Exosomal non-coding RNAs (Exo-ncRNAs) in cardiovascular health

https://doi.org/10.1016/j.yjmcc.2019.09.016Get rights and content

Abstract

Extracellular vesicles (EVs) play a role in the pathophysiological processes and in different diseases, including cardiovascular disease. Out of several categories of EVs, exosomes (smallest – 30 to 150 nm) are gaining most of the focus as the next generation of biomarkers and in therapeutic strategies. This is because exosomes can be differentiated from other types of EVs based on the expression of tetraspanin molecules on the surface. More importantly, exosomes can be traced back to the cell of origin by identifying the unique cellular marker(s) on the exosomal surface. Recently, several researchs have demonstrated an important and underappreciated mechanism of paracrine cell-cell communication involving exosomal transfer, and its subsequent functional impact on recipient cells. Exosomes are enriched in proteins, mRNAs, miRNAs, and other non-coding RNAs, which can potentially alter myocardial function. Additionally, different stages of tissue damage can also be identified by measuring these bioactive molecules in the circulation. There are several aspects of this new concept still unknown. Therefore, in this review, we have summarized the knowledge we have so far and highlighted the potential of this novel concept of next generation biomarkers and therapeutic intervention.

Introduction

Far from being completely understood, it is now known that genomic sequence can be transcribed into a wide variety of protein-coding RNAs (mRNAs) and non-coding RNAs (ncRNAs) [1]. While 80–95% of the prokaryotic genome is mapped to protein-coding sequences, only about 2% of the human genome codes for known proteins. However, at least 60–70% of the mammalian genome is transcribed on one or both strands [2]. Recently, it has been identified that ncRNAs can have different biological functions, such as involvement in chromosome dynamics, splicing, RNA editing, translational inhibition and mRNA degradation [2].

ncRNAs are classified by length into long ncRNAs (lncRNAs; > 200 nucleotides long), and small ncRNAs (< 200 nucleotides long). The small ncRNAs are divided into microRNAs (miRNAs), small interfering RNAs (siRNAs), small nuclear RNAs (snRNAs), small nucleolar RNAs (snoRNAs), and P-element-induced wimpy testis (piwi)-interacting RNAs (piwiRNAs) [3].

Long ncRNAs are more heterogeneous and difficult to classify and characterize compared to small ncRNAs. The 200 nucleotide long cut-off was selected based on RNA-purification protocols [4]. Considering their reduced abundance, lncRNAs were initially thought to be transcriptional noise resulting from low RNA polymerase fidelity [5]. LncRNAs predominantly localize into the nucleus and can function both in cis- or trans- forms at the transcription site [6]. In mammalian cells, relative lncRNA expression changes occur in a disease-, tissue and developmental stage-specific manner [7]. Unlike miRNAs or proteins, a specific lncRNA function can’t be easily inferred from its sequence or structure. These functions include epigenetic, transcriptional and post-transcriptional regulation [5]. LncRNAs are also classified by their structure as linear and circular lncRNAs (circRNAs) [8]. In the heart, lncRNAs have been implicated in the development of several diseases [[9], [10], [11]] as well as in normal tissue development [[10], [11], [12], [13]].

Circular RNAs (circRNAs) are the most recently discovered class of ncRNAs that form a covalently closed circle structure [14]. As a general characteristic, circRNAs are widespread exhibiting cell type-specific expression. CircRNAs are abundant and in some cases, the expression is higher than their related linear isoforms [15,16]. CircRNAs are involved in several physiological and pathological processes, mainly to down-regulate miRNAs by working as “sponges”. [8]. Considering the circular structure, circRNAs have significantly longer half-lives than linear RNA molecules. Mammalian cells can secret circRNAs. CircRNAs can be detected in plasma, making them candidates for disease biomarkers [14].

Competing endogenous RNAs (ceRNAs) contain miRNA response elements (MREs) and can communicate with and regulate each other by competing for shared binding sites for the same miRNAs [[17], [18], [19]]. All types of RNAs may compete with each other for microRNAs, acting as “miRNAs sponges”. It’s been shown that ceRNAs can be non-coding RNAs, mRNAs and/or pseudogenes [[17], [18], [19]]. Although there is still some discussion about the feasibility of ceRNA function in vivo [20], several lncRNAs have been postulated as ceRNAs of different miRNAs involved in the development of cardiac hypertrophy [11,[21], [22], [23], [24], [25]].

Among the small ncRNAs, miRNAs and siRNAs are the most studied molecules. miRNAs and siRNAs are short RNA fragments that are approximately 19–25 nucleotides long, derived either from hairpin or double-stranded RNA precursors (dsRNA), respectively. siRNAs are derived from dsRNA of hundreds or thousands of base pairs in length that are converted into 21–25 nt length dsRNAs. These dsRNAs associate with Argonaute 2 (Ago2) to bind to complementary mRNA targets [26]. On the other hand, miRNAs are derived from primitive forms of primary RNAs (pri-miRNAs) that are either encoded within introns of protein-coding genes or autonomous miRNA genes. Pri-miRNAs, contain multiple hairpin structures of which each hairpin structure is about 70 nucleotides long. Inside the nuclear component, Drosha (dsRNA-specific endonuclease) cuts these hairpin structures from pri-miRNA to form a pre-mature miRNAs (pre-miRNA). Pre-miRNAs are then exported to the cytosol by a nuclear protein called Exportin. Pre-miRNAs are spliced into 19–25 nucleotide long single strand RNAs (ssRNA) by another dsRNA-specific endonuclease, Dicer [[27], [28], [29]]. miRNAs form the RNA-induced silencing complex (RISC) with several other proteins, including Ago2 and Dicer [[27], [28], [29]]. RISC is the functional unit of both miRNA and siRNA. A seed sequence of 6–8 nucleotides in the 5’-end of the miRNAs and siRNAs binds to a complementary sequence on the 3’-untranslated region (UTR) of the target mRNA. Depending on whether the pairing of the seed sequence to the target mRNA is complete, or partial, the interaction results in mRNA degradation or translation inhibition, respectively [26]. Normally, siRNAs lead to mRNA degradation, and miRNAs inhibit the translational process. However, recently there were some ncRNAs, such as miR369-3 and Let-7, that were shown to increase mRNA translation by interacting with the UTRs of target mRNAs [[30], [31], [32]]. In addition, it’s been shown that one miRNA can silence several genes, and multiple miRNAs can regulate the same gene. In fact, an entire cellular pathway can be regulated by an individual miRNA or miRNA clusters, and the binding of several miRNAs to the same mRNA (in different MREs) can function in a combinatorial manner [27]. Interestingly, miRNAs have also been shown to target the 5′-UTR and the coding sequence of mRNAs as well as pseudogenes and lncRNAs [[33], [34], [35]]. Several miRNAs with known and even unknown functions have been observed in plasma, and are seen as promising tools for disease biomarkers [36,37].

There are also other types of small ncRNAs with known physiological and pathological roles. Particularly, Y RNAs are a class of small ncRNAs from approximately 100 nucleotides long with a stem-loop secondary structure, involved in RNA processing and quality control and also, in DNA replication [38]. Fragments of Y RNAs have been found in body fluids and are gaining interest as possible disease biomarkers.

Cellular release of molecules within membrane vesicles has been recognized for some time. Initially believed to be cell debris, it is now clear that cells release vesicles of varying sizes both through the endosomal pathway and by budding from the plasma membrane. These vesicles are collectively termed extracellular vesicles (EVs) [39]. EVs are small vesicles (30 nm–5  μm) released from the cell surface by membrane fusion of many different cell types into different body fluids that contain proteins, lipids, and RNA representative of the host cell [40]. EVs can be classified into apoptotic bodies (800–5000 nm diameter) that are released by cells undergoing programmed cell death; microvesicles or ectosomes (50–1000 nm diameter) that are produced by budding from the plasma membrane, and exosomes (30–150 nm diameter) from an endocytic origin [40]. EVs are a mechanism of horizontal transfer of intercellular information between cells mainly in the form of proteins and RNA molecules [41]. Both mRNAs and ncRNAs are present within EVs and can be transferred into recipient cells with functional consequences [39]. A single cell can release EVs of different sizes, with a different biogenesis mechanism and cargo. These can mediate autocrine, paracrine, and endocrine functions. EVs have different physiological roles, such as cellular migration and invasion, immunity, development, and nervous system communication. EVs have also been implicated in the pathogenesis of multiple diseases such as cancer and neurodegenerative diseases, among others (reviewed in [39]) and can be used as markers of disease and prognosis [3,27,42].

Exosomes are the best-characterized class of vesicles and represent a specific subset due to the intracellular origin from the endosome. Exosomes express certain molecular signatures, such as tetraspanins, heat shock proteins, GTPases, and endosomal proteins and markers, which also differentiate exosomes from other EVs [41,42]. Most importantly, exosomes express specific proteins from their parental cell type [41,43] allowing for the identification of the exosomal source for clinical implications. Different populations of exosomes released from the apical and basolateral cell surface not only demonstrate the heterogeneity of vesicles but also strongly suggest the existence of very specialized mechanisms to control the selective sorting of cargo into these vesicles [44].

The inward budding of the late endosomal membrane of multivesicular bodies (MVB) results in the formation of intraluminal vesicles. These vesicles are the origin of exosomes and are released by the cells through the fusion of the MVB with the cell membrane. All eukaryotic cell types secrete exosomes [42]. Based on their 30–150 nm size distribution, exosomes are separated at higher sedimentation speeds (100,000 ×g) compared to other EVs. Electron microscopy is one of the most robust methods that allows for the visualization of exosomes [41]. It has been shown that the mRNAs carried by exosomes can be translated into proteins in the target cell, while miRNAs can be functionally transferred to recipient cells, and subsequently silence gene expression, indicating that exosomes can act as a vector of genetic information [41,45].

Although cargo transfer from exosomes into the recipient cells is not completely understood, the general consensus is that exosomal content is internalized into recipient cells via endocytosis or into an endocytic compartment [39]. In this manner, several groups have been studying different proteins that could be involved in specifically direct exosomes to specific tissues or cell types [[46], [47], [48]]. Particularly, at least two recent papers proved that Cardiac Homing Peptide expressed in exosomes could increase exosome transfer to cardiac tissue, with beneficial effects on heart function [49,50]. Taking all this into account, exosomes are increasingly recognized as one of the major players in both local and distant cell-cell communication [44,45].

Exosomes have been shown to carry different classes of ncRNAs. The lipid bilayer of exosomes protects ncRNAs from degradation in the extracellular space and body fluids [51]. lncRNAs have been detected in exosomes from body fluids of cancer patients [52] and are capable of modulating angiogenesis and metastasis [52]. Other exosomal ncRNAs, such as miRNAs and piwiRNAs, can be used as potential cancer biomarkers [3]. Other types of small ncRNAs as transfer RNA, ribosomal RNA, small nuclear RNA, small nucleolar RNA and fragments of YRNAs and mitochondrial RNAs have been also detected in human exosomes [53]. Exosomal miRNAs modulate inflammatory responses and circRNAs can be used as biomarkers for cancer diagnosis [54] (reviewed in [3]).

Section snippets

ncRNAs in cardiovascular pathophysiology

Although research on ncRNAs in the cardiovascular system is in its infancy, it has already been shown that ncRNAs have a potential therapeutic application. Additionally, they are exciting targets as biomarkers in cardiovascular diseases [55,56]. Due to the growing interest in studying exosomal ncRNAs (Exo-ncRNAs), the information about ncRNAs involved in the pathophysiology of cardiovascular diseases is growing exponentially, especially regarding exosomal miRNAs (Exo-miRs) [[56], [57], [58],

Discussion

Even though mouse or rat studies could not necessarily be translated, several studies have shown some conservation of ncRNAs sequences across species, indicating that the possibility of extrapolating results may be individually considered [137]. In this matter, over a thousand long intervening ncRNAs are likely to have a conserved function in mammals [138] and most ncRNAs display positional conservation across species, indicating that the genomic location may be important [137]. Interestingly,

Conclusions

Several recent studies have focused on the role of ncRNAs in multiple disease pathologies. More recently, several groups are interested in dissecting the role of ncRNAs in cardiovascular diseases. This is due to the fact that ncRNAs are highly abundant in cells, and can influence several signaling pathways inside them. Interestingly, several ncRNAs can affect one particular cellular function, highlighting the importance of ncRNAs in the pathophysiology of the cardiac disease condition.

Acknowledgements

This work was supported by grants from the AHA, 14SDG18890049 and MSCRF, Mscrfd-4313 (S.D.), and Stimulating and Advancing ACCM Research (S.D.).

References (139)

  • S. Vasudevan et al.

    AU-rich-element-mediated upregulation of translation by FXR1 and Argonaute 2

    Cell

    (2007)
  • U.A. Orom et al.

    MicroRNA-10a binds the 5’UTR of ribosomal protein mRNAs and enhances their translation

    Mol. Cell

    (2008)
  • K.M. Danielson et al.

    Plasma circulating extracellular RNAs in left ventricular remodeling post-myocardial infarction

    EBioMedicine

    (2018)
  • M.P. Kowalski et al.

    Functional roles of non-coding Y RNAs

    Int. J. Biochem. Cell Biol.

    (2015)
  • S.L.N. Maas et al.

    Extracellular vesicles: unique intercellular delivery vehicles

    Trends Cell Biol.

    (2017)
  • S. Mathivanan et al.

    Exosomes: extracellular organelles important in intercellular communication

    J. Proteome

    (2010)
  • C. Emanueli et al.

    Exosomes and exosomal miRNAs in cardiovascular protection and repair

    Vasc. Pharmacol.

    (2015)
  • S. Rana et al.

    Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection

    Int. J. Biochem. Cell Biol.

    (2012)
  • R.A. Boon

    Non-coding RNAs in cardiovascular health and disease

    Noncoding RNA Res.

    (2018)
  • Y. Chen et al.

    MicroRNA-133 overexpression promotes the therapeutic efficacy of mesenchymal stem cells on acute myocardial infarction

    Stem Cell Res. Ther.

    (2017)
  • S. Das et al.

    Extracellular vesicle microRNA transfer in cardiovascular disease

    Cardiovasc. Pathol.

    (2015)
  • D.A. Chistiakov et al.

    Cardiac-specific miRNA in cardiogenesis, heart function, and cardiac pathology (with focus on myocardial infarction)

    J. Mol. Cell. Cardiol.

    (2016)
  • X. Wang et al.

    Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells

    J. Mol. Cell. Cardiol.

    (2014)
  • L.M. Ptaszek et al.

    Towards regenerative therapy for cardiac disease

    Lancet

    (2012)
  • K.H. Wu et al.

    Stem cell engraftment and survival in the ischemic heart

    Ann. Thorac. Surg.

    (2011)
  • A. Izarra et al.

    miR-133a enhances the protective capacity of cardiac progenitors cells after myocardial infarction

    Stem Cell Rep.

    (2014)
  • R.C. Lai et al.

    Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury

    Stem Cell Res.

    (2010)
  • Y. Wang et al.

    Exosomes/microvesicles from induced pluripotent stem cells deliver cardioprotective miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocardium

    Int. J. Cardiol.

    (2015)
  • B. Yu et al.

    Exosomes secreted from GATA-4 overexpressing mesenchymal stem cells serve as a reservoir of anti-apoptotic microRNAs for cardioprotection

    Int. J. Cardiol.

    (2015)
  • D. Sayed et al.

    MicroRNA-21 is a downstream effector of AKT that mediates its antiapoptotic effects via suppression of Fas ligand

    J. Biol. Chem.

    (2010)
  • L. Chen et al.

    Cardiac progenitor-derived exosomes protect ischemic myocardium from acute ischemia/reperfusion injury

    Biochem. Biophys. Res. Commun.

    (2013)
  • A.G. Ibrahim et al.

    Exosomes as critical agents of cardiac regeneration triggered by cell therapy

    Stem Cell Rep.

    (2014)
  • D. Hilfiker-Kleiner et al.

    A cathepsin D-cleaved 16 kDa form of prolactin mediates postpartum cardiomyopathy

    Cell

    (2007)
  • L.C. Heather et al.

    Metabolism, hypoxia and the diabetic heart

    J. Mol. Cell. Cardiol.

    (2011)
  • J.J. Quinn et al.

    Unique features of long non-coding RNA biogenesis and function

    Nat. Rev. Genet.

    (2016)
  • J.S. Mattick et al.

    Non-coding RNA

    Hum. Mol. Genet.

    (2006)
  • S. Anfossi et al.

    Clinical utility of circulating non-coding RNAs – an update

    Nat. Rev. Clin. Oncol.

    (2018)
  • P. Kapranov et al.

    RNA maps reveal new RNA classes and a possible function for pervasive transcription

    Science

    (2007)
  • T.R. Mercer et al.

    Long non-coding RNAs: insights into functions

    Nat. Rev. Genet.

    (2009)
  • X. Jiang et al.

    The emerging roles of long noncoding RNAs in common cardiovascular diseases

    Hypertens. Res.

    (2015)
  • L. Hobuss et al.

    Long non-coding RNAs: at the heart of cardiac dysfunction?

    Front. Physiol.

    (2019)
  • K. Ono et al.

    Long non-coding RNAs as key regulators of cardiovascular diseases

    Circ. J.

    (2018)
  • S. Greco et al.

    Long noncoding RNAs and cardiac disease

    Antioxid. Redox Signal.

    (2018)
  • J. Salzman et al.

    Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types

    PLoS One

    (2012)
  • S.P. Barrett et al.

    Circular RNAs: analysis, expression and potential functions

    Development

    (2016)
  • Y. Tay et al.

    The multilayered complexity of ceRNA crosstalk and competition

    Nature

    (2014)
  • K. Wang et al.

    The long noncoding RNA CHRF regulates cardiac hypertrophy by targeting miR-489

    Circ. Res.

    (2014)
  • X.H. Zhu et al.

    LncRNA MIAT enhances cardiac hypertrophy partly through sponging miR-150

    Eur. Rev. Med. Pharmacol. Sci.

    (2016)
  • Z. Li et al.

    Long noncoding RNA myocardial infarctionassociated transcript is associated with the microRNA1505p/P300 pathway in cardiac hypertrophy

    Int. J. Mol. Med.

    (2018)
  • P.D. Zamore et al.

    Ribo-gnome: the big world of small RNAs

    Science

    (2005)
  • Cited by (14)

    • Circular RNAs are a novel type of non-coding RNAs in ROS regulation, cardiovascular metabolic inflammations and cancers

      2021, Pharmacology and Therapeutics
      Citation Excerpt :

      CircHIPK3 has been reported to be transferred from cardiomyocytes into cardiac microvascular endothelial cells by exosomes and protect endothelial cells from oxidative damage and vascular dysfunction in vitro under oxidative stress conditions by sponging miR-29 (Wang et al., 2019b). Additionally, exosomal ncRNAs are involved in cardiovascular diseases and their potential use as disease biomarkers (Jaquenod De Giusti, Santalla, & Das, 2019). However, the roles of exo-circRNAs in accelerating and propagating cardiovascular and metabolic inflammation still require further study.

    • Extracellular vesicles: Potential impact on cardiovascular diseases

      2021, Advances in Clinical Chemistry
      Citation Excerpt :

      For this reason, the term “extracellular vesicle” is suggested by the ISEV as a generic name for all secreted vesicles, and also as a keyword in publications [26]. Recent reviews have categorized EVs into three groups [27–29]: exosomes, microvesicles, and apoptotic bodies. Exosomes are a homogenous population of EVs derived from the endocytic compartment.

    • Extracellular vesicles in cardiovascular disease

      2021, Advances in Clinical Chemistry
      Citation Excerpt :

      Over the past decade, more than 2600 miRNAs and 16,000 lncRNAs have been identified in the human genome [106,109]. Emer-ging evidence suggests that EVs serve as transport carriers of circulating ncRNAs by protecting them from degradation by RNases [110,111], therefore, the diagnostic value of ncRNAs in EVs may be superior to that in serum or plasma. The potential of EV ncRNAs as biomarkers for CVD, including ACS [59,112,113], atherosclerosis [60], HF [61–65], cardiac arrhythmia [66] and cerebrovascular diseases [67,68], has attracted tremendous interest in recent years (Table 1).

    • The pluripotent role of exosomes in mediating non-coding RNA in ventricular remodeling after myocardial infarction

      2020, Life Sciences
      Citation Excerpt :

      Exosomes are multivesicular bodies (MVBs) with lipid bilayer structure secreted by the fusion of MVBs and cell membranes. The diameter of an exosome is 30–150 nm [42], the membrane of which is rich in cholesterol, sphingomyelin, glycolipid and neuroamide [43]. Through the endocytosis pathway, a variety of bioactive substances in the cytoplasm are gathered in the intracellular body of the MVBs before being released to the extracellular environment through the fusion and lysis of the MVBs and the plasma membrane to form exosomes [44].

    • Extracellular vesicular MicroRNA-27a* contributes to cardiac hypertrophy in chronic heart failure

      2020, Journal of Molecular and Cellular Cardiology
      Citation Excerpt :

      Moreover, cardiac fibroblast-derived EVs in response to cardiac stress, such as Ang II induce cardiac hypertrophy through the down-regulation of PDLIM5 probably attributed to both miRNA-27a* and miRNA-21* (Figs. 7, 8), suggesting that cardiac fibroblast secreted miRNA*-enriched EVs play a role in cardiac hypertrophy in the CHF state. miRNAs, in particular circulating EV-enriched miRNAs have recently been developed as therapeutic targets and biomarkers of cardiovascular disorders [12,38,46,47]. Pharmacological inhibition of miR-21* has been shown to attenuate cardiac hypertrophy [16].

    View all citing articles on Scopus
    View full text