Circular RNAs in coronary heart disease: From molecular mechanism to promising clinical application (Review)
- Authors:
- Published online on: October 31, 2024 https://doi.org/10.3892/ijmm.2024.5452
- Article Number: 11
-
Copyright: © Fan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Coronary heart disease (CHD), a leading cause of morbidity and mortality globally, represents a significant public health challenge (1). CHD is primarily driven by atherosclerosis, an inflammatory process characterized by the accumulation of lipids, inflammatory cells and fibrous elements within the arterial wall, which leads to the formation of atherosclerotic plaques and further compromises blood flow and oxygen delivery to the myocardium, contributing to severe outcomes such as myocardial infarction and heart failure (2). The underlying mechanisms of CHD are multifactorial, involving dyslipidemia, endothelial dysfunction, oxidative stress and chronic inflammation (3). These factors collectively contribute to the initiation and progression of atherosclerosis, high-lighting the need for comprehensive approaches to understand and manage the disease. Furthermore, the heterogeneity of the disease, coupled with the complex interplay of genetic, environmental and lifestyle factors, complicates the identification of precise diagnostic markers and effective therapeutic targets (4).
Currently, circular RNAs (circRNAs) have emerged as a pivotal class of non-coding RNAs, attracting substantial attention in the field of cardiovascular research (5). Unlike linear RNAs, circRNAs are characterized by their covalently closed loop structure, which confers remarkable stability and resistance to exonuclease-mediated degradation (6). This unique structure not only enables circRNAs to function as microRNA (miRNA) sponges, which contain miRNA binding sites and regulate gene expression by sequestering miRNAs, but also allows them to interact with RNA-binding proteins (RBPs), participating in the regulation of gene expression (7). In CHD, emerging evidence suggests that circRNAs play critical roles in the regulation of vascular function, myocardial injury and inflammation (8-10). Several studies have identified differentially expressed circRNAs in patients with CHD, correlating with disease severity and clinical outcomes (11,12). These findings highlight the potential of circRNAs as non-invasive biomarkers for early diagnosis and prognosis of CHD. Furthermore, specific circRNA has been verified to participate in modulating key signaling pathways associated with atherosclerosis and myocardial angiogenesis, offering insights into novel therapeutic strategies (13,14).
This review aims to provide a comprehensive overview of the current understanding of circRNAs in CHD. It summarizes the molecular mechanisms by which circRNAs influence the pathophysiology of CHD, including their roles in cardiomyocyte death, endothelial injury, vascular dysfunction and inflammation. In addition, it explores the potential clinical applications of circRNAs as biomarkers and therapeutic targets, emphasizing the translational implications of circRNA research in improving patient outcomes.
Biogenesis and functions of circRNAs
CircRNAs are a unique class of non-coding RNAs found in various organisms, including plants and animals, as well as in certain plant and animal viruses, where they are linked to viral replication and survival (15). The stable structure contributes to their accumulation within cells and makes them play long-term regulatory roles (16). Irrespective of the size, a circRNA molecule may possess <100 nuclei and numerous bases, thereby rendering each element of the reverse shearing procedure deterministic (17). In general, circRNAs are comprised of 2-3 exons with a median size frequently exceeding 500 nucleotides, yet not surpassing 700 nucleotides (18). They exhibit a high degree of conservation across different species, with their expression typically being specific to certain cell types, tissues and stages of development (19). Hence, the presence in numerous species, abundance of information, tissue-specific nature and remarkable stability represent key attributes of circRNAs. Given these characteristics, circRNAs have the potential to serve as valuable indicators for specific disorders and novel targets for therapeutic interventions.
Biogenesis
The formation of circRNAs involves specific sequences and secondary structures within the precursor (pre)-mRNA, which facilitate the back-splicing process (20). Intronic complementary sequences and RBPs connect donor-splice and acceptor-splice sites by appropriate pairing of bases or interacting with definite motifs, playing crucial roles in circRNA biogenesis (21,22). RBPs such as Quaking and Muscleblind bind to flanking intronic sequences to bring splice sites into close proximity, promoting the back-splicing event (23,24). The resulting circRNAs can originate from three main sources: Exonic genes (exon-derived circRNA), intronic regions (intron-derived circRNA, such as pre-mRNA and pre-transfer RNA), or combinations of both exons and introns, leading to three primary types: Exonic circRNAs (EcircRNAs), circular intronic RNAs (ciRNAs) and exon-intron circRNAs (EIciRNAs) (25-27). The majority of identified circRNAs are derived from exonic regions, while ciRNAs and EIciRNAs only constitute a small fraction within this category (28).
The generation of endogenous circRNAs is characterized by notable inefficiency, a phenomenon attributed to the elongation process of RNA polymerase II and tightly regulated by cis elements (29,30). Various types of circRNAs are produced through distinct mechanisms (Fig. 1), primarily involving exon cyclization to generate cytoplasmic circRNAs (31). The creation of exon circRNAs predominantly occurs via two mechanisms: Lariat-driven circularization and circularization driven by introns containing reverse complementary sequences (32). Lariat-driven circularization involves the direct linkage of the 5′ and 3′ ends of the pre-mRNA to form a circular structure (33). Furthermore, this process is associated with exon skipping, where certain exons are omitted to facilitate circular structure formation (34). In addition, intron pairing loops facilitate circRNA formation by promoting base complementary pairing between introns (35). Typically, introns excised from the pre-mRNA are debranched and degraded by exonucleases. However, intron RNA formation is sustained by the presence of 7 GU sequences and 11 C sequences at the intron 5′ splice site within a lasso structure, preventing degradation and promoting circularization (36). The generation mechanism of exon-intron RNA mirrors that of EcircRNAs, with the distinction that the circular structure retains the intron component (14,16,37).
Owing to the advancing sophistication of contemporary biological information and RNA sequencing techniques, a substantial volume of transcriptional data has emerged, revealing the presence of circRNAs in various cell types within human organs (38). In the human heart, the percentage of expressed genes capable of generating circRNAs is 9% (17). It is evident that as human organs develop and certain diseases occur, the expression of circRNA undergoes changes (39). Research has demonstrated an elevation in circRNA expression levels in the developing heart (40,41). While the reason for this escalation remains elusive, several possible explanations have so far been proposed. For instance, a study utilizing cardiomyocytes derived from human-induced pluripotent stem cells revealed the dynamic regulation of circRNA expression under chronic and acute stress conditions (42). It should be noted that N-6 methylation (m6A) has the ability to impact circRNA biogenesis and cellular localization. Sites of m6A position in proximity to the start and stop codons of mRNAs can recruit the spliceosome and further facilitate back-splicing and circRNA generation, while specific nuclear reader proteins possess the capability to interact with m6A motifs, aiding in the export of circRNAs from the nucleus to the cytoplasm (43,44). Furthermore, various stimuli like high temperature or oxidative stress also affect circRNA levels (45). Therefore, the biogenesis, structure formation, post-transcriptional modifications and subcellular localization of circRNAs, which are intricately linked to their functional significance within the organism, deserve further investigation.
Despite their stability, circRNAs can still undergo degradation through various pathways, such as RNase L and Argonaute-dependent and independent mechanisms (46). CircRNAs also interact with RBPs, which can influence their stability. For instance, proteins like transcript factor II B-related factor 1 and KH-type splicing regulatory protein, which are regulated by signaling pathways such as PI3K, can mediate mRNA stability and potentially affect circRNA degradation (47). The m6A modifications on circRNAs further alter the interaction of circRNAs with RBPs, thereby influencing their stability and degradation pathways (48). Understanding the degradation pathways of circRNA can inform therapeutic strategies, particularly in diseases where circRNAs are dysregulated, such as CHD. Targeting specific degradation pathways could modulate circRNA levels and their associated biological effects.
Functions
In the last five years, investigations into circRNAs have facilitated a more thorough comprehension of RNA's functionality by researchers. The unique structural features of circRNAs confer specific biological functions upon them. Current research findings indicate that circRNAs exert a significant effect across various developmental phases and pathophysiological states by functioning as miRNA sponges, engaging with RBPs, serving as modulators of transcription or translation, impacting pre-mRNA splicing and contributing to protein translation (Fig. 1) (22,27,49-51).
One of the most well-documented functions of circRNAs is their ability to act as miRNA sponges or competitive endogenous RNAs (ceRNAs) (37,52). CircRNAs include a quantity of miRNA response elements that decrease the expression abundance of miRNA by competitive binding to miRNA, hindering its complementary pairing with the 3′-UTR of the downstream target mRNA, thereby alleviating the suppressive effect of miRNAs on mRNA and subsequently enhancing mRNA expression (53,54). However, the majority of circRNAs lack a substantial number of miRNA binding sites and their expression levels are inferior to those of the corresponding miRNAs (55). The first identified gene cerebellar degeneration-related protein 1 antisense RNA (CDR1as) is demonstrated to be responsible for the production of circRNA molecules. Circ-CDR1as possesses >70 specific target sites for miRNAs that effectively inhibit their functions, suggesting that circRNAs can serve as sponges for miRNAs (37).
Apart from miRNA sponges, circRNAs can also interact with RBPs and other regulatory proteins, regulating various cellular processes via affecting protein localization, stability and function (56,57). There are three prevalent manners of interaction. Primary among these is the role of circRNAs as sponges of proteins. Of note, it is reported that the introns encompassing circular forms of muscleblind (circ-MBL) are rich in MBL binding sites, which possess a specific affinity for MBL proteins, consequently leading to a reduction in the intracellular MBL protein levels (50). Another mode involves circRNAs acting as protein reservoirs. For instance, the abnormal presence of circ-forkhead box (FOX)O3 was verified to prompt cardiac fibroblast aging through interacting with various proteins linked to cellular stress responses, such as DNA binding inhibitor 1, E2F transcription factor 1, hypoxia inducible factor 1 and focal adhesion kinase, thereby sequestering these proteins from the cytoplasm (58). Furthermore, circRNAs can serve as scaffolds for proteins. Zeng et al (59) revealed that a circRNA derived from angiomotin-like 1 associates with 3-phosphoinositol-dependent protein kinase 1 (PDK1) and AKT1 to facilitate the phosphorylation of PDK1-dependent AKT1, conferring protection to the heart. In addition, circRNAs have the ability to attract proteins to specific sites, which allows circRNAs to act as scaffolds or guides, facilitating the localization of proteins to distinct regions of the genome. For instance, circ-FECR1 recruits ten-eleven translocation (TET)1 protein to the promoter region of its host gene Friend leukemia integration 1, triggering the demethylation of CpG sites and promoting active transcription (60).
Although the majority of circRNAs predominantly localize in the cytoplasm where they are employed as protein scaffolds or miRNA sponges, a minority of EIciRNAs are situated in the nucleus and play crucial roles in transcriptional processes. For instance, circRNA eukaryotic translation initiation factor 3 subunit J and circRNA poly(A)-binding protein-interacting protein 2 are confirmed to activate the promoter and modulate the transcription of their host genes in a cis-regulatory manner through binding to U1 small ribonucleoprotein and RNA polymerase II transcription complexes (27). However, the underlying mechanism of EIciRNAs regulating gene transcription has remained largely elusive. In addition, EIciRNAs can affect gene expression by regulating the splicing of their linear counterparts. Furthermore, circRNAs that encompass the start codon of the host gene function as 'mRNA traps' to hinder the expression of the parental gene. By the circularization of an exon that contains an ATG translation start site, circ-homeodomain-interacting protein kinase (HIPK)2 and circ-HIPK3 are generated to repress the synthesis of the HIPK2 and HIPK3 proteins (61).
Due to the absence of essential components for cap-dependent translation, specifically the 5′ cap and poly(A) tail, it is widely accepted that circRNAs are incapable of being translated into proteins. However, recent research has revealed that circRNAs containing internal ribosome entry site fragments (IRES) and adenine-uracil-guanine codon sites may serve as templates for protein translation under particular conditions (62). For instance, circ-F-box and WD-repeat domain-containing 7 (FBXW7) has a region called the open reading frame that starts the translation process with the help of IRES, regardless of the 5′-cap structure, elevating the expression of FBXW7 (63). Additional mechanisms triggering circRNA translation include m6A-methylation initiation of translation and rolling circle adaptation (64,65). It is worth noting that while circRNAs possess a certain translational capacity, their translation efficiency is limited by the unique circular structure of circRNAs. The roles of translation products, such as proteins and peptides, generated from circRNAs remain elusive, necessitating additional investigation.
In conclusion, circRNAs represent a fascinating and rapidly evolving field of study with broad implications for understanding gene regulation and disease mechanisms. Their unique properties and diverse functions underscore their potential as biomarkers and therapeutic targets. Continued research into circRNAs will undoubtedly uncover more about their roles in various diseases such as CHD, paving the way for innovative diagnostic and therapeutic approaches.
Roles of circRNAs in CHD
CircRNAs have emerged as crucial players in the pathogenesis of CHD. Research has identified numerous circRNAs that are differentially expressed in patients with CHD compared to healthy controls, suggesting their crucial role in disease progression. For instance, hsa_circRNA11783-2 and hsa_circ_0000563 have been found to influence the initiation and progression of CHD, with hsa_circ_0000563 affecting the pathological process via targeting hub genes like ribosomal protein subunit 3 and subunit 1 (66,67). Hsa_circ_0000563 has been verified to participate in the atherosclerotic changes in human coronary artery segments with severe atherosclerotic stenosis (68). Furthermore, the combination of hsa_circ_0001879 and hsa_circ_0004104 can discriminate patients with CHD from healthy controls; in addition, upregulation of hsa_circ_0004104 causes dysregulation of atherosclerosis-related genes in macrophages, indicating that hsa_circ_0004104 is involved in inflammatory response during atherosclerosis (69). Similarly, it has been demonstrated that the combination of smoking and high hsa_circ_0008507 expression results in the occurrence and development of CHD (70). In addition, the exosome-derived circRNA hsa_circ_0005540 has been identified as a potential diagnostic biomarker for CHD, with its expression levels being associated with disease severity and progression (71). The ceRNA networks involving circRNA-miRNA have been shown to regulate key cellular and molecular biological processes, which are implicated in CHD pathophysiology (72,73). Indeed, hsa_circ_0066439, hsa_circ_0081241 and hsa_circ_0122984 were found to regulate multiple signaling pathways to participate in the acute myocardial infarction through hsa-miR-1254 and hsa-miR-328-5p (74). Furthermore, 9 circRNAs (hsa_circ_0089378, hsa_circ_0083357, hsa_circ_0082824, hsa_circ_0068942, hsa_circ_0057576, hsa_circ_0054537, hsa_circ_0051172, hsa_circ_0032970 and hsa_circ_0006323) were identified to promote the expression of transient receptor potential melastatin-3, a calcium-permeable ion channel, by inhibiting hsa-miR-130a-3p in patients with CHD (75). However, these circRNAs were investigated by observational studies or bioinformatics, while molecular mechanistic evaluations warrant further investigation. Overall, the growing body of evidence highlights the multifaceted roles of circRNAs in CHD, from their involvement in cell proliferation and apoptosis to inflammation, providing new avenues for the diagnosis and treatment of this disease.
Of note, circRNAs have distinct cell-type-specific roles in cardiomyocytes, endothelial cells and vascular smooth muscle cells (VSMCs), contributing to CHD progression. In cardiomyocytes, circRNAs are involved in regulating cell apoptosis and hypertrophic responses, both of which are critical in the development of CHD (76). Furthermore, circRNAs in endothelial cells are key players in angiogenesis regulation, endothelial dysfunction and endothelial barrier inflammation (77). In addition to influencing the proliferation and migration of VSMCs, circRNAs regulate their phenotypic switch between contractile and synthetic phenotypes in response to vascular injury, affecting plaque formation and vascular stiffening (78).
Cardiomyocyte death
Cardiomyocyte death has a pivotal role in the progression of CHD by initiating and exacerbating a cascade of pathological events that compromise cardiac function. Various forms of cell death, including apoptosis, autophagy, necroptosis, pyroptosis and ferroptosis, are implicated in the deterioration of cardiomyocytes during CHD (79,80). The loss of cardiomyocytes weakens the contractile power of the heart, leading to impaired cardiac output and heart failure (81). This cell death is often triggered by ischemic events such as myocardial infarction, where the lack of oxygen and nutrients causes extensive cardiomyocyte damage and death (82). The failing cardiomyocyte is characterized by a complex interplay of abnormal signaling pathways, oxidative stress, impaired mitochondrial function and altered gene expression, all of which contribute to the vicious cycle of cardiac dysfunction and cell death (83). The irreversible loss of cardiomyocytes leads to fibrosis and scar formation, which exacerbate heart failure and limit the heart's ability to recover from injury (84). Thus, understanding the mechanisms of cardiomyocyte death is essential for developing targeted therapies to protect cardiomyocytes, reduce heart damage and improve clinical outcomes in CHD.
CircRNAs are involved in cardiomyocyte death through various mechanisms, primarily by acting as miRNA sponges, thereby influencing gene expression and cellular processes. For instance, circ-HIPK2 has been shown to interact with miR-485-5p to upregulate the expression of autophagy-related protein 101, which facilitates autophagy to accelerate cardiomyocyte apoptosis in H2O2-induced myocardial oxidative injury (8). Similarly, circ_0030235 is upregulated in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced H9c2 cells, a widely used rat cardiomyoblast cell line that can be induced to exhibit certain properties of cardiac muscle cells, and aggravates mitochondrial dysfunction and oxidative damage in cardiomyocytes by targeting miR-526b and thus inactivating the PI3K/AKT and MEK/ERK pathways (85). These findings indicate that circRNAs exert a promoting effect on cardiomyocyte death and disease progression. In addition, overexpression of circRNA-differentially expressed in normal cells and neoplasia domain containing 4 C (DENND4C) has been observed in OGD/R-stimulated H9c2 cells, and further mechanistic evaluation revealed that circ-DENND4C attenuates OGD/R-induced cardiomyocyte death by downregulation of miR-320 through activating the ERK and mTOR pathways (86). Consistent with this result, circ-LRP62–2 is confirmed to protect cardiomyocytes from hypoxia-induced apoptosis. This circRNA is downregulated in cardiomyocytes exposed to hypoxia, while its overexpression represses cell apoptosis. Mechanically, under hypoxia, circ-LRP62−2 recruits heterogeneous nuclear ribonucleoprotein M and further enhances the expression of fibroblast growth factor 9, facilitating hypoxia-adaption and viability of cardiomyocytes (87). Thus, circRNAs also exhibit protective effects on cardiomyocytes and delay disease progression. Overall, the multifaceted roles of circRNAs in cardiomyocyte death, through their interactions with miRNAs and other molecular targets, provide a comprehensive understanding of their contribution to cardiovascular pathophysiology and potential clinical applications.
Therefore, circRNAs have been implicated in regulating cardiomyocyte death by affecting processes such as cell proliferation and autophagy, further impacting the progression of CHD (Table I). Despite advances in therapeutic approaches, the limited regenerative capacity of adult cardiomyocytes poses a significant challenge in restoring heart function after extensive cell loss (88). In this regard, targeting specific circRNAs may improve cardiac outcomes, highlighting the potential to mitigate cardiomyocyte death. Understanding the molecular mechanisms by which circRNAs participate in different forms of cell death is essential for developing effective treatments to prevent cardiomyocyte loss and improve the prognosis for patients with CHD.
Endothelial cell injury
Endothelial cell injury is a critical factor in the development of CHD due to its multifaceted impacts on vascular function and integrity. The endothelium, which lines the inner walls of blood vessels, has a vital role in maintaining vascular tone, regulating hemostasis and controlling inflammation and thrombosis (89). When endothelial cells are damaged, several pathological processes are triggered. Firstly, endothelial injury enhances the permeability of the vascular intima, facilitating leukocyte adhesion and transmigration, which further exacerbates inflammation and promotes thrombus formation (90). Increased inflammatory mediators, such as cytokines and oxidized lipoproteins, lead to the overexpression of adhesion molecules, selectins and chemokines, which attract and retain leukocytes in the subendothelial space (91). This inflammatory cascade is compounded by oxidative stress, which not only damages endothelial cells directly but also disrupts the balance of pro- and anti-coagulant factors, shifting the endothelium towards a pro-thrombotic state (92,93). Furthermore, endothelial cell injury is linked to metabolic disturbances such as lipid metabolism disorders, which are significant risk factors for atherosclerosis. The damage to endothelial cells impairs their ability to regulate lipid levels, leading to the accumulation of lipids in the arterial walls and the formation of atherosclerotic plaques (94). Mechanical, chemical and biological stresses, including hypertension, hyperglycemia and infections, exacerbate endothelial dysfunction by inducing cell senescence, autophagy dysregulation and mitochondrial stress (95,96). Autophagy, a process crucial for cellular homeostasis, is often impaired in endothelial cells during aging and disease, further contributing to endothelial dysfunction (97). The persistent endothelial activation and injury, coupled with diminished repair capacity, create a vicious cycle that perpetuates vascular inflammation, thrombosis and atherosclerosis, ultimately leading to the development and progression of CHD (98).
CircRNAs participate in regulating endothelial cell function in CHD through complex molecular mechanisms involving the sponging of miRNAs and subsequent modulation of mRNA targets. For instance, hsa_circ_0000284 is downregulated in patients with CHD and oxidative stress-induced EA-hy926 endothelial cells, and overexpression of hsa_circ_0000284 leads to impaired cell proliferation and increased apoptosis by sponging miR-338-3p, thereby repressing the expression of E26 transformation-specific sequence-1, a transcription factor involved in endothelial cell function (9). Another circRNA, circ_0001785, is decreased in the circulating peripheral blood of patients with CHD but increased within atherosclerotic plaque tissue. This circRNA is implicated in delaying atherogenesis by alleviating aortic endothelial cell injury and the formation of intraplaque neovascularization, via modulating the miR-513a-5p/TGFBR3 axis (99). In addition, hsa_circ_0030042 has been shown to suppress oxidized low-density lipoprotein (ox-LDL)-mediated abnormal autophagy of endothelial cells and maintain plaque stability in high-fat-diet fed apolipoprotein E−/− mice by sponging the endogenous eukaryotic initiation factor 4A-III, which impedes its recruitment to beclin1 and FOXO1 mRNA (100). Furthermore, circRNA-0024103, which is highly expressed in patients with coronary chronic total occlusion, has been verified to accelerate tube formation and cell migration via the miR-495/Notch1 axis in endothelial cells. This circRNA also increased collateral formation of the ligated femoral artery in mice after hindlimb ischemia, which is related to myocardial perfusion improvement after revascularization (101).
Collectively, these studies highlight the multifaceted roles of circRNAs in regulating endothelial cell function through intricate ceRNA networks, influencing key processes such as angiogenesis, cell proliferation and apoptosis, thereby offering novel insights into the pathogenesis and potential therapeutic strategies for CHD (Table I). Further studies are needed to unravel the detailed mechanisms by which circRNAs regulate endothelial cell function and contribute to CHD. Understanding these mechanisms will provide insights into potential therapeutic targets.
VSMC apoptosis
VSMC apoptosis is involved in the development and progression of CHD by affecting vascular remodeling, plaque stability and inflammatory responses (102). The balance between VSMC proliferation and apoptosis is crucial in the pathogenesis of atherosclerosis, a primary underlying cause of CHD (103). VSMCs with a synthetic phenotype are characterized by increased migration and proliferation to repair the damage (104). This phenotypic switching is regulated by a network of factors, including transcription factors, growth factors and non-coding RNAs, which contribute to vascular aging and atherosclerosis (104). Early atherosclerotic lesions are characterized by intense apoptosis of VSMCs, which decreases as the disease progresses, leading to intimal hyperplasia and plaque formation (105). Clonal expansion of VSMCs, initiated by a small fraction of cells that proliferate and migrate to form oligoclonal neointima, is a hallmark of early atherosclerotic lesions, suggesting that selective VSMC activation drives disease progression (106). Furthermore, VSMCs can transdifferentiate into mesenchymal and myeloid-like phenotypes, contributing to the progression of organ fibrosis and potentially affecting other vital organs beyond the vascular system (107). The dynamic variations in VSMC phenotypes shape the atherosclerotic plaque microenvironment, leading to heterogeneous clinical outcomes in CHD (108). Overall, the injury-induced phenotypic switching and clonal expansion of VSMCs, along with the disequilibrium between cell proliferation and apoptosis, underscore the critical role of VSMCs in the pathogenesis of CHD, providing potential targets for therapeutic intervention.
Dysregulated circRNAs have been implicated in the phenotypes and function of VSMCs during CHD progression. For instance, hsa_circ_0000280 has been shown to inhibit VSMC proliferation and induce cell-cycle arrest by facilitating the interaction between human antigen R and cyclin-dependent kinase suppressor 1 mRNA, leading to cell cycle arrest at the G1/S checkpoint and reducing neointimal hyperplasia in vivo (109). Another circRNA, circ-LDLR, is downregulated in CHD tissues, and its upregulation represses proliferation and promotes apoptosis of VSMCs through the miR-26-5p/KDM6A axis (110). Furthermore, circ-MAP3K5 has been identified to exert antiproliferative effects on VSMCs, where circ-MAP3K5 acts as a master negative regulator of TET2-mediated VSMC differentiation by sequestering miR-22-3p and thus blocking the expression of TET2 (111). Thus, these findings indicate that circRNAs postpone CHD development via mitigating VSMC proliferation and differentiation. Furthermore, circ-SATB2 has been reported to promote the phenotypic switch of VSMCs from a contractile to a synthetic state through the miR-939/stromal interacting molecule 1 axis, which regulates VSMC phenotypic differentiation, proliferation, apoptosis and migration (112). In addition, hsa_circ_0031891 participates in atherosclerosis by promoting VSMC proliferation, migration and dedifferentiation through the miR-579-3p/high mobility group box 1 axis. Silencing hsa_circ_0031891 inhibits these processes, suggesting its role in the pathogenesis of atherosclerosis (113). Likewise, in platelet-derived growth factor subunit B-induced VSMCs, circ_0006251 is upregulated to facilitate VSMC proliferation and reduce their apoptosis by enhancing TET3 and protein phosphatase 1B expression through sponging miR-361-3p, thereby contributing to disease occurrence (114). Hence, circRNAs boost CHD progression by facilitating VSMC proliferation and differentiation.
Taken together, circRNAs are crucial regulators of VSMC phenotype and function in CHD, regulating cell proliferation, migration and phenotypic switching through complex molecular pathways, making them promising targets for future therapeutic strategies (Table I).
Inflammatory response
The inflammatory response is a complex biological process initiated by the immune system to protect the body against harmful stimuli such as pathogens, damaged cells or irritants (115). It involves the activation and recruitment of various immune cells, including macrophages, neutrophils and T cells, which release cytokines and other inflammatory mediators to eliminate the offending agents and promote tissue repair (116). However, chronic inflammation can have detrimental effects, particularly in the context of CHD. Inflammation has a pivotal role in the development and progression of CHD by contributing to endothelial dysfunction, atherosclerosis and plaque instability (117). Macrophages ingest ox-LDL, transforming into foam cells and forming the lipid-rich necrotic core of atheromas, while the secretion of cytokines and proteases by these cells further exacerbates the inflammatory process and weakens the fibrous cap, making plaques more prone to rupture (118,119). The macrophage-mediated innate immune response is crucial in both the initiation and resolution of inflammation following myocardial infarction, influencing cardiac remodeling and heart failure development (120). Consequently, the inflammatory response is a double-edged sword in CHD, being essential for initial defense and tissue repair but potentially harmful when it becomes chronic.
CircRNAs have a vital role in the inflammatory response associated with the development of CHD. For instance, hsa_circ_0007478 has been found to be upregulated in ox-LDL-stimulated macrophages, and its expression exacerbates lipid metabolism imbalance and foam cell formation through the miR-765/EFNA3 axis, along with interleukin (IL)-1β production and NLR family pyrin domain containing 3 inflammasome activation, a key player in cell pyroptosis and inflammation (10). Besides, in ox-LDL-stimulated endothelial cells and patients with CHD, circ-CHFR is upregulated to provoke atherosclerosis development by sponging miR-15b-5p, which in turn enhances the expression of GADD45G, thus triggering the secretion of atherosclerosis-associated cytokines, including IL-1β, IL-6 and tumor necrosis factor (TNF)-α (121). Consistently, silencing of circ_0004104, which is upregulated in patients with CHD, mitigates ox-LDL-mediated inflammatory injury in endothelial cells by the miR-100/TNFAIP8 axis, highlighting the deleterious effect of circ_0004104 in CHD pathogenesis (122). Therefore, circRNAs can promote endothelial cell injury and atherosclerosis progression by enhancing inflammatory responses. Moreover, the construction of circRNA-related ceRNA networks has revealed that hsa_circ_0126672 is involved in regulating inflammation-related pathways, such as the JAK/ STAT and Apelin signaling pathways, which are critical for atherosclerotic plaque progression and instability (123). By activating these signaling pathways, hsa_circ_0092576 induces vascular inflammation via the activation and proliferation of VSMCs, as well as mediates the accumulation of oxidized lipids and oxidative damage to endothelial cells, resulting in the development of atherosclerosis (124).
Therefore, circRNAs mediate the inflammatory response in CHD by regulating miRNA and mRNA interactions, influencing cell proliferation, oxidative stress and atherosclerosis, thereby contributing to disease development and progression (Table I). The circRNA-miRNA-mRNA regulatory network is crucial in controlling the inflammatory processes that contribute to atherosclerotic plaque formation and instability, highlighting the therapeutic potential of targeting these pathways. Hence, the complex interplay between circRNAs, miRNAs and mRNAs requires further elucidation to understand the precise mechanisms of action during CHD progression. Furthermore, the influence of environmental factors, such as smoking, on circRNA expression and their interaction with inflammatory response must be considered to develop personalized treatment strategies.
Applications of circRNAs for CHD
CircRNAs as biomarkers
CircRNAs have emerged as promising biomarkers for the diagnosis and treatment of cardiovascular disease due to their unique properties and regulatory roles in disease occurrence and development (20). Compared with linear RNAs, such as miRNAs and long non-coding RNAs, they are covalently closed-loop structures without free 5′ and 3′ ends, which makes them highly resistant to exonucleases and more stable in bodily fluids, enhancing their reliability as biomarkers (125). CircRNAs are abundant and exhibit tissue-specific expression, making them ideal candidates for clinical applications (126). CircRNAs have been detected in considerable quantities in various bodily fluids, such as plasma, serum and saliva (71,127-129). Furthermore, they exhibit a half-life of 48 h in bodily fluids, a duration that surpasses that of linear RNA (130). In addition, circRNAs exhibit a broad distribution within cells, as well as in extracellular regions, and the extracellular amount of circRNAs is thought to reflect, at least in part, their intracellular abundance. When cells undergo stress or pathological conditions, transcriptional upregulation of intracellular circRNAs may lead to an increased release of circRNAs into the extracellular space. It is hypothesized that the expression levels of intracellular circRNAs could impact the concentrations of extracellular circRNAs, but the exact relationship between these two pools of circRNAs remains to be fully elucidated (131). These distinctive attributes of circRNA substantiate their potential suitability as biomarkers.
CircRNAs have emerged as promising biomarkers for the early detection and prognosis of CHD due to their unique properties and regulatory roles in gene expression. Numerous studies have highlighted their diagnostic value, with varying degrees of efficacy. A meta-analysis of 16 studies involving 3,962 subjects revealed that circRNAs have a pooled receiver operating characteristic curve of 0.80, with sensitivity and specificity values of 0.77 and 0.68, respectively, indicating their potential as reliable biomarkers for CHD diagnosis (132). Besides, several dysregulated circRNAs, such as circ-HECTD1, circ-ZBTB46 and hsa_circ_0001445, have been detected in the peripheral blood of patients with CHD and their expression level was shown to be associated with laboratory parameters, such as hemoglobin, triglycerides and cholesterol levels, suggesting their potential as diagnostic markers (73,133). High-throughput sequencing has confirmed differentially expressed circRNAs in the plasma of patients with CHD, such as hsa_circ_0069972, hsa_circ_0021509, circ-RPRD1A and circ-HERPUD2, which show significant upregulation and serve as new biomarkers for the diagnosis of coronary artery disease (134,135). Specifically, exosome-derived circRNAs have been identified as potential biomarkers for CHD, with certain circRNAs such as hsa_circ_0001445, hsa_circ_0001360 and hsa_ circ_0000038 showing significant downregulation in patients with CHD (127,136). Furthermore, hsa_circ_0124644, hsa_circ_0001946, hsa_circ_0001785, hsa_circ_0000973, hsa_circ_0001741 and hsa_circ_0003922 in peripheral blood have shown their ability to discriminate between patients with CHD and healthy controls, with promising potential as diagnostic biomarkers for CHD (137-139). In addition, bioinformatics analyses have constructed circRNA-miRNA-mRNA regulatory networks, revealing the involvement of circRNAs such as circ-YOD1 in lipid metabolism and protein modification, further supporting their diagnostic and prognostic potential (140,141). Microarray data analysis has identified differentially expressed circRNAs in patients with CHD with varying severity, with hsa_circ_0016868, hsa_circ_0001364, hsa_circ_0006731 and circ-ANRIL emerging as promising biomarkers for early CHD diagnosis (11,12,142).
Collectively, these findings suggest that circRNAs hold substantial promise as biomarkers for CHD, offering a new avenue for the early diagnosis, prognosis and personalized treatment of CHD, thereby addressing unmet clinical needs such as timely diagnosis and effective monitoring of treatment responses. However, circRNA-based biomarkers are still undergoing experimental evaluation in CHD. Further integration of circRNA profiling with advanced bioinformatics, high-throughput sequencing and molecular biology techniques will hold great promise for the identification and validation of differentially expressed circRNAs in patients with CHD, enabling the construction of circRNA-miRNA-mRNA regulatory networks that provide insight into disease mechanisms and potential therapeutic targets. However, circRNAs can be present at low concentrations and distinguishing them from linear RNA counterparts remains a technical challenge. Thus, developing more sensitive, reliable and high-throughput detection techniques is essential. Besides, circRNAs are highly heterogeneous and their expression can vary widely across different cell types and conditions. In this regard, identifying a panel of circRNAs rather than relying on a single circRNA marker may enhance the diagnostic power.
CircRNA-targeting therapeutic strategies
Current strategies for targeting circRNAs in the treatment of CHD are multifaceted, leveraging their unique properties and regulatory roles in cardiovascular diseases. By designing synthetic circRNAs or using antisense oligonucleotides to inhibit specific circRNAs, researchers aim to modulate these regulatory networks to achieve therapeutic effects. For instance, targeting proatherogenic circRNAs such as circ_0002984 and circ_0029589 could potentially mitigate atherosclerosis and its associated risks in patients with CHD (143). However, solutions to increasing the stability of the antisense oligonucleotide and its efficiency should be further developed. In addition, gene therapy techniques are being explored to either upregulate protective circRNAs or downregulate harmful ones, thereby restoring the balance of circRNA-miRNA-mRNA interactions crucial for cardiovascular health (144). Physical exercise has also been suggested as a non-pharmacological strategy to modulate circRNA expression, offering a low-cost and accessible means to alleviate CHD symptoms (145). Furthermore, the development of circRNA synthesis and engineering delivery systems holds promise for their application in therapeutics. These systems aim to enhance the stability, specificity and delivery efficiency of circRNA-based treatments, potentially overcoming current challenges in circRNA therapy (146). Thus, the integration of circRNAs into the therapeutic landscape of CHD represents a promising frontier, with ongoing research aimed at optimizing these strategies for clinical application.
CircRNAs can influence patient responses to conventional CHD treatments by modulating the molecular pathways involved in disease mechanisms and drug efficacy. Dysregulation of circRNAs has been linked to cholesterol homeostasis, which alters the effectiveness of statins, leading to variable lipid-lowering responses (147). Antiplatelet agents like clopidogrel and ticagrelor are used to prevent thrombosis in patients with CHD. CircRNAs can regulate platelet activation and aggregation by controlling the expression of proteins involved in this process (148). Upregulated circRNAs, such as hsa_circ_0070675_CBC1, hsa-circ_13011-5_CBC1 and hsa-circ_6406-3_CBC1, are related to clopidogrel resistance in patients with CHD (149). Platelet-derived circFAM13B has been implicated in platelet aggregation processes, which affect the antiplatelet action of ticagrelor patients with CHD (150). Hence, circRNAs represent a key regulatory layer that can influence patient responses to conventional CHD therapies, making them promising targets for personalized medicine strategies in this disease.
Despite the promising potential of circRNAs as therapeutic targets, no clinical application or ongoing clinical trials has investigated circRNA-targeting therapeutic strategies in CHD, which face several challenges. One significant challenge is the accurate and sensitive detection of circRNAs, which is crucial for understanding their biological processes. Current methodologies for circRNA identification, including purification and sequencing methods, have limitations in sensitivity and specificity, which can hinder the precise characterization of circRNAs involved in CHD. In addition, the regulatory roles of circRNAs in cardiovascular diseases are complex, involving interactions with miRNAs and mRNAs in regulatory networks that are not yet fully understood. This complexity necessitates advanced technologies for identifying, validating and analyzing circRNAs, which are still under development. Furthermore, the involvement of circRNAs in various pathophysiological processes, such as atherogenesis and myocardial infarction, adds another layer of complexity. Specific circRNAs have been identified with either atheroprotective or proatherogenic effects, but translating these findings into therapeutic applications requires a deeper understanding of their mechanisms and interactions within the circRNA-miRNA-mRNA regulatory axis. Thus, the continued exploration of circRNA functions and their interactions within the cardiovascular system will likely yield novel insights and more effective treatments for CHD.
Conclusions and future perspectives
CircRNAs represent a promising frontier in cardiovascular research, offering novel insights into the molecular mechanisms of CHD. They play crucial roles in various cellular processes pertinent to CHD, ranging from endothelial dysfunction to inflammation, by acting as miRNA sponges and further regulating gene expression and signaling pathways (Fig. 2). They exhibit distinct expression profiles in patients with CHD compared to healthy controls, positioning them as valuable biomarkers and therapeutic targets. However, translating circRNA research into clinical practice requires concerted efforts to overcome current challenges. First, advancements in high-throughput sequencing technologies and bioinformatics tools are essential for accurate circRNA detection and quantification. Standardized methods for circRNA isolation, sequencing and analysis will improve reproducibility and facilitate cross-study comparisons. Furthermore, extensive in vivo studies are required to elucidate the precise mechanisms by which circRNAs influence CHD pathophysiology. As numerous circRNAs serve as miRNA sponges or regulate transcription and splicing, uncovering their exact molecular functions in different cell types within the cardiovascular system is critical for therapeutic targeting. Integrating single-cell sequencing approaches could unravel cell-type-specific roles of circRNAs, offering a more precise understanding of their contributions to disease. In addition, large-scale clinical studies are needed to validate the diagnostic and prognostic utility of circRNAs identified in preliminary research. Integrating circRNA biomarkers into existing diagnostic frameworks could enhance early detection and risk stratification in patients with CHD. CircRNAs could be used to monitor treatment responses, particularly in therapies that target molecular pathways involved in lipid metabolism, vascular remodeling and inflammation. Furthermore, developing circRNA-based therapies involves designing molecules that can specifically modulate circRNA activity. Antisense oligonucleotides and small interfering RNAs targeting circRNAs hold promise but require optimization for efficient delivery and minimal off-target effects. Developing safe and effective delivery systems, such as lipid nanoparticles or viral vectors, will be crucial for translating circRNA-targeting therapies into clinical practice. Furthermore, addressing the potential immunogenicity of these delivery platforms is essential to ensure safety and minimize adverse reactions. Exploring the therapeutic potential of circRNAs in combination with existing treatments could offer synergistic benefits, improving patient outcomes in CHD.
Availability of data and materials
Not applicable.
Authors' contributions
ZF and YY wrote the manuscript, XY revised the manuscript and YY designed the research. All authors have read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Abbreviations:
CDKN1A |
cyclin-dependent kinase suppressor 1 |
ceRNA |
competitive endogenous RNA |
CHD |
coronary heart disease |
ciRNA |
circular intronic RNA |
circRNA |
circular RNA |
EcircRNA |
exonic circRNA |
EIciRNA |
exon-intron circRNA |
ELAVL1 |
human antigen R |
FBXW7 |
F-box and WD-repeat domain-containing 7 |
FOXO1 |
forkhead box O1 |
HIPK |
homeodomain-interacting protein kinase |
HMGB1 |
high mobility group box 1 |
IL |
interleukin |
IRES |
internal ribosome entry site fragments |
LDL |
low-density lipoprotein |
miRNA |
microRNA |
m6A |
N-6 methylation |
NLRP3 |
NLR family pyrin domain containing 3 |
OGD/R |
oxygen-glucose deprivation/reoxygenation |
PDGF |
platelet-derived growth factor |
PDK1 |
3-phosphoinositol-dependent protein kinase 1 |
RBP |
RNA-binding protein |
STIM1 |
stromal interacting molecule 1 |
TET |
ten-eleven translocation |
TNF |
tumor necrosis factor |
VSMC |
vascular smooth muscle cell |
Acknowledgments
Not applicable.
Funding
This study is supported by the Youth Fund Project of Jiangxi Provincial Natural Science Foundation (grant no. 20224BAB216097).
References
Katta N, Loethen T, Lavie CJ and Alpert MA: Obesity and coronary heart disease: Epidemiology, Pathology, and coronary artery imaging. Curr Probl Cardiol. 46:1006552021. View Article : Google Scholar | |
Pothineni NVK, Subramany S, Kuriakose K, Shirazi LF, Romeo F, Shah PK and Mehta JL: Infections, atherosclerosis, and coronary heart disease. Eur Heart J. 38:3195–3201. 2017. View Article : Google Scholar : PubMed/NCBI | |
Dong Y, Chen H, Gao J, Liu Y, Li J and Wang J: Molecular machinery and interplay of apoptosis and autophagy in coronary heart disease. J Mol Cell Cardiol. 136:27–41. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hodel F, Xu ZM, Thorball CW, de La Harpe R, Letang-Mathieu P, Brenner N, Butt J, Bender N, Waterboer T, Marques-Vidal PM, et al: Associations of genetic and infectious risk factors with coronary heart disease. Elife. 12:e797422023. View Article : Google Scholar : PubMed/NCBI | |
Sygitowicz G and Sitkiewicz D: Involvement of circRNAs in the Development of Heart Failure. Int J Mol Sci. 23:141292022. View Article : Google Scholar : PubMed/NCBI | |
Ma XK, Zhai SN and Yang L: Approaches and challenges in genome-wide circular RNA identification and quantification. Trends Genet. 39:897–907. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hwang HJ and Kim YK: Molecular mechanisms of circular RNA translation. Exp Mol Med. 56:1272–1280. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zhou J, Li L, Hu H, Wu J, Chen H, Feng K and Ma L: Circ-HIPK2 accelerates cell apoptosis and autophagy in myocardial oxidative injury by sponging miR-485-5p and Targeting ATG101. J Cardiovasc Pharmacol. 76:427–436. 2020. View Article : Google Scholar : PubMed/NCBI | |
Dinh P, Tran C, Dinh T, Ali A and Pan S: Hsa_circRNA_0000284 acts as a ceRNA to participate in coronary heart disease progression by sponging miRNA-338-3p via regulating the expression of ETS1. J Biomol Struct Dyn. 42:5114–5127. 2024. View Article : Google Scholar | |
Ye B, Liang X, Zhao Y, Cai X, Wang Z, Lin S, Wang W, Shan P, Huang W and Huang Z: Hsa_circ_0007478 aggravates NLRP3 inflammasome activation and lipid metabolism imbalance in ox-LDL-stimulated macrophage via miR-765/EFNA3 axis. Chem Biol Interact. 368:1101952022. View Article : Google Scholar : PubMed/NCBI | |
Hou C, Gu L, Guo Y, Zhou Y, Hua L, Chen J, He S, Zhang S, Jia Q, Zhao C, et al: Association between circular RNA expression content and severity of coronary atherosclerosis in human coronary artery. J Clin Lab Anal. 34:e235522020. View Article : Google Scholar : PubMed/NCBI | |
Akan G, Nyawawa E, Nyangasa B, Turkcan MK, Mbugi E, Janabi M and Atalar F: Severity of coronary artery disease is associated with diminished circANRIL expression: A possible blood based transcriptional biomarker in East Africa. J Cell Mol Med. 28:e180932024. View Article : Google Scholar : | |
Cao Q, Guo Z, Du S, Ling H and Song C: Circular RNAs in the pathogenesis of atherosclerosis. Life Sci. 255:1178372020. View Article : Google Scholar : PubMed/NCBI | |
Ma X, Chen X, Mo C, Li L, Nong S and Gui C: The role of circRNAs in the regulation of myocardial angiogenesis in coronary heart disease. Microvasc Res. 142:1043622022. View Article : Google Scholar : PubMed/NCBI | |
Chen CK, Cheng R, Demeter J, Chen J, Weingarten-Gabbay S, Jiang L, Snyder MP, Weissman JS, Segal E, Jackson PK and Chang HY: Structured elements drive extensive circular RNA translation. Mol Cell. 81:4300–4318.e13. 2021. View Article : Google Scholar : PubMed/NCBI | |
Busa VF and Leung AKL: Thrown for a (stem) loop: How RNA structure impacts circular RNA regulation and function. Methods. 196:56–67. 2021. View Article : Google Scholar : PubMed/NCBI | |
Aufiero S, van den Hoogenhof MMG, Reckman YJ, Beqqali A, van der Made I, Kluin J, Khan MAF, Pinto YM and Creemers EE: Cardiac circRNAs arise mainly from constitutive exons rather than alternatively spliced exons. RNA. 24:815–827. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zheng Q, Bao C, Guo W, Li S, Chen J, Chen B, Luo Y, Lyu D, Li Y, Shi G, et al: Circular RNA profiling reveals an abundant circHIPK3 that regulates cell growth by sponging multiple miRNAs. Nat Commun. 7:112152016. View Article : Google Scholar : PubMed/NCBI | |
Jiang S, Fu R, Shi J, Wu H, Mai J, Hua X, Chen H, Liu J, Lu M and Li N: CircRNA-Mediated regulation of angiogenesis: A new chapter in cancer biology. Front Oncol. 11:5537062021. View Article : Google Scholar : PubMed/NCBI | |
Ding C and Zhou Y: Insights into circular RNAs: Biogenesis, function and their regulatory roles in cardiovascular disease. J Cell Mol Med. 27:1299–1314. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhang XO, Wang HB, Zhang Y, Lu X, Chen LL and Yang L: Complementary sequence-mediated exon circularization. Cell. 159:134–147. 2014. View Article : Google Scholar : PubMed/NCBI | |
Zang J, Lu D and Xu A: The interaction of circRNAs and RNA binding proteins: An important part of circRNA maintenance and function. J Neurosci Res. 98:87–97. 2020. View Article : Google Scholar | |
Montañés-Agudo P, van der Made I, Aufiero S, Tijsen AJ, Pinto YM and Creemers EE: Quaking regulates circular RNA production in cardiomyocytes. J Cell Sci. 136:sc2611202023. View Article : Google Scholar | |
Pamudurti NR, Patop IL, Krishnamoorthy A, Bartok O, Maya R, Lerner N, Ashwall-Fluss R, Konakondla JVV, Beatus T and Kadener S: circMbl functions in cis and in trans to regulate gene expression and physiology in a tissue-specific fashion. Cell Rep. 39:1107402022. View Article : Google Scholar : PubMed/NCBI | |
Kelly S, Greenman C, Cook PR and Papantonis A: Exon skipping is correlated with exon circularization. J Mol Biol. 427:2414–2417. 2015. View Article : Google Scholar : PubMed/NCBI | |
Monat C, Quiroga C, Laroche-Johnston F and Cousineau B: The Ll.LtrB intron from Lactococcus lactis excises as circles in vivo: insights into the group II intron circularization pathway. RNA. 21:1286–1293. 2015. View Article : Google Scholar : PubMed/NCBI | |
Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, et al: Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 22:256–264. 2015. View Article : Google Scholar : PubMed/NCBI | |
Werfel S, Nothjunge S, Schwarzmayr T, Strom TM, Meitinger T and Engelhardt S: Characterization of circular RNAs in human, mouse and rat hearts. J Mol Cell Cardiol. 98:103–107. 2016. View Article : Google Scholar : PubMed/NCBI | |
Liang D, Tatomer DC, Luo Z, Wu H, Yang L, Chen LL, Cherry S and Wilusz JE: The Output of Protein-Coding Genes Shifts to Circular RNAs When the Pre-mRNA Processing Machinery Is Limiting. Mol Cell. 68:940–954.e3. 2017. View Article : Google Scholar : PubMed/NCBI | |
García-Lerena JA, González-Blanco G, Saucedo-Cárdenas O and Valdés J: Promoter-Bound Full-Length Intronic Circular RNAs-RNA Polymerase II Complexes Regulate Gene Expression in the Human Parasite Entamoeba histolytica. Noncoding RNA. 8:122022.PubMed/NCBI | |
Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, Marzluff WF and Sharpless NE: Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA. 19:141–157. 2013. View Article : Google Scholar : | |
Gao Y, Wang J and Zhao F: CIRI: An efficient and unbiased algorithm for de novo circular RNA identification. Genome Biol. 16:42015. View Article : Google Scholar : PubMed/NCBI | |
Chen LL: The expanding regulatory mechanisms and cellular functions of circular RNAs. Nat Rev Mol Cell Biol. 21:475–490. 2020. View Article : Google Scholar : PubMed/NCBI | |
Conn SJ, Pillman KA, Toubia J, Conn VM, Salmanidis M, Phillips CA, Roslan S, Schreiber AW, Gregory PA and Goodall GJ: The RNA binding protein quaking regulates formation of circRNAs. Cell. 160:1125–1134. 2015. View Article : Google Scholar : PubMed/NCBI | |
Chen I, Chen CY and Chuang TJ: Biogenesis, identification, and function of exonic circular RNAs. Wiley Interdiscip Rev RNA. 6:563–579. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, Zhu S, Yang L and Chen LL: Circular intronic long noncoding RNAs. Mol Cell. 51:792–806. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK and Kjems J: Natural RNA circles function as efficient microRNA sponges. Nature. 495:384–388. 2013. View Article : Google Scholar : PubMed/NCBI | |
Vromman M, Anckaert J, Bortoluzzi S, Buratin A, Chen CY, Chu Q, Chuang TJ, Dehghannasiri R, Dieterich C, Dong X, et al: Large-scale benchmarking of circRNA detection tools reveals large differences in sensitivity but not in precision. Nat Methods. 20:1159–1169. 2023. View Article : Google Scholar : PubMed/NCBI | |
Haque S and Harries LW: Circular RNAs (circRNAs) in Health and Disease. Genes (Basel). 8:3532017. View Article : Google Scholar : PubMed/NCBI | |
Zhang Q, Sun W, Han J, Cheng S, Yu P, Shen L, Fan M, Tong H, Zhang H, Chen J and Chen X: The circular RNA hsa_ circ_0007623 acts as a sponge of microRNA-297 and promotes cardiac repair. Biochem Biophys Res Commun. 523:993–1000. 2020. View Article : Google Scholar : PubMed/NCBI | |
Szabo L, Morey R, Palpant NJ, Wang PL, Afari N, Jiang C, Parast MM, Murry CE, Laurent LC and Salzman J: Statistically based splicing detection reveals neural enrichment and tissue-specific induction of circular RNA during human fetal development. Genome Biol. 16:1262015. View Article : Google Scholar : PubMed/NCBI | |
Siede D, Rapti K, Gorska AA, Katus HA, Altmüller J, Boeckel JN, Meder B, Maack C, Völkers M, Müller OJ, et al: Identification of circular RNAs with host gene-independent expression in human model systems for cardiac differentiation and disease. J Mol Cell Cardiol. 109:48–56. 2017. View Article : Google Scholar : PubMed/NCBI | |
Wu J, Guo X, Wen Y, Huang S, Yuan X, Tang L and Sun H: N6-Methyladenosine Modification Opens a New Chapter in Circular RNA Biology. Front Cell Dev Biol. 9:7092992021. View Article : Google Scholar : PubMed/NCBI | |
Xu T, He B, Sun H, Xiong M, Nie J, Wang S and Pan Y: Novel insights into the interaction between N6-methyladenosine modification and circular RNA. Mol Ther Nucleic Acids. 27:824–837. 2022. View Article : Google Scholar : PubMed/NCBI | |
Du WW, Yang W, Chen Y, Wu ZK, Foster FS, Yang Z, Li X and Yang BB: Foxo3 circular RNA promotes cardiac senescence by modulating multiple factors associated with stress and senescence responses. Eur Heart J. 38:1402–1412. 2017. | |
Tao M, Zheng M, Xu Y, Ma S, Zhang W and Ju S: CircRNAs and their regulatory roles in cancers. Mol Med. 27:942021. View Article : Google Scholar : PubMed/NCBI | |
Graham JR, Hendershott MC, Terragni J and Cooper GM: mRNA degradation plays a significant role in the program of gene expression regulated by phosphatidylinositol 3-kinase signaling. Mol Cell Biol. 30:5295–5305. 2010. View Article : Google Scholar : PubMed/NCBI | |
Nisar S, Bhat AA, Singh M, Karedath T, Rizwan A, Hashem S, Bagga P, Reddy R, Jamal F, Uddin S, et al: Insights Into the Role of CircRNAs: Biogenesis, characterization, functional, and clinical impact in human malignancies. Front Cell Dev Biol. 9:6172812021. View Article : Google Scholar : PubMed/NCBI | |
Huang J, Yu S, Ding L, Ma L, Chen H, Zhou H, Zou Y, Yu M, Lin J and Cui Q: The Dual Role of Circular RNAs as miRNA Sponges in breast cancer and colon cancer. Biomedicines. 9:15902021. View Article : Google Scholar : PubMed/NCBI | |
Ashwal-Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, Hanan M, Evantal N, Memczak S, Rajewsky N and Kadener S: circRNA biogenesis competes with pre-mRNA splicing. Mol Cell. 56:55–66. 2014. View Article : Google Scholar : PubMed/NCBI | |
Huang A, Zheng H, Wu Z, Chen M and Huang Y: Circular RNA-protein interactions: Functions, mechanisms, and identification. Theranostics. 10:3503–3517. 2020. View Article : Google Scholar : PubMed/NCBI | |
Huang S, Li X, Zheng H, Si X, Li B, Wei G, Li C, Chen Y, Chen Y, Liao W, et al: Loss of Super-Enhancer-Regulated circRNA Nfix Induces Cardiac Regeneration After Myocardial Infarction in Adult Mice. Circulation. 139:2857–2876. 2019. View Article : Google Scholar : PubMed/NCBI | |
Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al: Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 495:333–338. 2013. View Article : Google Scholar : PubMed/NCBI | |
Ma B, Wang S, Wu W, Shan P, Chen Y, Meng J, Xing L, Yun J, Hao L, Wang X, et al: Mechanisms of circRNA/lncRNA-miRNA interactions and applications in disease and drug research. Biomed Pharmacother. 162:1146722023. View Article : Google Scholar : PubMed/NCBI | |
Misir S, Wu N and Yang BB: Specific expression and functions of circular RNAs. Cell Death Differ. 29:481–491. 2022. View Article : Google Scholar : PubMed/NCBI | |
Das A, Sinha T, Shyamal S and Panda AC: Emerging Role of Circular RNA-Protein Interactions. Noncoding RNA. 7:482021.PubMed/NCBI | |
Jiang MP, Xu WX, Hou JC, Xu Q, Wang DD and Tang JH: The Emerging Role of the Interactions between Circular RNAs and RNA-binding Proteins in Common Human Cancers. J Cancer. 12:5206–5219. 2021. View Article : Google Scholar : PubMed/NCBI | |
Du WW, Fang L, Yang W, Wu N, Awan FM, Yang Z and Yang BB: Induction of tumor apoptosis through a circular RNA enhancing Foxo3 activity. Cell Death Differ. 24:357–370. 2017. View Article : Google Scholar : | |
Zeng Y, Du WW, Wu Y, Yang Z, Awan FM, Li X, Yang W, Zhang C, Yang Q, Yee A, et al: A Circular RNA Binds To and Activates AKT phosphorylation and nuclear localization reducing apoptosis and enhancing cardiac repair. Theranostics. 7:3842–3855. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chen N, Zhao G, Yan X, Lv Z, Yin H, Zhang S, Song W, Li X, Li L, Du Z, et al: A novel FLI1 exonic circular RNA promotes metastasis in breast cancer by coordinately regulating TET1 and DNMT1. Genome Biol. 19:2182018. View Article : Google Scholar : PubMed/NCBI | |
Conte A and Pierantoni GM: Update on the Regulation of HIPK1, HIPK2 and HIPK3 Protein Kinases by microRNAs. Microrna. 7:178–186. 2018. View Article : Google Scholar : PubMed/NCBI | |
Abe N, Matsumoto K, Nishihara M, Nakano Y, Shibata A, Maruyama H, Shuto S, Matsuda A, Yoshida M, Ito Y and Abe H: Rolling circle translation of circular RNA in living human cells. Sci Rep. 5:164352015. View Article : Google Scholar : PubMed/NCBI | |
Ye F, Gao G, Zou Y, Zheng S, Zhang L, Ou X, Xie X and Tang H: circFBXW7 Inhibits Malignant Progression by Sponging miR-197-3p and Encoding a 185-aa Protein in Triple-Negative Breast Cancer. Mol Ther Nucleic Acids. 18:88–98. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yang Y, Fan X, Mao M, Song X, Wu P, Zhang Y, Jin Y, Yang Y, Chen LL, Wang Y, et al: Extensive translation of circular RNAs driven by N(6)-methyladenosine. Cell Res. 27:626–641. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu C, Wu X, Gokulnath P, Li G and Xiao J: The Functions and Mechanisms of Translatable Circular RNAs. J Pharmacol Exp Ther. 384:52–60. 2023. View Article : Google Scholar | |
Li X, Zhao Z, Jian D, Li W, Tang H and Li M: Hsa-circRNA11783-2 in peripheral blood is correlated with coronary artery disease and type 2 diabetes mellitus. Diab Vasc Dis Res. 14:510–515. 2017. View Article : Google Scholar : PubMed/NCBI | |
Guo N, Zhou H, Zhang Q, Fu Y, Jia Q, Gan X, Wang Y, He S, Li C, Tao Z, et al: Exploration and bioinformatic prediction for profile of mRNA bound to circular RNA BTBD7_hsa_ circ_0000563 in coronary artery disease. BMC Cardiovasc Disord. 24:712024. View Article : Google Scholar | |
Chen JX, Hua L, Zhao CH, Jia QW, Zhang J, Yuan JX, Zhang YJ, Jin JL, Gu MF, Mao ZY, et al: Quantitative proteomics reveals the regulatory networks of circular RNA BTBD7_hsa_ circ_0000563 in human coronary artery. J Clin Lab Anal. 34:e234952020. View Article : Google Scholar | |
Wang L, Shen C, Wang Y, Zou T, Zhu H, Lu X, Li L, Yang B, Chen J, Chen S, et al: Identification of circular RNA Hsa_ circ_0001879 and Hsa_circ_0004104 as novel biomarkers for coronary artery disease. Atherosclerosis. 286:88–96. 2019. View Article : Google Scholar : PubMed/NCBI | |
Sun Y, Chen R, Lin S, Xie X, Ye H, Zheng F, Lin J, Huang Q, Huang S, Ruan Q, et al: Association of circular RNAs and environmental risk factors with coronary heart disease. BMC Cardiovasc Disord. 19:2232019. View Article : Google Scholar : PubMed/NCBI | |
Wu WP, Pan YH, Cai MY, Cen JM, Chen C, Zheng L, Liu X and Xiong XD: Plasma-Derived Exosomal Circular RNA hsa_ circ_0005540 as a Novel Diagnostic Biomarker for Coronary Artery Disease. Dis Markers. 2020:31786422020. View Article : Google Scholar | |
Yu F, Tie Y, Zhang Y, Wang Z, Yu L, Zhong L and Zhang C: Circular RNA expression profiles and bioinformatic analysis in coronary heart disease. Epigenomics. 12:439–454. 2020. View Article : Google Scholar : PubMed/NCBI | |
Dinh P, Peng J, Tran T, Wu D, Tran C, Dinh T and Pan S: Identification of hsa_circ_0001445 of a novel circRNA-miRNA-mRNA regulatory network as potential biomarker for coronary heart disease. Front Cardiovasc Med. 10:11042232023. View Article : Google Scholar : PubMed/NCBI | |
Yin L, Tang Y and Jiang M: Research on the circular RNA bioinformatics in patients with acute myocardial infarction. J Clin Lab Anal. 35:e236212021. View Article : Google Scholar : | |
Pan RY, Liu P, Zhou HT, Sun WX, Song J, Shu J, Cui GJ, Yang ZJ and Jia EZ: Circular RNAs promote TRPM3 expression by inhibiting hsa-miR-130a-3p in coronary artery disease patients. Oncotarget. 8:60280–60290. 2017. View Article : Google Scholar : PubMed/NCBI | |
Yijian L, Weihan S, Lin Y, Heng Z, Yu W, Lin S, Shuo M, Mengyang L and Jianxun W: CircNCX1 modulates cardiomyocyte proliferation through promoting ubiquitination of BRG1. Cell Signal. 120:1111932024. View Article : Google Scholar : PubMed/NCBI | |
Kishore R, Garikipati VNS and Gonzalez C: Role of Circular RNAs in Cardiovascular Disease. J Cardiovasc Pharmacol. 76:128–137. 2020. View Article : Google Scholar : PubMed/NCBI | |
Maguire EM and Xiao Q: Noncoding RNAs in vascular smooth muscle cell function and neointimal hyperplasia. FEBS J. 287:5260–5283. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ma W, Wei S, Zhang B and Li W: Molecular Mechanisms of Cardiomyocyte Death in Drug-Induced Cardiotoxicity. Front Cell Dev Biol. 8:4342020. View Article : Google Scholar : PubMed/NCBI | |
Dorn GW II: Apoptotic and non-apoptotic programmed cardiomyocyte death in ventricular remodelling. Cardiovasc Res. 81:465–473. 2009. View Article : Google Scholar : | |
Nah J, Zablocki D and Sadoshima J: The roles of the inhibitory autophagy regulator Rubicon in the heart: A new therapeutic target to prevent cardiac cell death. Exp Mol Med. 53:528–536. 2021. View Article : Google Scholar : PubMed/NCBI | |
Martens MD, Karch J and Gordon JW: The molecular mosaic of regulated cell death in the cardiovascular system. Biochim Biophys Acta Mol Basis Dis. 1868:1662972022. View Article : Google Scholar | |
Xiang Q, Yi X, Zhu XH, Wei X and Jiang DS: Regulated cell death in myocardial ischemia-reperfusion injury. Trends Endocrinol Metab. 35:219–234. 2024. View Article : Google Scholar | |
Parrotta EI, Lucchino V, Scaramuzzino L, Scalise S and Cuda G: Modeling cardiac disease mechanisms using induced pluripotent stem cell-derived cardiomyocytes: progress, promises and challenges. Int J Mol Sci. 21:43542020. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Liu S, Ding L, Wang D, Li Q and Li D: Circ_0030235 knockdown protects H9c2 cells against OGD/R-induced injury via regulation of miR-526b. PeerJ. 9:e114822021. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Zhang T, Zhang W, Zou C, Zhang Q, Ma X and Zhu Y: Circular RNA-DENND4C in H9c2 cells relieves OGD/R-induced injury by down regulation of microRNA-320. Cell Cycle. 19:3074–3085. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ding W, Ding L, Lu Y, Sun W, Wang Y, Wang J, Gao Y and Li M: Circular RNA-circLRP6 protects cardiomyocyte from hypoxia-induced apoptosis by facilitating hnRNPM-mediated expression of FGF-9. FEBS J. 291:1246–1263. 2024. View Article : Google Scholar | |
Ko T and Nomura S: Manipulating cardiomyocyte plasticity for heart regeneration. Front Cell Dev Biol. 10:9292562022. View Article : Google Scholar : PubMed/NCBI | |
Theofilis P, Sagris M, Oikonomou E, Antonopoulos AS, Siasos G, Tsioufis C and Tousoulis D: Inflammatory mechanisms contributing to endothelial dysfunction. Biomedicines. 9:7812021. View Article : Google Scholar : PubMed/NCBI | |
Shaito A, Aramouni K, Assaf R, Parenti A, Orekhov A, Yazbi AE, Pintus G and Eid AH: Oxidative stress-induced endothelial dysfunction in cardiovascular diseases. Front Biosci (Landmark Ed). 27:1052022. View Article : Google Scholar : PubMed/NCBI | |
Gallo G and Savoia C: New insights into endothelial dysfunction in cardiometabolic diseases: Potential mechanisms and clinical implications. Int J Mol Sci. 25:29732024. View Article : Google Scholar : PubMed/NCBI | |
Huang Y, Song C, He J and Li M: Research progress in endothelial cell injury and repair. Front Pharmacol. 13:9972722022. View Article : Google Scholar : PubMed/NCBI | |
Xu S, Ilyas I, Little PJ, Li H, Kamato D, Zheng X, Luo S, Li Z, Liu P, Han J, et al: Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: From mechanism to pharmacotherapies. Pharmacol Rev. 73:924–967. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang R, Wang M, Ye J, Sun G and Sun X: Mechanism overview and target mining of atherosclerosis: Endothelial cell injury in atherosclerosis is regulated by glycolysis (Review). Int J Mol Med. 47:65–76. 2021. View Article : Google Scholar | |
Zha D, Wang S, Monaghan-Nichols P, Qian Y, Sampath V and Fu M: Mechanisms of endothelial cell membrane repair: Progress and Perspectives. Cells. 12:26482023. View Article : Google Scholar : PubMed/NCBI | |
Marzoog BA: Endothelial cell autophagy in the context of disease development. Anat Cell Biol. 56:16–24. 2023. View Article : Google Scholar : | |
Wang LP, Han RM, Wu B, Luo MY, Deng YH, Wang W, Huang C, Xie X and Luo J: Mst1 silencing alleviates hypertensive myocardial injury associated with the augmentation of microvascular endothelial cell autophagy. Int J Mol Med. 50:1462022. View Article : Google Scholar : PubMed/NCBI | |
Sobrevia L, Aiello EA and Contreras P: Mechanisms of endothelial dysfunction and cardiovascular system adaptation. Curr Vasc Pharmacol. 20:201–204. 2022. View Article : Google Scholar : PubMed/NCBI | |
Tong X, Dang X, Liu D, Wang N, Li M, Han J, Zhao J, Wang Y, Huang M, Yang Y, et al: Exosome-derived circ_0001785 delays atherogenesis through the ceRNA network mechanism of miR-513a-5p/TGFBR3. J Nanobiotechnology. 21:3622023. View Article : Google Scholar : PubMed/NCBI | |
Yu F, Zhang Y, Wang Z, Gong W and Zhang C: Hsa_ circ_0030042 regulates abnormal autophagy and protects atherosclerotic plaque stability by targeting eIF4A3. Theranostics. 11:5404–5417. 2021. View Article : Google Scholar : | |
Gao W, Li C, Yuan J, Zhang Y, Liu G, Zhang J, Shi H, Liu H and Ge J: Circ-MBOAT2 Regulates Angiogenesis via the miR-495/ NOTCH1 axis and associates with myocardial perfusion in patients with coronary chronic total occlusion. Int J Mol Sci. 25:7932024. View Article : Google Scholar | |
Wong D, Turner AW and Miller CL: Genetic insights into smooth muscle cell contributions to coronary artery disease. Arterioscler Thromb Vasc Biol. 39:1006–1017. 2019. View Article : Google Scholar : PubMed/NCBI | |
Low EL, Baker AH and Bradshaw AC: TGFβ, smooth muscle cells and coronary artery disease: A review. Cell Signal. 53:90–101. 2019. View Article : Google Scholar : | |
Cao G, Xuan X, Hu J, Zhang R, Jin H and Dong H: How vascular smooth muscle cell phenotype switching contributes to vascular disease. Cell Commun Signal. 20:1802022. View Article : Google Scholar : PubMed/NCBI | |
Milutinović A, Šuput D and Zorc-Pleskovič R: Pathogenesis of atherosclerosis in the tunica intima, media, and adventitia of coronary arteries: An updated review. Bosn J Basic Med Sci. 20:21–30. 2020. | |
Schnack L, Sohrabi Y, Lagache SMM, Kahles F, Bruemmer D, Waltenberger J and Findeisen HM: Mechanisms of trained innate immunity in oxLDL primed human coronary smooth muscle cells. Front Immunol. 10:132019. View Article : Google Scholar : PubMed/NCBI | |
Lacolley P, Regnault V, Segers P and Laurent S: Vascular smooth muscle cells and arterial stiffening: Relevance in development, aging, and disease. Physiol Rev. 97:1555–1617. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang G, Liu Z, Deng J, Liu L, Li Y, Weng S, Guo C, Zhou Z, Zhang L, Wang X, et al: Smooth muscle cell fate decisions decipher a high-resolution heterogeneity within atherosclerosis molecular subtypes. J Transl Med. 20:5682022. View Article : Google Scholar : PubMed/NCBI | |
Wang Z, Wang H, Guo C, Yu F, Zhang Y, Qiao L, Zhang H and Zhang C: Role of hsa_circ_0000280 in regulating vascular smooth muscle cell function and attenuating neointimal hyperplasia via ELAVL1. Cell Mol Life Sci. 80:32022. View Article : Google Scholar : PubMed/NCBI | |
Dai H, Zhao N and Zheng Y: CircLDLR modulates the proliferation and apoptosis of vascular smooth muscle cells in coronary artery disease through miR-26-5p/KDM6A Axis. J Cardiovasc Pharmacol. 80:132–139. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zeng Z, Xia L, Fan S, Zheng J, Qin J, Fan X, Liu Y, Tao J, Liu Y, Li K, et al: Circular RNA CircMAP3K5 Acts as a MicroRNA-22-3p Sponge to Promote Resolution of Intimal Hyperplasia Via TET2-Mediated smooth muscle cell differentiation. Circulation. 143:354–371. 2021. View Article : Google Scholar | |
Mao YY, Wang JQ, Guo XX, Bi Y and Wang CX: Circ-SATB2 upregulates STIM1 expression and regulates vascular smooth muscle cell proliferation and differentiation through miR-939. Biochem Biophys Res Commun. 505:119–125. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Li H, Zheng Z and Li Y: Hsa_circ_0031891 targets miR-579-3p to enhance HMGB1 expression and regulate PDGF-BB-induced human aortic vascular smooth muscle cell proliferation, migration, and dedifferentiation. Naunyn Schmiedebergs Arch Pharmacol. 397:1093–1104. 2024. View Article : Google Scholar | |
Zhong W, Wang L and Xiong L: Circ_0006251 mediates the proliferation and apoptosis of vascular smooth muscle cells in CAD via enhancing TET3 and PPM1B expression. Cell Mol Biol (Noisy-le-grand). 69:34–39. 2023. View Article : Google Scholar : PubMed/NCBI | |
Medina-Leyte DJ, Zepeda-García O, Domínguez-Pérez M, González-Garrido A, Villarreal-Molina T and Jacobo-Albavera L: Endothelial dysfunction, inflammation and coronary artery disease: Potential biomarkers and promising therapeutical approaches. Int J Mol Sci. 22:38502021. View Article : Google Scholar : PubMed/NCBI | |
Bazoukis G, Stavrakis S and Armoundas AA: Vagus nerve stimulation and inflammation in cardiovascular disease: A State-of-the-Art Review. J Am Heart Assoc. 12:e0305392023. View Article : Google Scholar : PubMed/NCBI | |
Bhattacharya P, Kanagasooriyan R and Subramanian M: Tackling inflammation in atherosclerosis: Are we there yet and what lies beyond? Curr Opin Pharmacol. 66:1022832022. View Article : Google Scholar : PubMed/NCBI | |
Prati F, Marco V, Paoletti G and Albertucci M: Coronary inflammation: Why searching, how to identify and treat it. Eur Heart J Suppl. 22(Suppl E): E121–E124. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S and Liu C: Macrophages in cardiovascular diseases: Molecular mechanisms and therapeutic targets. Signal Transduct Target Ther. 9:1302024. View Article : Google Scholar : PubMed/NCBI | |
Matter MA, Paneni F, Libby P, Frantz S, Stähli BE, Templin C, Mengozzi A, Wang YJ, Kündig TM, Räber L, et al: Inflammation in acute myocardial infarction: The good, the bad and the ugly. Eur Heart J. 45:89–103. 2024. View Article : Google Scholar : | |
Li Y and Wang B: Circular RNA circCHFR downregulation protects against oxidized low-density lipoprotein-induced endothelial injury via regulation of microRNA-15b-5p/growth arrest and DNA damage inducible gamma. Bioengineered. 13:4481–4492. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ji P, Song X and Lv Z: Knockdown of circ_0004104 alleviates oxidized low-density lipoprotein-induced vascular endothelial cell injury by regulating miR-100/TNFAIP8 Axis. J Cardiovasc Pharmacol. 78:269–279. 2021. View Article : Google Scholar : PubMed/NCBI | |
Rafiq M, Dandare A, Javed A, Liaquat A, Raja AA, Awan HM, Khan MJ and Naeem A: Competing Endogenous RNA Regulatory Networks of hsa_circ_0126672 in pathophysiology of coronary heart disease. Genes (Basel). 14:5502023. View Article : Google Scholar : PubMed/NCBI | |
Dandare A, Rafiq M, Liaquat A, Raja AA and Khan MJ: Identification of hsa_circ_0092576 regulatory network in the pathogenesis of coronary heart disease. Genes Dis. 10:26–28. 2023. View Article : Google Scholar : PubMed/NCBI | |
Liu X, Yao X and Chen L: Expanding roles of circRNAs in cardiovascular diseases. Noncoding RNA Res. 9:429–436. 2024. View Article : Google Scholar : PubMed/NCBI | |
Tang Y, Bao J, Hu J, Liu L and Xu DY: Circular RNA in cardiovascular disease: Expression, mechanisms and clinical prospects. J Cell Mol Med. 25:1817–1824. 2021. View Article : Google Scholar : | |
Vilades D, Martínez-Camblor P, Ferrero-Gregori A, Bär C, Lu D, Xiao K, Vea À, Nasarre L, Sanchez Vega J, Leta R, et al: Plasma circular RNA hsa_circ_0001445 and coronary artery disease: Performance as a biomarker. FASEB J. 34:4403–4414. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sonnenschein K, Wilczek AL, de Gonzalo-Calvo D, Pfanne A, Derda AA, Zwadlo C, Bavendiek U, Bauersachs J, Fiedler J and Thum T: Serum circular RNAs act as blood-based biomarkers for hypertrophic obstructive cardiomyopathy. Sci Rep. 9:203502019. View Article : Google Scholar | |
Bahn JH, Zhang Q, Li F, Chan TM, Lin X, Kim Y, Wong DT and Xiao X: The landscape of microRNA, Piwi-interacting RNA, and circular RNA in human saliva. Clin Chem. 61:221–230. 2015. View Article : Google Scholar : | |
Wang S, Zhang K, Tan S, Xin J, Yuan Q, Xu H, Xu X, Liang Q, Christiani DC, Wang M, et al: Circular RNAs in body fluids as cancer biomarkers: The new frontier of liquid biopsies. Mol Cancer. 20:132021. View Article : Google Scholar : PubMed/NCBI | |
Wang W, Sun L, Huang MT, Quan Y, Jiang T, Miao Z and Zhang Q: Regulatory circular RNAs in viral diseases: applications in diagnosis and therapy. RNA Biol. 20:847–858. 2023. View Article : Google Scholar : PubMed/NCBI | |
Li MZ, Zhang JN, Ren F, Yin DL, Zhao XH and Liu K: Diagnostic value of circRNA in coronary heart disease: A meta-analysis. Biomark Med. 17:667–677. 2023. View Article : Google Scholar : PubMed/NCBI | |
Fu Y, He S, Li C, Gan X, Wang Y, Zhou Y, Jiang R, Zhang Q, Pan Y, Zhou H, et al: Detailed profiling of m6A modified circRNAs and synergistic effects of circRNA and environmental risk factors for coronary artery disease. Eur J Pharmacol. 951:1757612023. View Article : Google Scholar : PubMed/NCBI | |
He S, Fu Y, Li C, Wang Y, Zhou H, Jiang R, Zhang Q, Jia Q, Chen X and Jia EZ: Interaction between the expression of hsa_circRPRD1A and hsa_circHERPUD2 and classical coronary risk factors promotes the development of coronary artery disease. BMC Med Genomics. 16:1312023. View Article : Google Scholar : PubMed/NCBI | |
Zhong Q, Jin S, Zhang Z, Qian H, Xie Y, Yan P, He W and Zhang L: Identification and verification of circRNA biomarkers for coronary artery disease based on WGCNA and the LASSO algorithm. BMC Cardiovasc Disord. 24:3052024. View Article : Google Scholar : PubMed/NCBI | |
Zhang W, Cui J, Li L, Zhu T and Guo Z: Identification of Plasma Exosomes hsa_circ_0001360 and hsa_circ_0000038 as key biomarkers of coronary heart disease. Cardiol Res Pract. 2024:55571432024. View Article : Google Scholar : PubMed/NCBI | |
Zhao Z, Li X, Gao C, Jian D, Hao P, Rao L and Li M: Peripheral blood circular RNA hsa_circ_0124644 can be used as a diagnostic biomarker of coronary artery disease. Sci Rep. 7:399182017. View Article : Google Scholar : PubMed/NCBI | |
Huang S, Zeng Z, Sun Y, Cai Y, Xu X, Li H and Wu S: Association study of hsa_circ_0001946, hsa-miR-7-5p and PARP1 in coronary atherosclerotic heart disease. Int J Cardiol. 328:1–7. 2021. View Article : Google Scholar | |
Tong X, Zhao X, Dang X, Kou Y and Kou J: circRNA, a novel diagnostic biomarker for coronary heart disease. Front Cardiovasc Med. 10:10706162023. View Article : Google Scholar : PubMed/NCBI | |
Ji WF, Chen JX, He S, Zhou YQ, Hua L, Hou C, Zhang S, Gan XK, Wang YJ, Zhou HX, et al: Characteristics of circular RNAs expression of peripheral blood mononuclear cells in humans with coronary artery disease. Physiol Genomics. 53:349–357. 2021. View Article : Google Scholar : PubMed/NCBI | |
Miao L, Yin RX, Zhang QH, Liao PJ, Wang Y, Nie RJ and Li H: A novel circRNA-miRNA-mRNA network identifies circ-YOD1 as a biomarker for coronary artery disease. Sci Rep. 9:183142019. View Article : Google Scholar : PubMed/NCBI | |
Ward Z, Schmeier S, Pearson J, Cameron VA, Frampton CM, Troughton RW, Doughty RN, Richards AM and Pilbrow AP: Identifying candidate circulating RNA markers for coronary artery disease by deep RNA-Sequencing in human plasma. Cells. 11:31912022. View Article : Google Scholar : PubMed/NCBI | |
Dergunova LV, Vinogradina MA, Filippenkov IB, Limborska SA and Dergunov AD: Circular RNAs variously participate in coronary atherogenesis. Curr Issues Mol Biol. 45:6682–6700. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Xu GE, Spanos M, Li G, Lei Z, Sluijter JPG and Xiao J: Circular RNAs in cardiovascular diseases: Regulation and therapeutic applications. Research (Wash D C). 6:00382023.PubMed/NCBI | |
Goina CA, Goina DM, Farcas SS and Andreescu NI: The role of circular RNA for early diagnosis and improved management of patients with cardiovascular diseases. Int J Mol Sci. 25:29862024. View Article : Google Scholar : PubMed/NCBI | |
Long Q, Lv B, Jiang S and Lin J: The landscape of circular RNAs in cardiovascular diseases. Int J Mol Sci. 24:45712023. View Article : Google Scholar : PubMed/NCBI | |
Chen W, Xu J, Wu Y, Liang B, Yan M, Sun C, Wang D, Hu X, Liu L, Hu W, et al: The potential role and mechanism of circRNA/miRNA axis in cholesterol synthesis. Int J Biol Sci. 19:2879–2896. 2023. View Article : Google Scholar : PubMed/NCBI | |
Neu CT, Gutschner T and Haemmerle M: Post-Transcriptional expression control in platelet biogenesis and function. Int J Mol Sci. 21:76142020. View Article : Google Scholar : PubMed/NCBI | |
Yu R, Yu Q, Li Z, Li J, Yang J, Hu Y, Zheng N, Li X, Song Y, Li J, et al: Transcriptome-wide map of N6-methyladenosine (m6A) profiling in coronary artery disease (CAD) with clopidogrel resistance. Clin Epigenetics. 15:1942023. View Article : Google Scholar : PubMed/NCBI | |
Zou Y, Wang Y, Yao Y, Wu Y, Lv C and Yin T: Platelet-derived circFAM13B associated with anti-platelet responsiveness of ticagrelor in patients with acute coronary syndrome. Thromb J. 22:532024. View Article : Google Scholar : PubMed/NCBI |