International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Cardiovascular diseases (CVDs), including heart failure (HF), hypertension, coronary heart disease (CHD), vascular calcification and others, are among the primary causes of health deterioration in humans worldwide. The Global Health Assessment 2019 report by the World Health Organization (1) indicated that heart disease has remained the leading cause of mortality globally for the past two decades. Improving the prognosis and course of treatment for CVDs requires early detection and diagnosis. Every year, there is an increase in the prevalence of CVDs despite advancements in therapy for controlling traditional risk factors. Furthermore, the number of CVD-associated mortalities is predicted to increase to ~23.6 million by 2030 as a result of aging and changing lifestyles (2,3). Thus, in-depth studies to broaden the current understanding of CVDs mechanisms, diagnosis and treatment are imperative.
Research on exosomes is part of a newly emerged field, which has attracted great attention due to their role in regulating intercellular communication. Exosomes are a subtype of extracellular vesicles with a typical size of ~100 nm, which can carry a range of lipids, carbohydrates, nucleic acids and proteins (4). Numerous CVDs are directly impacted by different circular RNAs (circRNAs), long noncoding RNAs (lncRNAs), messenger RNAs, short nucleolar RNAs and microRNAs (miRNA) transported by exosomes. Due to these properties, exosomes have also been proposed as potential diagnostic and/or prognostic biomarkers for CVD, as well as novel potential therapeutic modalities (5). Epigenetics regulates the function and expression levels of CVD-associated genes primarily through ncRNA regulation and DNA methylation, thereby influencing the progression of CVD (6). Exosome-mediated epigenetic regulation participates in the interaction between blood vessels and circulating cells, as well as in intercellular communication processes, and exosomes serve as biomarkers of cell activation (7).
The aim of the present study was to examine the molecular mechanism behind exosome-mediated epigenetic regulation in the development of CVDs, and to explore the potential roles of exosome-mediated epigenetic regulation in the diagnosis and treatment of CVDs.
Epigenetics is considered chromosomal changes that lead to stable genetic phenotypes in the absence of changes in the DNA sequence (8). Epigenetic markers are typically present in the first stages of CVD, and are valuable molecular markers for the diagnosis, evaluation and prediction of CVD response to treatment (9). Controlling these changed genes and proteins could also provide a novel focus for the management of heart disease, which is of considerable relevance for certain diseases that are becoming more common as the population ages and are yet challenging to treat clinically. This is because epigenetic modifications are reversible. In CVDs, through DNA methylation and ncRNA regulation, epigenetics primarily controls the function and expression level of associated genes, thus influencing the development of CVD (6,10).
Exosomes participate in intercellular communication among cells via direct communication with neighboring cells or via secretion of soluble factors under physiological and pathological conditions, which can extend the interaction over long distances (11,12). Exosome formation involves a number of processes, including synthesis of intraluminal vesicles, secretion and fusion with target cells, and cargo loading. The process of exosomal biogenesis commences with endosomal membrane invagination, resulting in the formation of multivesicular bodies (MVBs) in vivo (13). During this step, MVBs integrate proteins and cytoplasmic nucleic acids (14). On the one hand, when MVBs fuse with lysosomes, the encapsulated contents are degraded. When endosomes fuse with parental cell membranes, they are secreted in exosomes (15), as illustrated in Fig. 1. Since endosomes are the result of plasma membrane budding, the membrane protein orientation of the exosomes generated by this second invagination process is identical to that of the parental cells (16). Cargo sorting to exosomes is generally handled by endosomal sorting complexes required for transport (ESCRT) or sphingolipids (ESCRT-dependent or ESCRT-independent pathways). Cargo, such as proteins and RNA, can remain in the extracellular space unbroken if it is not part of the exosome lipid bilayer. Depending on the source, target and environment, target cells absorb exosomes through a variety of methods, such as membrane fusion, ligand receptor binding and endocytosis. Exosomes can enter adjacent target cells and bodily fluids, and even reach distant cells once they are secreted (8,9). This diagram illustrates the process of exosome biogenesis, including endosomal invagination, multivesicular body formation, cargo sorting, and exosome release. Exosomes facilitate intercellular communication by transferring genetic material (such as ncRNAs) to target cells through fusion, receptor binding or endocytosis. This process plays a key role in regulating gene expression and epigenetic changes in CVDs.
LncRNAs, circRNAs and miRNAs are examples of ncRNAs, which are generally not translated into proteins but function through the regulation of gene expression. Instead, they carry out their specific biological roles at the RNA level (6). MiRNAs are ~22 nucleotides in length, and are mainly transcribed by large primary miRNAs that are involved in the post-transcriptional regulation of gene expression, and are produced by RNA polymerase II. Pri-miRNAs comprise one or more hairpin (stem-loop) structures, each of which is consists of ~70 nucleotides (17). In addition to being the targets of epigenetic changes such as DNA methylation, miRNAs also function as histone deacetylase and DNA methyltransferase (DNMT) regulators (18). LncRNAs are noncoding RNAs that control gene expression patterns by modifying the accessibility of DNA and the structure of chromatin via molecular mechanisms such as scaffolding, decoy, guiding and signal transduction (19). CircRNAs are long, noncoding endogenous RNA molecules that have no 5′cap or 3′cluster (A) end, and have single-stranded covalently closed RNA rings that can be used as templates for protein synthesis. CircRNAs interact with miRNAs to regulate target gene expression and mRNA translation, and bind to functional proteins or RNA-binding proteins in order to control their transport and function (20,21), as illustrated in Table I.
The role of ncRNAs in CVDs has attracted great attention, particularly due to their potential in early disease detection and prevention. MiRNAs, lncRNAs and circRNAs offer new perspectives for understanding the progression of CVDs. Notably, lncRNAs can exert both protective and detrimental effects in CVD, and this dual regulatory role makes them a key focus on cardiovascular research, as illustrated in Fig. 2. First, miRNAs act as essential regulatory factors in diseases such as coronary heart disease, myocardial infarction and vascular calcification (22,23). Furthermore, miRNAs also play a critical role in atherosclerosis. In endothelial cells (ECs), miR-342-5p exerts an anti-atherosclerotic effect by suppressing inflammatory responses (24), whereas miR-92a promotes the progression of atherosclerosis (25). Similarly, in pulmonary arterial hypertension, miR-181a-5p and miR-324-5p confer protective effects by inhibiting pulmonary vascular remodeling, thereby slowing disease progression (26). Similar to miRNAs, lncRNAs in CVDs can exert both protective effects and promote disease progression. For instance, lncRNA LEENE has been shown to inhibit atherosclerosis in vitro through its interaction with miR-1 (23,27). In addition, specific lncRNAs, including APF, CAIF and Mirf, have been demonstrated to regulate cardiac autophagy, which may play a critical role during myocardial infarction-induced injury (28,29). Additionally, lncRNAs RNA-H19, LIPCAR and COL1A1 have been proposed as biomarkers for risk assessment and prediction of HF (30–32). Furthermore, circRNAs, as novel regulatory molecules, also play an important role in CVDs. For example, circRNAs can modulate the migration and proliferation of vascular smooth muscle cells (VSMCs) by interacting with miRNAs such as miRNA-939 and miRNA-221, thereby contributing to the development of atherosclerosis (33,34). In cardiomyocytes, circRNAs also play a crucial role, and are considered novel regulatory elements involved in hypertrophy, fibrosis, autophagy and apoptosis (35). Among them, mmu_circ_0005019 and circRNA CDYL have been identified as key regulators in the progression of HF (36,37). Notably, the overexpression of certain miRNAs may also lead to adverse effects. For example, excessive levels of miRNA-765, miRNA-483-3p and miRNA-143/145 can disrupt the renin-angiotensin system and increase the blood pressure (38). Similarly, circACTA2 interacts with miRNA-548F-5p, which targets the 3′-untranslated region of α-SMA mRNA to promote VSMC contraction and regulate vascular tone, thereby contributing to the development of hypertension (39). In summary, miRNAs, lncRNAs and circRNAs play multifaceted roles in CVDs. In-depth research on these RNA molecules may offer novel insights and potential biomarkers for the early diagnosis, treatment and prevention of CVDs.
miRNAs are the most abundant ncRNAs packaged within exosomes. Exosomal miRNAs derived from the heart and vascular system exhibit distinct expression patterns under various pathophysiological conditions. Their abundance and composition depend not only on the state of CVD but also on specific stimuli (40). The incorporation of miRNAs into exosomes relies on distinct selective packaging mechanisms, a process that is critical for their functional roles within the cardiovascular system (41). Previous studies in mouse models have further advanced the understanding of the role of miRNAs in cardiac development. Notably, cardiac-specific downregulation of the Dicer gene markedly disrupts miRNA synthesis in neonatal mice, resulting in atrial hypertrophy, cardiac dysfunction, ventricular remodeling and dilation, and ultimately early mortality (42,43). These findings underscore the critical importance of miRNAs in cardiac development and functional regulation. Among the numerous cardiac-specific miRNAs (44), miR-1 is particularly abundant, representing ~24% of the total miRNA population in the human heart and exhibiting pronounced enrichment in cardiac tissue (45). The miR-1 sequence is composed of two almost identical transcripts known as miR-1-1 and miR-1-2. While the loss of both transcripts results in mortality, the deletion of any of these transcripts causes defective cardiomyocyte differentiation and impaired cardiac electrical conduction (42,46). These findings indicate that miR-1 plays an indispensable role in normal cardiac development and functional maintenance. However, dysregulated expression of miRNAs, particularly abnormal levels within exosomes, may contribute to the onset of CVDs. For instance, excessive expression of certain miRNAs, such as miR-195 and miR-208, in vitro can lead to concentric ventricular hypertrophy and the development of heart failure (47,48). Therefore, measuring circulating miRNA levels and identifying specific exosomal miRNA signatures may provide critical information for the early diagnosis, monitoring and treatment of CVDs. Dysregulation of specific miRNAs, such as upregulation of miR-21 and miR-155 (49,50), or downregulation of miR-126 (51), is closely associated with the progression of cardiac atherosclerosis, and these miRNAs hold promise as potential biomarkers for cardiac defects, as summarized in Table I.
DNA methylation is a common modification in eukaryotic cells, which, by controlling DNA methyltransferases (DNMT), can transfer genetic information to the DNA of the offspring (52). Accumulating evidence has suggested that aberrant methylation status of candidate genes, such as AHRR (53), F2RL3 (54) and CDKN2B-AS1 (55), can be used as indicators to assess the progression of cardiovascular disorders, as illustrated in Fig. 2. The expression of potential genes linked to CHD, HF hypertension and other CVDs has been reported to be strongly correlated with DNA methylation (56), as illustrated in Table II.
Specifically, alterations at multiple gene methylation sites have been demonstrated to be strongly associated with CVDs. Previous studies have identified that three DNA methylation areas, including the SLC9A1 (57), SLC1A5 (58) and TNRC6C genes (59), are associated with CVDs. Among them, the methylation of one CpG (CG22304262) in SLC1A5 is strongly associated with incident CHD, and shows significant associations with diabetes, blood pressure and all-cause mortality (60–62). The primary role of SLC1A5 in the heart is to regulate glutamine uptake. However, in patients with HF, reduced SLC1A5 expression leads to impaired glutamine absorption, resulting in disrupted cardiac glutamine storage and an imbalance in glutamine homeostasis, which ultimately compromises myocardial function and health (63). In addition to SLC1A5 methylation, previous epigenomic studies revealed broader associations between differential DNA methylation patterns and atherosclerosis as well as CHD (64). For instance, specific loci such as AHRR (cg05575921) and F2RL3 (cg03636183) in blood-derived DNA have been linked to both smoking exposure and increased risk of incident CHD (65,66). Similarly, epigenome-wide studies have identified methylation signatures related to chronic inflammation and lipid metabolism that correlate with atherosclerosis progression (67,68). Importantly, these epigenetic markers (e.g., methylation of AHRR and LINE-1 elements) have been shown to provide greater predictive value for CHD than traditional risk factors alone (69). Additionally, alterations in the methylation levels of genes associated with acute myocardial infarction (AMI), such as Csf1r, Ptpn6, Map3k14 and Col6a1, further support the mechanistic role of DNA methylation in cardiovascular pathophysiology (70). Previous studies have shown that upregulation of thioredoxin-interacting protein is associated with myocardial ischemia/reperfusion (MI/R) injury (71), potentially through aberrant activation of the m6A methyltransferase complex component WTAP (72). In vivo experiments further demonstrated that exosome-mediated delivery of WTAP small interfering RNA effectively alleviated MI/R injury (73).
Furthermore, DNA methylation mediated by DNMT3a plays a critical role in maintaining the structural and metabolic homeostasis of human cardiomyocytes (74), thus underscoring its relevance in CVDs. Previous research has shown that DNMT3a deficiency alters the expression of contractile protein genes in cardiomyocytes, leading to mitochondrial damage and glucose metabolism disorders (75), which may represent a critical therapeutic target for cardiac diseases. Furthermore, extracellular vesicles in the circulation of patients with acute coronary syndrome are enriched in DNMTs, particularly showing markedly increased expression of the de novo methyltransferases DNMT3A and DNMT3B. These enzymes can modulate the methylome and signaling pathways of recipient cells, thereby contributing to CVDs (76). Further in vitro studies revealed that downregulation of DNMTs not only participates in the physiological regulation of cardiomyocytes but also plays a crucial role in pathological conditions such as cardiac hypertrophy (77). Notably, the DNMT inhibitor RG108 has been shown to attenuate pressure overload-induced cardiac hypertrophy in animal models, possibly by modulating epigenetic mechanisms in non-myocyte cells (78). A previous study showed that selenium supplementation exerts beneficial effects in alleviating HF symptoms (79). By reducing cardiomyocyte death and reactive oxygen species production, selenium not only helps mitigate HF, but also indirectly improves the epigenetic state of cardiomyocytes by inhibiting the DNMT2-mediated methylation of the glutathione peroxidase 1 gene promoter (80).
Beyond genes that directly affect cardiac health, DNA methylation also plays a critical role in other physiological systems. In mouse models, the loss of the Wnt receptor LRP6 in aortic VSMCs leads to increased methylation of GTPase-activating protein-binding proteins, which may be linked to the transcriptional responses that trigger arterial calcification (81). Furthermore, DNA methylation patterns in the placenta have also been found to be associated with maternal blood pressure and CVDs. A previous study revealed that mothers with a family history of hypertension exhibited lower global placental DNA methylation levels in placental samples, which as accompanied by higher mean arterial pressure, thus highlighting the potential role of placental epigenetics in the development of CVDs (82). Additionally, in the field of cancer research, DNMTs have been shown to play a crucial role in mediating tumor drug resistance and progression through exosome-based delivery mechanisms (83–85).
In summary, DNA methylation, as a key epigenetic regulatory mechanism, is likely to play an important role in the onset and progression of CVDs. However, the mechanisms underlying exosome-mediated DNA methylation in the cardiovascular system remain to be fully elucidated. Although these pathways have been extensively studied in cancer, such as hepatocellular carcinoma (86), colorectal cancer (87) and acute myeloid leukemia (88), as well as in other diseases including Alzheimer's disease (89), type 2 diabetes (90) and systemic lupus erythematosus (91), direct evidence in CVDs is relatively limited, highlighting the urgent need for systematic investigations to clarify their specific roles and clinical implications. Future research focusing on the role of exosomes in epigenetic regulation may provide novel insights into the pathogenesis of CVDs.
The homeostasis of the cardiovascular system is maintained through the interplay of various cell types, including cardiac fibroblasts, ECs, VSMCs, cardiomyocytes, immune cells and resident stem cells (92). These cells release exosomes that mediate intercellular signaling, thereby contributing to tissue repair, remodeling and the regulation of pathological responses in the cardiovascular system. Notably, exosomes are not merely structural vesicles, but functional carriers that transport epigenetic information and influence gene expression in recipient cells. Consequently, exosome-mediated epigenetic regulatory mechanisms are increasingly recognized as critical contributors to the onset and progression of CVDs. Previous studies have demonstrated that exosomes of different cellular origins, such as ECs, erythrocytes, platelets and leukocytes, exhibit donor cell-specific profiles of RNAs, proteins and modifying enzymes, thereby reflecting the physiological or pathological state of their source cells (93,94). For example, exosomes derived from ECs are essential for the phenotypic transition of VSMCs (95), while exosomal miRNA profiles in the plasma of atherosclerotic patients have been investigated as potential early biomarkers (59,60). These findings suggest that pathological alterations in the cellular microenvironment not only influence exosome abundance and secretion dynamics, but also profoundly reshape their epigenetic regulatory cargo.
CVDs, including HF, hypertension, atherosclerosis, atrial fibrillation and AMI, continue to be the primary causes worldwide rates of morbidity and mortality, despite continuous advancements in the clinical diagnosis and treatment of heart disease. Exosomes are involved in the movement and interchange of molecules that signal, which makes them a crucial component in controlling the course of CVDs, as shown in Table III. Furthermore, ncRNAs found in exosomes function as novel regulators in dyslipidemia, which raises the risk of CVD caused by atherosclerosis. For example, miR-26a is markedly downregulated in exosomes from obese mice and overweight individuals, and its restoration reverses multiple markers associated with dysregulated lipid metabolism, suggesting a pivotal role for exosomal miRNAs in the cardiometabolic axis. Importantly, exosome-mediated epigenetic mechanisms exhibit substantial heterogeneity across different CVD subtypes. For instance, in HF, exosomal ncRNAs may contribute to the regulation of myocardial autophagy and fibrosis, whereas, in atherosclerosis, they are more likely to influence inflammatory cell infiltration and endothelial repair processes. Exosomes associated with hypertension are often enriched in miRNAs involved in vascular tone regulation, whereas those related to AMI tend to carry factors that modulate cardiomyocyte apoptosis and stress responses. These differences underscore the importance of analyzing exosomal epigenetic functions in a disease-specific context, which represents a promising direction for precision cardiovascular therapy. Accordingly, the following section aimed to systematically explore the roles of exosomal ncRNAs and DNA methylation-related factors in the pathophysiological mechanisms of four major CVD subtypes, namely HF, hypertension, atherosclerosis and myocardial infarction, with the aim of providing a theoretical basis for understanding their regulatory networks and clinical potential.
In HF, exosomal miRNAs play a pivotal role in regulating myocardial remodeling, and exhibit potential as both biomarkers and therapeutic targets, as shown in Table III. In a rat model of chronic HF, exosomal miR-124 derived from human umbilical cord mesenchymal stem cells was found to target PRSS23 and suppress associated fibrotic signaling pathways, thereby reducing the expression of the endothelial marker CD31, alleviating myocardial ischemic injury, and promoting angiogenesis and cellular repair (96). A clinical study also highlighted the critical role of exosomal miRNAs in HF (97). Previous studies have shown that patients with HF and reduced ejection fraction exhibit elevated levels of exosomal miR-92b-5p (97,98). This miRNA correlates positively with left atrial diameter, systolic blood pressure and left ventricular end-diastolic diameter, while it correlates negatively with fractional shortening and left ventricular ejection fraction (97), suggesting its potential involvement in the structural and functional remodeling of the left-side of the heart. In addition, miR-425 and miR-744 have also exhibited beneficial effects in mitigating myocardial fibrosis. In angiotensin-induced models, overexpression of these two miRNAs effectively suppresses collagen production and fibrotic processes in fibroblasts, whereas their downregulation is accompanied by significant upregulation of α-SMA and COL1, further confirming their critical regulatory role in cardiac fibrosis (99). Therefore, miR-425 and miR-744, as exosome-delivered epigenetic regulators, represent promising therapeutic targets and potential novel tools for reversing cardiac remodeling in HF. Notably, the regulatory effects of exosomal miRNAs may be bidirectional. Previous research has reported that serum exosomal miR-27a levels are significantly reduced in patients with HF, and this decrease is closely associated with poor prognosis (100). Additionally, aberrant expression of miR-126 in peripheral blood mononuclear cells of patients with chronic HF has been shown to exacerbate cardiac injury (101). Collectively, exosomal miRNAs modulate myocardial fibrosis through multiple targets and mechanisms, thus playing a critical role in the structural and functional remodeling of the heart.
Hypertension is a major risk factor for cardiovascular events such as coronary artery disease, HF, stroke and myocardial infarction (102,103). In addition, its onset and progression are also governed by a variety of molecular and cellular processes. In recent years, exosome-mediated ncRNA signaling has been recognized as a key epigenetic regulatory mechanism linking these pathological processes (104,105). According to previous studies, numerous cellular and molecular processes, such as the renin-angiotensin-aldosterone system, endothelial dysfunction, vascular remodeling, oxidative stress, angiogenesis, VSMC proliferation and inflammation, are regulated by a number of exosomal ncRNAs, which ultimately lead to the development of hypertension (77,106).
Exosomal miRNAs can influence the development and occurrence of hypertension in a positive or negative manner. In spontaneously hypertensive rats (SHRs), compared with Wistar-Kyoto rats, 23 exosomal miRNAs were upregulated and 4 were downregulated, indicating a systemic alteration in circulating exosomal miRNA profiles under hypertensive conditions (107). For example, miR-155-5p was significantly downregulated in exosomes derived from adventitial fibroblasts of SHR aortas (1). This miRNA could suppress ACE/Ang II signaling, reduce oxidative stress and inflammation, and thereby inhibit VSMCs migration in SHRs (108). On the other hand, certain exosomal miRNAs have been shown to exert pro-hypertensive effects. Exosomal miR-27a derived from THP-1 monocytes could inhibit the phosphorylation of endothelial nitric oxide synthase in mesenteric arteries, and reduce Mas receptor expression in ECs, thereby impairing vasodilation and elevating blood pressure (109). Following Ang II treatment, the levels of intercellular adhesion molecule-1 in THP-1 cells increased, whereas exosomal miR-17 could downregulate its expression, thus modulating endothelial inflammatory responses and indirectly affecting the vascular tone (110). In addition, exosomal miR-106b-5p, released from macrophages, could be transferred to glomerular mesangial cells, where it suppressed the transcription factors Pde3b and E2f1, thus mediating tissue responses associated with inflammatory hypertension (111). A previous epidemiological study reported that extracellular vesicle-derived miRNAs, particularly miR-199a/b and miR-223-3p, exhibited significant interactions with elevated systolic blood pressure, thus potentially contributing to hypertension through mechanisms involving vascular function, inflammatory responses and oxidative stress (112). In summary, miRNAs play a critical regulatory role in the onset and progression of hypertension. They modulate vascular tone and remodeling through multiple pathways, such as exosomal miR-155 (113), which impairs endothelial nitric oxide signaling and promotes vascular inflammation, and exosomal miR-505 (114), which enhances vascular smooth muscle cell proliferation and migration, reflecting their unique and complex functional characteristics in hypertensive pathology. However, current research on miRNAs in hypertension remains largely preclinical (115), with a lack of systematic validation in large populations and limited progress in translational applications.
Atherosclerosis is a long-term immunoinflammatory disease characterized by the accumulation of lipid-rich particles in arterial plaques, which is one of the main causes of CVDs, with serious clinical consequences such as myocardial infarction and stroke. In recent years (116,117), exosome-mediated regulation of ncRNAs has been recognized as one of the key epigenetic mechanisms driving the development and progression of atherosclerosis (AS), as shown in Table III. Exosomal ncRNAs primarily regulate lipid metabolism, vascular inflammation and cell survival, which have crucial regulatory functions in the onset and advancement of atherosclerosis.
Numerous studies have demonstrated that, in both patients and animal models of atherosclerosis, miRNAs, serving as key epigenetic molecules within exosomes, can significantly influence the pathological progression of AS through changes in their expression levels (116,118). In patients with unstable angina, 28 miRNAs were upregulated, among which miR-19b showed the most pronounced increase. This miRNA may inhibit the transcriptional activity of the STAT3 signaling pathway, thereby suppressing the proliferation, migration and angiogenesis of ECs, ultimately delaying the formation and progression of unstable plaques (41). Dysfunction of ECs can lead to atherosclerosis by causing EC senescence, inflammation, hyperpermeability, oxidative stress and vasodilation abnormalities (119). Previous studies have shown that exosomal ncRNAs play a major role in this process. Human smooth muscle cells in the aorta produce miR-221/222, which inhibits autophagy in these cells by regulating the PTEN/Akt axis (120,121). Thrombin-stimulated platelet exosomes showed enhanced miR-21, miR-339 and miR-223 levels, and miR-223 could inhibit the expression of intercellular adhesion molecule-1 (ICAM-1) in inflammatory processes by regulating the MAPK/NF-κB axis (122). In addition, exosomal miR-146a modulates inflammatory responses in ECs by inhibiting interleukin-1 receptor-associated kinase 1 (123), whereas miR-4532 exacerbates EC injury through its interaction with SP1 (124). A previous study showed that upregulation of miR-155 can regulate the interaction between ECs and VSMCs, inhibiting EC migration, proliferation and reendothelialization (113). This, in turn, weakens the endothelial barrier, increases vascular wall permeability and promotes the progression of atherosclerotic plaque (125).
In addition, the pathophysiology of atherosclerosis is regulated by exosomal miRNAs derived from non-cardiomyocyte cell types, such as endothelial cell-derived exosomal miR-92a that promotes vascular inflammation (126), macrophage-derived exosomal miR-146a that modulates inflammatory signaling (127), and vascular smooth muscle cell-derived exosomal miR-221/222 that enhances cell proliferation and migration (128). Macrophages in particular possess the unique ability to engulf aggregated and oxidized low-density lipoprotein via micropinocytosis and scavenger receptor-mediated uptake, subsequently transforming into lipid-laden foam cells that constitute the core of atherosclerotic plaques. Exosomal miR-33a-5p, secreted by these cells, can regulate ABCA1 expression and promote apoA1-mediated cholesterol efflux, thereby affecting foam cell formation (129). miR-16-5p accelerates inflammation and oxidative stress in atherosclerosis by downregulating SMAD7, and promotes apoptosis (130), thereby facilitating the progression of atherosclerotic lesions. Endothelial dysfunction is one of the key initiating factors in the early stages of atherosclerosis. Previous in vitro studies have shown that circ_0124644 promotes endothelial injury by modulating the miR-149-5p/PAPP-A axis, highlighting its pathogenic role in the progression of AS and its potential as a target for clinical intervention (131). In addition, the miR-203-3p/cathepsin S pathway, which functions along the p38/MAPK axis, has been shown to reverse atherosclerosis (132). Neutrophil extracellular traps (NETs) have also been identified as contributors to the amplification of immune-inflammatory responses in AS (133,134). NETs are a network of densified chromatin, nuclear histones and granular antimicrobial proteins that are released by neutrophils in response to microbial antigens (135). Exosomal miR-146a released from macrophages upon oxidized low-density lipoprotein (oxLDL) stimulation can induce the formation of NETs and promote the progression of atherosclerosis (136). Following oxLDL treatment, human umbilical vein ECs (HUVECs) exhibit enhanced NF-κB pathway activation, which upregulates miR-505 expression. MiR-505 then targets SIRT3, leading to increased reactive oxygen species production and elevated NET formation (88). VSMCs participate in plaque formation during the early stages of AS and contribute to plaque stability in the later stages.
In addition to the aforementioned miRNAs, lncRNAs are also important epigenetic regulators, and have been shown to be significantly upregulated in both animal models and patients with atherosclerosis. Previous studies have shown that exosomal lncRNA LIPCAR regulates CDK2 and participates in the phenotypic transformation of VSMCs (137,138). MiR-106a-3p modulates apoptosis by targeting CASP9 (139), and the LINC01005/miR-128-3p/KLF4 axis influences VSMC migration and proliferation (140). These lncRNAs mediate intercellular communication via exosomes, and serve as key regulatory nodes in the evolution and stability of atherosclerotic plaques. In THP-1 cells, lncRNA GAS5 can induce and regulate apoptosis in both THP-1 monocytes and ECs (141). By contrast, EC-derived lncRNA MALAT1 exhibits bidirectional effects: On one hand, it promotes M2 macrophage polarization and protects against atherosclerosis (142), while, on the other hand, it may also facilitate the formation of NETs, thereby accelerating the progression of atherosclerosis (143). In addition, several studies have found that exosomal circRNA-0006896 is closely associated with the pathogenesis of atherosclerosis (144,145). It is significantly upregulated in patients with unstable atherosclerotic plaques, and its levels are positively correlated with triglycerides, low-density lipoprotein and C-reactive protein. Mechanistically, circRNA-0006896 may promote EC proliferation and migration by activating the NF-κB signaling pathway (144,146). Previous in vitro studies have shown that exosome levels are significantly elevated in the serum of patients with coronary artery disease, which can exacerbate inflammatory responses and apoptosis in HUVECs, thereby promoting endothelial injury and atherosclerosis progression (147,148). Conversely, exosomal circ_0001785 can alleviate endothelial damage through the miR-513a-5p/TGFBR3 axis, thus exhibiting a protective effect (149).
In summary, exosome-derived miRNAs, lncRNAs and circRNAs in atherosclerosis profoundly participate in immune inflammation, apoptosis, lipid metabolism and plaque stability by targeting multiple key signaling pathways, including MAPK, NF-κB, STAT3 and p38. These epigenetic factors not only hold promise as diagnostic biomarkers for AS, but also offer new strategies for intervening in plaque progression and enhancing therapeutic precision.
AMI is typically caused by sudden coronary artery occlusion and is characterized by myocardial cell necrosis and loss of function, which can have severe clinical consequences (150). Previous studies have shown that exosomal miRNAs play a pivotal regulatory role in the onset and progression of myocardial infraction (MI) by modulating a variety of pathological processes, including autophagy, inflammation, angiogenesis and apoptosis (151,152), as shown in Table III.
Multiple studies have found that the exosomal miRNA expression profile derived from cardiomyocytes is significantly altered in patients with MI. Notably, exosomal miR-125b, miR-499, miR-133, miR-22, miR-21 and miR-301, which originate from cardiomyocytes, are markedly upregulated following MI (153,154). These miRNAs are encapsulated within cardiomyocyte-derived exosomes and are broadly expressed in damaged myocardial regions. By inhibiting cardiomyocyte death, modulating local inflammatory responses and promoting ischemic tissue repair, they help alleviate pathological injury following MI and may partially contribute to the prevention of complications such as atherosclerosis (155,156). Furthermore, inflammation-associated miRNAs, such as miR-146a and miR-25-3p, are upregulated in the immune environment induced by MI. Notably, exosomal miR-301 is also elevated and has been shown to suppress autophagy in cardiomyocytes (109,157). At the translational level, exosomal miR-499 and miR-133a are significantly elevated in patients with MI compared to those with stable coronary artery disease or acute coronary syndrome, demonstrating their potential utility for clinical diagnosis and disease stratification (94). In addition, miR-26b-5p promotes ferroptosis by targeting SLC7A11, and its downregulation in exosomes from patients with AMI helps attenuate myocardial injury (158), thus offering new perspectives for anti-ferroptosis therapies.
In addition to those derived from cardiomyocytes, exosomal ncRNAs from other cell types also play important roles in the pathology of MI. For example, during ischemic episodes, the expression of miR-22 in mesenchymal stem cell-derived exosomes is reduced (159); however, in vivo supplementation of this miRNA can significantly attenuate cardiac fibrosis (160), highlighting its therapeutic potential after AMI. High expression of miR-21 in mesenchymal stem cell-derived exosomes can significantly inhibit EC apoptosis and promote angiogenesis, thereby exerting a protective effect on cardiomyocytes (161). In addition, lncRNAs are involved in the epigenetic regulation of MI. LncRNA BC002059 has been shown to reduce infarct size in mice with coronary artery ligation (162). Mechanistically, BC002059 exerts its cytoprotective effects by influencing the downstream target gene ABHD10 and subsequently regulating the expression of miR-19b-3p (163). The roles of other lncRNAs in the process of MI should not be overlooked, such as MALAT and H19 (164,165). After AMI, the levels of MALAT1 in exosomes released from cardiomyocytes are significantly increased under hyperoxic conditions. MALAT1 enhances neovascularization by suppressing miR-92a expression (117), thereby contributing to ischemic tissue repair. Atorvastatin treatment has been shown to induce an increase in lncRNA H19 release from mesenchymal stem cells (118). By contrast, under hypoxic conditions, cardiomyocyte-derived exosomal lncRNA AK139128 regulates cardiac fibrosis by affecting the proliferation, migration and apoptosis of cardiac fibroblasts (165). These findings suggest that exosomal lncRNAs not only play functional roles in response to ischemic injury, but may also serve as important molecular targets for the treatment of MI.
Overall, exosomal ncRNAs play critical roles in myocardial infarction as well as other CVDs, including hypertension, atherosclerosis and HF. Previous research in both animal models and clinical samples has identified numerous potential therapeutic targets, such as exosomal miR-92b-5p regulating cardiomyocyte apoptosis (166), lncRNA H19 enhancing the efficacy of mesenchymal stem cell-derived exosomes in myocardial infarction (167). However, there is considerable heterogeneity in the mechanisms, target pathways and functional roles of ncRNAs among different types of CVDs. In HF, exosomal miRNAs primarily regulate fibroblast activity and influence myocardial remodeling, demonstrating potential for anti-fibrotic effects and cardiac structural modulation. By contrast, exosomal ncRNAs associated with atherosclerosis are more focused on regulating endothelial function and lipid metabolism, reflecting microenvironment-specific regulatory characteristics within atherosclerotic lesions. By comparison, those involved in hypertension predominantly participate in the regulation of vascular tone and remodeling.
Although these findings reveal distinct mechanisms by which exosomal ncRNAs act in different CVDs, as shown in Table III, their clinical translation still faces important challenges. The majority of previous studies rely on animal models and in vitro experiments, with a lack of validation in large-scale human samples. Future research urgently requires multicenter clinical data to further clarify the specific mechanisms of exosomal ncRNAs in cardiovascular pathology.
Prognostic biomarkers may be employed for identification of diseases and/or prognostic indicators. The most crucial and essential qualities of effective biomarkers are their sensitivity, specificity, stability and relative noninvasive detection. Notably, recent studies (166,168) are focusing on exosomes as possible biomarkers due to their easy and affordable detection in multiple bodily fluids, and their ability to function as a protective transport system against enzymatic degradation. Exosomes possess the capacity to serve as helpful biomarkers for the early detection of CVDs, since their cargo for epigenetic pathways varies depending on the type of CVD (169,170). Therefore, exosomes may be used as biomarkers to assist diagnosis according to their abnormal manifestations in different CVDs.
Previous studies have shown that exosomal ncRNAs exhibit diverse biological functions within the cardiovascular system. Dicer's heart-specific downregulation in young mice degrades miRNAs, particularly miR-1, miR-133a, miR-208 and miR-499 (171,172), resulting in atrial expansion, ventricular remodeling and enlargement, reduced cardiac function, and early mortality (173,174). By contrast, overexpression of certain specific miRNAs in vitro can induce cardiac hypertrophy and HF (44,45), indicating that the balance of miRNA expression is essential for maintaining cardiac homeostasis (173). Therefore, exosomal miRNAs and other epigenetic cargos not only reflect disease states but may also serve as potential targets for therapeutic intervention.
As therapeutic agents, exosomes hold promise as viable delivery vehicles due to a range of advantages, including minimal immunogenicity and toxicity, high biocompatibility, efficient bio-barrier permeability and natural availability (174). It has been attempted to alter exosomes through genetic engineering for medicinal uses (175,176). The potential to use exosomes as carriers to deliver certain cargo to particular tissues or organs is made possible by the capacity to change isolated and purified exosomes with target molecules and surface-loaded mimics or inhibitors of epigenetic payload (172,177). This gene regulation points to exosome epigenetic cargo as a potentially effective treatment strategy for a range of human diseases.
Previous animal studies have demonstrated the therapeutic potential of exosomes in various CVD models (168,178,179). Engineered exosomes enriched with low-density lipoprotein receptor mRNA have been shown to effectively alleviate hypercholesterolemia in mice (180). Additionally, mesenchymal stem/stromal cell (MSC)-derived exosomes can reduce cardiomyocyte apoptosis after myocardial infarction by suppressing SOCS2 signaling via miRNA-185 (181–183). A previous study further confirmed that pharmacological agents can modulate exosomes to intervene in the development and progression of CVDs (165). Furthermore, a previous study found that phenol can attenuate inflammatory responses in HUVECs, which deaccelerates the progression of atherosclerosis, and decreases the expression levels of STAT3 and phosphorylated STAT3 in HUVECs by increasing the quantity of exosome miR-223 in HUVECs treated with THP-1 and exosomes (184). In an I/R injury model in mice, the injection of cortical diaphysial-derived exosomes decreased the size of the infarct (185). On the contrary, chrysin reduced miR-92a expression in HCAEC cells and accelerated the progression of atherosclerosis (186). Furthermore, exosomal miR-21-3p derived from nicotine-treated macrophages could bind to PTEN, thereby promoting VSMC migration and proliferation, and exacerbating the progression of atherosclerosis (187). In addition, exosomes have shown a promising potential in promoting cardiac regeneration. Multiple studies using models of ischemic heart disease have demonstrated that injection of mesenchymal stem cell-derived exosomes can effectively improve myocardial remodeling, enhance cardiomyocyte survival (188,189), and promote EC proliferation, thereby partially restoring cardiac function (190,191). This ‘cell-free therapy’ offers a safer and more controllable alternative to traditional cell transplantation.
In recent years, exosome-based therapeutic strategies have attracted increasing attention for the treatment of CVDs (192). Exosomes are nanoscale, membrane-bound vesicles secreted by cells, and their unique structural and functional properties make them highly promising as therapeutic carriers (176,193). The bilayer lipid membrane of exosomes effectively protects encapsulated bioactive molecules, such as miRNAs and proteins, enhancing their stability and biological activity in vivo (194). For example, compared with free curcumin, curcumin encapsulated in exosomes exhibits higher plasma concentrations and greater anti-inflammatory effects (195). Furthermore, as non-living particulate structures, exosomes offer greater controllability compared to viral vectors or live cell systems, and lack tumorigenic potential (196). In multiple studies, exosomes have demonstrated a favorable safety profile (197,198).
However, despite the numerous advantages of exosomes as therapeutic tools, their clinical application still faces notable challenges. First, safety concerns cannot be ignored. While exosomes derived from autologous cells are generally considered to have low immunogenicity, artificially engineered exosome delivery systems may still pose immunogenic risks, follow unintended pathways away from target recipient cell types, and result in unexpected off-target effects (199). More importantly, exosomes lack specific biodistribution in vivo. Once cleared from the bloodstream, exosomes tend to accumulate in organs such as the lungs, spleen and liver (200,201), which can result in off-target effects, reduced therapeutic efficacy and even potential toxicity. Due to the tendency of RNA to aggregate or degrade, loading specific RNA cargos into exosomes using conventional electroporation or sonication techniques presents remarkable technical challenges, and the efficacy of these approaches remains uncertain (202). In addition, there may be a risk of infection when these particles enter the human bloodstream. More challenging is the substantial variability in exosomal drug loading capacity and the lack of uniformity, making it difficult to optimize exosome dosing (140). Furthermore, the optimal therapeutic time window for exosome administration in vivo has not yet been determined, which further increases the uncertainty of clinical translation (203). Therefore, before advancing exosomes as RNA delivery vehicles for clinical use, it is essential to ensure their safety in vivo and to minimize immune responses or off-target effects. In addition, optimization of loading techniques is needed to improve RNA stability and achieve a uniform cargo distribution.
Furthermore, the industrialization and clinical translation of exosomes face important technical bottlenecks. Currently, ultracentrifugation is the most commonly used isolation method for exosomes; however, it is labor-intensive, inefficient, and has limited yield, thus making it difficult to meet the demands of large-scale clinical applications. On one hand, there is still a lack of unified and standardized Good Manufacturing Practice-grade manufacturing processes (204). On the other hand, exosomes derived from different cell sources show significant variations in composition, function and efficacy, leading to poor batch-to-batch reproducibility and substantial challenges in quality control. Furthermore, delivery efficiency remains one of the most critical issues. Exosomes are commonly administered for the treatment of CVDs via intravenous, intracoronary or intramyocardial injection; however, these approaches are often associated with procedural complexity or low cardiac retention rates in clinical practice. Although surface engineering modifications can enhance targeted delivery (205), exosomes still face challenges such as off-target effects, rapid clearance and the need for rigorous safety validation in the complex human physiological environment.
In summary, exosomes hold great therapeutic promise for CVDs, particularly due to their advantages in targeting specificity, delivery efficiency and epigenetic regulation. However, several key issues must be addressed before exosome-based therapies can be translated into clinical practice, including ensuring safety, improving manufacturing processes and enhancing consistency in therapeutic efficacy.
The present study aimed to review in detail the exosome-mediated epigenetic mechanisms, such as exosomal ncRNAs and exosome-mediated DNA methylation, and their role in CVDs. Concurrently, the present study examined the function of exosomal phylogenetic pathways in CVD detection and treatment. The miRNA cargo of exosomes is of particular importance, as these molecules have the capacity to control the expression of proteins. At present, research on the mechanisms by which exosomes influence cardiovascular epigenetics is still ongoing, with current studies mainly focusing on exosomal ncRNAs. Targeted approaches for the treatment of CVDs are feasible, as exosomes are capable of delivering therapeutic agents directly to damaged cells and tissues. The application of exosomes as delivery vehicles for CVD therapy is highly promising. Both animal studies and preliminary clinical trials have shown encouraging results, suggesting that exosome-based therapies could represent a breakthrough in CVD treatment. Given that CVD remains a leading cause of mortality worldwide, this strategy is particularly meaningful for high-risk populations, including those with comorbidities such as diabetes and hypertension, or individuals with a history of angina or myocardial infarction.
However, the clinical application of exosomes still faces multiple challenges. First, there is a lack of standardized protocols for exosome isolation, characterization and cargo detection, resulting in poor reproducibility of experimental results and efficacy validation across studies, which limits translational progress. Second, the low loading efficiency and instability of functional RNAs compromise the stability and controllability of exosome-based delivery systems. In addition, the non-specific biodistribution of exosomes in vivo leads to risks of rapid clearance and off-target effects, which highlights the need for improved delivery strategies such as surface modification and magnetic guidance to enhance targeting. Particularly in the cardiovascular field, achieving safe, stable and effective therapeutic delivery in complex microenvironments requires further systematic research and validation.
However, as research on exosomes continues to advance, their prospects in precision medicine remain highly promising. In particular, exosomes offer novel insights into epigenetic regulation and intercellular communication underlying the pathogenesis and progression of CVDs. Future studies should focus on both mechanistic exploration and the establishment of technical standards to facilitate the critical transition from laboratory research to clinical application.
Not applicable.
The present study was supported by the Jilin Province Science and Technology Development Plan Project (grant no. YDZJ202201ZYTS209); the Regional Joint Fund Project of National Natural Science Foundation of China (grant no. U24A20779); and the Science and Technology Development Plan of Jilin Province (grant no. YDZJ202301ZYTS337).
Not applicable.
Conceptualization and study design: LZ and JL. Writing of the first manuscript draft: YZ. Literature search, figure reparation (created with BioRender) and manuscript editing: WJ and YL. Data curation, writing, reviewing and editing: YZ and SZ. Supervision and project administration: LZ and JL. All authors contributed to the critical revision of the manuscript, read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
CVDs |
cardiovascular diseases |
|
HF |
heart failure |
|
CHD |
coronary heart disease |
|
circRNAs |
circular RNAs |
|
lncRNAs |
long noncoding RNAs |
|
MI/R |
myocardial ischemia/reperfusion |
|
miRNAs |
microRNAs |
|
ncRNAs |
noncoding RNAs |
|
MVBs |
multivesicular bodies |
|
ESCRT |
endosomal sorting complexes required for transport |
|
DNMT |
DNA methyltransferase |
|
ECs |
endothelial cells |
|
VSMCs |
vascular smooth muscle cells |
|
AMI |
acute myocardial infarction |
|
SHRs |
spontaneously hypertensive rats |
|
NETs |
neutrophilic extranuclear traps |
|
HUVECs |
human umbilical vein endothelial cells |
|
GBD 2019 Viewpoint Collaborators, . Five insights from the global burden of disease study 2019. Lancet. 396:1135–1159. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Shah R, Patel T and Freedman JE: Circulating extracellular vesicles in human disease. N Engl J Med. 379:958–966. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Théry C, Witwer KW, Aikawa E, Alcaraz JM, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, et al: Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 7:15357502018. View Article : Google Scholar : PubMed/NCBI | |
|
Sahoo S, Adamiak M, Mathiyalagan P, Kenneweg F, Kafert-Kasting S and Thum T: Therapeutic and diagnostic translation of extracellular vesicles in cardiovascular diseases. Circulation. 143:1426–1449. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Martin-Ventura JL, Roncal C, Orbe J and Blanco-Colio LM: Role of extracellular vesicles as potential diagnostic and/or therapeutic biomarkers in chronic cardiovascular diseases. Front Cell Dev Biol. 10:8138852022. View Article : Google Scholar : PubMed/NCBI | |
|
Shi Y, Zhang H, Huang S, Yin L, Wang F, Luo P and Huang H: Epigenetic regulation in cardiovascular disease: Mechanisms and advances in clinical trials. Signal Transduct Target Ther. 7:2002022. View Article : Google Scholar : PubMed/NCBI | |
|
Khan MI, Alsayed RKME, Choudhry H and Ahmad A: Exosome-mediated response to cancer therapy: Modulation of epigenetic machinery. Int J Mol Sci. 23:62222022. View Article : Google Scholar : PubMed/NCBI | |
|
Nasir A, Bullo MMH, Ahmed Z, Imtiaz A, Yaqoob E, Jadoon M, Ahmed H, Afreen A and Yaqoob S: Nutrigenomics: Epigenetics and cancer prevention: A comprehensive review. Crit Rev Food Sci Nutr. 60:1375–1387. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Schiano C, Benincasa G, Franzese M, Della Mura N, Pane K, Salvatore M and Napoli C: Epigenetic-sensitive pathways in personalized therapy of major cardiovascular diseases. Pharmacol Ther. 210:1075142020. View Article : Google Scholar : PubMed/NCBI | |
|
Hulshoff MS, Xu X, Krenning G and Zeisberg EM: Epigenetic regulation of Endothelial-to-mesenchymal transition in chronic heart disease. Arterioscler Thromb Vasc Biol. 38:1986–1996. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Chang W and Wang J: Exosomes and their noncoding RNA cargo are emerging as new modulators for diabetes mellitus. Cells. 8:8532019. View Article : Google Scholar : PubMed/NCBI | |
|
Mathieu M, Martin-Jaular L, Lavieu G and Théry C: Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 21:9–17. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Shao H, Im H, Castro CM, Breakefield X, Weissleder R and Lee H: New technologies for analysis of extracellular vesicles. Chem Rev. 118:1917–1950. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Raiborg C and Stenmark H: The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins. Nature. 458:445–452. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Li S, Li L, Li M, Guo C, Yao J and Mi S: Exosome and exosomal microRNA: Trafficking, sorting, and function. Genomics Proteomics Bioinformatics. 13:17–24. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Kalluri R: The biology and function of exosomes in cancer. J Clin Invest. 126:1208–1215. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Poddar S, Kesharwani D and Datta M: Interplay between the miRNome and the epigenetic machinery: Implications in health and disease. J Cell Physiol. 232:2938–2945. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Petrovic N and Ergun S: miRNAs as potential treatment targets and treatment options in cancer. Mol Diagn Ther. 22:157–168. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Quinn JJ and Chang HY: Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 17:47–62. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lou Z, Zhou R, Su Y, Liu C, Ruan W, Jeon CO, Han X, Lin C and Jia B: Minor and major circRNAs in virus and host genomes. J Microbiol. 59:324–331. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yang T, Long T, Du T, Chen Y, Dong Y and Huang ZP: Circle the cardiac remodeling with circRNAs. Front Cardiovasc Med. 8:7025862021. View Article : Google Scholar : PubMed/NCBI | |
|
Hall IF, Climent M, Viviani Anselmi C, Papa L, Tragante V, Lambroia L, Farina FM, Kleber ME, März W, Biguori C, et al: rs41291957 controls miR-143 and miR-145 expression and impacts coronary artery disease risk. EMBO Mol Med. 13:e140602021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang F, Chen Q, Wang W, Ling Y, Yan Y and Xia P: Hepatocyte-derived extracellular vesicles promote endothelial inflammation and atherogenesis via microRNA-1. J Hepatol. 72:156–166. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Hou Z, Qin X, Hu Y, Zhang X, Li G, Wu J, Li J, Sha J, Chen J, Xia J, et al: Longterm Exercise-derived exosomal miR-342-5p: A novel exerkine for cardioprotection. Circ Res. 124:1386–1400. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Chang YJ, Li YS, Wu CC, Wang KC, Huang TC, Chen Z and Chien S: Extracellular MicroRNA-92a mediates endothelial Cell-macrophage communication. Arterioscler Thromb Vasc Biol. 39:2492–2504. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sindi HA, Russomanno G, Satta S, Abdul-Salam VB, Jo KB, Qazi-Chaudhry B, Ainscough AJ, Szulcek R, Jan Bogaard H, Morgan CC, et al: Therapeutic potential of KLF2-induced exosomal microRNAs in pulmonary hypertension. Nat Commun. 11:11852020. View Article : Google Scholar : PubMed/NCBI | |
|
Miao Y, Ajami NE, Huang TS, Lin FM, Lou CH, Wang YT, Li S, Kang J, Munkacsi H, Maurya MR, et al: Enhancer-associated long non-coding RNA LEENE regulates endothelial nitric oxide synthase and endothelial function. Nat Commun. 9:2922018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang K, Liu CY, Zhou LY, Wang JX, Wang M, Zhao B, Zhao WK, Xu S, Fan LH, Zhang XJ, et al: APF lncRNA regulates autophagy and myocardial infarction by targeting miR-188-3p. Nat Commun. 6:67792015. View Article : Google Scholar : PubMed/NCBI | |
|
Liang H, Su X, Wu Q, Shan H, Lv L, Yu T, Zhao X, Sun J, Yang R, Zhang L, et al: LncRNA 2810403D21Rik/Mirf promotes ischemic myocardial injury by regulating autophagy through targeting Mir26a. Autophagy. 16:1077–1091. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Viereck J, Bührke A, Foinquinos A, Chatterjee S, Kleeberger JA, Xiao K, Janssen-Peters H, Batkai S, Ramanujam D, Kraft T, et al: Targeting muscle-enriched long non-coding RNA H19 reverses pathological cardiac hypertrophy. Eur Heart J. 41:3462–3474. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Omura J, Habbout K, Shimauchi T, Wu WH, Breuils-Bonnet S, Tremblay E, Martineau S, Nadeau V, Gagnon K, Mazoyer F, et al: Identification of long noncoding RNA H19 as a new biomarker and therapeutic target in right ventricular failure in pulmonary arterial hypertension. Circulation. 142:1464–1484. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Hua X, Wang YY, Jia P, Xiong Q, Hu Y, Chang Y, Lai S, Xu Y, Zhao Z, Song J, et al: Multi-level transcriptome sequencing identifies COL1A1 as a candidate marker in human heart failure progression. BMC Med. 18:22020. View Article : Google Scholar : PubMed/NCBI | |
|
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 | |
|
Shen L, Hu Y, Lou J, Yin S, Wang W, Wang Y, Xia Y and Wu W: CircRNA-0044073 is upregulated in atherosclerosis and increases the proliferation and invasion of cells by targeting miR-107. Mol Med Rep. 19:3923–3932. 2019.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 | |
|
Wu N, Li C, Xu B, Xiang Y, Jia X, Yuan Z, Wu L, Zhong L and Li Y: Circular RNA mmu_circ_0005019 inhibits fibrosis of cardiac fibroblasts and reverses electrical remodeling of cardiomyocytes. BMC Cardiovasc Disord. 21:3082021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M, Wang Z, Cheng Q, Wang Z, Lv X, Wang Z and Li N: Circular RNA (circRNA) CDYL induces myocardial regeneration by ceRNA after myocardial infarction. Med Sci Monit. 26:e9231882020.PubMed/NCBI | |
|
Leimena C and Qiu H: Non-Coding RNA in the pathogenesis, progression and treatment of hypertension. Int J Mol Sci. 19:9272018. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Yang Z, Zheng B, Zhang XH, Zhang ML, Zhao XS, Zhao HY, Suzuki T and Wen JK: A novel regulatory mechanism of smooth muscle α-Actin expression by NRG-1/circACTA2/miR-548f-5p Axis. Circ Res. 121:628–635. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yang K, Zhou Q, Qiao B, Shao B, Hu S, Wang G, Yuan W and Sun Z: Exosome-derived noncoding RNAs: Function, mechanism, and application in tumor angiogenesis. Mol Ther Nucleic Acids. 27:983–997. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Qian Z, Shen Q, Yang X, Qiu Y and Zhang W: The role of extracellular vesicles: An epigenetic view of the cancer microenvironment. Biomed Res Int. 2015:6491612015. View Article : Google Scholar : PubMed/NCBI | |
|
Radhakrishna U, Albayrak S, Zafra R, Baraa A, Vishweswaraiah S, Veerappa AM, Mahishi D, Saiyed N, Mishra NK, Guda C, et al: Placental epigenetics for evaluation of fetal congenital heart defects: Ventricular Septal Defect (VSD). PLoS One. 14:e02002292019. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Geng Q, Chen H, Zhang J, Cao C, Zhang F, Song J, Liu C and Liang W: The potential inhibitory effects of miR-19b on vulnerable plaque formation via the suppression of STAT3 transcriptional activity. Int J Mol Med. 41:859–867. 2018.PubMed/NCBI | |
|
Small EM and Olson EN: Pervasive roles of microRNAs in cardiovascular biology. Nature. 469:336–342. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Li Q, Song XW, Zou J, Wang GK, Kremneva E, Li XQ, Zhu N, Sun T, Lappalainen P, Yuan WJ, et al: Attenuation of microRNA-1 derepresses the cytoskeleton regulatory protein twinfilin-1 to provoke cardiac hypertrophy. J Cell Sci. 123:2444–2452. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Heidersbach A, Saxby C, Carver-Moore K, Huang Y, Ang YS, de Jong PJ, Ivey KN and Srivastava D: microRNA-1 regulates sarcomere formation and suppresses smooth muscle gene expression in the mammalian heart. Elife. 2:e013232013. View Article : Google Scholar : PubMed/NCBI | |
|
Van Rooij E, Sutherland LB, Liu N, Williams AH, McAnally J, Gerard RD, Richardson JA and Olson EN: A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc Natl Acad Sci USA. 103:18255–18260. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Benzoni P, Nava L, Giannetti F, Guerini G, Gualdoni A, Bazzini C, Milanesi R, Bucchi A, Baruscotti M and Barbuti A: Dual role of miR-1 in the development and function of sinoatrial cells. J Mol Cell Cardiol. 157:104–112. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ji R, Cheng Y, Yue J, Yang J, Liu X, Chen H, Dean DB and Zhang C: MicroRNA expression signature and antisense-mediated depletion reveal an essential role of MicroRNA in vascular neointimal lesion formation. Circ Res. 100:1579–1588. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Nazari-Jahantigh M, Wei Y, Noels H, Akhtar S, Zhou Z, Koenen RR, Heyll K, Gremse F, Kiessling F, Grommes J, et al: MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J Clin Invest. 122:4190–202. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Zernecke A, Bidzhekov K, Noels H, Shagdarsuren E, Gan L, Denecke B, Hristov M, Köppel T, Jahantigh MN, Lutgens E, et al: Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal. 2:ra812009. View Article : Google Scholar : PubMed/NCBI | |
|
Mattei AL, Bailly N and Meissner A: DNA methylation: A historical perspective. Trends Genet. 38:676–707. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu L, Zhu C, Wang J, Yang R and Zhao X: The association between DNA methylation of 6p21.33 and AHRR in blood and coronary heart disease in Chinese population. BMC Cardiovasc Disord. 22:3702022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Yang R, Burwinkel B, Breitling LP, Holleczek B, Schöttker B and Brenner H: F2RL3 methylation in blood DNA is a strong predictor of mortality. Int J Epidemiol. 43:1215–1225. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Guarrera S, Fiorito G, Onland-Moret NC, Russo A, Agnoli C, Allione A, Di Gaetano C, Mattiello A, Ricceri F, Chiodini P, et al: Gene-specific DNA methylation profiles and LINE-1 hypomethylation are associated with myocardial infarction risk. Clin Epigenetics. 7:1332015. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang W, Song M, Qu J and Liu GH: Epigenetic modifications in cardiovascular aging and diseases. Circ Res. 123:773–786. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Nakamura TY, Iwata Y, Arai Y, Komamura K and Wakabayashi S: Activation of Na+/H+ exchanger 1 is sufficient to generate Ca2+ signals that induce cardiac hypertrophy and heart failure. Circ Res. 103:891–899. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Chen S, Ma J, Chi J, Zhang B, Zheng X, Chen J and Liu J: Roles and potential clinical implications of tissue transglutaminase in cardiovascular diseases. Pharmacol Res. 177:1060852022. View Article : Google Scholar : PubMed/NCBI | |
|
Westerman K, Sebastiani P, Jacques P, Liu S, DeMeo D and Ordovás JM: DNA methylation modules associate with incident cardiovascular disease and cumulative risk factor exposure. Clin Epigenetics. 11:1422019. View Article : Google Scholar : PubMed/NCBI | |
|
GTEx Consortium; Laboratory, Data Analysis & Coordinating Center (LDACC)-Analysis Working Group; Statistical Methods groups-Analysis Working Group; Enhancing GTEx (eGTEx) groups; NIH Common Fund; NIH/NCI; NIH/NHGRI; NIH/NIMH; NIH/NIDA; Biospecimen Collection Source Site-NDRI et al., . Genetic effects on gene expression across human tissues. Nature. 550:204–213. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Walaszczyk E, Luijten M, Spijkerman AMW, Bonder MJ, Lutgers HL, Snieder H, Wolffenbuttel BHR and van Vliet-Ostaptchouk JV: DNA methylation markers associated with type 2 diabetes, fasting glucose and HbA1c levels: A systematic review and replication in a case-control sample of the Lifelines study. Diabetol. 61:354–368. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Richard MA, Huan T, Ligthart S, Gondalia R, Jhun MA, Brody JA, Irvin MR, Marioni R, Shen J, Tsai PC, et al: DNA methylation analysis identifies loci for blood pressure regulation. American. J Human Genet. 101:888–902. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Kennel PJ, Liao X, Saha A, Ji R, Zhang X, Castillero E, Brunjes D, Takayama H, Naka Y, Thomas T, et al: Impairment of myocardial glutamine homeostasis induced by suppression of the amino acid carrier SLC1A5 in failing myocardium. Circ Heart Fail. 12:e0063362019. View Article : Google Scholar : PubMed/NCBI | |
|
Fernández-Sanlés A, Sayols-Baixeras S, Curcio S, Subirana I, Marrugat J and Elosua R: DNA Methylation and Age-independent cardiovascular risk, an Epigenome-wide approach: The REGICOR study (REgistre GIroní del COR). Arterioscler Thromb Vasc Biol. 38:645–652. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Guarrera S, Fiorito G, Onland-Moret NC, Russo A, Agnoli C, Allione A, Di Gaetano C, Mattiello A, Ricceri F, Chiodini P, et al: Gene-specific DNA methylation profiles and LINE-1 hypomethylation are associated with myocardial infarction risk. Clin Epigenetics. 7:1332015. View Article : Google Scholar : PubMed/NCBI | |
|
Shenker NS, Polidoro S, van Veldhoven K, Sacerdote C, Ricceri F, Birrell MA, Belvisi MG, Brown R, Vineis P and Flanagan JM: Epigenome-wide association study in the European Prospective Investigation into Cancer and Nutrition (EPIC-Turin) identifies novel genetic loci associated with smoking. Hum Mol Genet. 22:843–851. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zaina S, Heyn H, Carmona FJ, Varol N, Sayols S, Condom E, Ramírez-Ruz J, Gomez A, Gonçalves I, Moran S and Esteller M: DNA methylation map of human atherosclerosis. Circ Cardiovasc Genet. 7:692–700. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Ligthart S, Marzi C, Aslibekyan S, Mendelson MM, Conneely KN, Tanaka T, Colicino E, Waite LL, Joehanes R, Guan W, et al: DNA methylation signatures of chronic low-grade inflammation are associated with complex diseases. Genome Biol. 17:2552016. View Article : Google Scholar : PubMed/NCBI | |
|
Navas-Acien A, Domingo-Relloso A, Subedi P, Riffo-Campos AL, Xia R, Gomez L, Haack K, Goldsmith J, Howard BV, Best LG, et al: Blood DNA methylation and incident coronary heart disease: Evidence from the strong heart study. JAMA Cardiol. 6:1237–1246. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Luo X, Hu Y, Shen J, Liu X, Wang T, Li L and Li J: Integrative analysis of DNA methylation and gene expression reveals key molecular signatures in acute myocardial infarction. Clin Epigenetics. 14:462022. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshioka J, Imahashi K, Gabel SA, Chutkow WA, Burds AA, Gannon J, Schulze PC, MacGillivray C, London RE, Murphy E and Lee RT: Targeted deletion of thioredoxin-interacting protein regulates cardiac dysfunction in response to pressure overload. Circ Res. 101:1328–1338. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Dorn LE, Lasman L, Chen J, Xu X, Hund TJ, Medvedovic M, Hanna JH, van Berlo JH and Accornero F: The N6-methyladenosine mRNA methylase METTL3 controls cardiac homeostasis and hypertrophy. Circulation. 139:533–545. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yin T, Wang N, Jia F, Wu Y, Gao L, Zhang J and Hou R: Exosome-based WTAP siRNA delivery ameliorates myocardial ischemia-reperfusion injury. Eur J Pharm Biopharm. 197:1142182024. View Article : Google Scholar : PubMed/NCBI | |
|
Madsen A, Krause J, Höppner G, Hirt MN, Tan WLW, Lim I, Hansen A, Nikolaev VO, Foo RSY, Eschenhagen T and Stenzig J: Hypertrophic signaling compensates for contractile and metabolic consequences of DNA methyltransferase 3A loss in human cardiomyocytes. J Mol Cell Cardiol. 154:115–123. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Madsen A, Höppner G, Krause J, Hirt MN, Laufer SD, Schweizer M, Tan WLW, Mosqueira D, Anene-Nzelu CG, Lim I, et al: An important role for DNMT3A-Mediated DNA methylation in cardiomyocyte metabolism and contractility. Circulation. 142:1562–1578. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Schiano C, Balbi C, Burrello J, Ruocco A, Infante T, Fiorito C, Panella S, Barile L, Mauro C, Vassalli G and Napoli C: De novo DNA methylation induced by circulating extracellular vesicles from acute coronary syndrome patients. Atherosclerosis. 354:41–52. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Fang X, Poulsen RR, Wang-Hu J, Shi O, Calvo NS, Simmons CS, Rivkees SA and Wendler CC: Knockdown of DNA methyltransferase 3a alters gene expression and inhibits function of embryonic cardiomyocytes. FASEB J. 30:3238–3255. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Stenzig J, Schneeberger Y, Löser A, Peters BS, Schaefer A, Zhao RR, Ng SL, Höppner G, Geertz B, Hirt MN, et al: Pharmacological inhibition of DNA methylation attenuates pressure overload-induced cardiac hypertrophy in rats. J Mol Cell Cardiol. 120:53–63. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Bomer N, Grote Beverborg N, Hoes MF, Streng KW, Vermeer M, Dokter MM, IJmker J, Anker SD, Cleland JGF, Hillege HL, et al: Selenium and outcome in heart failure. Eur J Heart Fail. 22:1415–1423. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu H, Wang X, Meng X, Kong Y, Li Y, Yang C, Guo Y, Wang X, Yang H, Liu Z and Wang F: Selenium supplementation improved cardiac functions by suppressing DNMT2-Mediated GPX1 Promoter DNA methylation in AGE-induced heart failure. Oxid Med Cell Longev. 2022:54029972022. View Article : Google Scholar : PubMed/NCBI | |
|
Ramachandran B, Stabley JN, Cheng SL, Behrmann AS, Gay A, Li L, Mead M, Kozlitina J, Lemoff A, Mirzaei H, et al: A GTPase-activating protein-binding protein (G3BP1)/antiviral protein relay conveys arteriosclerotic Wnt signals in aortic smooth muscle cells. J Biol Chem. 293:7942–7968. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Putra SED, Reichetzeder C, von Websky K, Neuber C, Halle H, Kleuser B, Krämer BK and Hocher B: Association between placental global DNA methylation and blood pressure during human pregnancy. J Hypertens. 40:1002–1009. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Shao H, Chung J, Lee K, Balaj L, Min C, Carter BS, Hochberg FH, Breakefield XO, Lee H, Weissleder R, et al: Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat Commun. 6:69992015. View Article : Google Scholar : PubMed/NCBI | |
|
Yu W, Zhang L, Wei Q and Shao A: O6-Methylguanine-DNA methyltransferase (MGMT): Challenges and new opportunities in glioma chemotherapy. Front Oncol. 9:15472019. View Article : Google Scholar : PubMed/NCBI | |
|
Cao YL, Zhuang T, Xing BH, Li N and Li Q: Exosomal DNMT1 mediates cisplatin resistance in ovarian cancer. Cell Biochem Funct. 35:296–303. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Chen M and Wong CM: The emerging roles of N6-methyladenosine (m6A) deregulation in liver carcinogenesis. Mol Cancer. 19:442020. View Article : Google Scholar : PubMed/NCBI | |
|
Li T, Hu PS, Zuo Z, Lin JF, Li X, Wu QN, Chen ZH, Zeng ZL, Wang F, Zheng J, et al: METTL3 facilitates tumor progression via an mA-IGF2BP2-dependent mechanism in colorectal carcinoma. Mol Cancer. 18:1122019. View Article : Google Scholar : PubMed/NCBI | |
|
Vu LP, Pickering BF, Cheng Y, Zaccara S, Nguyen D, Minuesa G, Chou T, Chow A, Saletore Y, MacKay M, et al: The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med. 23:1369–1376. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Han M, Liu Z, Xu Y, Liu X, Wang D, Li F, Wang Y and Bi J: Abnormality of m6A mRNA methylation is involved in Alzheimer's disease. Front Neurosci. 14:982020. View Article : Google Scholar : PubMed/NCBI | |
|
Fang J, Wu X, He J, Zhang H, Chen X, Zhang H, Novakovic B, Qi H and Yu X: RBM15 suppresses hepatic insulin sensitivity of offspring of gestational diabetes mellitus mice via m6A-mediated regulation of CLDN4. Mol Med. 29:232023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Wang X, Huang M, Luo A, Liu S, Cai M, Li W, Yuan S, Zheng Z, Liu X and Tang C: METTL3 facilitates kidney injury through promoting IRF4-mediated plasma cell infiltration via an m6A-dependent manner in systemic lupus erythematosus. BMC Med. 22:5112024. View Article : Google Scholar : PubMed/NCBI | |
|
Henning RJ: Cardiovascular exosomes and MicroRNAs in cardiovascular physiology and pathophysiology. J Cardiovasc Transl Res. 14:195–212. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Boulanger CM, Loyer X, Rautou PE and Amabile N: Extracellular vesicles in coronary artery disease. Nat Rev Cardiol. 14:259–272. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Waldenström A and Ronquist G: Role of exosomes in myocardial remodeling. Circ Res. 114:315–324. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Lao KH, Zeng L and Xu Q: Endothelial and smooth muscle cell transformation in atherosclerosis. Curr Opin Lipidol. 26:449–456. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Gao D, Wang S, Lin H, Wang Y and Xu W: Exosomal microRNA-1246 from human umbilical cord mesenchymal stem cells potentiates myocardial angiogenesis in chronic heart failure. Cell Biol Int. 45:2211–2225. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wu T, Chen Y, Du Y, Tao J, Li W, Zhou Z and Yang Z: Circulating exosomal miR-92b-5p is a promising diagnostic biomarker of heart failure with reduced ejection fraction patients hospitalized for acute heart failure. J Thorac Dis. 10:6211–6220. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wu T, Chen Y, Du Y, Tao J, Zhou Z and Yang Z: Serum exosomal MiR-92b-5p as a potential biomarker for acute heart failure caused by dilated cardiomyopathy. Cell Physiol Biochem. 46:1939–1950. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L, Liu J, Xu B, Liu YL and Liu Z: Reduced exosome miR-425 and miR-744 in the plasma represents the progression of fibrosis and heart failure. Kaohsiung J Med Sci. 34:626–633. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Xie Y, Hang JZ, Zhang N and Liu G: Clinical significance of MiR-27a expression in serum exosomes in patients with heart failure. Cell Mol Biol (Noisy-le-grand). 67:324–331. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Natrus L, Labudzynskyi D, Muzychenko P, Chernovol P and Klys Y: Plasma-derived exosomes implement miR-126-associated regulation of cytokines secretion in PBMCs of CHF patients in vitro. Acta Biomed. 93:e20220662022.PubMed/NCBI | |
|
Fuchs FD and Whelton PK: High blood pressure and cardiovascular disease. Hypertension. 75:285–292. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Bress AP, Colantonio LD, Cooper RS, Kramer H, Booth JN III, Odden MC, Bibbins-Domingo K, Shimbo D, Whelton PK, Levitan EB, et al: Potential cardiovascular disease events prevented with adoption of the 2017 American college of Cardiology/American heart association blood pressure guideline. Circulation. 139:24–36. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gan L, Guo X, Dong S and Sun C: The biology of exosomes and exosomal non-coding RNAs in cardiovascular diseases. Front Pharmacol. 16:15293752025. View Article : Google Scholar : PubMed/NCBI | |
|
Bhaskara M, Anjorin O and Wang M: Mesenchymal stem Cell-derived exosomal microRNAs in cardiac regeneration. Cells. 12:28152023. View Article : Google Scholar : PubMed/NCBI | |
|
Jayaseelan VP and Arumugam P: Exosomal microRNAs as a promising theragnostic tool for essential hypertension. Hypertens Res. 43:74–75. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ren XS, Tong Y, Qiu Y, Ye C, Wu N, Xiong XQ, Wang JJ, Han Y, Zhou YB, Zhang F, et al: MiR155-5p in adventitial fibroblasts-derived extracellular vesicles inhibits vascular smooth muscle cell proliferation via suppressing angiotensin-converting enzyme expression. J Extracell Vesicles. 9:16987952019. View Article : Google Scholar : PubMed/NCBI | |
|
Wu N, Ye C, Zheng F, Wan GW, Wu LL, Chen Q, Li YH, Kang YM and Zhu GQ: MiR155-5p inhibits cell migration and oxidative stress in vascular smooth muscle cells of spontaneously hypertensive rats. Antioxidants (Basel). 9:2042020. View Article : Google Scholar : PubMed/NCBI | |
|
Zou X, Wang J, Chen C, Tan X, Huang Y, Jose PA, Yang J and Zeng C: Secreted monocyte miR-27a, via mesenteric arterial Mas receptor-eNOS pathway, causes hypertension. Am J Hypertens. 33:31–42. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Osada-Oka M, Shiota M, Izumi Y, Nishiyama M, Tanaka M, Yamaguchi T, Sakurai E, Miura K and Iwao H: Macrophage-derived exosomes induce inflammatory factors in endothelial cells under hypertensive conditions. Hypertens Res. 40:353–360. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Oh J, Matkovich SJ, Riek AE, Bindom SM, Shao JS, Head RD, Barve RA, Sands MS, Carmeliet G, Osei-Owusu P, et al: Macrophage secretion of miR-106b-5p causes renin-dependent hypertension. Nat Commun. 11:47982020. View Article : Google Scholar : PubMed/NCBI | |
|
Rodosthenous RS, Kloog I, Colicino E, Zhong J, Herrera LA, Vokonas P, Schwartz J, Baccarelli AA and Prada D: Extracellular vesicle-enriched microRNAs interact in the association between long-term particulate matter and blood pressure in elderly men. Environ Res. 167:640–649. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ren Y and Zhang H: Emerging role of exosomes in vascular diseases. Front Cardiovasc Med. 10:10909092023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Hu L, Li Q, Ma J and Li H: Exosome-encapsulated miR-505 from ox-LDL-treated vascular endothelial cells aggravates atherosclerosis by inducing NET formation. Acta Biochim Biophys Sin (Shanghai). 51:1233–1241. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Shaheen N, Shaheen A, Diab RA and Desouki MT: MicroRNAs (miRNAs) role in hypertension: Pathogenesis and promising therapeutics. Ann Med Surg (Lond). 86:319–328. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Han J, Kang X, Su Y, Wang J, Cui X, Bian Y and Wu C: Plasma exosomes from patients with coronary artery disease promote atherosclerosis via impairing vascular endothelial junctions. Sci Rep. 14:298132024. View Article : Google Scholar : PubMed/NCBI | |
|
Guo B, Zhuang TT, Li CC, Li F, Shan SK, Zheng MH, Xu QS, Wang Y, Lei LM, Tang KX, et al: MiRNA-132/212 encapsulated by adipose tissue-derived exosomes Worsen atherosclerosis progression. Cardiovasc Diabetol. 23:3312024. View Article : Google Scholar : PubMed/NCBI | |
|
Wehbe Z, Wehbe M, Al Khatib A, Dakroub AH, Pintus G, Kobeissy F and Eid AH: Emerging understandings of the role of exosomes in atherosclerosis. J Cell Physiol. 240:e314542025. 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 | |
|
Alidadi M, Hjazi A, Ahmad I, Mahmoudi R, Sarrafha M, Reza Hosseini-Fard S and Ebrahimzade M: Exosomal non-coding RNAs: Emerging therapeutic targets in atherosclerosis. Biochem Pharmacol. 212:1155722023. View Article : Google Scholar : PubMed/NCBI | |
|
Li L, Wang Z, Hu X, Wan T, Wu H, Jiang W and Hu R: Human aortic smooth muscle cell-derived exosomal miR-221/222 inhibits autophagy via a PTEN/Akt signaling pathway in human umbilical vein endothelial cells. Biochem Biophys Res Commun. 479:343–350. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Tan M, Xiang Q, Zhou Z and Yan H: Thrombin-activated platelet-derived exosomes regulate endothelial cell expression of ICAM-1 via microRNA-223 during the thrombosis-inflammation response. Thromb Res. 154:96–105. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhong X, Gao W, Wu R, Liu H and Ge J: Dendritic cell exosome-shuttled miRNA146a regulates exosome-induced endothelial cell inflammation by inhibiting IRAK-1: A feedback control mechanism. Mol Med Rep. 20:5315–5323. 2019.PubMed/NCBI | |
|
Liu P, Wang S, Wang G, Zhao M, Du F, Li K, Wang L, Wu H, Chen J, Yang Y and Su G: Macrophage-derived exosomal miR-4532 promotes endothelial cells injury by targeting SP1 and NF-κB P65 signalling activation. J Cell Mol Med. 26:5165–5180. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng B, Yin WN, Suzuki T, Zhang XH, Zhang Y, Song LL, Jin LS, Zhan H, Zhang H, Li JS and Wen JK: Exosome-Mediated miR-155 transfer from smooth muscle cells to endothelial cells induces endothelial injury and promotes atherosclerosis. Mol Ther. 25:1279–1294. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Loyer X, Potteaux S, Vion AC, Guérin CL, Boulkroun S, Rautou PE, Ramkhelawon B, Esposito B, Dalloz M, Paul JL, et al: Inhibition of microRNA-92a prevents endothelial dysfunction and atherosclerosis in mice. Circ Res. 114:434–443. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Masson JJR, Cherry CL, Murphy NM, Sada-Ovalle I, Hussain T, Palchaudhuri R, Martinson J, Landay AL, Billah B, Crowe SM and Palmer CS: Polymorphism rs1385129 Within Glut1 Gene SLC2A1 is linked to poor CD4+ T cell recovery in Antiretroviral-Treated HIV+ individuals. Front Immunol. 9:9002018. View Article : Google Scholar : PubMed/NCBI | |
|
Lee MA, Raad N, Song MH, Yoo J, Lee M, Jang SP, Kwak TH, Kook H, Choi EK, Cha TJ, et al: The matricellular protein CCN5 prevents adverse atrial structural and electrical remodelling. J Cell Mol Med. 24:11768–11778. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Stamatikos A, Knight E, Vojtech L, Bi L, Wacker BK, Tang C and Dichek DA: Exosome-mediated transfer of Anti-miR-33a-5p from transduced endothelial cells enhances macrophage and vascular smooth muscle cell cholesterol efflux. Hum Gene Ther. 31:219–232. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen F, Li J, She J, Chen T and Yuan Z: Exosomal microRNA-16-5p from macrophage exacerbates atherosclerosis via modulating mothers against decapentaplegic homolog 7. Microvasc Res. 142:1043682022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang G, Li Y, Liu Z, Ma X, Li M, Lu Q, Li Y, Lu Z, Niu L, Fan Z and Lei Z: Circular RNA circ_0124644 exacerbates the ox-LDL-induced endothelial injury in human vascular endothelial cells through regulating PAPP-A by acting as a sponge of miR-149-5p. Mol Cell Biochem. 471:51–61. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lin B, Xie W, Zeng C, Wu X, Chen A, Li H, Jiang R and Li P: Transfer of exosomal microRNA-203-3p from dendritic cells to bone marrow-derived macrophages reduces development of atherosclerosis by downregulating Ctss in mice. Aging (Albany NY). 13:15638–15658. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Adamidis PS, Pantazi D, Moschonas IC, Liberopoulos E and Tselepis AD: Neutrophil extracellular traps (NETs) and atherosclerosis: Does hypolipidemic treatment have an effect? J Cardiovasc Dev Dis. 11:722024.PubMed/NCBI | |
|
Sano M, Maejima Y, Nakagama S, Shiheido-Watanabe Y, Tamura N, Hirao K, Isobe M and Sasano T: Neutrophil extracellular traps-mediated Beclin-1 suppression aggravates atherosclerosis by inhibiting macrophage autophagy. Front Cell Dev Biol. 10:8761472022. View Article : Google Scholar : PubMed/NCBI | |
|
Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y and Zychlinsky A: Neutrophil extracellular traps kill bacteria. Science. 303:1532–1535. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang YG, Song Y, Guo XL, Miao RY, Fu YQ, Miao CF and Zhang C: Exosomes derived from oxLDL-stimulated macrophages induce neutrophil extracellular traps to drive atherosclerosis. Cell Cycle. 18:2674–268. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Li D, Chen H, Wei X and Xu X: Expression of Long Noncoding RNA LIPCAR promotes cell proliferation, cell migration, and change in phenotype of vascular smooth muscle cells. Med Sci Monit. 25:7645–7651. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hu N, Zeng X, Tang F and Xiong S: Exosomal long non-coding RNA LIPCAR derived from oxLDL-treated THP-1 cells regulates the proliferation of human umbilical vein endothelial cells and human vascular smooth muscle cells. Biochem Biophys Res Commun. 575:65–72. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Zhang WL, Gu JJ, Sun YQ, Cui HZ, Bu JQ and Chen ZY: Exosome-mediated miR-106a-3p derived from ox-LDL exposed macrophages accelerated cell proliferation and repressed cell apoptosis of human vascular smooth muscle cells. Eur Rev Med Pharmacol Sci. 24:7039–7050. 2020.PubMed/NCBI | |
|
Zhang Z, Yi D, Zhou J, Zheng Y, Gao Z, Hu X, Ying G, Peng X and Wen T: Exosomal LINC01005 derived from oxidized low-density lipoprotein-treated endothelial cells regulates vascular smooth muscle cell phenotypic switch. Biofactors. 46:743–753. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Yang W, Guo Y, Chen W, Zheng P, Zeng J and Tong W: Exosomal lncRNA GAS5 regulates the apoptosis of macrophages and vascular endothelial cells in atherosclerosis. PLoS One. 12:e01854062017. View Article : Google Scholar : PubMed/NCBI | |
|
Huang C, Han J, Wu Y, Li S, Wang Q, Lin W and Zhu J: Exosomal MALAT1 derived from oxidized low-density lipoprotein-treated endothelial cells promotes M2 macrophage polarization. Mol Med Rep. 18:509–515. 2018.PubMed/NCBI | |
|
Gao H, Wang X, Lin C, An Z, Yu J, Cao H, Fan Y and Liang X: Exosomal MALAT1 derived from ox-LDL-treated endothelial cells induce neutrophil extracellular traps to aggravate atherosclerosis. Biol Chem. 401:367–376. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wen Y, Chun Y, Lian ZQ, Yong ZW, Lan YM, Huan L, Xi CY, Juan LS, Qing ZW, Jia C and Ji ZH: circRNA-0006896-miR1264-DNMT1 axis plays an important role in carotid plaque destabilization by regulating the behavior of endothelial cells in atherosclerosis. Mol Med Rep. 23:3112021. View Article : Google Scholar : PubMed/NCBI | |
|
Lu Z, Tang H, Li S, Zhu S, Li S and Huang Q: Role of circulating exosomes in cerebrovascular diseases: A comprehensive review. Curr Neuropharmacol. 21:1575–1593. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yu Z, Huang Q, Zhang Q, Wu H and Zhong Z: CircRNAs open a new era in the study of cardiovascular disease (review). Int J Mol Med. 47:49–64. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang P, Liang T, Wang X, Wu T, Xie Z, Yu Y and Yu H: Serum-derived exosomes from patients with coronary artery disease induce endothelial injury and inflammation in human umbilical vein endothelial cells. Int Heart J. 62:396–406. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zarà M, Campodonico J, Cosentino N, Biondi ML, Amadio P, Milanesi G, Assanelli E, Cerri S, Biggiogera M, Sandrini L, et al: Plasma exosome profile in ST-Elevation myocardial infarction patients with and without Out-of-Hospital cardiac arrest. Int J Mol Sci. 22:80652021. 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 | |
|
Byrne RA, Rossello X, Coughlan JJ, Barbato E, Berry C, Chieffo A, Claeys MJ, Dan GA, Dweck MR, Galbraith M, et al: 2023 ESC Guidelines for the management of acute coronary syndromes. Eur Heart J. 44:3720–382. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ong SG, Lee WH, Huang M, Dey D, Kodo K, Sanchez-Freire V, Gold JD and Wu JC: Cross talk of combined gene and cell therapy in ischemic heart disease: role of exosomal microRNA transfer. Circulation. 130 (Suppl 1):S60–S69. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Saparov A, Ogay V, Nurgozhin T, Chen WCW, Mansurov N, Issabekova A and Zhakupova J: Role of the immune system in cardiac tissue damage and repair following myocardial infarction. Inflamm Res. 66:739–751. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Yang R, Guo B, Zhang H, Zhang H, Liu S and Li Y: Exosomal miR-301 derived from mesenchymal stem cells protects myocardial infarction by inhibiting myocardial autophagy. Biochem Biophys Res Commun. 514:323–332. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
De Rosa S, Fichtlscherer S, Lehmann R, Assmus B, Dimmeler S and Zeiher AM: Transcoronary concentration gradients of circulating microRNAs. Circulation. 124:1936–1944. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Peng Y, Zhao JL, Peng ZY, Xu WF and Yu GL: Exosomal miR-25-3p from mesenchymal stem cells alleviates myocardial infarction by targeting pro-apoptotic proteins and EZH2. Cell Death Dis. 11:3172020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W, Zheng Y, Wang M, Yan M, Jiang J and Li Z: Exosomes derived miR-126 attenuates oxidative stress and apoptosis from ischemia and reperfusion injury by targeting ERRFI1. Gene. 690:75–80. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Pan J, Alimujiang M, Chen Q, Shi H and Luo X: Exosomes derived from miR-146a-modified adipose-derived stem cells attenuate acute myocardial infarction-induced myocardial damage via downregulation of early growth response factor 1. J Cell Biochem. 120:4433–4443. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Li H, Ding J, Liu W, Wang X, Feng Y, Guan H and Chen Z: Plasma exosomes from patients with acute myocardial infarction alleviate myocardial injury by inhibiting ferroptosis through miR-26b-5p/SLC7A11 axis. Life Sci. 322:1216492023. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Y, Huang W, Wani M, Yu X and Ashraf M: Ischemic preconditioning potentiates the protective effect of stem cells through secretion of exosomes by targeting Mecp2 via miR-22. PLoS One. 9:e886852014. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Huang W, Xu R, Nie Y, Cao X, Meng J, Xu X, Hu S and Zheng Z: MicroRNA-24 regulates cardiac fibrosis after myocardial infarction. J Cell Mol Med. 16:2150–2160. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Wang K, Jiang Z, Webster KA, Chen J, Hu H, Zhou Y, Zhao J, Wang L, Wang Y, Zhong Z, et al: Enhanced Cardioprotection by human endometrium mesenchymal stem cells driven by exosomal MicroRNA-21. Stem Cells Transl Med. 6:209–222. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Liao B, Dong S, Xu Z, Gao F, Zhang S and Liang R: MiR-19b-3p regulated by BC002059/ABHD10 axis promotes cell apoptosis in myocardial infarction. Biol Direct. 17:202022. View Article : Google Scholar : PubMed/NCBI | |
|
Shyu KG, Wang BW, Fang WJ, Pan CM and Lin CM: Hyperbaric oxygen-induced long non-coding RNA MALAT1 exosomes suppress MicroRNA-92a expression in a rat model of acute myocardial infarction. J Cell Mol Med. 24:12945–12954. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Huang P, Wang L, Li Q, Tian X, Xu J, Xu J, Xiong Y, Chen G, Qian H, Jin C, et al: Atorvastatin enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction via up-regulating long non-coding RNA H19. Cardiovasc Res. 116:353–367. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L and Zhang J: Exosomal lncRNA AK139128 Derived from hypoxic cardiomyocytes promotes apoptosis and inhibits cell proliferation in cardiac fibroblasts. Int J Nanomedicine. 15:3363–3376. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng M, Yang J, Zhao X, Zhang E, Zeng Q, Yu Y, Yang L, Wu B, Yi G, Mao X, et al: Circulating myocardial microRNAs from infarcted hearts are carried in exosomes and mobilise bone marrow progenitor cells. Nat Commun. 10:9592019. View Article : Google Scholar : PubMed/NCBI | |
|
Huang P, Wang L, Li Q, Tian X, Xu J, Xu J, Xiong Y, Chen G, Qian H, Jin C, et al: Atorvastatin enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction via up-regulating long non-coding RNA H19. Cardiovasc Res. 116:353–367. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Liu Y, Liu H and Tang WH: Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 9:192019. View Article : Google Scholar : PubMed/NCBI | |
|
Xu H, Du X, Xu J, Zhang Y, Tian Y, Liu G, Wang X, Ma M, Du W, Liu Y, et al: Pancreatic β cell microRNA-26a alleviates type 2 diabetes by improving peripheral insulin sensitivity and preserving β cell function. PLoS Biol. 18:e30006032020. View Article : Google Scholar : PubMed/NCBI | |
|
Fu Q, Jiang H, Wang Z, Wang X, Chen H, Shen Z, Xiao L, Guo X and Yang T: Injury factors alter miRNAs profiles of exosomes derived from islets and circulation. Aging (Albany NY). 10:3986–3999. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
da Costa Martins PA, Bourajjaj M, Gladka M, Kortland M, van Oort RJ, Pinto YM, Molkentin JD and De Windt LJ: Conditional dicer gene deletion in the postnatal myocardium provokes spontaneous cardiac remodeling. Circulation. 118:1567–1576. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Chen JF, Murchison EP, Tang R, Callis TE, Tatsuguchi M, Deng Z, Rojas M, Hammond SM, Schneider MD and Selzman CH: Targeted deletion of Dicer in the heart leads to dilated cardiomyopathy and heart failure. Proc Natl Acad Sci USA. 105:2111–2116. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO, Landthaler M, et al: A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 129:1401–1414. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020. View Article : Google Scholar : PubMed/NCBI | |
|
Liang Y, Duan L, Lu J and Xia J: Engineering exosomes for targeted drug delivery. Theranostics. 11:3183–3195. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Vader P, Mol EA, Pasterkamp G and Schiffelers RM: Extracellular vesicles for drug delivery. Adv Drug Deliv Rev. 106:148–156. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
O'Brien K, Breyne K, Ughetto S, Laurent LC and Breakefield XO: RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. 21:585–606. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Wang M, Yu Y, Li C and Zhang C: Advances in the study of exosomes derived from mesenchymal stem cells and cardiac cells for the treatment of myocardial infarction. Cell Commun Signal. 21:2022023. View Article : Google Scholar : PubMed/NCBI | |
|
Das D, Jothimani G, Banerjee A, Dey A, Duttaroy AK and Pathak S: A brief review on recent advances in diagnostic and therapeutic applications of extracellular vesicles in cardiovascular disease. Int J Biochem Cell Biol. 173:1066162024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Z, Zhao P, Zhang Y, Wang J, Wang C, Liu Y, Yang G and Yuan L: Exosome-based Ldlr gene therapy for familial hypercholesterolemia in a mouse model. Theranostics. 11:2953–2965. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang L, Yang L, Ding Y, Jiang X, Xia Z and You Z: Human umbilical cord mesenchymal stem cells-derived exosomes transfers microRNA-19a to protect cardiomyocytes from acute myocardial infarction by targeting SOX6. Cell Cycle. 19:339–353. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Zhou J, Zhang O, Wu X, Guan X, Xue Y, Li S, Zhuang X, Zhou B, Miao G and Zhang L: Bone marrow mesenchymal stem cells-derived exosomal microRNA-185 represses ventricular remolding of mice with myocardial infarction by inhibiting SOCS2. Int Immunopharmacol. 80:1061562020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao J, Li X, Hu J, Chen F, Qiao S, Sun X, Gao L, Xie J and Xu B: Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc Res. 115:1205–1216. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Li C, Wu H, Xie X, Sun Y and Dai M: Paeonol attenuated inflammatory response of endothelial cells via stimulating Monocytes-Derived exosomal MicroRNA-223. Front Pharmacol. 9:11052018. View Article : Google Scholar : PubMed/NCBI | |
|
Schena GJ, Murray EK, Hildebrand AN, Headrick AL, Yang Y, Koch KA, Kubo H, Eaton D, Johnson J, Berretta R, et al: Cortical bone stem cell-derived exosomes' therapeutic effect on myocardial ischemia-reperfusion and cardiac remodeling. Am J Physiol Heart Circ Physiol. 321:H1014–H1029. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lin CM, Wang BW, Pan CM, Fang WJ, Chua SK, Cheng WP and Shyu KG: Chrysin boosts KLF2 expression through suppression of endothelial cell-derived exosomal microRNA-92a in the model of atheroprotection. Eur J Nutr. 60:4345–4355. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu J, Liu B, Wang Z, Wang D, Ni H, Zhang L and Wang Y: Exosomes from nicotine-stimulated macrophages accelerate atherosclerosis through miR-21-3p/PTEN-mediated VSMC migration and proliferation. Theranostics. 9:6901–6919. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabé-Heider F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, et al: Evidence for cardiomyocyte renewal in humans. Science. 324:98–102. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Smith RR, Barile L, Cho HC, Leppo MK, Hare JM, Messina E, Giacomello A, Abraham MR and Marbán E: Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation. 115:896–908. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Teerlink JR, Metra M, Filippatos GS, Davison BA, Bartunek J, Terzic A, Gersh BJ, Povsic TJ, Henry TD, Alexandre B, et al: Benefit of cardiopoietic mesenchymal stem cell therapy on left ventricular remodelling: Results from the congestive heart failure cardiopoietic regenerative therapy (CHART-1) study. Eur J Heart Fail. 19:1520–1529. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Bartunek J, Terzic A, Behfar A and Wijns W: Clinical experience with regenerative therapy in heart failure: Advancing care with cardiopoietic stem cell interventions. Circ Res. 122:1344–1346. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Zhao M, Liu S, Guo J, Lu Y, Cheng J and Liu J: Macrophage-derived extracellular vesicles: Diverse mediators of pathology and therapeutics in multiple diseases. Cell Death Dis. 11:9242020. View Article : Google Scholar : PubMed/NCBI | |
|
EL Andaloussi S, Mäger I, Breakefield XO and Wood MJ: Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 12:347–357. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Sun D, Zhuang X, Xiang X, Liu Y, Zhang S, Liu C, Barnes S, Grizzle W, Miller D and Zhang HG: A Novel nanoparticle drug delivery system: The Anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 18:1606–1614. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Sherman LS, Shaker M, Mariotti V and Rameshwar P: Mesenchymal stromal/stem cells in drug therapy: New perspective. Cytotherapy. 19:19–27. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Ubanako P, Mirza S, Ruff P and Penny C: Exosome-mediated delivery of siRNA molecules in cancer therapy: Triumphs and challenges. Front Mol Biosci. 11:14479532024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun L, Xu R, Sun X, Duan Y, Han Y, Zhao Y, Qian H, Zhu W and Xu W: Safety evaluation of exosomes derived from human umbilical cord mesenchymal stromal cell. Cytotherapy. 18:413–422. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Fu W, Li T, Chen H, Zhu S and Zhou C: Research progress in Exosome-based nanoscale drug carriers in tumor therapies. Front Oncol. 12:9192792022. View Article : Google Scholar : PubMed/NCBI | |
|
Smyth T, Kullberg M, Malik N, Smith-Jones P, Graner MW and Anchordoquy TJ: Biodistribution and delivery efficiency of unmodified tumor-derived exosomes. J Control Release. 199:145–155. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Frolova L and Li ITS: Targeting capabilities of native and bioengineered extracellular vesicles for drug delivery. Bioengineering (Basel). 9:4962022. View Article : Google Scholar : PubMed/NCBI | |
|
Jeyaram A, Lamichhane TN, Wang S, Zou L, Dahal E, Kronstadt SM, Levy D, Parajuli B, Knudsen DR, Chao W and Jay SM: Enhanced loading of functional miRNA cargo via pH gradient modification of extracellular vesicles. Mol Ther. 28:975–985. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Perocheau D, Touramanidou L, Gurung S, Gissen P and Baruteau J: Clinical applications for exosomes: Are we there yet? Br J Pharmacol. 178:2375–2392. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lamparski HG, Methadamani A, Yao JY, Patel S, Hsu DH, Ruegg C and Le PJ: Production and characterization of clinical grade exosomes derived from dendritic cells. J Immunol Methods. 270:211–226. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Li D, Liu Z, Zhou Y, Chu D, Li X, Jiang X, Hou D, Chen X and Chen Y: Targeted exosome-mediated delivery of opioid receptor Mu siRNA for the treatment of morphine relapse. Sci Rep. 5:175432015. View Article : Google Scholar : PubMed/NCBI |