Cell Mol Med Res
Cellular and Molecular Medicine Research, ISSN 2817-6359 online, Open Access
Article copyright, the authors; Journal compilation copyright, Cell Mol Med Res and Elmer Press Inc
Journal website https://www.thecmmr.org

Review

Volume 1, Number 1, September 2023, pages 8-11


Long Non-Coding RNAs as Regulators of Angiogenesis: A Mini-Review

Qiuwang Zhang

Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada

Manuscript submitted May 23, 2023, accepted June 26, 2023, published online August 30, 2023
Short title: Long Non-Coding RNAs
doi: https://doi.org/10.14740/cmmr15e

Abstract▴Top 

While only about 5-10% mammalian transcripts act as mRNAs, the vast majority of them do not have protein-coding capability, of which, a class of non-coding RNAs with a length of over 200 nucleotides are defined as long non-coding RNAs (lncRNAs). It has been shown that lncRNAs interact with RNA, DNA, or proteins through diverse mechanisms to regulate gene expression, thereby controlling a wide range of biological processes. Angiogenesis, a process of new blood vessel formation from pre-existing ones, occurs under both physiological and pathological conditions. It is involved in many diseases. In this article, lncRNA regulatory roles in angiogenesis and their therapeutic potentials were reviewed and discussed.

Keywords: Long non-coding RNAs; Angiogenesis; Endothelial cells; Cell proliferation; Tube formation

Introduction▴Top 

Transcripts of eukaryotic genomes differ in size and protein-coding capability. Genome-wide studies have revealed that only 5-10% mammalian transcripts function as mRNAs while the vast majority of them do not have protein-coding capability [1-3]. Based on the size, a class of non-coding RNAs with a length of over 200 nucleotides are defined as long non-coding RNAs (lncRNAs) [4, 5]. It is known lncRNAs regulate gene expression through a variety of mechanisms involving chromatin remodeling, the regulation of splicing and the control of microRNA function [5-8]. Studies have shown dysregulation of lncRNAs in many disease states, implication of lncRNAs in disease pathogenesis [9-15].

Angiogenesis is a process of new vessel formation from pre-existing ones, which is controlled by multiple angiogenic molecules and signaling pathways. It occurs under both physiological and pathological conditions. Angiogenesis is involved in wounding healing, tumor growth and metastasis, inflammation and many other disorders. Targeting abnormal angiogenesis is an important therapeutic strategy for various diseases [16-18]. In this article, several recently emerged angiogenic lncRNAs and their potentials as therapeutic targets were reviewed and discussed.

LncRNA Maternally Expressed Gene 3 (MEG3)▴Top 

Encoded by the MEG3, LncRNA MEG3 is a tumor suppressor [19]. Recently, several lines of evidence suggest a role for MEG3 in angiogenesis [20-23]. It is shown that the embryonic brain of MEG3–/– mice has remarkably elevated VEGF-A and VEGF-receptor 1 mRNA levels compared with that of wildtype littermates, indicating greater angiogenic activity in the brain after MEG3 knockout [20]. Indeed, MEG3-null embryos have a higher cortical microvessel density [20]. Increased expression of MEG3 in senescent human umbilical vein endothelial cells (HUVECs) is detected by RNA deep sequencing technology, and MEG3 elevation is associated with reduced HUVEC sprouting activity [21]. When MEG3 expression is suppressed, the impaired angiogenic activity of senescent HUVECs is restored. The inhibitory effect of MEG3 in angiogenesis has been further validated in a murine hind-limb ischemic model, as significantly increased new vessels and markedly improved blood flow are observed in the ischemic hind-limb after MEG3 silencing [21]. In line with these findings, Qiu et al have reported that knockdown of MEG3 in RF/6A endothelial cells promotes cell proliferation, migration and tube formation, and the mechanistic study suggests MEG3 impedes RF/6A cell angiogenic activity by blocking the activation of the PI3K-Akt signaling pathway [22]. More recently, another group demonstrates that MEG3 overexpression significantly suppresses endothelial proliferation and in vitro angiogenesis, whereas knockdown of MEG3 has the opposite effect [23]. These data indicate MEG3 is a negative regulator of angiogenesis.

LncRNA Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1)▴Top 

The MALAT1, an LncRNA that was originally described to be associated with metastasis of lung cancers [14], exhibits pro-angiogenic properties [10, 24-28]. MALAT1 deficiency results in impaired HUVEC proliferation, which is in accord with in vivo findings, as the number of proliferating endothelial cells and the vessel density in the retina of MALAT1-/- mice were significantly lower than those of wildtype controls [24]. Li et al have shown that suppression of MALAT1 markedly compromises the ability of HUVECs to form capillary-like structures [25]. MALAT1 knockdown also leads to decreased proliferation of retinal RF/6A endothelial cells through the regulation of the p38/MAPK signaling pathway [26]. In human umbilical cord mesenchymal stem cells (SMCs), MALAT1 up-regulates VEGF expression, and the conditioned medium from SMCs overexpressing MALAT1 enhances HUVEC tube formation [10]. Tumor angiogenesis plays a pivotal role in tumor growth and metastasis. LncRNA MALAT1 promoting tumor angiogenesis has also been reported [25, 27, 28]. Two groups have found lncRNA MALAT1 promotes tumor angiogenesis by mediating the production of fibroblast growth factor-2 (FGF-2). Huang et al reported that LncRNA MALAT1 up-regulated FGF-2 in tumor-associated macrophages, thereby boosting angiogenesis and furthering thyroid cancer cell migration and invasion [27]. Augmented expression of LncRNA MALAT1 has been detected in human neuroblastoma cells under hypoxic conditions, which is associated with dramatically increased production of FGF-2 from neuroblastoma cells, leading to robust angiogenesis contributing to tumor growth [28].

Other Angiogenic LncRNAs▴Top 

LncRNA MANTIS has been shown to be elevated in endothelial cells isolated from glioblastoma but reduced in pulmonary artery endothelial cells from lungs of patients with end stage idiopathic pulmonary arterial hypertension [29]. Knockdown of MANTIS in endothelial cells leads to attenuated cell migration, proliferation and tube formation. Thus, MANTIS positively maintains endothelial angiogenic capacity [29]. LncRNA IGF2AS expression is augmented in myocardial microvascular endothelial (mMVE) cells isolated from rats with type 2 diabetes. Inhibition of lncRNA IGF2AS in mMVE cells increases cell proliferation through up-regulating insulin-like growth factor 2 and VEGF [30]. LncRNA MIAT has emerged as an angiogenic activator, as repression of MIAT compromises the proliferative ability of endothelial cells [31]. High glucose can induce the expression of MIAT in multiple endothelial cells including human microvascular endothelial cells, HUVECs and retinal endothelial cells (RF/6A), suggesting MIAT might be involved in diabetes mellitus-induced microvascular dysfunction [31]. SENCR is a vascular-enriched lncRNA and its expression is diminished in endothelial cells isolated from patients with critical limb ischemia or premature coronary artery disease [32]. Overexpression of SENCR in HUVECs stimulates cell migration and promotes tube formation through upregulation of proangiogenic chemokines CCL5 and CX3CL1 [32]. Two important pro-angiogenic lncRNAs LINC00323-003 and MIR503HG are induced in endothelial cells by hypoxia. Silencing LINC00323-003 or MIR503HG results in defective HUVEC proliferation. However, HUVECs deficient in LINC00323-003 or MIR503HG differ in their ability to form capillary-like structures. Knockdown of LINC00323-003 leads to reduced tube formation in HUVECs, while MIR503HG deficiency does not affect tube formation significantly [33].

Angiogenic LncRNAs as Therapeutic Targets▴Top 

The research field of angiogenic lncRNAs is evolving. The mechanisms governing lncRNA angiogenic actions remain largely elusive, impeding the investigation of angiogenic lncRNAs as therapeutic targets. Indeed, there are only a limited number of studies that have explored angiogenic lncRNAs for therapeutic purposes. MEG3 knockdown has been examined for the treatment of stroke in a rat ischemic stroke model. In vivo application of lentiviral particles expressing hairpin RNA to suppress MEG3 increases microvessel density in the ischemic region and reduces brain lesion [34]. The Gapmer antisense oligonucleotide can specifically and effectively cleave target RNAs including lncRNAs in vivo and appears as a promising therapeutic agent [35]. The locked nucleic acid Gapmer directed against MALAT1 has been therapeutically tested in a mouse model of hind-limb ischemia. Intraperitoneal injection of the Gapmers is able to inhibit MALAT1 expression in both control and ischemic muscle tissues, and MALAT1 repression significantly reduces capillary density and blood flow in the ischemic muscles [24]. These data suggest angiogenesis can be enhanced or blocked by modulating lncRNA expression, which could be potentially applied for the treatment of angiogenesis-related diseases.

Conclusions▴Top 

LncRNAs plays an important role in angiogenesis. Targeting lncRNAs as a novel therapeutic approach for angiogenesis-related diseases seems promising. However, numerous issues are to be solved such as how to avoid off-site effect of therapeutic RNA molecules, how to efficiently deliver in vivo therapeutic molecules, and how to minimize the adverse effects, which warrants further studies.

Conflicts of Interest

None.


References▴Top 
  1. Bertone P, Stolc V, Royce TE, Rozowsky JS, Urban AE, Zhu X, Rinn JL, et al. Global identification of human transcribed sequences with genome tiling arrays. Science. 2004;306(5705):2242-2246.
    doi pubmed
  2. Cheng J, Kapranov P, Drenkow J, Dike S, Brubaker S, Patel S, Long J, et al. Transcriptional maps of 10 human chromosomes at 5-nucleotide resolution. Science. 2005;308(5725):1149-1154.
    doi pubmed
  3. Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, Stadler PF, et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 2007;316(5830):1484-1488.
    doi pubmed
  4. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136(4):629-641.
    doi pubmed
  5. Lee JT. Epigenetic regulation by long noncoding RNAs. Science. 2012;338(6113):1435-1439.
    doi pubmed
  6. Postepska-Igielska A, Giwojna A, Gasri-Plotnitsky L, Schmitt N, Dold A, Ginsberg D, Grummt I. LncRNA Khps1 regulates expression of the proto-oncogene SPHK1 via triplex-mediated changes in chromatin structure. Mol Cell. 2015;60(4):626-636.
    doi pubmed
  7. Imig J, Brunschweiger A, Brummer A, Guennewig B, Mittal N, Kishore S, Tsikrika P, et al. miR-CLIP capture of a miRNA targetome uncovers a lincRNA H19-miR-106a interaction. Nat Chem Biol. 2015;11(2):107-114.
    doi pubmed
  8. Wang K, Liu F, Zhou LY, Long B, Yuan SM, Wang Y, Liu CY, et al. The long noncoding RNA CHRF regulates cardiac hypertrophy by targeting miR-489. Circ Res. 2014;114(9):1377-1388.
    doi pubmed
  9. Li X, Song Y, Liu F, Liu D, Miao H, Ren J, Xu J, et al. Long Non-Coding RNA MALAT1 Promotes Proliferation, Angiogenesis, and Immunosuppressive Properties of Mesenchymal Stem Cells by Inducing VEGF and IDO. J Cell Biochem. 2017.
    doi
  10. Liu X, Lv R, Zhang L, Xu G, Bi J, Gao F, Zhang J, et al. Long noncoding RNA expression profile of infantile hemangioma identified by microarray analysis. Tumour Biol. 2016.
    doi
  11. Yan B, Tao ZF, Li XM, Zhang H, Yao J, Jiang Q. Aberrant expression of long noncoding RNAs in early diabetic retinopathy. Invest Ophthalmol Vis Sci. 2014;55(2):941-951.
    doi pubmed
  12. Vausort M, Wagner DR, Devaux Y. Long noncoding RNAs in patients with acute myocardial infarction. Circ Res. 2014;115(7):668-677.
    doi pubmed
  13. Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, Morgan TE, Finch CE, et al. Expression of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feed-forward regulation of beta-secretase. Nat Med. 2008;14(7):723-730.
    doi pubmed pmc
  14. Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, Tidow N, et al. MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 2003;22(39):8031-8041.
    doi pubmed
  15. Polesskaya OO, Haroutunian V, Davis KL, Hernandez I, Sokolov BP. Novel putative nonprotein-coding RNA gene from 11q14 displays decreased expression in brains of patients with schizophrenia. J Neurosci Res. 2003;74(1):111-122.
    doi pubmed
  16. Wang J, Chen J, Guo Y, Wang B, Chu H. Strategies targeting angiogenesis in advanced non-small cell lung cancer. Oncotarget. 2017.
  17. Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov. 2017.
    doi pubmed
  18. Missiaen R, Morales-Rodriguez F, Eelen G, Carmeliet P. Targeting endothelial metabolism for anti-angiogenesis therapy: A pharmacological perspective. Vascul Pharmacol. 2017;90:8-18.
    doi pubmed
  19. Zhou Y, Zhang X, Klibanski A. MEG3 noncoding RNA: a tumor suppressor. J Mol Endocrinol. 2012;48(3):R45-53.
    doi pubmed pmc
  20. Gordon FE, Nutt CL, Cheunsuchon P, Nakayama Y, Provencher KA, Rice KA, Zhou Y, et al. Increased expression of angiogenic genes in the brains of mouse meg3-null embryos. Endocrinology. 2010;151(6):2443-2452.
    doi pubmed pmc
  21. Boon RA, Hofmann P, Michalik KM, Lozano-Vidal N, Berghauser D, Fischer A, Knau A, et al. Long Noncoding RNA Meg3 Controls Endothelial Cell Aging and Function: Implications for Regenerative Angiogenesis. J Am Coll Cardiol. 2016;68(23):2589-2591.
    doi pubmed
  22. Qiu GZ, Tian W, Fu HT, Li CP, Liu B. Long noncoding RNA-MEG3 is involved in diabetes mellitus-related microvascular dysfunction. Biochem Biophys Res Commun. 2016;471(1):135-141.
    doi pubmed
  23. He C, Yang W, Yang J, Ding J, Li S, Wu H, Zhou F, et al. Long Noncoding RNA MEG3 Negatively Regulates Proliferation and Angiogenesis in Vascular Endothelial Cells. DNA Cell Biol. 2017;36(6):475-481.
    doi pubmed
  24. Michalik KM, You X, Manavski Y, Doddaballapur A, Zornig M, Braun T, John D, et al. Long noncoding RNA MALAT1 regulates endothelial cell function and vessel growth. Circ Res. 2014;114(9):1389-1397.
    doi pubmed
  25. Li Y, Wu Z, Yuan J, Sun L, Lin L, Huang N, Bin J, et al. Long non-coding RNA MALAT1 promotes gastric cancer tumorigenicity and metastasis by regulating vasculogenic mimicry and angiogenesis. Cancer Lett. 2017;395:31-44.
    doi pubmed
  26. Liu JY, Yao J, Li XM, Song YC, Wang XQ, Li YJ, Yan B, et al. Pathogenic role of lncRNA-MALAT1 in endothelial cell dysfunction in diabetes mellitus. Cell Death Dis. 2014;5:e1506.
    doi pubmed pmc
  27. Huang JK, Song WH, Lu BY, Huang YB, Dong HM, Ma XK, Zhu ZZ, et al. LncRNA-MALAT1 promotes angiogenesis of thyroid cancer by modulating tumor-associated macrophage FGF2 protein secretion. J Cell Biochem. 2017.
    doi
  28. Tee AE, Liu B, Song R, Li J, Pasquier E, Cheung BB, Jiang C, et al. The long noncoding RNA MALAT1 promotes tumor-driven angiogenesis by up-regulating pro-angiogenic gene expression. Oncotarget. 2016;7(8):8663-8675.
    pubmed pmc
  29. Leisegang MS, Fork C, Josipovic I, Richter F, Preussner J, Hu J, Miller MJ, et al. Long noncoding RNA MANTIS facilitates endothelial angiogenic function. Circulation. 2017.
    doi pubmed
  30. Zhao Z, Liu B, Li B, Song C, Diao H, Guo Z, Li Z, et al. Inhibition of long noncoding RNA IGF2AS promotes angiogenesis in type 2 diabetes. Biomed Pharmacother. 2017;92:445-450.
    doi pubmed
  31. Yan B, Yao J, Liu JY, Li XM, Wang XQ, Li YJ, Tao ZF, et al. lncRNA-MIAT regulates microvascular dysfunction by functioning as a competing endogenous RNA. Circ Res. 2015;116(7):1143-1156.
    doi pubmed
  32. Boulberdaa M, Scott E, Ballantyne M, Garcia R, Descamps B, Angelini GD, Brittan M, et al. A role for the long noncoding RNA SENCR in commitment and function of endothelial Cells. Mol Ther. 2016;24(5):978-990.
    doi pubmed pmc
  33. Fiedler J, Breckwoldt K, Remmele CW, Hartmann D, Dittrich M, Pfanne A, Just A, et al. Development of long noncoding RNA-based strategies to modulate tissue vascularization. J Am Coll Cardiol. 2015;66(18):2005-2015.
    doi pubmed pmc
  34. Liu J, Li Q, Zhang KS, Hu B, Niu X, Zhou SM, Li SG, et al. Downregulation of the long non-coding RNA Meg3 promotes angiogenesis after ischemic brain injury by activating notch signaling. Mol Neurobiol. 2016.
    doi pubmed pmc
  35. Kasuya T, Hori S, Watanabe A, Nakajima M, Gahara Y, Rokushima M, Yanagimoto T, et al. Ribonuclease H1-dependent hepatotoxicity caused by locked nucleic acid-modified gapmer antisense oligonucleotides. Sci Rep. 2016;6:30377.
    doi pubmed pmc


This article is distributed under the terms of the Creative Commons Attribution Non-Commercial 4.0 International License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.


Cellular and Molecular Medicine Research is published by Elmer Press Inc.

 

Browse  Journals  

 

Journal of Clinical Medicine Research

Journal of Endocrinology and Metabolism

Journal of Clinical Gynecology and Obstetrics

 

World Journal of Oncology

Gastroenterology Research

Journal of Hematology

 

Journal of Medical Cases

Journal of Current Surgery

Clinical Infection and Immunity

 

Cardiology Research

World Journal of Nephrology and Urology

Cellular and Molecular Medicine Research

 

Journal of Neurology Research

International Journal of Clinical Pediatrics

 

 
       
 

Cellular and Molecular Medicine Research, quarterly, ISSN 2817-6359 (Online), published by Elmer Press Inc.                     
The content of this site is intended for health care professionals.
This is an open-access journal distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License, which permits unrestricted
non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Creative Commons Attribution license (Attribution-NonCommercial 4.0 International CC-BY-NC 4.0)


This journal follows the International Committee of Medical Journal Editors (ICMJE) recommendations for manuscripts submitted to biomedical journals,
the Committee on Publication Ethics (COPE) guidelines, and the Principles of Transparency and Best Practice in Scholarly Publishing.

website: www.thecmmr.org   editorial contact: editor@thecmmr.org
Address: 9225 Leslie Street, Suite 201, Richmond Hill, Ontario, L4B 3H6, Canada

© Elmer Press Inc. All Rights Reserved.


Disclaimer: The views and opinions expressed in the published articles are those of the authors and do not necessarily reflect the views or opinions of the editors and Elmer Press Inc. This website is provided for medical research and informational purposes only and does not constitute any medical advice or professional services. The information provided in this journal should not be used for diagnosis and treatment, those seeking medical advice should always consult with a licensed physician.