affiliation(s)
- Faezeh Malakoti
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student's Research committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Niloufar Targhazeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haniye Karimzadeh
- Department of Clinical Biochemistry, School of Pharmacy & Isfahan Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Erfan Mohammadi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.,Drugs Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Drugs Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Cuesta-Mateos C, Terrón F, Herling M. CCR7 in Blood Cancers - Review of Its Pathophysiological Roles and the Potential as a Therapeutic Target. Front Oncol 2021; 11:736758. [PMID: 34778050 PMCID: PMC8589249 DOI: 10.3389/fonc.2021.736758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
According to the classical paradigm, CCR7 is a homing chemokine receptor that grants normal lymphocytes access to secondary lymphoid tissues such as lymph nodes or spleen. As such, in most lymphoproliferative disorders, CCR7 expression correlates with nodal or spleen involvement. Nonetheless, recent evidence suggests that CCR7 is more than a facilitator of lymphatic spread of tumor cells. Here, we review published data to catalogue CCR7 expression across blood cancers and appraise which classical and novel roles are attributed to this receptor in the pathogenesis of specific hematologic neoplasms. We outline why novel therapeutic strategies targeting CCR7 might provide clinical benefits to patients with CCR7-positive hematopoietic tumors.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto la Princesa (IIS-IP), Madrid, Spain.,Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Fernando Terrón
- Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Marco Herling
- Clinic of Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
12
|
Sabol M, Calleja-Agius J, Di Fiore R, Suleiman S, Ozcan S, Ward MP, Ozretić P. (In)Distinctive Role of Long Non-Coding RNAs in Common and Rare Ovarian Cancers. Cancers (Basel) 2021; 13:cancers13205040. [PMID: 34680193 PMCID: PMC8534192 DOI: 10.3390/cancers13205040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 02/05/2023] Open
Abstract
Rare ovarian cancers (ROCs) are OCs with an annual incidence of fewer than 6 cases per 100,000 women. They affect women of all ages, but due to their low incidence and the potential clinical inexperience in management, there can be a delay in diagnosis, leading to a poor prognosis. The underlying causes for these tumors are varied, but generally, the tumors arise due to alterations in gene/protein expression in cellular processes that regulate normal proliferation and its checkpoints. Dysregulation of the cellular processes that lead to cancer includes gene mutations, epimutations, non-coding RNA (ncRNA) regulation, posttranscriptional and posttranslational modifications. Long non-coding RNA (lncRNA) are defined as transcribed RNA molecules, more than 200 nucleotides in length which are not translated into proteins. They regulate gene expression through several mechanisms and therefore add another level of complexity to the regulatory mechanisms affecting tumor development. Since few studies have been performed on ROCs, in this review we summarize the mechanisms of action of lncRNA in OC, with an emphasis on ROCs.
Collapse
Affiliation(s)
- Maja Sabol
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia;
| | - Jean Calleja-Agius
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta; (J.C.-A.); (R.D.F.); (S.S.)
| | - Riccardo Di Fiore
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta; (J.C.-A.); (R.D.F.); (S.S.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Sherif Suleiman
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta; (J.C.-A.); (R.D.F.); (S.S.)
| | - Sureyya Ozcan
- Department of Chemistry, Middle East Technical University (METU), 06800 Ankara, Turkey;
- Cancer Systems Biology Laboratory (CanSyl), Middle East Technical University (METU), 06800 Ankara, Turkey
| | - Mark P. Ward
- Department of Histopathology, Trinity St James’s Cancer Institute, Emer Casey Molecular Pathology Laboratory, Trinity College Dublin and Coombe Women’s and Infants University Hospital, D08 RX0X Dublin, Ireland;
| | - Petar Ozretić
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia;
- Correspondence: ; Tel.: +385-(1)-4571292
| |
Collapse
|
13
|
Xie W, Sun H, Li X, Lin F, Wang Z, Wang X. Ovarian cancer: epigenetics, drug resistance, and progression. Cancer Cell Int 2021; 21:434. [PMID: 34404407 PMCID: PMC8369623 DOI: 10.1186/s12935-021-02136-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/03/2021] [Indexed: 03/05/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common malignant tumors in women. OC is associated with the activation of oncogenes, the inactivation of tumor suppressor genes, and the activation of abnormal cell signaling pathways. Moreover, epigenetic processes have been found to play an important role in OC tumorigenesis. Epigenetic processes do not change DNA sequences but regulate gene expression through DNA methylation, histone modification, and non-coding RNA. This review comprehensively considers the importance of epigenetics in OC, with a focus on microRNA and long non-coding RNA. These types of RNA are promising molecular markers and therapeutic targets that may support precision medicine in OC. DNA methylation inhibitors and histone deacetylase inhibitors may be useful for such targeting, with a possible novel approach combining these two therapies. Currently, the clinical application of such epigenetic approaches is limited by multiple obstacles, including the heterogeneity of OC, insufficient sample sizes in reported studies, and non-optimized methods for detecting potential tumor markers. Nonetheless, the application of epigenetic approaches to OC patient diagnosis, treatment, and prognosis is a promising area for future clinical investigation.
Collapse
Affiliation(s)
- Weiwei Xie
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Huizhen Sun
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Xiaoduan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feikai Lin
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China
| | - Ziliang Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Xinhua Hospital, 1665 Kongjiang Road, Yangpu District, Shanghai, China.
| |
Collapse
|
14
|
李 玉, 陈 婕, 罗 倩, 谈 勇. Research progress in lncRNA and its action as ceRNA in ovarian function as well as the relevant diseases. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:745-752. [PMID: 34382592 PMCID: PMC10930132 DOI: 10.11817/j.issn.1672-7347.2021.200622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Indexed: 11/03/2022]
Abstract
Long chain non-encoding RNA (lncRNA) can affect gene expression through transcription, post transcriptional regulation and epigenetic modification, and it is involved in regulating ovarian physiological function. LncRNA, as a competitive endogenous RNA, can affect the expression of target gene mRNA by competitively binding microRNA (miRNA), which are called lncRNA/miRNA/mRNA regulatory network. It plays an important role in the regulation of ovarian physiological function and the occurrence and development of ovarian reproductive disorders, expecting to become a new target and diagnostic index for the treatment of reproductive disorders.
Collapse
Affiliation(s)
| | | | | | - 勇 谈
- 谈勇,, ORCID: 0000-0003-3629-1789
| |
Collapse
|
15
|
Radu MR, Prădatu A, Duică F, Micu R, Creţoiu SM, Suciu N, Creţoiu D, Varlas VN, Rădoi VE. Ovarian Cancer: Biomarkers and Targeted Therapy. Biomedicines 2021; 9:693. [PMID: 34207450 PMCID: PMC8235073 DOI: 10.3390/biomedicines9060693] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/04/2021] [Accepted: 06/16/2021] [Indexed: 12/25/2022] Open
Abstract
Ovarian cancer is one of the most common causes of death in women as survival is highly dependent on the stage of the disease. Ovarian cancer is typically diagnosed in the late stage due to the fact that in the early phases is mostly asymptomatic. Genomic instability is one of the hallmarks of ovarian cancer. While ovarian cancer is stratified into different clinical subtypes, there still exists extensive genetic and progressive diversity within each subtype. Early detection of the disorder is one of the most important steps that facilitate a favorable prognosis and a good response to medical therapy for the patients. In targeted therapies, individual patients are treated by agents targeting the changes in tumor cells that help them grow, divide and spread. Currently, in gynecological malignancies, potential therapeutic targets include tumor-intrinsic signaling pathways, angiogenesis, homologous-recombination deficiency, hormone receptors, and immunologic factors. Ovarian cancer is usually diagnosed in the final stages, partially due to the absence of an effective screening strategy, although, over the times, numerous biomarkers have been studied and used to assess the status, progression, and efficacy of the drug therapy in this type of disorder.
Collapse
Affiliation(s)
- Mihaela Raluca Radu
- Fetal Medicine Excellence Research Center, Alessandrescu-Rusescu National Institute for Mother and Child Health, 020395 Bucharest, Romania; (M.R.R.); (A.P.); (F.D.); (N.S.)
| | - Alina Prădatu
- Fetal Medicine Excellence Research Center, Alessandrescu-Rusescu National Institute for Mother and Child Health, 020395 Bucharest, Romania; (M.R.R.); (A.P.); (F.D.); (N.S.)
| | - Florentina Duică
- Fetal Medicine Excellence Research Center, Alessandrescu-Rusescu National Institute for Mother and Child Health, 020395 Bucharest, Romania; (M.R.R.); (A.P.); (F.D.); (N.S.)
| | - Romeo Micu
- Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Sanda Maria Creţoiu
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Nicolae Suciu
- Fetal Medicine Excellence Research Center, Alessandrescu-Rusescu National Institute for Mother and Child Health, 020395 Bucharest, Romania; (M.R.R.); (A.P.); (F.D.); (N.S.)
- Division of Obstetrics, Gynecology and Neonatology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Obstetrics and Gynecology, Alessandrescu-Rusescu National Institute for Mother and Child Health, Polizu Clinical Hospital, 020395 Bucharest, Romania;
| | - Dragoş Creţoiu
- Fetal Medicine Excellence Research Center, Alessandrescu-Rusescu National Institute for Mother and Child Health, 020395 Bucharest, Romania; (M.R.R.); (A.P.); (F.D.); (N.S.)
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Valentin Nicolae Varlas
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, 01171 Bucharest, Romania
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 030167 Bucharest, Romania
| | - Viorica Elena Rădoi
- Department of Obstetrics and Gynecology, Alessandrescu-Rusescu National Institute for Mother and Child Health, Polizu Clinical Hospital, 020395 Bucharest, Romania;
- Department of Medical Genetics, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
16
|
Non-Coding RNAs as Biomarkers of Tumor Progression and Metastatic Spread in Epithelial Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13081839. [PMID: 33921525 PMCID: PMC8069230 DOI: 10.3390/cancers13081839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Despite advances in cancer research in recent years, efficient predictive biomarkers of tumor progression and metastatic spread for ovarian cancer are still missing. Therefore, we critically address recent findings in the field of non-coding RNAs (microRNAs and long non-coding RNAs) and DNA methylation in ovarian cancer patients as promising novel biomarkers of ovarian cancer progression. Abstract Ovarian cancer is one of the most common causes of death among gynecological malignancies. Molecular changes occurring in the primary tumor lead to metastatic spread into the peritoneum and the formation of distant metastases. Identification of these changes helps to reveal the nature of metastases development and decipher early biomarkers of prognosis and disease progression. Comparing differences in gene expression profiles between primary tumors and metastases, together with disclosing their epigenetic regulation, provides interesting associations with progression and metastasizing. Regulatory elements from the non-coding RNA families such as microRNAs and long non-coding RNAs seem to participate in these processes and represent potential molecular biomarkers of patient prognosis. Progress in therapy individualization and its proper targeting also rely upon a better understanding of interactions among the above-listed factors. This review aims to summarize currently available findings of microRNAs and long non-coding RNAs linked with tumor progression and metastatic process in ovarian cancer. These biomolecules provide promising tools for monitoring the patient’s response to treatment, and further they serve as potential therapeutic targets of this deadly disease.
Collapse
|
17
|
Elevated long noncoding RNA MALAT-1 expression is predictive of poor prognosis in patients with breast cancer: a meta-analysis. Biosci Rep 2021; 40:225866. [PMID: 32700729 PMCID: PMC7419804 DOI: 10.1042/bsr20200215] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 07/08/2020] [Accepted: 07/21/2020] [Indexed: 12/29/2022] Open
Abstract
Accumulating evidence indicates that aberrant regulation of metastasis-associated lung adenocarcinoma transcript 1 (MALAT-1), a long noncoding RNA (lncRNA), plays a vital role in tumorigenesis. However, its association with breast cancer has not been systematically evaluated. In the current study, a meta-analysis was conducted to clarify the association between MALAT-1 and the prognosis and clinicopathological features of breast cancer. Relevant literature published in several databases was searched. Hazard ratio (HR) and odds ratio (OR) with 95% confidence interval (CI) were calculated to evaluate the effect of MALAT-1 expression on the survival outcomes and clinicopathological features of breast cancer. A total of 12 studies involving 4106 patients were identified. Pooled HR demonstrated that elevated MALAT-1 expression significantly predicted unfavorable overall survival (HR = 2.06, 95% CI: 1.66–2.56, P<0.0001) in patients with breast cancer. Subgroup analysis stratified by cancer type, sample size, and method of variance analysis also showed statistically significant associations. Additionally, the HR of patients with up-regulated MALAT-1 expression concerning disease-free survival (DFS), recurrence-free survival (RFS), and disease-specific survival (DSS) was 1.91 (95% CI: 1.53–2.39, P<0.0001). Further, elevated MALAT-1 expression was positively correlated with the progesterone receptor (PR) status (OR = 1.47, 95% CI: 1.18–1.82). Thus, MALAT-1 is a promising biomarker for predicting survival outcomes in patients with breast cancer.
Collapse
|
18
|
Qiao FH, Tu M, Liu HY. Role of MALAT1 in gynecological cancers: Pathologic and therapeutic aspects. Oncol Lett 2021; 21:333. [PMID: 33692865 DOI: 10.3892/ol.2021.12594] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 02/02/2021] [Indexed: 12/15/2022] Open
Abstract
Gynecological cancers, including breast, ovarian, uterine, vaginal, cervical and vulvar cancers are among the major threats to modern life, particularly to female health. Long non-coding RNAs (lncRNAs) play critical roles in normal development of organisms, as well as the tumorigenesis process, and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a large infrequently spliced lncRNA, which have been implicated in different gynecological cancers. MALAT1 is overexpressed in breast, ovarian, cervical and endometrial cancers, which initiates cancer progression by inducing changes in the expression of several anti-apoptotic and epithelial-to-mesenchymal transition-related genes. Targeting MALAT1 is an important strategy to combat gynecological cancers, and application of RNA-interference technology and chemotherapeutic process are crucial to target and minimize MALAT1 activity. The present review discusses the role of MALAT1 in gynecological cancers, and potential strategies to target this lncRNA to develop cancer therapeutics. However, further clinical studies are required to determine the prognostic potential of MALAT1 in gynecological cancers.
Collapse
Affiliation(s)
- Feng-Hua Qiao
- Department of Gynecology, Second People's Hospital of Jingmen, Jingmen, Hubei 448000, P.R. China
| | - Min Tu
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen, Hubei 448000, P.R. China
| | - Hong-Yan Liu
- Department of Gynecology, Maternal and Child Health Hospital of Jingmen, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
19
|
Lambrou GI, Adamaki M, Hatziagapiou K, Vlahopoulos S. Gene Expression and Resistance to Glucocorticoid-Induced Apoptosis in Acute Lymphoblastic Leukemia: A Brief Review and Update. Curr Drug Res Rev 2021; 12:131-149. [PMID: 32077838 DOI: 10.2174/2589977512666200220122650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/29/2019] [Accepted: 01/23/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Resistance to glucocorticoid (GC)-induced apoptosis in Acute Lymphoblastic Leukemia (ALL), is considered one of the major prognostic factors for the disease. Prednisolone is a corticosteroid and one of the most important agents in the treatment of acute lymphoblastic leukemia. The mechanics of GC resistance are largely unknown and intense ongoing research focuses on this topic. AIM The aim of the present study is to review some aspects of GC resistance in ALL, and in particular of Prednisolone, with emphasis on previous and present knowledge on gene expression and signaling pathways playing a role in the phenomenon. METHODS An electronic literature search was conducted by the authors from 1994 to June 2019. Original articles and systematic reviews selected, and the titles and abstracts of papers screened to determine whether they met the eligibility criteria, and full texts of the selected articles were retrieved. RESULTS Identification of gene targets responsible for glucocorticoid resistance may allow discovery of drugs, which in combination with glucocorticoids may increase the effectiveness of anti-leukemia therapies. The inherent plasticity of clinically evolving cancer justifies approaches to characterize and prevent undesirable activation of early oncogenic pathways. CONCLUSION Study of the pattern of intracellular signal pathway activation by anticancer drugs can lead to development of efficient treatment strategies by reducing detrimental secondary effects.
Collapse
Affiliation(s)
- George I Lambrou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| | - Maria Adamaki
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| | - Kyriaki Hatziagapiou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Athens, Greece
| |
Collapse
|
20
|
Dai C, Xu P, Liu S, Xu S, Xu J, Fu Z, Cao J, Lv M, Zhou J, Liu G, Zhang H, Jia X. Long noncoding RNA ZEB1-AS1 affects paclitaxel and cisplatin resistance by regulating MMP19 in epithelial ovarian cancer cells. Arch Gynecol Obstet 2020; 303:1271-1281. [PMID: 33151424 DOI: 10.1007/s00404-020-05858-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/21/2020] [Indexed: 01/27/2023]
Abstract
PURPOSE The long noncoding RNA (lncRNA) ZEB1-AS1 is reported overexpressed in sensitive ovarian cancer cells A2780 compared with paclitaxel (PTX)-and cisplatin (DDP)- resistant. However, the function and mechanism of ZEB1-AS1 in EOC cells still unknown. METHODS We used quantitative real-time PCR (qPCR) to detect ZEB1-AS1 expression in A2780 and A2780/R cells. A combination of siRNA, plasmids, CCK8 and flow cytometry was used to detect the effect of ZEB1-AS1 on ovarian cancer cell A2780 PTX and DDP resistance. Transcriptome sequencing, qPCR, and western blot were used for further mechanistic studies. RESULTS ZEB1-AS1 depletion using siRNA in chemosensitive A2780 cells significantly increased PTX and DDP resistance. In contrast, ZEB1-AS1 overexpression in PTX- and DDP-resistant A2780/resistant (A2780/R) cells reversed the observed drug resistance. Thus, ZEB1-AS1 plays an important role in PTX and DDP resistance in EOC cells. However, quantitative real-time PCR (qPCR) and western blot results suggested that ZEB1-AS1 did not regulate chemoresistance through regulation of ZEB1 protein. We used sequencing to detect mRNA expression changes in A2780 cells after ZEB1-AS1 silencing. The results indicated that MMP19 was the likely downstream factor of ZEB1-AS1. We further examined whether ZEB1-AS1 played an important role in chemoresistance by silencing MMP19 in ZEB1-AS1-overexpressing cells. CCK8 assay results suggested that MMP19 knockdown promoted ZEB1-AS1-induced chemoresistance to PTX and DDP in A2780 cells. CONCLUSION This study is the first to reveal that ZEB1-AS1 plays a pivotal role in cancer chemoresistance.
Collapse
Affiliation(s)
- Chencheng Dai
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.,Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Pengfei Xu
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Siyu Liu
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.,Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Sujuan Xu
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Juan Xu
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.,Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Ziyi Fu
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Jian Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Mingming Lv
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Juan Zhou
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Guangquan Liu
- Nanjing Maternal and Child Health Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.,Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Huilin Zhang
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.
| |
Collapse
|
21
|
Fu S, Wang Y, Li H, Chen L, Liu Q. Regulatory Networks of LncRNA MALAT-1 in Cancer. Cancer Manag Res 2020; 12:10181-10198. [PMID: 33116873 PMCID: PMC7575067 DOI: 10.2147/cmar.s276022] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/03/2020] [Indexed: 12/18/2022] Open
Abstract
Long noncoding (lnc)RNAs are a group of RNAs with a length greater than 200 nt that do not encode a protein but play an essential role in regulating the expression of target genes in normal biological contexts as well as pathologic processes including tumorigenesis. The lncRNA metastasis-associated lung adenocarcinoma transcript (MALAT)-1 has been widely studied in cancer. In this review, we describe the known functions of MALAT-1; its mechanisms of action; and associated signaling pathways and their clinical significance in different cancers. In most malignancies, including lung, colorectal, thyroid, and other cancers, MALAT-1 functions as an oncogene and is upregulated in tumors and tumor cell lines. MALAT-1 has a distinct mechanism of action in each cancer type and is thus at the center of large gene regulatory networks. Dysregulation of MALAT-1 affects cellular processes such as alternative splicing, epithelial–mesenchymal transition, apoptosis, and autophagy, which ultimately results in the abnormal cell proliferation, invasion, and migration that characterize cancers. In other malignancies, such as glioma and endometrial carcinoma, MALAT-1 functions as a tumor suppressor and thus forms additional regulatory networks. The current evidence indicates that MALAT-1 and its associated signaling pathways can serve as diagnostic or prognostic biomarker or therapeutic target in the treatment of many cancers.
Collapse
Affiliation(s)
- Shijian Fu
- The First Affiliated Hospital of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Yanhong Wang
- Department of Laboratory Medicine, Yuebei People's Hospital of Shaoguan, The Affiliated Hospital of Shantou University, Shaoguan 512025, People's Republic of China
| | - Hang Li
- The First Affiliated Hospital of Harbin Medical University, Harbin 150081, People's Republic of China
| | - Leilei Chen
- Department of Cardiology, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Capital Medical University, Beijing 100029, People's Republic of China
| | - Quanzhong Liu
- Department of Medical Genetics, Harbin Medical University, Harbin 150081, People's Republic of China
| |
Collapse
|
22
|
Wu Y, Sarkissyan M, Ogah O, Kim J, Vadgama JV. Expression of MALAT1 Promotes Trastuzumab Resistance in HER2 Overexpressing Breast Cancers. Cancers (Basel) 2020; 12:E1918. [PMID: 32708561 PMCID: PMC7409266 DOI: 10.3390/cancers12071918] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is associated with cancer progression. Our study examined the role of MALAT1 in breast cancer and the mechanisms involved in the regulation of MALAT1. METHODS In vitro cell and in vivo animal models were used to examine the role of MALAT1 in breast cancer. The interaction of FOXO1 (Forkhead Box O1) at the promoter region of MALAT1 was investigated by chromatin immunoprecipitation (ChIP) assay. RESULTS The data shows an elevated expression of MALAT1 in breast cancer tissues and cells compared to non-cancer tissues and cells. The highest level of MALAT1 was observed in metastatic triple-negative breast cancer and trastuzumab-resistant HER2 (human epidermal growth factor receptor 2) overexpressing (HER2+) cells. Knockdown of MALAT1 in trastuzumab-resistant HER2+ cells reversed epithelial to mesenchymal transition-like phenotype and cell invasiveness. It improved the sensitivity of the cell's response to trastuzumab. Furthermore, activation of Akt by phosphorylation was associated with the upregulation of MALAT1. The transcription factor FOXO1 regulates the expression of MALAT1 via the PI3/Akt pathway. CONCLUSIONS We show that MALAT1 contributes to HER2+ cell resistance to trastuzumab. Targeting the PI3/Akt pathway and stabilizing FOXO1 translocation could inhibit the upregulation of MALAT1.
Collapse
Affiliation(s)
- Yanyuan Wu
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 East 120th Street, Los Angeles, CA 90059, USA; (M.S.); (O.O.); (J.K.)
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Marianna Sarkissyan
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 East 120th Street, Los Angeles, CA 90059, USA; (M.S.); (O.O.); (J.K.)
| | - Ochanya Ogah
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 East 120th Street, Los Angeles, CA 90059, USA; (M.S.); (O.O.); (J.K.)
| | - Juri Kim
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 East 120th Street, Los Angeles, CA 90059, USA; (M.S.); (O.O.); (J.K.)
| | - Jaydutt V. Vadgama
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, 1731 East 120th Street, Los Angeles, CA 90059, USA; (M.S.); (O.O.); (J.K.)
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
23
|
Qi FF, Yang Y, Zhang H, Chen H. Long non-coding RNAs: Key regulators in oxaliplatin resistance of colorectal cancer. Biomed Pharmacother 2020; 128:110329. [PMID: 32502843 DOI: 10.1016/j.biopha.2020.110329] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed malignancies in the world with high relapse and mortality rates. Although oxaliplatin (OXA), a platinum-based anticancer drug, is widely used in CRC treatment, the resulting chemoresistance dramatically attenuates the drug efficacy and increases the failure rate of this therapy. Thus, the study on OXA-induced chemoresistance is extremely urgent. In recent years, emerging evidence has shown that lncRNAs play irreplaceable roles in drug resistance. However, we only have a limited knowledge of the lncRNAs that are closely related to oxaliplatin resistance in CRC. In present study, we identify and characterize these lncRNAs, including their functions, underlying mechanisms and possible applications.
Collapse
Affiliation(s)
- Fang-Fang Qi
- Department of Histology and Embryology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yunyao Yang
- Department of Histology and Embryology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Haowen Zhang
- Department of Histology and Embryology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Hongping Chen
- Department of Histology and Embryology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, PR China; Jiangxi Key Laboratory of Experimental Animals, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
24
|
García-Guede Á, Vera O, Ibáñez-de-Caceres I. When Oxidative Stress Meets Epigenetics: Implications in Cancer Development. Antioxidants (Basel) 2020; 9:antiox9060468. [PMID: 32492865 PMCID: PMC7346131 DOI: 10.3390/antiox9060468] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide and it can affect any part of the organism. It arises as a consequence of the genetic and epigenetic changes that lead to the uncontrolled growth of the cells. The epigenetic machinery can regulate gene expression without altering the DNA sequence, and it comprises methylation of the DNA, histones modifications, and non-coding RNAs. Alterations of these gene-expression regulatory elements can be produced by an imbalance of the intracellular environment, such as the one derived by oxidative stress, to promote cancer development, progression, and resistance to chemotherapeutic treatments. Here we review the current literature on the effect of oxidative stress in the epigenetic machinery, especially over the largely unknown ncRNAs and its consequences toward cancer development and progression.
Collapse
Affiliation(s)
- Álvaro García-Guede
- Epigenetics Laboratory, INGEMM, Hospital La PAZ. 28046 Madrid, Spain; (Á.G.-G.); (I.I.-d.-C.)
- Experimental Therapies and Novel Biomarkers in Cancer, Instituto de Investigación Sanitaria del Hospital La Paz. IdiPAZ, 28046 Madrid, Spain
| | - Olga Vera
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Correspondence:
| | - Inmaculada Ibáñez-de-Caceres
- Epigenetics Laboratory, INGEMM, Hospital La PAZ. 28046 Madrid, Spain; (Á.G.-G.); (I.I.-d.-C.)
- Experimental Therapies and Novel Biomarkers in Cancer, Instituto de Investigación Sanitaria del Hospital La Paz. IdiPAZ, 28046 Madrid, Spain
| |
Collapse
|
25
|
Colvin EK, Howell VM, Mok SC, Samimi G, Vafaee F. Expression of long noncoding RNAs in cancer-associated fibroblasts linked to patient survival in ovarian cancer. Cancer Sci 2020; 111:1805-1817. [PMID: 32058624 PMCID: PMC7226184 DOI: 10.1111/cas.14350] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 02/01/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant cell type in the tumor microenvironment and are responsible for producing the desmoplastic reaction that is a poor prognostic factor in ovarian cancer. Long non-coding RNAs (lncRNAs) have been shown to play important roles in cancer. However, very little is known about the role of lncRNAs in the tumor microenvironment. We aimed to identify lncRNAs expressed in ovarian CAFs that were associated with patient survival and used computational approaches to predict their function. Increased expression of 9 lncRNAs and decreased expression of 1 lncRNA in ovarian CAFs were found to be associated with poorer overall survival. A "guilt-by-association" approach was used to predict the function of these lncRNAs. In particular, MIR155HG was predicted to play a role in immune response. Further investigation revealed high MIR155HG expression to be associated with higher infiltrates of immune cell subsets. In conclusion, these data indicate expression on several lncRNAs in CAFs are associated with patient survival and are likely to play an important role in regulating CAF function.
Collapse
Affiliation(s)
- Emily K Colvin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Sydney, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Viive M Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Sydney, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Samuel C Mok
- Division of Surgery, Department of Gynecologic Oncology and Reproductive Medicine Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Goli Samimi
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
26
|
Pal S, Garg M, Pandey AK. Deciphering the Mounting Complexity of the p53 Regulatory Network in Correlation to Long Non-Coding RNAs (lncRNAs) in Ovarian Cancer. Cells 2020; 9:E527. [PMID: 32106407 PMCID: PMC7140525 DOI: 10.3390/cells9030527] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/05/2020] [Accepted: 02/15/2020] [Indexed: 12/22/2022] Open
Abstract
Amongst the various gynecological malignancies affecting female health globally, ovarian cancer is one of the predominant and lethal among all. The identification and functional characterization of long non-coding RNAs (lncRNAs) are made possible with the advent of RNA-seq and the advancement of computational logarithm in understanding human disease biology. LncRNAs can interact with deoxyribonucleic acid (DNA), ribonucleic acid (RNA), proteins and their combinations. Moreover, lncRNAs regulate orchestra of diverse functions including chromatin organization and transcriptional and post-transcriptional regulation. LncRNAs have conferred their critical role in key biological processes in human cancer including tumor initiation, proliferation, cell cycle, apoptosis, necroptosis, autophagy, and metastasis. The interwoven function of tumor-suppressor protein p53-linked lncRNAs in the ovarian cancer paradigm is of paramount importance. Several lncRNAs operate as p53 regulators or effectors and modulates a diverse array of functions either by participating in various signaling cascades or via interaction with different proteins. This review highlights the recent progress made in the identification of p53 associated lncRNAs while elucidating their molecular mechanisms behind the altered expression in ovarian cancer tumorigenesis. Moreover, the development of novel clinical and therapeutic strategies for targeting lncRNAs in human cancers harbors great promise.
Collapse
Affiliation(s)
- Sonali Pal
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India;
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida 201313, India;
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology, Amity University Haryana, Panchgaon, Manesar, Haryana 122413, India;
| |
Collapse
|
27
|
Ghafouri-Fard S, Esmaeili M, Shoorei H, Taheri M. A comprehensive review of the role of long non-coding RNAs in organs with an endocrine function. Biomed Pharmacother 2020; 125:110027. [PMID: 32106365 DOI: 10.1016/j.biopha.2020.110027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/09/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are transcripts with sizes larger than 200 nucleotides and no/ small open reading frame that cannot produce functional proteins. The number of these transcripts surpasses the number of coding genes. LncRNAs regulate many aspects of cell functions such as proliferation, cell cycle transition and differentiation; so their dysregulation has pervasive effects on cell phenotype. Increasing numbers of these transcripts have been shown to participate in the pathogenesis of cancer. In the current review, we summarize recent findings regarding the role of lncRNAs in tumors originated from organs which have an endocrine function. We mostly focused on adrenal, pancreas and pituitary gland as prototypes of these organs. Moreover, we presented the obtained data of the role of lncRNAs in prostate, ovarian and testicular cancers. Recent data highly supports the role of lncRNAs in the pathogenesis of cancers originated from these organs. Moreover, certain genomic loci within lncRNAs have been shown to be associated with risk of these cancers. Diagnostic and prognostic role of some lncRNAs in these cancers have been evaluated recently. Taken together, lncRNAs are putative biomarkers for cancers originated from organs which have an endocrine function.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadhosein Esmaeili
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
MiRNA-Based Inspired Approach in Diagnosis of Prostate Cancer. ACTA ACUST UNITED AC 2020; 56:medicina56020094. [PMID: 32102477 PMCID: PMC7074198 DOI: 10.3390/medicina56020094] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/10/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022]
Abstract
Prostate cancer is one of the most encountered cancer diseases in men worldwide and in consequence it requires the improvement of therapeutic strategies. For the clinical diagnosis, the standard approach is represented by solid biopsy. From a surgical point of view, this technique represents an invasive procedure that may imply several postoperative complications. To overcome these impediments, many trends are focusing on developing liquid biopsy assays and on implementing them in clinical practice. Liquid samples (blood, urine) are rich in analytes, especially in transcriptomic information provided by genetic markers. Additionally, molecular characterization regarding microRNAs content reveals outstanding prospects in understanding cancer progression mechanisms. Moreover, these analytes have great potential for prostate cancer early detection, more accurate prostate cancer staging and also for decision making respecting therapy schemes. However, there are still questionable topics and more research is needed to standardize liquid biopsy-based techniques.
Collapse
|
29
|
Abildgaard C, Do Canto LM, Steffensen KD, Rogatto SR. Long Non-coding RNAs Involved in Resistance to Chemotherapy in Ovarian Cancer. Front Oncol 2020; 9:1549. [PMID: 32039022 PMCID: PMC6985280 DOI: 10.3389/fonc.2019.01549] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/20/2019] [Indexed: 12/26/2022] Open
Abstract
Ovarian cancer (OC) accounts for more than 150,000 deaths worldwide every year. Patients are often diagnosed at an advanced stage with metastatic dissemination. Although platinum- and taxane-based chemotherapies are effective treatment options, they are rarely curative and eventually, the disease will progress due to acquired resistance. Emerging evidence suggests a crucial role of long non-coding RNAs (lncRNAs) in the response to therapy in OC. Transcriptome profiling studies using high throughput approaches have identified differential expression patterns of lncRNAs associated with disease recurrence. Furthermore, several aberrantly expressed lncRNAs in resistant OC cells have been related to increased cell division, improved DNA repair, up-regulation of drug transporters or reduced susceptibility to apoptotic stimuli, supporting their involvement in acquired resistance. In this review, we will discuss the key aspects of lncRNAs associated with the development of resistance to platinum- and taxane-based chemotherapy in OC. The molecular landscape of OC will be introduced, to provide a background for understanding the role of lncRNAs in the acquisition of malignant properties. We will focus on the interplay between lncRNAs and molecular pathways affecting drug response to evaluate their impact on treatment resistance. Additionally, we will discuss the prospects of using lncRNAs as biomarkers or targets for precision medicine in OC. Although there is still plenty to learn about lncRNAs and technical challenges to be solved, the evidence of their involvement in OC and the development of acquired resistance are compelling and warrant further investigation for clinical applications.
Collapse
Affiliation(s)
- Cecilie Abildgaard
- Department of Clinical Genetics, Lillebaelt Hospital-University Hospital of Southern Denmark, Vejle, Denmark.,Department of Clinical Oncology, Lillebaelt Hospital-University Hospital of Southern Denmark, Vejle, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Luisa M Do Canto
- Department of Clinical Genetics, Lillebaelt Hospital-University Hospital of Southern Denmark, Vejle, Denmark
| | - Karina D Steffensen
- Department of Clinical Oncology, Lillebaelt Hospital-University Hospital of Southern Denmark, Vejle, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Silvia R Rogatto
- Department of Clinical Genetics, Lillebaelt Hospital-University Hospital of Southern Denmark, Vejle, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
30
|
Sherif IO, Al-Shaalan NH, Sabry D. Ginkgo Biloba Extract Alleviates Methotrexate-Induced Renal Injury: New Impact on PI3K/Akt/mTOR Signaling and MALAT1 Expression. Biomolecules 2019; 9:biom9110691. [PMID: 31684190 PMCID: PMC6920877 DOI: 10.3390/biom9110691] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/28/2019] [Accepted: 11/02/2019] [Indexed: 12/15/2022] Open
Abstract
Renal injury induced by the chemotherapeutic agent methotrexate (MTX) is a serious adverse effect that has limited its use in the treatment of various clinical conditions. The antioxidant activity of Ginkgo biloba extract (GB) was reported to mitigate renal injury induced by MTX. Our research was conducted to examine the nephroprotective role of GB versus MTX-induced renal injury for the first time through its impact on the regulation of phosphatidylinositol 3-kinase/protein kinase B/ mammalian target of rapamycin (PI3K/Akt/mTOR) signaling together with the renal level of TGF-β mRNA and long non-coding RNA-metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) expression. A group of adult rats was intraperitoneally (ip) injected with MTX 20 mg/kg as a single dose to induce kidney injury (MTX group). The other group of rats was orally administered with GB 60 mg/kg every day for 10 days (GB+ MTX group). The MTX increased the serum creatinine and urea levels, renal TGF-β mRNA and MALAT1 expression, in addition to dysregulation of the PI3K/Akt/mTOR signaling when compared with normal control rats that received saline only (NC group). Moreover, renal damage was reported histopathologically in the MTX group. The GB ameliorated the renal injury induced by MTX and reversed the changes of these biochemical analyses. The involvement of PI3K/Akt/mTOR signaling and downregulation of TGF-β mRNA and MALAT1 renal expressions were firstly reported in the nephroprotective molecular mechanism of GB versus MTX-induced renal injury.
Collapse
Affiliation(s)
- Iman O Sherif
- Emergency Hospital, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Nora H Al-Shaalan
- Chemistry Department, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia.
| | - Dina Sabry
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
31
|
Wen J, Chen L, Tian H, Li J, Zhang M, Cao Q, Zhang W, Chen S, Shi L. Effect of MALAT1 Polymorphisms on Papillary Thyroid Cancer in a Chinese Population. J Cancer 2019; 10:5714-5721. [PMID: 31788131 PMCID: PMC6879318 DOI: 10.7150/jca.28887] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/05/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Long noncoding RNA MALAT1 has been previously reported in the carcinogenesis of several tumors, and its potential functional polymorphisms have also been investigated in various diseases. However, the relationship between these polymorphisms and the susceptibility of thyroid cancer has still been largely unknown. In the present study, we aimed to explore the association between MALAT1 polymorphisms and thyroid cancer (TC) susceptibility, as well as potential biological function in TC. Methods: We conducted a case-control study with 1134 papillary thyroid cancer (PTC) patients and 1228 controls to evaluate the potential correlation between MALAT1 genetic variations (single nucleotide polymorphism, SNP) and the risk of PTC. More detailed molecular mechanisms were explored by luciferase assay, cell counting kit-8 (CCK-8), and flow cytometry. Results: MALAT1 SNP rs619586 was identified as a significantly protective factor of PTC susceptibility (P = 0.017, OR= 0.76, 95%CI = 0.60-0.95). Further functional experiments of rs619586 indicated that G allele of rs619586 could significantly decrease MALAT1expression, reduce PTC proliferation, and directly increase PTC apoptosis. Conclusions: Our findings suggested that MALAT1 SNP rs619586 could serve as a potential indicator for PTC susceptibility and pathogenesis.
Collapse
Affiliation(s)
- Jing Wen
- Department of Ultrasonics, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.,Department of Pathophysiology, the Institute of Basic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Liang Chen
- Department of General Surgery, Wujiang NO.1 People's Hospital, Suzhou 215200, China
| | - Hua Tian
- Department of acute infectious disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Ji Li
- Department of Pathophysiology, the Institute of Basic Medicine, Guizhou Medical University, Guiyang 550004, China.,Central Laboratory, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Miao Zhang
- Department of Endocrinology, the Hospital Affiliated to Guizhou Medical University, Guiyang 550004, China
| | - Qing Cao
- College of Medicine, Henan University of Science and Technology, Luoyang 471023, China
| | - Wei Zhang
- Department of Thyroid Surgery, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Shi Chen
- Department of Public Health Sciences, University of North Carolina Charlotte, Charlotte, NC 28223, USA
| | - Lixin Shi
- Department of Pathophysiology, the Institute of Basic Medicine, Guizhou Medical University, Guiyang 550004, China.,Department of Endocrinology, the Hospital Affiliated to Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
32
|
Hong CH, Lin SH, Lee CH. CCL21 Induces mTOR-dependent MALAT1 Expression, Leading to Cell Migration in Cutaneous T-Cell Lymphoma. In Vivo 2019; 33:793-800. [PMID: 31028199 DOI: 10.21873/invivo.11541] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mycosis fungoides (MF) is indolent, but may disseminate to leukemia. We reported that C-C motif chemokine ligand 21 (CCL21) is associated with MF invasion and progression. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long noncoding RNA, is associated with several cancer types, however, how it interacts with CCL21 to regulate MF progression, remains unclear. MATERIALS AND METHODS Expression of long noncoding RNAs MALAT1, antisense noncoding RNA in the INK4 locus (ANRIL), Hox antisense intergenic RNA (HOTAIR), highly up-regulated in liver cancer RNA (HULC), and leukemia-associated non-coding insulin-like growth factor 1 receptor activator RNA 1 (LUNAR1) in tissues from MF was studied using polymerase chain reaction and RNA interference in MF cell line MyLa were used to address this question. RESULTS Expression of MALAT1 was selectively increased in MF tissues. C-C Chemokine receptor type 7 (CCR7) expression was found to be increased in MyLa cells. CCL21 was found not only to mediate migration, but also to enhance MALAT1 and mammalian target of rapamycin (mTOR) activation in MyLa cells. Knockdown of MALAT1 abrogated CCL21-mediated migration, but not mTOR activation. In contrast, mTOR inhibition reduced CCL21-mediated migration and MALAT1 expression. CONCLUSION CCL21 induced mTOR activation in MyLa cells, followed by expression of MALAT1, causing cell migration. MALAT1 and mTOR are potential therapeutic targets for MF.
Collapse
Affiliation(s)
- Chien-Hui Hong
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, R.O.C.,Department of Dermatology, National Yang-Ming University College of Medicine, Taipei, Taiwan, R.O.C
| | - Shang-Hung Lin
- Department of Dermatology, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, R.O.C
| | - Chih-Hung Lee
- Department of Dermatology, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, R.O.C.
| |
Collapse
|
33
|
Siddique H, Al-Ghafari A, Choudhry H, AlTurki S, Alshaibi H, Al Doghaither H, Alsufiani H. Long Noncoding RNAs as Prognostic Markers for Colorectal Cancer in Saudi Patients. Genet Test Mol Biomarkers 2019; 23:509-514. [DOI: 10.1089/gtmb.2018.0308] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Halima Siddique
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayat Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Cancer and Mutagenesis Unit, and King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Cancer and Mutagenesis Unit, and King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Suzan AlTurki
- University Medical Services Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda Alshaibi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hadeil Alsufiani
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
34
|
Gao G, Cheng X, Wei Q, Chen W, Huang W. Long noncoding RNA MALAT‐1 inhibits apoptosis and matrix metabolism disorder in interleukin‐1β‐induced inflammation in articular chondrocytes via the JNK signaling pathway. J Cell Biochem 2019; 120:17167-17179. [PMID: 31111559 DOI: 10.1002/jcb.28977] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 03/28/2019] [Accepted: 04/08/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Gui‐Cheng Gao
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Xi‐Gao Cheng
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Qiang‐Qiang Wei
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Wei‐Cai Chen
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| | - Wen‐Zhou Huang
- Department of Orthopedics The Second Affiliated Hospital of Nanchang University Nanchang China
| |
Collapse
|
35
|
Zeng S, Liu S, Feng J, Gao J, Xue F. Upregulation of lncRNA AB073614 functions as a predictor of epithelial ovarian cancer prognosis and promotes tumor growth in vitro and in vivo. Cancer Biomark 2019; 24:421-428. [PMID: 30909184 DOI: 10.3233/cbm-182160] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Saitian Zeng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Gynecology and Obstetrics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, China
| | - Shikai Liu
- Department of Gynecology and Obstetrics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, China
| | - Jing Feng
- Department of Gynecology and Obstetrics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, China
| | - Jiefan Gao
- Department of Gynecology and Obstetrics, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, Hebei 061000, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
36
|
Wang J, Zhang X, Chen W, Hu X, Li J, Liu C. Regulatory roles of long noncoding RNAs implicated in cancer hallmarks. Int J Cancer 2019; 146:906-916. [PMID: 30873588 DOI: 10.1002/ijc.32277] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 02/18/2019] [Accepted: 03/11/2019] [Indexed: 12/16/2022]
Abstract
Cancer cells acquire numerous biological properties (designated "cancer hallmarks"), such as cell survival and energy metabolism, that facilitate tumor growth and metastatic dissemination during development. To date, eight hallmarks of cancer have been identified that provide a logical framework for understanding the remarkable diversity of neoplastic diseases, as proposed by Douglas Hanahan and Robert A. Weinberg. Long noncoding RNAs (lncRNAs), a category of transcripts widely demonstrated to exert significant regulatory effects on biological processes, have attracted considerable research attention due to their association with the occurrence and development of cancer. The mechanisms by which lncRNAs exert their functions require elucidation to optimize their potential utility as alternative biomarkers and therapeutic targets during tumor occurrence and progression. In this review, we have discussed recent research progress on lncRNAs involved in various cancer hallmarks and their related mechanisms of action, with a view to providing an updated picture of their immense therapeutic potential in the fight against cancer.
Collapse
Affiliation(s)
- Jun Wang
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuan Zhang
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wen Chen
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, China.,State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiang Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Jing Li
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Changning Liu
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, China.,State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
37
|
Qu C, Dai C, Guo Y, Qin R, Liu J. Long noncoding RNA SNHG15 serves as an oncogene and predicts poor prognosis in epithelial ovarian cancer. Onco Targets Ther 2018; 12:101-111. [PMID: 30588034 PMCID: PMC6304080 DOI: 10.2147/ott.s182657] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective This study aims to investigate the functional role of long noncoding RNA SNHG15 in epithelial ovarian cancer (EOC). Materials and methods The expression of SNHG15 was measured in EOC cells and tissues using qRT-PCR. The correlation of SNHG15 expression and the clinicopathological characters was statistically analyzed. The prognosis of patients with different clinical features in the high/low SNHG15 expression groups were calculated. Moreover, univariate and multivariate Cox regression analyses were performed to identify the risk factors for poor overall survival (OS) and progression-free survival (PFS). The effect of SNHG15 on the migration and invasion was evaluated using Transwell and Matrigel, respectively. The proliferation ability of EOC cells was tested using colony formation and MTT assay. The influence of SNHG15 on the cisplatin resistance was detected by measuring cell inhibition rate and cell viability. Results SNHG15 was upegulated in EOC cells and tissues. High SNHG15 expression was correlated with EOC progression and predicted poor OS and PFS in different subgroups of EOC patients. Moreover, multivariate Cox regression analysis defined high SNHG15 expression as an independent risk factor for poor OS and PFS. Furthermore, functional assays showed that the overexpression of SNHG15 promoted migration and invasion, while the loss of SNHG15 suppressed migration and invasion. Furthermore, the proliferation of EOC cells was improved after the ectopic expression of SNHG15, which was suppressed with SNHG15 deficiency. In addition, cisplatin-resistant EOC cells were established for detecting the effect of SNHG15 on EOC chemoresistance. The results showed that cisplatin-resistant EOC cells exhibited much higher levels of SNHG15 expression than controls, and SNHG15 contributed to the chemoresistance of EOC cells. Conclusion This study confirms that SNHG15 contributes to the migration, invasion, proliferation, and chemoresistance of EOC. SNHG15 may serve as a potential therapeutic target and prognostic biomarker of EOC patients.
Collapse
Affiliation(s)
- Chong Qu
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, People's Republic of China
| | - Chunmei Dai
- Department of School Hospital, Changchun University of Chinese Medicine, Changchun 130033, Jilin, People's Republic of China
| | - Yahua Guo
- Department of Obstetrics and Gynaecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, People's Republic of China,
| | - Rui Qin
- Department of Obstetrics and Gynaecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, People's Republic of China,
| | - Junbao Liu
- Department of Obstetrics and Gynaecology, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, People's Republic of China,
| |
Collapse
|
38
|
Wang JY, Lu AQ, Chen LJ. LncRNAs in ovarian cancer. Clin Chim Acta 2018; 490:17-27. [PMID: 30553863 DOI: 10.1016/j.cca.2018.12.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/30/2022]
Abstract
Ovarian cancer is one of the most common gynecologic malignancies and has a poor prognosis. Recently, long noncoding RNAs (lncRNAs) have been identified as key regulators of cancer development. Studies have shown that the dysregulation of lncRNAs is frequently observed in ovarian cancer and greatly contributes to malignant phenotypical changes. In this review, we provide perspectives on the involvement of lncRNAs in the proliferation, apoptosis, cell cycle, migration, invasion, metastasis and drug resistance of ovarian cancer based on recent discoveries. Then, we discuss the role of lncRNAs in predicting the prognosis of ovarian cancer. Finally, we provide insight into the potential of lncRNAs for evaluating the diagnosis and prognosis of ovarian cancer.
Collapse
Affiliation(s)
- Jin-Yan Wang
- Department of Obstetrics and Gynecology, Zhangjiagang First People's Hospital, Zhangjiagang 215600, Jiangsu, PR China; Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Ai-Qing Lu
- Zhangjiagang Hospital of Traditional Chinese Medicine, Zhangjiagang 215600, PR China
| | - Li-Juan Chen
- Department of Obstetrics and Gynecology, Zhangjiagang First People's Hospital, Zhangjiagang 215600, Jiangsu, PR China.
| |
Collapse
|
39
|
Zhu S, Wang J, He Y, Meng N, Yan GR. Peptides/Proteins Encoded by Non-coding RNA: A Novel Resource Bank for Drug Targets and Biomarkers. Front Pharmacol 2018; 9:1295. [PMID: 30483132 PMCID: PMC6243196 DOI: 10.3389/fphar.2018.01295] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are defined as RNA molecules that do not encode proteins, but recent evidence has proven that peptides/proteins encoded by ncRNAs do indeed exist and usually contain less than 100 amino acids. These peptides/proteins play an important role in regulating tumor energy metabolism, epithelial to mesenchymal transition of cancer cells, the stability of the c-Myc oncoprotein, and the ubiquitination and degradation of proliferating cell nuclear antigen (PCNA). These peptides/proteins represent promising drug targets for fighting against tumor growth or biomarkers for predicting the prognosis of cancer patients. In this review, we summarize the characteristics of peptides/proteins that have recently been identified as putative ncRNA translation products and their outlook for small molecule peptide drugs, drug targets, and biomarkers.
Collapse
Affiliation(s)
- Song Zhu
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Jizhong Wang
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Yutian He
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Nan Meng
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Guang-Rong Yan
- Biomedicine Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
40
|
Qiu JJ, Lin XJ, Tang XY, Zheng TT, Lin YY, Hua KQ. Exosomal Metastasis‑Associated Lung Adenocarcinoma Transcript 1 Promotes Angiogenesis and Predicts Poor Prognosis in Epithelial Ovarian Cancer. Int J Biol Sci 2018; 14:1960-1973. [PMID: 30585260 PMCID: PMC6299373 DOI: 10.7150/ijbs.28048] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/15/2018] [Indexed: 12/21/2022] Open
Abstract
Exosomes mediate cell-cell crosstalk in cancer progression by transferring their molecular cargos, including long noncoding RNAs (lncRNAs). Metastasis‑associated lung adenocarcinoma transcript 1 (MALAT1) is a well-known lncRNA associated with cancer angiogenesis and metastasis. However, the presence of MALAT1 in exosomes and the roles and clinical values of exosomal MALAT1 in epithelial ovarian cancer (EOC) remain unknown. The present study focused on the crosstalk between EOC cells and endothelial cells mediated by exosomal MALAT1 and aimed to explore the roles of exosomes and exosomal MALAT1 in EOC angiogenesis and to reveal the clinical relevance and prognostic predictive value of serum exosomal MALAT1 in EOC. We observed that MALAT1 was increased in both metastatic EOC cells and their secreted exosomes. Exosomal MALAT1 derived from EOC cells was transferred to recipient human umbilical vein endothelial cells (HUVECs) via exosomes. In vitro and in vivo experiments demonstrated that MALAT1 knockdown impaired the exosome-mediated proangiogenic activity of HUVECs through certain key angiogenesis-related genes. Clinically, elevated serum exosomal MALAT1 was highly correlated with an advanced and metastatic phenotype of EOC and was an independent predictive factor for EOC overall survival (OS). Moreover, a prognostic nomogram model we constructed showed a good prediction of the probability of 3-year OS of EOC patients according to the c-index (0.751, 95% confidence interval [CI]=0.691-0.811) and calibration curve. Collectively, our data provide a novel mechanism by which EOC cells transfer MALAT1 via exosomes to recipient HUVECs and influence HUVECs by stimulating angiogenesis-related gene expression, eventually promoting angiogenesis. Additionally, circulating exosomal MALAT1 can serve as a promising serum-based, noninvasive predictive biomarker for EOC prognosis.
Collapse
Affiliation(s)
- Jun-Jun Qiu
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai 200011, China.,Department of Obstetrics and Gynaecology of Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai 200011, China
| | - Xiao-Jing Lin
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai 200011, China.,Department of Obstetrics and Gynaecology of Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai 200011, China
| | - Xiao-Yan Tang
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai 200011, China.,Department of Obstetrics and Gynaecology of Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai 200011, China
| | - Ting-Ting Zheng
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai 200011, China.,Department of Obstetrics and Gynaecology of Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai 200011, China
| | - Ying-Ying Lin
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Ke-Qin Hua
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai 200011, China.,Department of Obstetrics and Gynaecology of Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai 200011, China
| |
Collapse
|
41
|
Panoutsopoulou K, Avgeris M, Scorilas A. miRNA and long non-coding RNA: molecular function and clinical value in breast and ovarian cancers. Expert Rev Mol Diagn 2018; 18:963-979. [DOI: 10.1080/14737159.2018.1538794] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Affiliation(s)
- Konstantina Panoutsopoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Margaritis Avgeris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
42
|
Engqvist H, Parris TZ, Rönnerman EW, Söderberg EMV, Biermann J, Mateoiu C, Sundfeldt K, Kovács A, Karlsson P, Helou K. Transcriptomic and genomic profiling of early-stage ovarian carcinomas associated with histotype and overall survival. Oncotarget 2018; 9:35162-35180. [PMID: 30416686 PMCID: PMC6205557 DOI: 10.18632/oncotarget.26225] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/01/2018] [Indexed: 12/28/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy in the western world. Despite recent efforts to characterize ovarian cancer using molecular profiling, few targeted treatment options are currently available. Here, we examined genetic variants, fusion transcripts, SNP genotyping, and gene expression patterns for early-stage (I and II) ovarian carcinomas (n=96) in relation to clinicopathological characteristics and clinical outcome, thereby identifying novel genetic features of ovarian carcinomas. Furthermore, mutation frequencies of specific genetic variants and/or their gene expression patterns were associated with histotype and overall survival, e.g. SLC28A2 (mucinous ovarian carcinoma histotype), ARCN1 (low expression in 0-2 year survival group), and tumor suppressor MTUS1 (mutation status and overall survival). The long non-coding RNA MALAT1 was identified as a highly promiscuous fusion transcript in ovarian carcinoma. Moreover, gene expression deregulation for 23 genes was associated with tumor aggressiveness. Taken together, the novel biomarkers identified here may improve ovarian carcinoma subclassification and patient stratification according to histotype and overall survival.
Collapse
Affiliation(s)
- Hanna Engqvist
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth Werner Rönnerman
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Sahlgrenska University Hospital, Department of Clinical Pathology and Genetics, Gothenburg, Sweden
| | - Elin M V Söderberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jana Biermann
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claudia Mateoiu
- Sahlgrenska University Hospital, Department of Clinical Pathology and Genetics, Gothenburg, Sweden
| | - Karin Sundfeldt
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anikó Kovács
- Sahlgrenska University Hospital, Department of Clinical Pathology and Genetics, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
43
|
Xue Z, Zhu X, Teng Y. Long non‑coding RNA CASC2 inhibits progression and predicts favorable prognosis in epithelial ovarian cancer. Mol Med Rep 2018; 18:5173-5181. [PMID: 30320385 DOI: 10.3892/mmr.2018.9550] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 08/13/2018] [Indexed: 11/05/2022] Open
Affiliation(s)
- Zhuowei Xue
- Department of Obstetrics and Gynaecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Xiaolu Zhu
- Department of Obstetrics and Gynaecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yincheng Teng
- Department of Obstetrics and Gynaecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
44
|
TUG1 promotes prostate cancer progression by acting as a ceRNA of miR-26a. Biosci Rep 2018; 38:BSR20180677. [PMID: 29967294 PMCID: PMC6167503 DOI: 10.1042/bsr20180677] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/15/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023] Open
Abstract
Previous studies have demonstrated that taurine-upregulated gene 1 (TUG1) was aberrantly expressed and involved in multiple types of cancer; however, the expression profile and potential role of TUG1 in prostate cancer (PCa) remains unclear. The aim of the present study was to evaluate the expression and function of TUG1 in PCa. In the present study, we analyzed TUG1 expression levels of PCa patients in tumor and adjacent normal tissue by real-time quantitative PCR. Knockdown of TUG1 by RNAi was performed to explore its roles in cell proliferation, migration, and invasion. Here we report, for the first time, that TUG1 promotes tumor cell migration, invasion, and proliferation in PCa by working in key aspects of biological behaviors. TUG1 could negatively regulate the expression of miR-26a in PCa cells. The bioinformatics prediction revealed putative miR-26a-binding sites within TUG1 transcripts. In conclusion, our study suggests that long non-coding RNA (lncRNA) TUG1 acts as a functional oncogene in PCa development.
Collapse
|
45
|
Cooper DR, Wang C, Patel R, Trujillo A, Patel NA, Prather J, Gould LJ, Wu MH. Human Adipose-Derived Stem Cell Conditioned Media and Exosomes Containing MALAT1 Promote Human Dermal Fibroblast Migration and Ischemic Wound Healing. Adv Wound Care (New Rochelle) 2018; 7:299-308. [PMID: 30263873 PMCID: PMC6158770 DOI: 10.1089/wound.2017.0775] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/08/2018] [Indexed: 12/15/2022] Open
Abstract
Objective: Chronically ill patients heal recalcitrant ulcerative wounds more slowly. Human adipose-derived stem cells (hADSCs) play an important role in tissue regeneration and exosomes secreted by hADSC contribute to their paracrine signaling. In addition to cytokines, lipids and growth factors, hADSC secrete mRNA, miRNA, and long noncoding (lnc) RNA into exosomes. In this study we examined the role of lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1), an abundant lncRNA in exosomes from conditioned media (CM), on cell migration and ischemic wound healing. Approach: CM and isolated exosomes from hADSC were applied to human dermal fibroblast (HDF) in scratch assays and electric cell-substrate impedance sensing (ECIS) assays. CM was also applied to a rat model of ischemic wound healing and wound closure was followed. Results: CM stimulated cell migration of HDFs in vitro by 48%. CM stimulated the closure of ischemic wounds in a rat model 50% faster than unconditioned media. The depletion of MALAT1 in adipose-derived stem cell (ADSC) CM significantly reduced cell migration. Since MALAT1 is secreted into exosomes, a purified population of exosomes was applied to HDF where they enhanced cell migration in a similar manner to FGF-2 or basic fibroblast growth factor (bFGF) in ECIS wound healing assays. The uptake of exosomes by HDF was shown using dynasore, an inhibitor that blocks clathrin- and caveolin-dependent endocytosis. Depletion of MALAT1 in hADSC with antisense oligonucleotides resulted in exosomes without MALAT1. These exosomes had an effect similar to the unconditioned, control media in ECIS assays. Innovation: Exosomes contain lncRNA MALAT1 and other factors that have the potential to stimulate HDF cell migration and angiogenesis involved in wound healing without applying stem cells to wounds. Conclusion: Our results show the potential of using topically applied ADSC-derived exosomes containing MALAT1 for treating ischemic wounds. This allows for harnessing the power of stem cell paracrine signaling capabilities without applying the cells.
Collapse
Affiliation(s)
- Denise R. Cooper
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Chunyan Wang
- Department of Physiology and Pharmacology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Rehka Patel
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Andrea Trujillo
- Department of Physiology and Pharmacology, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Niketa A. Patel
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Jamie Prather
- Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Lisa J. Gould
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Physiology and Pharmacology, University of South Florida Morsani College of Medicine, Tampa, Florida
- Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Mack H. Wu
- Research Service, James A. Haley Veterans Hospital, Tampa, Florida
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
- Surgery, University of South Florida Morsani College of Medicine, Tampa, Florida
| |
Collapse
|
46
|
Lekka E, Hall J. Noncoding RNAs in disease. FEBS Lett 2018; 592:2884-2900. [PMID: 29972883 PMCID: PMC6174949 DOI: 10.1002/1873-3468.13182] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/18/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022]
Abstract
Noncoding RNAs are emerging as potent and multifunctional regulators in all biological processes. In parallel, a rapidly growing number of studies has unravelled associations between aberrant noncoding RNA expression and human diseases. These associations have been extensively reviewed, often with the focus on a particular microRNA (miRNA) (family) or a selected disease/pathology. In this Mini‐Review, we highlight a selection of studies in order to demonstrate the wide‐scale involvement of miRNAs and long noncoding RNAs in the pathophysiology of three types of diseases: cancer, cardiovascular and neurological disorders. This research is opening new avenues to novel therapeutic approaches.
Collapse
Affiliation(s)
- Evangelia Lekka
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Switzerland
| | - Jonathan Hall
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Switzerland
| |
Collapse
|
47
|
Bai L, Wang A, Zhang Y, Xu X, Zhang X. Knockdown of MALAT1 enhances chemosensitivity of ovarian cancer cells to cisplatin through inhibiting the Notch1 signaling pathway. Exp Cell Res 2018; 366:161-171. [PMID: 29548748 DOI: 10.1016/j.yexcr.2018.03.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 12/14/2022]
Abstract
Long non-coding RNAs (lncRNAs) are critical regulators in chemoresistance of various tumors including ovarian cancer. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been reported to be upregulated and contributed to ovarian cancer tumorigenesis. The aim of this study was to explore the roles of MALAT1 and the underlying molecular regulatory mechanism in the chemoresistance of ovarian cancer cells. Our data demonstrated that MALAT1 and Notch1 mRNA were upregulated in ovarian cancer tissues, as well as cisplatin (CDDP)-resistant ovarian cancer cells. A positive correlation between MALAT1 and Notch1 mRNA expression was observed. MALAT1 knockdown significantly attenuated CDDP resistance, and enhanced CDDP-induced apoptosis in CDDP-resistant ovarian cancer cells. MALAT1 knockdown enhanced CDDP-induced apoptosis in vivo, as indicated by upregulation of Bax protein expression and downregulation of Bcl-2 protein expression. Additionally, MALAT1 knockdown inhibited the Notch1 pathway and ABCC1 expression in CDDP-resistant ovarian cancer cells. MALAT1 was demonstrated to interact with Notch1. Notch1 knockdown attenuated CDDP resistance, and downregulated the protein expression of ABCC1 in ovarian cancer cells. Taken together, our findings suggested that knockdown of MALAT-1 enhanced chemosensitivity of ovarian cancer cells to CDDP through inhibiting Notch1 signaling pathway.
Collapse
MESH Headings
- Adenocarcinoma, Mucinous/drug therapy
- Adenocarcinoma, Mucinous/metabolism
- Adenocarcinoma, Mucinous/secondary
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Proliferation
- Cisplatin/pharmacology
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/secondary
- Drug Resistance, Neoplasm
- Endometrial Neoplasms/drug therapy
- Endometrial Neoplasms/metabolism
- Endometrial Neoplasms/secondary
- Female
- Follow-Up Studies
- Humans
- Lymphatic Metastasis
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Prognosis
- RNA, Long Noncoding/genetics
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lin Bai
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China.
| | - Aihua Wang
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China
| | - Yali Zhang
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China
| | - Xiaofeng Xu
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China
| | - Xiao Zhang
- Department of Obstetrics and Gynecology, The First People's Hospital of Shangqiu, No. 292 Kaixuan Nan Road, Suiyang District, Shangqiu 476100, China
| |
Collapse
|
48
|
Amodio N, Raimondi L, Juli G, Stamato MA, Caracciolo D, Tagliaferri P, Tassone P. MALAT1: a druggable long non-coding RNA for targeted anti-cancer approaches. J Hematol Oncol 2018; 11:63. [PMID: 29739426 PMCID: PMC5941496 DOI: 10.1186/s13045-018-0606-4] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023] Open
Abstract
The deeper understanding of non-coding RNAs has recently changed the dogma of molecular biology assuming protein-coding genes as unique functional biological effectors, while non-coding genes as junk material of doubtful significance. In the last decade, an exciting boom of experimental research has brought to light the pivotal biological functions of long non-coding RNAs (lncRNAs), representing more than the half of the whole non-coding transcriptome, along with their dysregulation in many diseases, including cancer.In this review, we summarize the emerging insights on lncRNA expression and functional role in cancer, focusing on the evolutionary conserved and abundantly expressed metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) that currently represents the best characterized lncRNA. Altogether, literature data indicate aberrant expression and dysregulated activity of MALAT1 in human malignancies and envision MALAT1 targeting as a novel treatment strategy against cancer.
Collapse
Affiliation(s)
- Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy.
| | - Lavinia Raimondi
- IRCSS Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopedic Institute, Palermo, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy
| | - Maria Angelica Stamato
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy
| | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100, Catanzaro, Italy.
| |
Collapse
|
49
|
Role of lncRNAs in ovarian cancer: defining new biomarkers for therapeutic purposes. Drug Discov Today 2018; 23:1635-1643. [PMID: 29698834 DOI: 10.1016/j.drudis.2018.04.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/23/2018] [Accepted: 04/18/2018] [Indexed: 01/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) are a class of noncoding RNA, involved in regulation of diverse physiological and pathological processes. Ovarian cancer is the leading cause of death among all gynecological malignancies in the world and its underlying mechanism is still unclear. LncRNAs exhibit multiple biological functions in various stages of ovarian cancer development. We will discuss and summarize the new and important lncRNAs and their involvement in disease, which might represent promising therapeutic targets. Therapeutic intervention based on silencing or functional inhibition of target lncRNAs will be beneficial for ovarian cancer patients.
Collapse
|
50
|
Vera O, Rodriguez-Antolin C, de Castro J, Karreth FA, Sellers TA, Ibanez de Caceres I. An epigenomic approach to identifying differential overlapping and cis-acting lncRNAs in cisplatin-resistant cancer cells. Epigenetics 2018; 13:251-263. [PMID: 29436261 PMCID: PMC5997141 DOI: 10.1080/15592294.2018.1436364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are critical regulators of cell biology whose alteration can lead to the development of diseases such as cancer. The potential role of lncRNAs and their epigenetic regulation in response to platinum treatment are largely unknown. We analyzed four paired cisplatin-sensitive/resistant non-small cell lung cancer and ovarian cancer cell lines. The epigenetic landscape of overlapping and cis-acting lncRNAs was determined by combining human microarray data on 30,586 lncRNAs and 20,109 protein coding mRNAs with whole-genome bisulfite sequencing. Selected candidate lncRNAs were further characterized by PCR, gene-ontology analysis, and targeted bisulfite sequencing. Differential expression in response to therapy was observed more frequently in cis-acting than in overlapping lncRNAs (78% vs. 22%, fold change ≥1.5), while significantly altered methylation profiles were more commonly associated with overlapping lncRNAs (29% vs. 8%; P value <0.001). Moreover, overlapping lncRNAs contain more CpG islands (CGIs) (25% vs. 17%) and the majority of CGI-containing overlapping lncRNAs share these CGIs with their associated coding genes (84%). The differences in expression between sensitive and resistant cell lines were replicated in 87% of the selected candidates (P<0.05), while our bioinformatics approach identifying differential methylation was confirmed in all of the selected lncRNAs (100%). Five lncRNAs under epigenetic regulation appear to be involved in cisplatin resistance (AC091814.2, AC141928.1, RP11-65J3.1-002, BX641110, and AF198444). These novel findings provide new insights into epigenetic mechanisms and acquired resistance to cisplatin that highlight specific lncRNAs, some with unknown function, that may signal strategies in epigenetic therapies.
Collapse
Affiliation(s)
- Olga Vera
- a Cancer Epigenetics Laboratory, INGEMM , La Paz University Hospital , Madrid , Spain.,b Biomarkers and Experimental Therapeutics in Cancer , IdiPAZ , Madrid , Spain
| | - Carlos Rodriguez-Antolin
- a Cancer Epigenetics Laboratory, INGEMM , La Paz University Hospital , Madrid , Spain.,b Biomarkers and Experimental Therapeutics in Cancer , IdiPAZ , Madrid , Spain
| | - Javier de Castro
- a Cancer Epigenetics Laboratory, INGEMM , La Paz University Hospital , Madrid , Spain.,b Biomarkers and Experimental Therapeutics in Cancer , IdiPAZ , Madrid , Spain
| | - Florian A Karreth
- c Department of Molecular Oncology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , USA
| | - Thomas A Sellers
- d Department of Cancer Epidemiology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , USA
| | - Inmaculada Ibanez de Caceres
- a Cancer Epigenetics Laboratory, INGEMM , La Paz University Hospital , Madrid , Spain.,b Biomarkers and Experimental Therapeutics in Cancer , IdiPAZ , Madrid , Spain
| |
Collapse
|