Abstract
Extracellular noncoding RNAs (ncRNAs) play crucial roles in intercellular communications. The process of ncRNA secretion is highly regulated, with specific ncRNA profiles produced under different physiological and pathological circumstances. These ncRNAs are transported primarily via extracellular vesicles (EVs) from their origin cells to target cells, utilising both endocrine and paracrine pathways. The intercellular impacts of extracellular ncRNAs are essential for maintaining homeostasis and the pathogenesis of various diseases. Given the unique aspects of extracellular ncRNAs, we propose the term “RNAkine” to describe these recently-identified secreted factors. In this article, we explore the roles of RNAkines as intercellular modulators, particularly in their ability to regulate metabolism and influence tumorigenesis, highlighting their definition and importance as a distinct class of secreted factors.
Keywords: Extracellular noncoding RNA, RNAkine, Metabolism, Cancer
Noncoding RNAs Are Secreted Regulatory Factors in Intercellular Communications
Historically, it was assumed that RNAs functioned solely within the cells where they were produced. However, a paradigm shift has occurred in recent years, as emerging evidence demonstrates that noncoding RNAs (ncRNAs) also play regulatory roles in distant target cells, influencing specific regulatory networks. This shift began with a significant discovery in 2007 and 2008, when several research groups independently reported finding mRNAs and microRNAs (miRNAs) (see Glossary) in the extracellular space [1, 2]. Our team and other researchers conducted ground-breaking studies that confirmed the presence of circulating miRNAs and revealed their unique expression patterns in the bloodstream, which are associated with specific diseases [3–5]. These findings position circulating miRNAs as promising diagnostic and prognostic biomarkers (Box 1). Inspired by the discovery of circulating miRNAs, further studies have explored other types of ncRNAs in circulation, such as long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), tRNA-derived small RNAs (tsRNAs), and small nucleolar RNA (snoRNAs) [6]. Like miRNAs, the expression patterns of these ncRNAs are indicative of physiological or pathological states, reinforcing their potential as diagnostic biomarkers [7, 8].
Glossary.
- Alcoholic Hepatitis (AH)
alcohol-associated liver disease characterized by acute liver inflammation caused by heavy alcohol use.
- Circular RNA (circRNA)
RNA transcript that forms a close loop with both ends covalently connected. The structure makes circRNA resistant to exonuclease. CircRNA influences gene expression via diverse ways: including competing miRNAs as a sponge, regulating mRNA transcription, splicing and stability.
- Chemokine
small signalling molecules that regulate immune responses by acting as chemoattractant to guide the migration of cells. They are small proteins or peptides that comprise four main subfamilies: CXC, CC, CX3X, and XC. They accomplish the function by binding with G protein-coupled receptors (GPCR).
- Cytokines
proteins or peptides made by certain immune cells or non-immune cells (endothelial cells and fibroblasts) and affect the immune system. Examples are interferons (IFNs), interleukins (ILs), lymphokines and tumour necrosis factors (TNFs). They exert a wide range of immunoregulatory actions by interacting with cytokine receptors, activating signal transduction.
- DICER
a crucial enzyme for miRNA biogenesis, which recognizes and cleaves the pre-miRNA loop hairpin, generating miRNA duplex with a length of ∼ 22-nt.
- Epithelial-mesenchymal-transition (EMT)
a process known as trans-differentiation of epithelial cells into mesenchymal cells that is involved in development, wound healing, fibrosis and cancer progression.
- EV/exosomal miRNA
miRNA that are transferred via EV/exosome.
- Exosome
main subpopulation of EV with a diameter ranging from 30-150 nm. Exosomes are originated intracellularly from endocytic pathway. The inward budding of endosomal membrane yields small vesicular structures within the endosome lumen to form multivesicular bodies (MVB). Exosomes are released extracellularly when MVB fuse with plasma membrane.
- Extracellular RNA
RNA presented in extracellular space, including in various body fluids (e.g., serum plasma, saliva, urine and milk). They are presented by enclosed in EVs, or associated with lipoproteins (e.g., HDL) and RNA-binding proteins (e.g., AGO2). Many types of RNA species are found presenting in extracellular space, such as mRNA, miRNA, lncRNA, rRNA, tRNA, tsRNA, snoRNA, snRNA, mitochondrial RNAs, and artificially designed siRNA or guide RNA (such as Cas9-associated RNA).
- Extracellular vesicle (EV)
Heterogeneous group of membrane-bound particles naturally released by cells into extracellular space. EV contains various types of cytosolic material, including miRNA, mRNA, lipids, and proteins.
- Growth factor
cytokines or hormones that regulate cell differentiation and proliferation. Examples include epidermal growth factor (EGF), fibroblast growth factor (FGF), and vascular endothelial growth factor (VEFG).
- Hormone
signalling molecules that are produced by glands throughout the body, including eicosanoids, steroids, amino acid derivatives, protein/peptides, and gases. They are released in response to specific stimuli into circulation, and act in paracrine, endocrine or autocrine manner. The signalling types of hormones include endocrine, paracrine, autocrine and intracrine. They regulate fundamental physiological functions, including growth and development, metabolism, and reproduction.
- Long noncoding RNA (lncRNA)
a class of > 200-nt-long RNA transcripts that are not encoding proteins. LncRNA can regulate gene expression by multiples ways, including acting sponge of a miRNAs, regulating epigenetic modifications, protein/RNA stability, and translation.
- MicroRNA (miRNA)
small noncoding regulatory RNAs (19-22 nucleotides) that post-transcriptionally regulate gene expression by binding to specific mRNAs, leading to either mRNA degradation or inhibition of mRNA translation.
- Microvesicle
main subpopulation of EV with a diameter ranging from 50-1000 nm. They are derived from outward budding of the plasma membrane.
- Myoblast
the mononucleated quiescent satellite cells that defined as the precursor of muscle cells.
- Myogenesis
the formation of skeletal muscular tissues.
- Myotube
a long liner multinucleated fibre formed through the fusion of developmental myoblasts.
- Secreted RNA
is a broad term used to describe a class of RNA molecules that are actively released by cells into the extracellular space.
- Small nucleolar RNA (snoRNA)
a family of conserved noncoding RNAs that have essential roles in guiding the chemical modifications and processing of other RNA, such as rRNA.
- Tumour microenvironment (TME)
the environment located in the vicinity of the tumour that are composed of multiple stroma cells, such as immune cells, fibroblasts, and endothelial cells, as well as noncellular components, such as extracellular matrix (ECM) and signalling molecules.
- tRNA-derived small RNA (tsRNA)
noncoding RNAs with 18-40 nucleotides in length that are derived from precursor or mature tRNAs.
Box 1. The potential and challenge for circulating ncRNA as disease fingerprints.
Circulating ncRNAs have an untapped potential as valuable biomarkers for disease diagnosis, prognosis, and prediction due to their stable presence in the bloodstream and association with various diseases [108]. Ideal biomarkers are specific, sensitive, non-invasive, stable, and reproducible. Circulating ncRNAs meet some of these criteria, such as stability in the bloodstream and non-invasive accessibility. However, specificity, sensitivity and reproducibility challenges still need to be addressed to establish circulating ncRNAs as reliable disease fingerprints.
Various studies have demonstrated differential expression of miRNAs, lncRNAs, circRNAs and tsRNAs in the plasma or serum of individuals with a wide range of diseases, including obesity [109], type 2 diabetes [109, 110], cardiovascular disease [111, 112], neurodegenerative disease [108, 113] and various types of cancer [108, 114, 115]. The sensitivity and specificity of some ncRNAs as disease biomarkers have been significantly strengthened. For example, the upregulation of miR-222 in obesity and overweight has been confirmed by three independent investigations [116–118], and the levels of miR-122 are increased in the plasma or serum of individuals with obesity and insulin resistance and decreased in those who underwent surgery-induced weight loss [119, 120]. Therefore, independent studies repeated on similar or related sample cohorts provide more substantial evidence of the association between circulating ncRNAs and diseases.
Despite the promising potential of circulating ncRNAs as biomarkers, several challenges persist. Most studies focus on circulating miRNAs as biomarkers, but inconsistencies in altered miRNA levels across studies raise concerns about their reproducibility. Establishing a standardised procedure for assessing circulating miRNAs is essential. Different methods and systems, such as qPCR with various probes, TaqMan low-density array, small RNA sequencing or microRNA sequencing, have been employed to measure circulating miRNA levels. Additionally, normalising quantitative data of circulating miRNA poses a significant challenge for cross-sample comparison due to the lack of internal control for circulating miRNA. Some studies use circulating miRNAs that do not change in a specific disease as endogenous controls, such as miR-16 and miR-106, utilised for metabolic disease [109]. Their applicability is limited since they have not yet been shown to remain unaltered across various types of disease. Another approach involves spiking in an exogenous miRNA sequence (e.g., Caenorhabditis elegans miR-39) as an external control, which fails to reflect individual-specific variations [121]. Therefore, establishing a standardised assessment process, including RNA isolation, quality control, and determination and quantification methods, is imperative to ensure the reproducibility of circulating miRNA detection.
The discovery of extracellular ncRNAs has sparked broad interest in their functional roles. Notably, their circulation in the bloodstream is just one part of their journey. Valadi et al. pioneered the model that mRNAs and miRNAs travel through the extracellular space encapsulated within extracellular vesicles (EVs) released by cells. They found that these RNAs, facilitated by EVs, shuttle between cells, introducing an alternative mechanism of gene-based intercellular communication [2]. Furthermore, Zhang et al. demonstrated the selective packaging of various extracellular miRNAs into EVs. They provided ground-breaking evidence of the functional impacts of miRNAs in recipient cells by showing how monocyte-secreted miR-150 regulates the mRNA expression of c-Myb in recipient endothelial cells (ECs) [9]. EVs, consisting mainly of microvesicles and exosomes [10], play a crucial role in this process. They enable the stable transfer of diverse ncRNAs, ensuring their functionality within recipient cells far from their origin [2, 6, 9]. Moreover, extracellular ncRNAs can travel independently of EVs and are often associated with stabilising proteins, such as high-density lipoprotein (HDL) [11], or the RNA-binding protein (RBP) Argonaute2 (Ago2) complex [12]. As such, extracellular ncRNAs have emerged as alternative secreted factors of transmitting intercellular information, paralleling the roles of hormones and cytokines mediating intercellular communications.
Traditionally, inter-tissue and inter-organ communications have been attributed to signalling molecules, such as cytokines, chemokines, growth factors, and hormones, integral to orchestrating physiological responses and maintaining whole-body homeostasis. In a way, extracellular ncRNAs meet the functional criteria of these conventional secreted factors. First, the secretion of extracellular ncRNAs is a regulated process. It is evident from the finding that the compositions of extracellular ncRNAs differ substantially from their cellular counterparts [13]. Moreover, they are selectively secreted in response to specific stimuli, demonstrating a precise stimulus-secretion coupling mechanism (Box 2) [9, 14, 15]. Such regulated secretion enables ncRNAs to be released in a manner that corresponds with physiological or pathological states. Second, extracellular ncRNAs share similarities with conventional secreted factors in transmitting information between organs, tissues, and cells via paracrine, endocrine, or autocrine methods. However, unlike conventional signalling molecules that typically bind to receptors and initiate signalling pathways, most extracellular ncRNAs operate within recipient cells as endogenous RNAs. Encapsulated extracellular miRNAs, for instance, directly regulate the expression of various types of molecules, including transcription factors (TFs) [9, 16], functional enzymes [17], signalling receptors [18, 19] and associated molecules [14, 20], and growth factors [21, 22]. Additionally, encapsulated lncRNAs and circRNAs often function as a competitive endogenous RNAs (ceRNAs) for miRNAs [23, 24], modulating mRNA expression by “sponging” miRNAs, thereby affecting downstream pathways (often represented as lncRNA or circRNA /mRNA/pathway axes) [25]. Some ncRNAs can also act as ligands, triggering Toll-like receptors (TLRs). For example, liver-secreted miR-122, which circulates independently of EVs, binds to TLR-7 and activates the inflammatory response in alveolar macrophages [26]. Moreover, EV-delivered miR-29 and miR-21 from cancer cells can bind to TLR7 and TLR8 residing in endosomes of immune cells after internalisation [27]. These interactions illustrate that extracellular ncRNAs convey a complex language, signalling recipient cells about the homeostatic changes in origin cells.
Box 2. Selective and regulated secretion of extracellular ncRNA.
The secretion of extracellular ncRNAs is a finely regulated process rather than a result of a passive cellular leakage. This notion is supported by the substantial variations observed in the spectra of extracellular RNAs, including miRNAs, lncRNAs, and circRNAs across diverse physiological and pathological conditions [6, 7, 109, 114]. Although the precise mechanisms behind ncRNA loading and packaging into EVs under specific conditions remain unclear, one promising mechanism involves sequence motif-determined sorting of EV ncRNAs. These motifs, responsible for ncRNA secretion, are termed EXOmotif, while those responsible for ncRNA retention are termed CELLmotif or CLmotif [13, 122]. Sequence motifs are recognised by specific RNA-binding proteins (RBPs) that act as the machinery for localising ncRNAs into EVs. Heterogeneous nuclear ribonucleoproteins (hnRNPs), a family of RBPs, have been identified as crucial players in ncRNA sorting. For instance, SUMOylated hnRNPA2B1 recognises motif GGAG, localising specific ncRNAs into EVs, such as miR-198 [122]. In addition, HNRNPA1 and HnRNPA2B1 can recognise the lncRNAs ELNAT1 and LNMAT2, mediating their packaging into EVs [123]. Notably, the expression of many RBPs is regulated by pathological states. Increases in hnRNPA2B1 expression have been observed in lung, pancreatic adenocarcinoma, and breast cancer cells [106]. Therefore, unravelling the regulatory axis responsible for disease-related RBP-ncRNA secretion may shed light on how the secretion of ncRNAs can accurately respond to specific biological stimuli. This mechanism may explain why the secretion of ncRNAs follows a stimulus-response pattern similar to that of conventional hormones or cytokines.
Comprehensive studies have yielded valuable insights into the implications of extracellular ncRNA-mediated crosstalk in various physiological processes and diseases, as summarised herein. However, despite the growing recognition of unique functional characteristics and relevance of extracellular ncRNAs, there remains a lack of universally accepted terminology for unequivocally identifying these molecules, which adds complexity and uncertainty to their understanding. Given their distinctive attributes, we propose considering extracellular ncRNAs as a distinct category of functionally relevant secreted molecules. To better reflect their broad yet selective spectrum of activity and signalling property during intercellular communications, we introduce the term “RNAkine”. This unified and distinctive nomenclature highlights the efficiency of extracellular ncRNAs as regulators in a diverse range of essential biological and pathological processes.
In this article, we aim to provide an in-depth review of findings that offer valuable insights into the concept and significance of RNAkines, including miRNAs, circRNAs and lncRNAs. Our primary focus will be on elucidating their relevance to two crucial areas: metabolism and tumorigenesis. By diving into these specific contexts, we hope to emphasise the necessity and rationale behind introducing the term “RNAkine” as a distinct nomenclature for these secreted molecules.
RNAkines Play Significant Roles in Contributing to Metabolism
RNAkines can function within recipient cells, even when originating from distant sources, suggesting their involvement in endocrine cell-to-cell and organ-to-organ communications. Several major organs, including the pancreas, adipose tissue, skeletal muscle, and liver, are integral components of metabolic networks. These organs rely on RNAkines to maintain interorgan coordination, which is critical in both metabolic homeostasis and disturbances (as illustrated in Figure 1 and summarised in Table 1).
Figure 1. RNAkine-mediated metabolic regulatory network.
The diagram illustrates the RNAkine-mediated metabolic regulatory network involving various secreted organs, including the pancreas, adipose tissue, skeletal muscle, and liver, which play critical roles in interorgan communication and metabolic regulation. The pancreas utilises RNAkines to regulate glucose homeostasis. Adipose tissue-secreted RNAkines are involved in regulating insulin sensitivity and glucose homeostasis. Skeletal muscle-secreted RNAkines contribute to the mechanisms underlying the metabolic benefits associated with physical activity. Liver-secreted RNAkines play pathological roles in various types of liver disease and pulmonary inflammation. This figure was generated using BioRender (https://biorender.com/).
Table 1. Functional RNAkines mediating crosstalk in metabolism.
RNAkine | Donor | Recipient | Target | Effect | Mechanism | Ref. |
---|---|---|---|---|---|---|
miR-29a/b/c | Pancreatic β cell | Liver | p85α | Impairing insulin sensitivity | Inhibiting PI3K/AKT signalling pathway | [14] |
miR-26 | Pancreatic β cell | Liver, VAT, BAT | ACSL3, ACSL4, TCF7L2, PCK1, GSKβ, PKCδ, PKC θ | Promoting insulin secretion and β cell replication (autocrine); improving peripheral insulin sensitivity (endocrine) | Activating insulin signalling pathway, inhibiting gluconeogenesis and lipid synthesis. | [17] |
MiR-29 | Pancreatic β cell | Monocyte/macrophage | TRAF3 | Inducing inflammation in T2DM | Increasing chemotaxis of Monocytes and macrophages | [20] |
miR-99b | BAT | Liver | Fgf21 | Affecting insulin tolerance | Down-regulating Fgf21 mRNA and circulating FGF21 | [21] |
miR-378a-3p | BAT | Liver | p110α | Improving glucose output during cold exposure. | Inhibiting PI3K/AKT signalling pathway | [39] |
miR-27 | WAT | Skeletal muscle | PPARγ | Impairing insulin sensitivity | Impairing insulin signalling pathway Glut 4 expression | [32] |
miR-222 | WAT | Liver, Skeletal muscle | IRS1 | Impairing insulin sensitivity | Impairing insulin signalling pathway | [33] |
miR-155 | WAT | Macrophages in intestine lamina propria | -- | Promoting DSS-induced colitis | Promoting M1 differentiation. | [34] |
miR-9-3p | WAT | Neuron | BDNF | Damaging synapse and cognitive | Impairing formation, maintenance, and plasticity of synapse | [22] |
miR-155 | ATM | Skeletal muscle, Liver | PPARγ | Impairing insulin sensitivity | Impairing insulin signalling pathway | [16] |
miR-29a | ATM | Adipocyte, Myocyte, Hepatocyte | PPARδ | Impairing insulin sensitivity | Impairing insulin signalling pathway | [40] |
miR-690 | M2 | Adipose tissue, Liver, Skeletal muscle | NADK | Improving insulin sensitivity | Improving insulin responses | [41] |
miR-34a | Adipose tissue | Macrophage in adipose tissue | KLF4 | Promoting obesity-induced adipose inflammation | Inhibiting M2 polarization | [34] |
Circ_sxc (in Drosophila) | Adipose tissue | Brain | Dme-miR-87-3p | Ensuring brain function | Sponging miR-87-3p and in turn regulating neurological receptor ligand proteins | [66] |
LncSNHG9 | Adipose tissue | EC | TRADD | Preventing EC dysfunction. | Inhibiting inflammation and apoptosis | [65] |
miR-22, miR-181a, miR-133a | Myotubes | Myoblasts | SIRT1 | Promoting myogenesis | Contributing to the commitment of myoblasts in the process of differentiation. | [50] |
miR-133a, miR-133b | Skeletal muscle | Liver | -- | Improving insulin sensitivity | Inhibiting gluconeogenesis through upregulating FoxO1 | [53] |
miR-29b-3p | Myotubes (Atrophy) | Neuron cells | c-FOS, BCL-2, RIT1, and LAMC1 | Inhibiting neuronal differentiation | Activating c-FOS/HIF1α-AS2 pathway and neuronal differentiation-related genes. | [54] |
miR-130a | Myotubes | ECs | -- | Improving the proliferation, migration and tube formation of ECs | Activating NF-κB | [55] |
miR-192 | Hepatocytes | HSCs | -- | Promoting HSC activation | Enhancing various of fibrosis-associated markers such as α-SMA, TGF-β, Col1a1. | [56] |
miR-107 | Hepatocytes | HSCs CD4+ lymphocyte | DKK1 Foxp1 | Promoting HSC activation | Activating HSCs by activating Wnt signalling, and increasing the expression of IL-9 | [57] |
miR-1297 | Hepatocytes | HSCs | PTEN | Promoting HSC activation | Contributing to the activation and proliferation of HSCs via regulating PTEN/PI3K/AKT signalling pathway | [58] |
miR-122-5p | Hepatocytes | Monocyte | -- | Inducing inflammation | Promoting the M1 polarisation | [59] |
miR-192-5p | Hepatocytes | Macrophage | Rictor | Inducing inflammation | Activating M1 by modulating Rictor/Akt/FoxO1 signalling and promoting NAFLD progression | [60] |
miR-122 | Hepatocytes | Monocyte | HO-1 | Inducing inflammation | Sensitising monocytes to LPS stimulation and increased levels of pro-inflammatory cytokines. | [61] |
miR-122 (EV-independent) | Hepatocytes | Alveolar macrophage | -- | Inducing inflammation in HPS | Activating inflammatory responses trough binding to TLR. | [26] |
miR-122-5p | Hepatocytes | Endothelial | AGPAT1 | Promoting angiogenesis | Enhancing fatty acid utilisation. | [63] |
LncRNA-H19 | Cholangiocytes | Hepatocytes | SHP | Promoting cholestatic liver injury | Supressing SHP expression | [67] |
LncRNA-H19 | Cholangiocytes | HSC | -- | Promoting HSC activation | -- | [68] |
LncRNA-H19 | Cholangiocytes | Macrophage (Kupffer cells) | -- | Promoting M1 polarization | -- | [69] |
LncRNA MT1DP | Liver | Kidney | -- | Reinforcing nephrotoxicity of Cadmium. | -- | [70] |
VAT: visceral adipose tissue, BAT: brown adipose tissue, WAT: white adipose tissue, EC: endothelial cell, HSC: hepatic stellate cells, HPS: hepatopulmonary syndrome, ACSL: acyl-CoA synthetase long-chain, TCF7L2: transcription factor 7 like 2, PCK: phosphoenolpyruvate carboxykinase (PEPCK), PKC: protein kinase, TRAF3: tumour necrosis factor receptor (TNFR)-associated factor 3, FGF21: fibroblast growth factor 21, PPAR: peroxisome proliferator-activated receptor, IRS: insulin receptor substrate, BNDF: brain-derived neurotrophic factor, NADK: NAD+ kinase, KLF4: Kruppel-like factor 4, TRADD: TNFR type 1-associated death domain protein, SIRT1: the silent information regulator sirtuin 1, LAMC: lamin C, DKK1: Dickkopf-1, PTEN: phosphatase and tensin homolog, HO-1: heme oxygenase-1, AGPAT1: 1-acetylglycerol-3-phosphate O-acyltransferase 1, SHP: secondary hyperparathyroidism.
Pancreas-secreted RNAkines
The discovery of RNAkines secreted by pancreatic β cells in recent years has expanded our understanding of the regulation of glucose homeostasis, challenging the long-standing belief that insulin is the sole pancreatic β cell-derived hormone responsible for this function. This ground-breaking revelation, which occurred in 2018, just before the 100th anniversary of the discovery of insulin, has revealed that β cells secrete increased RNAkines in response to stimuli such as K+, glucose, and arginine, which also trigger insulin secretion [28]. These stimuli induce the secretion of distinct patterns of RNAkines [28], indicating a tight control mechanism over RNAkine secretion induced by well-established insulinotropic triggers.
As speculated, RNAkines secreted from pancreatic β cells play significant roles in regulating glucose metabolism. Cellular miR-29 family members (miR-29a/b/c) can be considered “diabetes-related” miRNAs since increased miR-29 levels in adipose tissues [29] and skeletal muscle [30] promote insulin resistance. When there are high levels of free fatty acids (FFAs) in the bloodstream, which can result from physiological conditions, such as fasting, or pathological situations, such as a high-fat diet (HFD) or obesity, β cells release miR-29a/b/c into circulation in response to elevated FFA levels. These secreted miR-29 molecules are taken up by the liver, where they target p85α, promoting insulin resistance and disrupting systemic glucose homeostasis. Notably, genetic deletion of miR-29a/b/c in β cells reduces the circulating levels of miR-29 and improves HFD-induced hepatic insulin resistance [14]. Therefore, secreted miR-29 may act cooperatively with other obesity-associated factors to promote obesity-induced insulin resistance.
Furthermore, increased cellular miR-29a/b/c in β cells promotes chemotaxis, facilitating the recruitment of monocytes and macrophages [20]. These miR-29 molecules are also secreted and taken up by recruited macrophages, further promoting pro-inflammatory cytokine secretion [20]. As a result, both cellular and secreted miR-29 from β cells cooperate to trigger and promote inflammation in type 2 diabetes [20]. Additionally, miR-26a is another miRNA secreted by pancreatic β cells that improves hepatic insulin sensitivity [17]. Decreased levels of miR-26a in the blood of individuals with type 2 diabetes are associated with impaired insulin sensitivity [17]. These discoveries highlight the different roles of RNAkines in regulating glucose metabolism and the pathogenesis of insulin resistance in type 2 diabetes.
Adipose tissue-secreted RNAkines
Adipose tissue is a multifaceted organ responsible for energy storage and the secretion of various bioactive adipokines, including hormones, signalling lipids, and inflammatory mediators [31]. Recent studies have highlighted the significant role of adipose tissue as a major source of circulating miRNAs since disrupting miRNA biogenesis in adipose tissues by deleting Dicer, a crucial protein for miRNA biogenesis, significantly reduces circulating miRNA levels [21].
Adipose tissue-secreted RNAkines play crucial roles in endocrine and paracrine communications with other organs, including the liver, skeletal muscle, colon and brain, modulating multiple biological processes. In obesity, the upregulation of secreted miR-27a disrupts glucose metabolism in skeletal muscle and leads to insulin resistance [32]. Additionally, miR-222 secreted from gonadal white adipose tissue (WAT) enters the skeletal muscle and liver, reducing insulin sensitivity by targeting insulin receptor substrate 1 (IRS1) [33].
The impacts of adipose tissue-secreted RNAkines extend beyond metabolic regulation. For instance, colitis can be exacerbated by miR-155 secreted from adipose tissues of mice fed a HFD [34]. Interestingly, HFD treatment shifts the profile of RNAkines from an anti-inflammatory to a pro-inflammatory phenotype, resulting in increased miR-155 expression [34]. Moreover, adipocyte-secreted EVs can traverse the blood-brain barrier (BBB) in a membrane protein-dependent manner and interact with neurons, particularly in the hippocampus [22]. In diabetes, adipose tissue-brain EV trafficking facilitates the transfer of miR-9-3p into the brain, causing synaptic damage and cognitive impairment by targeting brain-derived neurotrophic factor (BDNF) [22]. Therefore, adipocyte-secreted RNAkines provide a crucial link between diabetes and associated cognitive impairment [22].
Adipocyte-secreted RNAkines also play significant roles in regulating the biological function of cells within their local microenvironment. For example, adipocyte-secreted miR-34a inhibits the polarisation of M2 macrophage in adipose tissue, contributing to obesity-induced adipose inflammation by connecting lipid-overloaded adipocytes with inflammatory immune cells in obese adipose tissue [35].
Brown adipose tissue (BAT), a critical organ for thermogenesis, also functions as an endocrine organ, secreting conventional regulatory molecules, such as peptides and lipids, and RNAkines [36]. For example, BAT secretes miR-99b, which downregulates Fgf21 mRNA levels in the liver. The disruption of miR-99b secretion in mice with adipose tissue-specific Dicer knockout (AdicerKO) leads to upregulated Fgf21 expression in the liver [21], which can be reversed by administering exosomes loaded with overexpressed miR-99b in AdicerKO mice [21]. During cold exposure, activated BAT consumes substantial circulating glucose to fuel thermogenesis. In response to increased nutrient demand, the secretome of BAT plays a direct role in upregulating hepatic glucose output. For instance, IL-6 mediates hyperglycaemia under both acute (3 h) [37] and chronic (6 days) [38] cold exposure. BAT also secretes RNAkines, such as miR-378a-3p, in response to prolonged cold exposure, reprogramming systemic glucose metabolism by promoting hepatic gluconeogenesis [39]. This mechanism helps to maintain systemic glucose homeostasis, preventing hypoglycaemia when facing cold challenge [39]. It is also interesting to speculate how BAT-secreted RNAkines and other conventional secreted factors (such as IL-6) have co-dependent effects in response to different extents of cold exposure.
Macrophages within adipose tissue, or adipose-associated macrophages (ATMs), also secrete RNAkines that regulate insulin sensitivity. In obese mice, increased secretion of miR-155 from ATM impairs insulin sensitivity and glucose tolerance in insulin-target tissues, such as the skeletal muscle and liver [16]. Similarly, ATM-secreted miR-29a promotes insulin resistance in adipocytes, myocytes, and hepatocytes [40]. Conversely, anti-inflammatory M2-like macrophages secrete miR-690, which systemically improves insulin sensitivity [41].
Skeletal muscle-secreted RNAkines
Skeletal muscle, traditionally known for its role in movement and energy storage, has emerged as a dynamic endocrine organ capable of secreting factors into circulation in response to various environmental and physiological challenges [42–44]. Recent research has unveiled the presence of functional RNAkines secreted by skeletal muscles. One group of miRNAs in muscles (known as myomiRs), including miR-1, miR-133a, miR-133b, miR-206, miR-208a, miR-208b, miR-486, and miR-499, are secreted into circulation via exosomes [45, 46]. Interestingly, the patterns of RNAkines secreted by muscles depend on the type of exercise performed. For instance, various exercises, such as downhill exercise [47], swimming training [46], and acute aerobic exercise [48], can result in different myomiR profiles in circulating exosomes. However, the mechanism underlying the selectivity of released RNAkines during different types of exercise is still unclear. Of note, the elevation in the levels of RNAkines is more likely to be observed immediately after exercise, regardless of the duration [49].
The secretion of RNAkines by muscle has garnered scientific interest due to their critical role in myogenesis. Muscle cells, particularly myoblasts and myotubes, communicate through these RNAkines, enhancing muscle cell development and differentiation. C2C12 myotubes, for instance, secrete RNAkines that regulate crucial signalling pathways, such as the Wnt signalling pathway, in myoblasts, facilitating muscle cell differentiation [50]. Exercise-induced RNAkines target genes in the MAPK pathway, thereby promoting muscle cell differentiation and growth [51]. Additionally, acute aerobic exercise-induced RNAkines, such as miR-206, miR-133b, and miR-181a-5p, have shown promise in preventing muscle dystrophy [52].
Moreover, RNAkines secreted by skeletal muscles during physical activity positively impact metabolic health. High-intensity interval training (HIIT), for example, induces the release of miR-133a and miR-133b from muscles, which improves hepatic insulin sensitivity by targeting the transcription factor Forkhead box O1 (FoxO1) in the liver [53]. Administration of exosomes loaded with miR-133a and miR-133b to mice enhances insulin sensitivity, highlighting the systemic metabolic influence of muscle-secreted RNAkines [53]. Additionally, long-term swimming training results in elevated levels of miR-342-5p in circulating exosomes, which inhibits cardiomyocyte apoptosis under hypoxia/reoxygenation conditions [46].
Furthermore, muscle-secreted RNAkines play a role in establishing communication between muscles and the brain. For instance, miR-29b-3p, upregulated in muscle cells in various types of atrophy, has been found to disrupt neuronal differentiation [54]. Ageing mice exhibit elevated levels of miR-29b-3p in both muscle and blood and in vitro studies have shown that atrophic muscle cells secrete miR-29b-3p, which downregulates neuronal-related genes and inhibits neuronal differentiation when taken up by neuronal SH-SY5Y cells [54]. Additionally, muscle-secreted RNAkines impact the function of ECs. For example, C2C12 myotube-secreted miR-130a promotes EC proliferation, migration, and tube formation by activating the nuclear factor-kappa B (NF-κB) pathway [55], suggesting an angiogenic role of muscle-secreted RNAkines and their involvement in skeletal muscle-mediated capillarisation.
Liver-secreted RNAkines
The liver, a vital organ with numerous functions, including metabolism, detoxification, digestion, synthesis, and storage, also releases RNAkines under various pathophysiological conditions. In liver disorders characterised by fibrosis, RNAkines mediate communication between hepatocytes and hepatic stellate cells (HSCs), a key player in liver fibrogenesis. Hepatocytes release RNAkines that can induce the activation of HSCs. For instance, in response to palmitic acid (PA) treatment, hepatocytes increase exosome production and modify miRNA profiles in exosomes. This leads to elevated expression of miR-192, which stimulates the expression of fibrotic genes in HSCs [56]. Additionally, PA treatment induces the secretion of miR-107 from hepatocytes, triggering HSC activation by targeting DKK1 and activating Wnt signalling [57].
RNAkines released by hepatocytes also contribute to metabolic-associated fatty liver disease (MAFLD) pathogenesis. In response to lipotoxicity, hepatocytes release miR-1297, which promotes HSC activation and proliferation by modulating the PTEN/PI3K/AKT signalling pathway [58]. Inflammation is closely linked to the development of liver disease, with RNAkines mediating communication between hepatocytes and immune cells. Cholesterol, a significant contributor to MAFLD, induces the increased release of miR-122-5p from hepatocytes, which leads to M1 polarisation of macrophages and inflammation [59]. Lipotoxic injury results in the release of miR-192-5p, which activates macrophages and contributes to lipotoxicity-induced MAFLD by regulating the Rictor/Akt/Foxo1 pathway [60]. In alcoholic hepatitis (AH), hepatocyte-secreted miR-122 sensitises monocytes to LPS by targeting the HO-1 pathway, which exacerbates inflammation [61]. Inhibiting miR-122 may hold therapeutic potential for treating inflammation-related liver diseases [61]. Finally, hepatocyte-secreted miR-107 is delivered to CD4+ lymphocytes, which enhances the expression of IL-9 by modulating Foxp1[57].
Furthermore, liver-secreted RNAkines can impact distant organs. In hepatopulmonary syndrome (HPS), characterised by pulmonary vascular abnormalities, oedema, and dyspnoea, liver-secreted RNAkines contribute to its pathogenesis. Hepatocyte-secreted miR-194 enhances pulmonary angiogenesis, a critical process in HPS development [62]. Liver injury induces pulmonary inflammation through miR-122 secretion [26]. Interestingly, circulating miR-122, independent of EV transport, increases in individuals with liver damage [26]. Alveolar macrophages take up circulating miR-122, which triggers inflammatory responses by directly binding to TLRs [26]. Depleting miR-122 in mouse livers abolishes pulmonary inflammation and tissue damage [26]. However, liver-secreted RNAkines can also have beneficial roles. For example, exercise induces the release of miR-122-5p from the liver and enhances angiogenesis and wound healing [63]. However, this enhancement of angiogenesis may be a concern in the context of anticancer treatment strategies.
Other types of RNAkines contributing to metabolic regulation and disease
Compared to those of miRNAkines, investigations of other types of RNAkines secreted by significant organs are still emerging. For instance, lncRNA-p3134, elevated in the serum of individuals with type 2 diabetes, plays a beneficial role in glucose homeostasis by promoting β cell function [64]. Adipocyte-secreted lncSNHG9 has protective properties, such as its roles in alleviating inflammation and apoptosis in ECs, which are achieved by suppressing the expression of TNF receptor type 1-associated death domain (TRADD) [65]. Interestingly, the levels of secreted lncSNHG9 are reduced in obese individuals with endothelial dysfunction, highlighting its role in preserving vascular physiology [65]. In a Drosophila study, adipose-derived circ_sxc maintains normal brain neuronal synaptic signalling [66]. The liver also contributes to lncRNAkine secretion, with cholangiocyte-enriched lncRNA-H19 being elevated in serum exosomes from a liver injury mouse model and individuals with cirrhosis. This lncRNA can be transferred to hepatocytes, suppressing the small heterodimer partner (SHP) at both transcriptional and posttranscriptional levels, thereby promoting cholestatic injury [67]. Furthermore, cholangiocyte-secreted lncRNA-H19 promotes fibrosis by activating HSCs [68]. Under cholestatic conditions, cholangiocyte-derived lncRNA-H19 can also activate M1 polarisation of Kupffer cells [69]. These findings collectively demonstrate the multifaceted regulatory functions of cholangiocyte-secreted lncRNA-H19 in liver diseases. In another context, the liver-secreted lncRNA MT1DP (metallothionein 1D pseudogene) can reach distant kidneys, where it reinforces the nephrotoxic effects of cadmium exposure [70].
Cancer Cell-Derived RNAkines as Modulators of Tumour Microenvironment
Tumorigenesis extends beyond the proliferation of cancer cells and involves multiple alterations within the tumour microenvironment (TME), which comprises ECs, cancer-associated fibroblasts (CAFs), and immune cells [71]. RNAkines, encompassing miRNAs, circRNAs, and lncRNAs, have emerged as modulators in the reciprocal interactions between cancer cells and nonmalignant host cells within the TME. These RNAkines govern critical processes, including angiogenesis, immune evasion, metastasis, and metabolic reprogramming [72]. Beyond their local effects, cancer cell-secreted RNAkines can also impact distant organs, such as skeletal muscle [73] and pancreas [74], orchestrating systemic metabolic adaptations to support tumour growth (as illustrated in Figure 2 and summarised in Table 2).
Figure 2. RNAkine-mediated crosstalk between malignant cancer cells and nonmalignant cells within the TME.
Cancer cell-secreted RNAkines play crucial roles in various processes of tumorigenesis, including angiogenesis, migration, metastasis, immune envision and metabolic reprogramming. 1) RNAkines regulate EC migration, promoting angiogenesis; 2-3) RNAkines facilitate the formation of a metastatic niche in distant tissues. CAFs can also secrete RNAkines, contributing to the migration and progression of tumour cells, 4) RNAkines induce immune evasion by expanding Tregs and M2 polarisation; 5-6) RNAkines initiate metabolic reprogramming in both adjacent cells and distant organs. This figure highlights the complex RNAkine-mediated interplay between cancer cells and cells within the TME.
Table 2. Pathological RNAkines mediating the crosstalk between cancer cells and cells within the TME.
RNAkine | Stimuli | Donor | Recipient | Target | Effect | Mechanism | Ref. |
---|---|---|---|---|---|---|---|
miR-210 | TIMP-1 | LUAD | ECs (HUVECs) | FGFRL1, E2F3, VMP-1, RAD52, SDHD | Promoting angiogenesis | Activating PI3K/AKT signalling pathway. | [76] |
miR-9 | MYC OCT4 | Glioma, medulloblastoma | ECs | S1P1, COL18A, THBS2, PTCH1, PHD3 | Promoting angiogenesis and inflammation | [77, 78] | |
miR-25-3p | - | CRC | ECs | KLF2, KLF4 | Promoting angiogenesis | Up-regulating the expression of VEGFR2, ZO-1, occluding and Caludin 5 | [79] |
miR-205 | - | OvCa | ECs | PTEN | Promoting angiogenesis | Targeting PTEN-AKT pathway | [80] |
miR-150 | AGE | Monocytes | ECs Macrophage | c-Myb | Promoting angiogenesis | Regulating c-Myc in ECs or promoting VEGF secretion from M2. | [9, 81, 82] |
miR-1247-3p | - | HCC | Fibroblast | B4GALT3 | Promoting metastasis | Converting normal fibroblasts to cancer-associated fibroblasts by activating β1-integrin-NF-κB signalling | [85] |
lncRNA LNMAT2 | - | BCa | HLEC | PROX1 | Lymphatic metastasis | Epigenetically upregulating PROX1 expression. | [86] |
miR-92a-3p | - | HCC (Huh7 with selective pressure) | Epithelial cells | PTEN | Promoting EMT | Regulating Akt/Snail signalling. | [84] |
miR-214 | - | Various types of Cancer cells | T cells | PTEN | Treg expansion | Increasing population of CD4+CD25highFoxp3+ regulatory T cells (Treg) and mediating immune evasion. | [88] |
circGSE1 | - | HCC | T cells | miR-324-5p | Treg expansion | Silencing miR-324-5p/TGFBR1/Smad3 axis. | [89] |
miR-25-3p miR-130b-3p miR-425-5p | CXCL12/CXCR4 | CRC | Macrophage | PTEN | M2 polarisation | Activating PI3K/AKT signalling pathway. | [92] |
miR-21-3p miR-125b-5p miR-181d-5p | Hypoxia | EOC | Macrophage | SOCS4 SOCS5 | M2 polarisation | Increasing the phosphorylation of STAT3 | [94] |
miR-310a-3p | Hypoxia | PC | Macrophage | PTEN | M2 polarisation | Activating PTEN/PI3Kγ signalling pathway and promoting the secretion of Arginase, IL-10 and TGF-β | [94] |
Circ0048117 | Hypoxia | ESCC | Macrophage | miR-140 | M2 polarisation | Activating TLR4 pathway and promoting the secretion of Arg1, IL-10 and TGF-β | [96] |
miR-23a-3p | - | HCC | Macrophage | PD-L1 | Inhibiting T cell function | Upregulating PD-L1 in macrophage | [90] |
circRNA_002178 | - | LUAD | T cells | miR-34 | Inhibiting T cell function | Inducing PD-L1 expression via sponging miR-34 | [91] |
miR-122 | - | BC | Stomal fibroblast | PKM | Metabolic reprogramming | Suppressing glucose uptake of non-tumour cells in premetastatic niche to facilitate disease progression | [97] |
miR-105 | MYC | BC | CAF | MYC | Metabolic reprogramming | Enhancing glucose and glutamine metabolism (nutrient replete) or converting metabolic waste into energy-rich metabolites (nutrient deprived) | [98] |
miR-122 | - | BC | Skeletal muscle | O-GlcNAc transferase | Promoting skeletal muscle proteolysis | [73] | |
miR-122 | - | BC | Pancreas | PKM | Systematic glucose reprogramming | Impairing insulin secretion and systemic glucose homeostasis. | [74] |
TIMP-1: Tissue inhibitor of metalloproteinases-1, EC: endothelial cells, HLEC, human lymphatic endothelial cells, LUAD: Lung adenocarcinoma, CRC: Colorectal cancer, OvCa: ovarian cancer, HCC: hepatocellular carcinoma, miR-23a-3p: Epithelial ovarian cancer, PC: pancreatic cancer, ESCC: esophageal cell squamous carcinoma, BCa : bladder cancer, BC: Breast cancer.
Evidence suggests that cancer cell-secreted RNAkines regulate EC function to favour angiogenesis across various cancers. Factors associated with tumorigenesis, such as hypoxia [75], tissue inhibitor of metalloproteinases-1 (TIMP-1) [76] and MYC [77, 78], can modulate the release of proangiogenic RNAkines. These RNAkines can enhance angiogenesis by upregulating proangiogenic factors. For example, colorectal cancer (CRC)-derived miR-25-3p upregulates vascular endothelial growth factor receptor 2 (VEGFR2), ZO-1, occludin, and claudin 5 by targeting KLF2 and KLF4 [79]. LncRNA-H19 from CRC competes with miR-138 for its target, the HIF-1α, activating the HIF-1α/VEGF axis. Additionally, ovarian cancer cell-secreted miR-205 augments angiogenesis by regulating the PTEN-AKT pathway [80]. It is not only cancer cells that contribute to angiogenesis by releasing RNAkines. Monocytes secrete miR-150 to enhance EC migration, tube formation, and angiogenesis [9, 81] by triggering VEGF secretion [82].
Tumour metastasis is a formidable challenge in cancer therapy and is often a leading cause of mortality. Cancer cells release RNAkines to create a conducive environment for metastasis [83]. Cancer cells with low metastatic potential can acquire an enhanced metastatic capacity by assimilating RNAkines released from highly metastatic cancer cells [84]. Highly metastatic hepatocellular carcinoma (HCC) cells secrete miR-1247-3p, which transforms normal fibroblasts into CAFs by activating the β1-integrin-NF-κB signalling pathway, creating a supportive niche for tumour metastasis [85]. Cancer cell-secreted RNAkines also contribute to metastasis by regulating the epithelial-mesenchymal-transition (EMT) process, exemplified by miR-92a-3p released from highly metastatic cancer cells [84]. Furthermore, RNAkines mediate the crosstalk between cancer cells and ECs to facilitate metastasis. LNMAT2, a lncRNA selectively loaded into exosomes via its interaction with hnRNPA2B1, can be internalised by human lymphatic endothelial cells (HLECs), epigenetically upregulating prospero homeobox 1 (PROX1) expression to enhance lymphatic metastasis [86]. Another example involves circRNA-100,338 secreted from HCC, which regulates the EC function and angiogenesis, ultimately enhancing metastasis [87].
Cancer cells employ various strategies to manipulate the local immune response within the TME, and RNAkines play crucial roles in these processes. A characteristic feature of tumour immune evasion is the expanded population of CD4+CD25highFoxp3+ regulatory T cells (Tregs). MiR-214, ubiquitously secreted by various types of human cancers and mouse tumour models, promotes Treg expansion by suppressing phosphatase and tensin homologue (PTEN) in recipient T cells [88]. Similarly, circGSE1 derived from HCC induces Treg expansion by regulating the miR-324-5p/TGFBR1/Smad3 axis, leading to a reduced immune response [89]. RNAkines also impact immune checkpoint molecules. Endoplasmic reticulum (ER) stress in HCC cells triggers the secretion of miR-23a-3p, which upregulates the expression of programmed death-ligand 1 (PD-L1) in macrophages [90]. Tumour-derived circRNA-002178 promotes the expression of programmed cell death protein 1 (PD-1) by sponging miR-28-5p in T cells, inducing immune evasion [91]. Cancer cell-secreted RNAkines govern tumour-associated-macrophage (TAM) polarisation (M2) within the TME by regulating PTEN. These RNAkines include miR-25-3p, miR-425-5p, and miR-130-3p [92], and their upregulation can be triggered by activating the CXCL12/CXCR4 axis in CRC cells [92]. M2 polarisation significantly contributes to tumour progression, aiding in tumour initiation, angiogenesis, and metastasis [93]. Hypoxia also plays a role in M2 polarisation through cancer cell-secreted RNAkines, including miR-21-3p [94], miR-125b-5p [94], miR-181d-5p [94], miR-301a-3p [95] and circ0048117 [96].
Metabolic reprogramming is another hallmark of cancer, with RNAkines participating in metabolic adaptation within the TME and distant organs. For instance, breast cancer (BC) cell-secreted miR-122 inhibits the glycolytic enzyme pyruvate kinase, suppressing glucose uptake by non-tumour cells in the pre-metastatic niche [97]. BC cell-secreted miR-105 reprograms the glucose and glutamine metabolism of CAFs, fuelling neighbouring cancer cells [98]. Interestingly, reprogrammed CAFs can convert metabolic waste into energy-rich metabolites [98]. BC cell-secreted miR-122 also has systemic effects on metabolism via its induction of proteolysis in skeletal muscle [73] and disruption of insulin secretion from the pancreas [74]. These insights partially elucidate the mechanisms underlying metabolic syndrome during cancer. RNAkines secreted from nonmalignant cells within the TME also promote glucose reprogramming in cancer cells. For example, TAM-secreted lncRNAs can regulate aerobic glycolysis in BC cells by stabilising HIF-1α [99].
The current evidence suggests that RNAkines form a complex intercellular signalling network. Broadly speaking, RNAkines operate in concert with conventional secreted factors, potentially regulating biological processes to varying degrees. Conventional secreted factors often work for the central control of the whole-body homeostasis. For example, leptin adjusts energy stores in response to overall energy balance of the body [100]. During tumorigenesis, host-derived cytokines can systematically regulate the immune system to suppress tumour formation [101]. In contrast, RNAkines typically work for the functionality of local tissues where they are produced or taken up. Examples include pancreatic islet-secreted RNAkine that regulates insulin sensitivity [17], BAT-secreted RNAkine that recruits fuel for thermogenesis [39], and cancer cell-derived RNAkines that promote tumorigenesis. This suggests a hierarchical interplay between RNAkines and conventional secreted factors within organisms. More efforts are necessary to fully elucidate the nuanced and distinct roles by conventional secreted factors and RNAkines.
Challenges in RNAkine Research
Research into RNAkines has revealed their signalling properties, but scientists continue to face significant challenges. First, there is a lack of comprehensive documentation regarding the concentrations of RNAkines in circulation and in recipient cells, which adds uncertainty to their causative effects in recipient cells. However, it is noteworthy that some RNAkines have been reported to reach concentrations in the picomolar range (10-12 M) in the bloodstream, comparable to those of cytokines. For example, miR-16, a widely expressed RNAkine, maintains an approximate concentration of 2.6 pM in the human bloodstream [102]. miR-378a-3p, which is significantly increased in response to cold exposure, can reach a concentration of 0.1 nM in the bloodstream [39]. The concentration of RNAkines in recipient cells is also a significant concern. Some studies offer exact functional concentrations of RNAkines in recipient cells. For example, miR-378a-3p is increased from ∼102 fmol/g liver tissue to ∼182 fmol/g liver tissue upon cold exposure [39]. When converted to copies per liver cell, the miR-378a-3p level reaches approximately 300 copies per liver cell (157 million cells per g of mouse liver [103]), reaching the reported functional miRNA levels (> 100 copies per cell [104]). In the future, establishing a common standard for assessing RNAkines in circulation is necessary to address this challenge.
Second, the mechanisms controlling stimuli-induced RNAkine secretion remain largely unknown. Various stimuli can induce the secretion of RNAkine via different mechanisms, including 1) regulating RNAkine transcription by targeting specific TFs [77, 78, 98], 2) modulating the secretory machinery involved in EV biogenesis, such as ceramide [105], and 3) altering the expression of specific RBPs that sort RNAkines into the extracellular space, such as hnRNPA2B1 [106]. A deeper understanding of these mechanisms can enhance our appreciation of the physiological and pathological significance of RNAkines.
Why Is New Terminology Needed for RNAs Outside the Cell?
The introduction of a new term, namely “RNAkine”, to describe a diverse class of ncRNAs actively secreted by cells in response to homeostatic changes and their role as mediators in intercellular communications, homeostasis and pathological processes addresses a critical need within the field of RNA research. Alternative terms, such as “EV-RNAs/exosomal RNAs” [107], highlighting their spatial localisation, have been proposed,. The term “secreted RNAs” refers to RNA that functions exclusively outside the cells that produce them [9]. While these terms have their merits, they fall short of capturing the unique characteristics of extracellular ncRNA. These characteristics include their selective release and potent signalling properties, which are essential to be considered bona fide factors of intercellular communications. Additionally, the field anticipates the discovery of RNAkines with other associations or biological sources, such as exogenous ncRNAs produced by other species, that may not fit neatly into existing categories.
The rationale behind coining the term “RNAkine” lies in the shared biological roles with conventional secreted factors known for their significant signalling capabilities. As noted above, RNAkines are distinct in their selective production and secretion in response to specific stimuli. They are crucial in conveying information between cells and organs, initiating cellular responses in recipient cells, and impacting gene expression, cell growth, immune responses, and metabolism. The suffix “-kine”, originating from the Greek word for “movement” or “motion”, aptly reflects the ability of the ncRNA to act at a distance. Thus, “RNAkine” effectively leverages this suffix to denote this unique characteristic.
More importantly, the term “RNAkine” helps differentiate functional extracellular ncRNAs from conventional secreted factors in several ways. First, the fact that extracellular ncRNAs are nucleic acids represents a significant qualitative distinction from cytokines or hormones, justifying the need for specialised nomenclature. Second, “RNAkine” utilises an alternative method for transferring information. They can simultaneously target different components of biological networks, leading to coordinated and integrated responses similar to the robust synergy seen in hormonal secretory patterns within complex biological processes. Third, “RNAkine” emphasises their different but co-dependent roles with conventional secreted factors in contributing to specific physiological processes or diseases. Finally, the term “RNAkine” encompasses various types of RNA molecules, creating a self-contained category that distinguishes them from non-functional extracellular RNAs or intracellular RNAs. (Figure 3). Thus, “RNAkine” provides a concise, comprehensive and precise representation of this emerging class of extracellular ncRNAs. This nomenclature fosters consensus and holds the potential for widespread acceptance within the scientific and clinical communities. Consequently, it assists scientists and clinicians in effectively exploring and discussing the roles played by these recently identified secreted factors in various critical biological processes.
Figure 3. Classifications of secreted factors.
In addition to conventional secreted factors, cells also secrete ncRNA molecules (RNAkines). The molecular types of secreted factors are presented in bold, and representative examples or subfamilies, such as chemokines, are listed. This figure provides an overview of the diverse types of secreted factors contributing to intercellular signalling. This figure was created using BioRender (https://biorender.com/)
Concluding Remarks and Future Perspectives
In summary, exploring RNAkines has unveiled their previously unrecognised roles in various biological processes, from metabolic homeostasis, exercise-mediated benefits, insulin resistance, and tumorigenesis. These discoveries highlight the potential of RNAkines as promising therapeutic targets for many diseases due to their substantial involvement in these critical biological pathways.
Despite substantial progress in comprehending RNAkines, numerous challenges and avenues for future research remain (see Outstanding Questions). It is imperative to pinpoint organ- and cell-specific RNAkines and unravel the intricate mechanisms governing their selective packaging, secretion and functional roles. While evidence suggests that the uptake of RNAkines by recipient organs or cells is a selective process, the mechanisms underpinning this phenomenon, particularly in cases where miRNAs function endocrinologically, remain predominantly uncharted. Consequently, we eagerly await further studies and well-designed investigations to illuminate the aetiopathogenic mechanism mediated by RNAkines. The field of RNAkine research harbours immense potential for advancing our comprehension of intercellular communication and its repercussions on various physiological and pathological processes.
Acknowledgements
This work was supported by the CAMS Innovation Fund for Medical Sciences (No. CIFMS-2021-12M-5-015).
Footnotes
Declaration of Interests:
The authors declare no competing interests.
References
- 1.Skog J, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–1476. doi: 10.1038/ncb1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Valadi H, et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659. doi: 10.1038/ncb1596. [DOI] [PubMed] [Google Scholar]
- 3.Chen X, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18:997–1006. doi: 10.1038/cr.2008.282. [DOI] [PubMed] [Google Scholar]
- 4.Lawrie CH, et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Brit J Haematol. 2008;141:672–675. doi: 10.1111/j.1365-2141.2008.07077.x. [DOI] [PubMed] [Google Scholar]
- 5.Mitchell PS, et al. Circulating microRNAs as stable blood-based markers for cancer detection. P Natl Acad Sci USA. 2008;105:10513–10518. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.O’Brien K, et al. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Bio. 2020;21:585–606. doi: 10.1038/s41580-020-0251-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Xu YX, et al. Exosomal ncRNAs: Novel therapeutic target and biomarker for diabetic complications. Pharmacological researchxs. 2022;178 doi: 10.1016/j.phrs.2022.106135. [DOI] [PubMed] [Google Scholar]
- 8.Zeuschner P, et al. Non-coding RNAs as biomarkers in liquid biopsies with a special emphasis on extracellular vesicles in urological malignancies. Expert review of molecular diagnostics. 2020;20:151–167. doi: 10.1080/14737159.2019.1665998. [DOI] [PubMed] [Google Scholar]
- 9.Zhang Y, et al. Secreted monocytic miR-150 enhances targeted endothelial cell migration. Mol Cell. 2010;39:133–144. doi: 10.1016/j.molcel.2010.06.010. [DOI] [PubMed] [Google Scholar]
- 10.van Niel G, et al. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat Rev Mol Cell Biol. 2022;23:369–382. doi: 10.1038/s41580-022-00460-3. [DOI] [PubMed] [Google Scholar]
- 11.Vickers KC, et al. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nature Cell Biology. 2011;13:423–U182. doi: 10.1038/ncb2210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Arroyo JD, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. P Natl Acad Sci USA. 2011;108:5003–5008. doi: 10.1073/pnas.1019055108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Garcia-Martin R, et al. MicroRNA sequence codes for small extracellular vesicle release and cellular retention. Nature. 2022;601:1264. doi: 10.1038/s41586-021-04234-3. + [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Li J, et al. Pancreatic beta cells control glucose homeostasis via the secretion of exosomal miR-29 family. J Extracell Vesicles. 2021;10:e12055. doi: 10.1002/jev2.12055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Muller G, et al. Microvesicles released from rat adipocytes and harboring glycosylphosphatidylinositol-anchored proteins transfer RNA stimulating lipid synthesis. Cell Signal. 2011;23:1207–1223. doi: 10.1016/j.cellsig.2011.03.013. [DOI] [PubMed] [Google Scholar]
- 16.Ying W, et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell. 2017;171:372–384.:e312. doi: 10.1016/j.cell.2017.08.035. [DOI] [PubMed] [Google Scholar]
- 17.Xu H, et al. Pancreatic beta cell microRNA-26a alleviates type 2 diabetes by improving peripheral insulin sensitivity and preserving beta cell function. PLoS biology. 2020;18:e3000603. doi: 10.1371/journal.pbio.3000603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yin Z, et al. Macrophage-derived exosomal microRNA-501-3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3-mediated TGF-beta signaling pathway. J Exp Clin Cancer Res. 2019;38:310. doi: 10.1186/s13046-019-1313-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Aucher A, et al. MicroRNAs transfer from human macrophages to hepato-carcinoma cells and inhibit proliferation. J Immunol. 2013;191:6250–6260. doi: 10.4049/jimmunol.1301728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Sun Y, et al. Expression of miRNA-29 in Pancreatic beta Cells Promotes Inflammation and Diabetes via TRAF3. Cell Rep. 2021;34:108576. doi: 10.1016/j.celrep.2020.108576. [DOI] [PubMed] [Google Scholar]
- 21.Thomou T, et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature. 2017;542:1264. doi: 10.1038/nature21365. + [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wang J, et al. Extracellular vesicles mediate the communication of adipose tissue with brain and promote cognitive impairment associated with insulin resistance. Cell metabolism. 2022;34:1264. doi: 10.1016/j.cmet.2022.08.004. + [DOI] [PubMed] [Google Scholar]
- 23.Misir S, et al. Specific expression and functions of circular RNAs. Cell Death Differ. 2022;29:481–491. doi: 10.1038/s41418-022-00948-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bridges MC, et al. LNCcation: lncRNA localization and function. J Cell Biol. 2021;220 doi: 10.1083/jcb.202009045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Xu Z, et al. Role of exosomal non-coding RNAs from tumor cells and tumor-associated macrophages in the tumor microenvironment. Mol Ther. 2022;30:3133–3154. doi: 10.1016/j.ymthe.2022.01.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wang Y, et al. Injured liver-released miRNA-122 elicits acute pulmonary inflammation via activating alveolar macrophage TLR7 signaling pathway. Proc Natl Acad Sci U S A. 2019;116:6162–6171. doi: 10.1073/pnas.1814139116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Fabbri M, et al. MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. P Natl Acad Sci USA. 2012;109:E2110–E2116. doi: 10.1073/pnas.1209414109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhang AM, et al. Islet beta cell: An endocrine cell secreting miRNAs. Biochem Bioph Res Co. 2018;495:1648–1654. doi: 10.1016/j.bbrc.2017.12.028. [DOI] [PubMed] [Google Scholar]
- 29.He A, et al. Overexpression of micro ribonucleic acid 29, highly up-regulated in diabetic rats, leads to insulin resistance in 3T3-L1 adipocytes. Molecular endocrinology. 2007;21:2785–2794. doi: 10.1210/me.2007-0167. [DOI] [PubMed] [Google Scholar]
- 30.Massart J, et al. Altered miR-29 Expression in Type 2 Diabetes Influences Glucose and Lipid Metabolism in Skeletal Muscle. Diabetes. 2017;66:1807–1818. doi: 10.2337/db17-0141. [DOI] [PubMed] [Google Scholar]
- 31.Lee MW, et al. Adipose Tissue-Derived Signatures for Obesity and Type 2 Diabetes: Adipokines, Batokines and MicroRNAs. J Clin Med. 2019;8 doi: 10.3390/jcm8060854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yu Y, et al. Adipocyte-Derived Exosomal MiR-27a Induces Insulin Resistance in Skeletal Muscle Through Repression of PPARgamma. Theranostics. 2018;8:2171–2188. doi: 10.7150/thno.22565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Li D, et al. Gonadal white adipose tissue-derived exosomal MiR-222 promotes obesity-associated insulin resistance. Aging. 2020;12:22719–22743. doi: 10.18632/aging.103891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wei MY, et al. Visceral Adipose Tissue Derived Exosomes Exacerbate Colitis Severity via Pro-inflammatory MiRNAs in High Fat Diet Fed Mice. ACS nano. 2020;14:5099–5110. doi: 10.1021/acsnano.0c01860. [DOI] [PubMed] [Google Scholar]
- 35.Pan Y, et al. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. Journal of Clinical Investigation. 2019;129:834–849. doi: 10.1172/JCI123069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yang FT, Stanford KI. Batokines: Mediators of Inter-Tissue Communication (a Mini-Review) Curr Obes Rep. 2022;11:1–9. doi: 10.1007/s13679-021-00465-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Qing H, et al. Origin and Function of Stress-Induced IL-6 in Murine Models. Cell. 2020;182:372. doi: 10.1016/j.cell.2020.05.054. + [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Egecioglu E, et al. Interleukin-6 is important for regulation of core body temperature during long-term cold exposure in mice. Biomedical Reports. 2018;9:206–212. doi: 10.3892/br.2018.1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Xu J, et al. Cold-activated brown fat-derived extracellular vesicle-miR-378a-3p stimulates hepatic gluconeogenesis in male mice. Nat Commun. 2023;14:5480. doi: 10.1038/s41467-023-41160-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Liu T, et al. Adipose tissue macrophage-derived exosomal miR-29a regulates obesity-associated insulin resistance. Biochem Biophys Res Commun. 2019;515:352–358. doi: 10.1016/j.bbrc.2019.05.113. [DOI] [PubMed] [Google Scholar]
- 41.Ying W, et al. MiR-690, an exosomal-derived miRNA from M2-polarized macrophages, improves insulin sensitivity in obese mice. Cell metabolism. 2021;33:781–790.:e785. doi: 10.1016/j.cmet.2020.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kjaer M, et al. Hormonal and metabolic responses to electrically induced cycling during epidural anesthesia in humans. J Appl Physiol (1985) 1996;80:2156–2162. doi: 10.1152/jappl.1996.80.6.2156. [DOI] [PubMed] [Google Scholar]
- 43.Pedersen BK, et al. Searching for the exercise factor: is IL-6 a candidate? J Muscle Res Cell Motil. 2003;24:113–119. doi: 10.1023/a:1026070911202. [DOI] [PubMed] [Google Scholar]
- 44.Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nature reviews Endocrinology. 2012;8:457–465. doi: 10.1038/nrendo.2012.49. [DOI] [PubMed] [Google Scholar]
- 45.Estebanez B, et al. Effects of exercise on exosome release and cargo in in vivo and ex vivo models: A systematic review. J Cell Physiol. 2021;236:3336–3353. doi: 10.1002/jcp.30094. [DOI] [PubMed] [Google Scholar]
- 46.Hou ZX, et al. Longterm Exercise-Derived Exosomal miR-342-5p A Novel Exerkine for Cardioprotection. Circulation Research. 2019;124:1386–1400. doi: 10.1161/CIRCRESAHA.118.314635. [DOI] [PubMed] [Google Scholar]
- 47.Yin X, et al. Time-Course Responses of Muscle-Specific MicroRNAs Following Acute Uphill or Downhill Exercise in Sprague-Dawley Rats. Frontiers in Physiology. 2019;10 doi: 10.3389/fphys.2019.01275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Guescini M, et al. Muscle Releases Alpha-Sarcoglycan Positive Extracellular Vesicles Carrying miRNAs in the Bloodstream. Plos One. 2015;10 doi: 10.1371/journal.pone.0125094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.D’Souza RF, et al. Circulatory exosomal miRNA following intense exercise is unrelated to muscle and plasma miRNA abundances. Am J Physiol-Endoc M. 2018;315:E723–E733. doi: 10.1152/ajpendo.00138.2018. [DOI] [PubMed] [Google Scholar]
- 50.Forterre A, et al. Myotube-derived exosomal miRNAs downregulate Sirtuin1 in myoblasts during muscle cell differentiation. Cell Cycle. 2014;13:78–89. doi: 10.4161/cc.26808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Oliveira GP, Jr, et al. Effects of Acute Aerobic Exercise on Rats Serum Extracellular Vesicles Diameter, Concentration and Small RNAs Content. Front Physiol. 2018;9:532. doi: 10.3389/fphys.2018.00532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Wang H, Wang B. Extracellular vesicle microRNAs mediate skeletal muscle myogenesis and disease. Biomed Rep. 2016;5:296–300. doi: 10.3892/br.2016.725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Castano C, et al. Delivery of muscle-derived exosomal miRNAs induced by HIIT improves insulin sensitivity through down-regulation of hepatic FoxO1 in mice. Proc Natl Acad Sci U S A. 2020;117:30335–30343. doi: 10.1073/pnas.2016112117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yang CP, et al. Muscle atrophy-related myotube-derived exosomal microRNA in neuronal dysfunction: Targeting both coding and long noncoding RNAs. Aging Cell. 2020;19:e13107. doi: 10.1111/acel.13107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Nie Y, et al. Skeletal muscle-derived exosomes regulate endothelial cell functions via reactive oxygen species-activated nuclear factor-kappaB signalling. Exp Physiol. 2019;104:1262–1273. doi: 10.1113/EP087396. [DOI] [PubMed] [Google Scholar]
- 56.Lee YS, et al. Exosomes derived from palmitic acid-treated hepatocytes induce fibrotic activation of hepatic stellate cells. Sci Rep. 2017;7:3710. doi: 10.1038/s41598-017-03389-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wang W, et al. Exosomes secreted by palmitic acid-treated hepatocytes promote LX-2 cell activation by transferring miRNA-107. Cell Death Discov. 2021;7:174. doi: 10.1038/s41420-021-00536-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Luo X, et al. Lipotoxic hepatocyte-derived exosomal miR-1297 promotes hepatic stellate cell activation through the PTEN signaling pathway in metabolic-associated fatty liver disease. World journal of gastroenterology. 2021;27:1419–1434. doi: 10.3748/wjg.v27.i14.1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhao Z, et al. Cholesterol impairs hepatocyte lysosomal function causing M1 polarization of macrophages via exosomal miR-122-5p. Exp Cell Res. 2020;387:111738. doi: 10.1016/j.yexcr.2019.111738. [DOI] [PubMed] [Google Scholar]
- 60.Liu XL, et al. Lipotoxic Hepatocyte-Derived Exosomal MicroRNA 192-5p Activates Macrophages Through Rictor/Akt/Forkhead Box Transcription Factor O1 Signaling in Nonalcoholic Fatty Liver Disease. Hepatology. 2020;72:454–469. doi: 10.1002/hep.31050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Momen-Heravi F, et al. Exosomes derived from alcohol-treated hepatocytes horizontally transfer liver specific miRNA-122 and sensitize monocytes to LPS. Sci Rep. 2015;5:9991. doi: 10.1038/srep09991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Fan G, et al. Anti-Inflammatory Activity of Tanshinone IIA in LPS-Stimulated RAW264.7 Macrophages via miRNAs and TLR4-NF-kappaB Pathway. Inflammation. 2016;39:375–384. doi: 10.1007/s10753-015-0259-1. [DOI] [PubMed] [Google Scholar]
- 63.Lou J, et al. Exercise promotes angiogenesis by enhancing endothelial cell fatty acid utilization via liver-derived extracellular vesicle miR-122-5p. J Sport Health Sci. 2021 doi: 10.1016/j.jshs.2021.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ruan YT, et al. Circulating LncRNAs Analysis in Patients with Type 2 Diabetes Reveals Novel Genes Influencing Glucose Metabolism and Islet β-Cell Function. Cellular Physiology and Biochemistry. 2018;46:335–350. doi: 10.1159/000488434. [DOI] [PubMed] [Google Scholar]
- 65.Song YB, et al. SNHG9, delivered by adipocyte-derived exosomes, alleviates inflammation and apoptosis of endothelial cells through suppressing TRADD expression. European Journal of Pharmacology. 2020;872 doi: 10.1016/j.ejphar.2020.172977. [DOI] [PubMed] [Google Scholar]
- 66.Li QF, et al. Adipose Tissue Exosome circ_sxc Mediates the Modulatory of Adiposomes on Brain Aging by Inhibiting Brain dme-miR-87-3p. Mol Neurobiol. 2023 doi: 10.1007/s12035-023-03516-3. [DOI] [PubMed] [Google Scholar]
- 67.Li XJY, et al. Cholangiocyte-derived exosomal long noncoding RNA H19 promotes cholestatic liver injury in mouse and humans. Hepatology. 2018;68:599–615. doi: 10.1002/hep.29838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Liu RP, et al. Cholangiocyte-Derived Exosomal Long Noncoding RNA H19 Promotes Hepatic Stellate Cell Activation and Cholestatic Liver Fibrosis. Hepatology. 2019;70:1317–1335. doi: 10.1002/hep.30662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Li XJY, et al. Cholangiocyte-Derived Exosomal lncRNA H19 Promotes Macrophage Activation and Hepatic Inflammation under Cholestatic Conditions. Cells. 2020;9 doi: 10.3390/cells9010190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gao M, et al. Liver-derived exosome-laden lncRNA MT1DP aggravates cadmium-induced nephrotoxicity. Environ Pollut. 2020;258 doi: 10.1016/j.envpol.2019.113717. [DOI] [PubMed] [Google Scholar]
- 71.Hinshaw DC, Shevde LA. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer research. 2019;79:4557–4566. doi: 10.1158/0008-5472.CAN-18-3962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nature medicine. 2013;19:1423–1437. doi: 10.1038/nm.3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Yan W, et al. Cancer-cell-secreted miR-122 suppresses O-GlcNAcylation to promote skeletal muscle proteolysis. Nat Cell Biol. 2022;24:793–804. doi: 10.1038/s41556-022-00893-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cao M, et al. Cancer-cell-secreted extracellular vesicles suppress insulin secretion through miR-122 to impair systemic glucose homeostasis and contribute to tumour growth. Nat Cell Biol. 2022 doi: 10.1038/s41556-022-00919-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bister N, et al. Hypoxia and extracellular vesicles: A review on methods, vesicular cargo and functions. J Extracell Vesicles. 2020;10:e12002. doi: 10.1002/jev2.12002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Cui H, et al. Tissue inhibitor of metalloproteinases-1 induces a pro-tumourigenic increase of miR-210 in lung adenocarcinoma cells and their exosomes. Oncogene. 2015;34:3640–3650. doi: 10.1038/onc.2014.300. [DOI] [PubMed] [Google Scholar]
- 77.Chen X, et al. MiR-9 promotes tumorigenesis and angiogenesis and is activated by MYC and OCT4 in human glioma. J Exp Clin Cancer Res. 2019;38:99. doi: 10.1186/s13046-019-1078-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Yao X, et al. MiR-9 Promotes Angiogenesis via Targeting on Sphingosine-1-Phosphate Receptor 1. Frontiers in cell and developmental biology. 2020;8:755. doi: 10.3389/fcell.2020.00755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zeng Z, et al. Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat Commun. 2018;9:5395. doi: 10.1038/s41467-018-07810-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.He L, et al. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics. 2019;9:8206–8220. doi: 10.7150/thno.37455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Li J, et al. Microvesicle-mediated transfer of microRNA-150 from monocytes to endothelial cells promotes angiogenesis. J Biol Chem. 2013;288:23586–23596. doi: 10.1074/jbc.M113.489302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Liu Y, et al. Microvesicle-delivery miR-150 promotes tumorigenesis by up-regulating VEGF, and the neutralization of miR-150 attenuate tumor development. Protein Cell. 2013;4:932–941. doi: 10.1007/s13238-013-3092-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Steeg PS. Targeting metastasis. Nature reviews Cancer. 2016;16:201–218. doi: 10.1038/nrc.2016.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Yang B, et al. High-metastatic cancer cells derived exosomal miR92a-3p promotes epithelial-mesenchymal transition and metastasis of low-metastatic cancer cells by regulating PTEN/Akt pathway in hepatocellular carcinoma. Oncogene. 2020;39:6529–6543. doi: 10.1038/s41388-020-01450-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Fang T, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018;9:191. doi: 10.1038/s41467-017-02583-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Chen CH, et al. Exosomal long noncoding RNA promotes lymphatic metastasis in bladder cancer. Journal of Clinical Investigation. 2020;130:404–421. doi: 10.1172/JCI130892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Huang XY, et al. Exosomal circRNA-100338 promotes hepatocellular carcinoma metastasis via enhancing invasiveness and angiogenesis. J Exp Clin Canc Res. 2020;39 doi: 10.1186/s13046-020-1529-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Yin Y, et al. Tumor-secreted miR-214 induces regulatory T cells: a major link between immune evasion and tumor growth. Cell Res. 2014;24:1164–1180. doi: 10.1038/cr.2014.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Huang MY, et al. Exosomal circGSE1 promotes immune escape of hepatocellular carcinoma by inducing the expansion of regulatory T cells. Cancer Science. 2022;113:1968–1983. doi: 10.1111/cas.15365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Liu J, et al. Endoplasmic Reticulum Stress Causes Liver Cancer Cells to Release Exosomal miR-23a-3p and Up-regulate Programmed Death Ligand 1 Expression in Macrophages. Hepatology. 2019;70:241–258. doi: 10.1002/hep.30607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang JF, et al. circRNA-002178 act as a ceRNA to promote PDL1/PD1 expression in lung adenocarcinoma. Cell Death & Disease. 2020;11 doi: 10.1038/s41419-020-2230-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Wang D, et al. Exosome-encapsulated miRNAs contribute to CXCL12/CXCR4-induced liver metastasis of colorectal cancer by enhancing M2 polarization of macrophages. Cancer letters. 2020;474:36–52. doi: 10.1016/j.canlet.2020.01.005. [DOI] [PubMed] [Google Scholar]
- 93.Boutilier AJ, Elsawa SF. Macrophage Polarization States in the Tumor Microenvironment. Int J Mol Sci. 2021;22 doi: 10.3390/ijms22136995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chen X, et al. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer letters. 2018;435:80–91. doi: 10.1016/j.canlet.2018.08.001. [DOI] [PubMed] [Google Scholar]
- 95.Wang X, et al. Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kgamma to Promote Pancreatic Cancer Metastasis. Cancer research. 2018;78:4586–4598. doi: 10.1158/0008-5472.CAN-17-3841. [DOI] [PubMed] [Google Scholar]
- 96.Lu QJ, et al. Hypoxic Tumor-Derived Exosomal Circ0048117 Facilitates M2 Macrophage Polarization Acting as miR-140 Sponge in Esophageal Squamous Cell Carcinoma. Oncotargets Ther. 2020;13:11883–11897. doi: 10.2147/OTT.S284192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Fong MY, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 2015;17:183–194. doi: 10.1038/ncb3094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Yan W, et al. Cancer-cell-secreted exosomal miR-105 promotes tumour growth through the MYC-dependent metabolic reprogramming of stromal cells. Nat Cell Biol. 2018;20:597–609. doi: 10.1038/s41556-018-0083-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Chen F, et al. Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nature Cell Biology. 2019;21:498. doi: 10.1038/s41556-019-0299-0. + [DOI] [PubMed] [Google Scholar]
- 100.Myers MG, Jr, et al. Obesity and leptin resistance: distinguishing cause from effect. Trends Endocrinol Metab. 2010;21:643–651. doi: 10.1016/j.tem.2010.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Dranoff G. Cytokines in cancer pathogenesis and cancer therapy. Nature reviews Cancer. 2004;4:11–22. doi: 10.1038/nrc1252. [DOI] [PubMed] [Google Scholar]
- 102.Liang H, et al. Effective detection and quantification of dietetically absorbed plant microRNAs in human plasma. J Nutr Biochem. 2015;26:505–512. doi: 10.1016/j.jnutbio.2014.12.002. [DOI] [PubMed] [Google Scholar]
- 103.Falconer DS, et al. Cell numbers and cell sizes in organs of mice selected for large and small body size. Genet Res. 1978;31:287–301. doi: 10.1017/s0016672300018061. [DOI] [PubMed] [Google Scholar]
- 104.Brown BD, et al. Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state. Nature biotechnology. 2007;25:1457–1467. doi: 10.1038/nbt1372. [DOI] [PubMed] [Google Scholar]
- 105.Kosaka N, et al. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 2010;285:17442–17452. doi: 10.1074/jbc.M110.107821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Fabbiano F, et al. RNA packaging into extracellular vesicles: An orchestra of RNA-binding proteins? Journal of Extracellular Vesicles. 2020;10 doi: 10.1002/jev2.12043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ma HZ, et al. MicroRNA-Mediated Cancer Metastasis Regulation Heterotypic Signals in the Microenvironment. Curr Pharm Biotechno. 2014;15:455–458. doi: 10.2174/1389201015666140516112042. [DOI] [PubMed] [Google Scholar]
- 108.Li C, et al. Roles and mechanisms of exosomal non-coding RNAs in human health and diseases. Signal Transduct Tar. 2021;6 doi: 10.1038/s41392-021-00779-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ji CB, Guo XR. The clinical potential of circulating microRNAs in obesity. Nature Reviews Endocrinology. 2019;15:731–743. doi: 10.1038/s41574-019-0260-0. [DOI] [PubMed] [Google Scholar]
- 110.Liu QQ, et al. Serum exosomal hsa_circRNA_0001842 is the potential biomarker for diagnosing lower limb vascular disease in type 2 diabetes mellitus. J Genet. 2023;102 [PubMed] [Google Scholar]
- 111.Jiang Y, et al. Peripheral blood miRNAs as a biomarker for chronic cardiovascular diseases. Sci Rep-Uk. 2014;4 doi: 10.1038/srep05026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Hu XY, et al. Exosomal circular RNAs: Biogenesis, effect, and application in cardiovascular diseases. Frontiers in cell and developmental biology. 2022;10 doi: 10.3389/fcell.2022.948256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kumar P, et al. Circulating miRNA Biomarkers for Alzheimer’s Disease. Plos One. 2013;8 doi: 10.1371/journal.pone.0069807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Qu HG, et al. Circulating miRNAs: Promising Biomarkers of Human Cancer. Asian Pac J Cancer P. 2011;12:1117–1125. [PubMed] [Google Scholar]
- 115.Jin FF, et al. A novel class of tsRNA signatures as biomarkers for diagnosis and prognosis of pancreatic cancer. Molecular Cancer. 2021;20 doi: 10.1186/s12943-021-01389-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ortega FJ, et al. Targeting the Circulating MicroRNA Signature of Obesity. Clin Chem. 2013;59:781–792. doi: 10.1373/clinchem.2012.195776. [DOI] [PubMed] [Google Scholar]
- 117.Al-Rawaf HA. Circulating microRNAs and adipokines as markers of metabolic syndrome in adolescents with obesity. Clinical nutrition. 2019;38:2231–2238. doi: 10.1016/j.clnu.2018.09.024. [DOI] [PubMed] [Google Scholar]
- 118.Cui XW, et al. Change in circulating microRNA profile of obese children indicates future risk of adult diabetes. Metabolism-Clinical and Experimental. 2018;78:95–105. doi: 10.1016/j.metabol.2017.09.006. [DOI] [PubMed] [Google Scholar]
- 119.Shah R, et al. Extracellular RNAs Are Associated With Insulin Resistance and Metabolic Phenotypes. Diabetes care. 2017;40:546–553. doi: 10.2337/dc16-1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Brandt S, et al. Circulating levels of miR-122 and nonalcoholic fatty liver disease in pre-pubertal obese children. Pediatr Obes. 2018;13:175–182. doi: 10.1111/ijpo.12261. [DOI] [PubMed] [Google Scholar]
- 121.Mirra P, et al. Circulating miRNAs as intercellular messengers, potential biomarkers and therapeutic targets for Type 2 diabetes. Epigenomics-Uk. 2015;7:653–667. doi: 10.2217/epi.15.18. [DOI] [PubMed] [Google Scholar]
- 122.Villarroya-Beltri C, et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nature Communications. 2013;4 doi: 10.1038/ncomms3980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Corrado C, et al. Molecular Mediators of RNA Loading into Extracellular Vesicles. Cells. 2021;10 doi: 10.3390/cells10123355. [DOI] [PMC free article] [PubMed] [Google Scholar]