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 and cerebrovascular diseases (CCVDs), encompassing ischemic and hemorrhagic conditions affecting the heart and brain, are primarily caused by adverse lifestyle factors such as smoking, physical inactivity, poor diet, hypertension, hyperlipidemia and inadequate glycemic control (1-4). These diseases have become the leading cause of death worldwide (5,6). Coronary heart disease is the predominant type of cardiovascular disease (CVD). Additionally, conditions such as heart failure (HF), myocardial hypertrophy and myocarditis also fall under the spectrum of CVDs (7). Cerebrovascular diseases include ischemic stroke (IS) and hemorrhagic stroke (6). In China, the burden of CCVDs is particularly severe. The 2022 China Cardiovascular Health and Disease Report (8) indicated that there were ~330 million patients with CVD, with CCVDs accounting for two out of every five deaths. The incidence is rising, particularly among individuals aged 30-50 years (9,10). CCVDs continue to pose a global health challenge, contributing to high rates of morbidity and mortality, while placing substantial medical and financial strain on healthcare systems (9). Although pharmacological and surgical interventions can improve vascular conditions, they fall short in promoting tissue regeneration and restoring function in areas affected by CCVDs (11). Thus, identifying novel therapeutic targets is crucial for the effective management of these diseases.
MicroRNA-155 (miR-155/miRNA-155) is a highly conserved non-coding single-stranded RNA encoded by the B-cell integration cluster (BIC) gene located on chromosome 21 (12). As a key member of the miRNA family, miR-155 regulates post-transcriptional processes by binding to the 3'-untranslated region (3'-UTR) of target gene mRNAs through complementary base pairing (12). As a multifunctional regulatory molecule, miR-155 is involved in a wide range of biological processes, including immune responses, inflammation, cell proliferation and apoptosis, vascular homeostasis, and tumorigenesis (13-15). Dysregulation of miR-155 expression is closely associated with the onset and progression of various pathological conditions (such as brain injury, pulmonary fibrosis and fatty liver disease) (16-18).
miR-155 serves a role in the pathogenesis and progression of CCVDs, including atherosclerosis (AS), myocardial hypertrophy, myocardial infarction (MI), ischemia-reperfusion (I/R), HF, stroke and aneurysm (19-24). The mechanisms of action of miR-155 include the regulation of inflammation, immunity, angiogenesis and other key processes (25-28). Currently, miR-155 is considered to be a promising diagnostic marker and therapeutic target for CCVDs (29-32). The present review explores the biogenesis, regulation and function of miR-155, summarizes the molecular mechanisms by which miR-155 influences the progression of various CCVDs, and examines therapeutic strategies targeting miR-155, with the aim of contributing to the understanding of CCVD pathogenesis and advancing precision treatment approaches. Compared with previous studies, to the best of our knowledge, the present review is the first to systematically integrate the dual regulatory roles of miR-155 in both CVDs and cerebrovascular diseases, elucidating its contradictory functions in processes such as inflammation and autophagy through distinct target genes [for example, endothelial nitric oxide synthase (eNOS) and suppressor of cytokine signaling 1 (SOCS1)]. The present review also compiles data on miR-155 inhibitors (such as cobomarsen) from the oncology field, exploring their cross-disciplinary therapeutic potential, and proposes an innovative biomimetic nanocarrier strategy [such as vascular cell adhesion molecule-1 (VCAM-1) antibody-modified exosomes] to address existing technological gaps in prior research.
The majority of miRNAs are generated through two primary steps: First, in the nucleus, the primary transcript (pri-miRNA) is processed by RNA polymerase II and the double-stranded RNA-binding protein complex Drosha/DiGeorge syndrome critical region gene 8 (DGCR8) to produce the miRNA precursor (pre-miRNA). Second, in the cytoplasm, the pre-miRNA is further processed by the type III RNA nuclease and the transactivation-responsive RNA-binding protein complex Dicer/TAR RNA-binding protein to generate the mature miRNA form (33). The human miR-155 gene is located within the third exon of the BIC gene on chromosome 21. The mature sequence of miR-155 is highly conserved across evolution, specifically targeting the 3'-UTR of target gene mRNAs through its seed sequence (comprising nucleotides 2-8 at the 5' end), thereby mediating post-transcriptional gene silencing or mRNA degradation (34). Argonaute protein 2 binds to the miR-155 double-stranded complex, forming the core of the RNA-induced silencing complex and producing single-stranded DNA molecules (34). The precursor miRNA hairpin has two arms that can generate biologically active mature miRNAs. The miRNA derived from the 5' arm is referred to as miR-155-5p, while that from the 3' arm is referred to as miR-155-3p. Notably, miR-155-5p exhibits higher biological activity (34) (Fig. 1). Additionally, miR-155 is expressed not only in hematopoietic cells but also in a wide range of tissues, including reproductive tissues, fibroblasts, epithelial tissues and the central nervous system (35-37).
miR-155 is a highly conserved miRNA, and its expression is tightly regulated at multiple levels, including transcriptional regulation, post-transcriptional modifications and epigenetic control (38-40) (Fig. 2). The physiological expression of miR-155 is essential for immune homeostasis, while pathological dysregulation contributes to the progression of various diseases, including AS, stroke and hyperlipidemia (41-43).
The transcription of miR-155 primarily relies on the synergistic action of cis-regulatory sequences and trans-regulatory elements in the promoter region (12,44). Activation of toll like receptors (TLRs) or cytokines (such as TNF-α and IFN-γ) within inflammatory signaling pathways induces miR-155 expression by activating transcription factors such as NF-κB and adaptor protein complex-1 (AP-1), which directly bind to the miR-155 promoter region (38,39). In B and T cells, antigen receptor (B cell receptor/T cell receptor) signaling activates downstream transcription factors (such as STAT3) through the PI3K/AKT or MAPK pathways, promoting miR-155 expression (36,45). Additionally, the transcriptional activity of the BIC gene can be dynamically regulated by DNA methylation and histone acetylation. For example, reduced methylation of the miR-155 promoter region in tumors is closely associated with its upregulation (46).
The maturation of miR-155 involves the cleavage of pri-miR-155 to pre-miR-155 and cytoplasmic transport of pre-miR-155, a process regulated by RNA binding proteins such as KH-type splicing regulatory protein (KSRP). KSRP binds to the stem-loop structure of pri-miR-155 and recruits the Drosha-DGCR8 complex to promote the processing of pri-miR-155 into pre-miR-155 (37). Lineage protein 28 (LIN28) family proteins (LIN28A/B), as RNA-binding proteins, can selectively block the maturation of miRNAs such as let-7 by inhibiting the processing of pri-miRNAs by Drosha/Dicer complexes (47). We hypothesized that LIN28 may regulate miR-155 through a similar mechanism, although further research is needed to confirm this. Additionally, competitive endogenous RNAs (ceRNAs) can indirectly modulate the functional activity of miR-155 by sequestering it via the 'sponge effect'. For example, long non-coding RNAs (lncRNAs) such as feline leukemia virus subgroup C receptor 1-antisense RNA 1 can bind to miR-155, thereby reducing its inhibition of target genes (40). Lu et al (48) identified that circNr1h4 acted as a ceRNA interacting with miR-155-5p, subsequently influencing the pathological progression of renal damage in salt-sensitive hypertensive mouse models.
In disease environments, the expression of miR-155 is often dysregulated due to imbalanced regulatory networks (49). Specifically, the activation of cancer-related genes (such as MYC) or suppression of tumor-suppressor genes (such as TP53) within the tumor microenvironment can trigger persistently elevated levels of miR-155, enhancing cellular proliferation and metastasis (50-52). For instance, overexpression of the BIC gene increases miR-155 levels in diffuse large B-cell lymphoma (53). During chronic inflammatory conditions, sustained inflammatory stimuli [such as high levels of oxidized low-density lipoprotein (ox-LDL) in AS] maintain miR-155 expression, promoting eNOS production and exacerbating tissue damage through continuous activation of the NF-κB signaling pathway (54). Furthermore, in patients with rheumatoid arthritis, synovial fibroblasts aberrantly activate miR-155 via the Janus kinase (JAK)-STAT pathway, leading to the release of inflammatory mediators, including IL-6 and TNF-α (55). Wang et al (56) demonstrated that C1q tumor necrosis factor related protein 12 (CTRP12) could mitigate AS by enhancing reverse cholesterol transport and reducing vasculitis through the miR-155-5p/liver X receptor α (LXRα) pathway. However, it remains unclear whether CTRP12 directly regulates miR-155-5p (56). A study (57) utilizing cerebral I/R injury (IRI) mouse models has demonstrated that upregulated expression levels of lncRNA opioid receptor Mu 1 (Oprm1) alleviated cellular apoptosis induced by cerebral IRI via the Oprm1/miR-155/GATA binding protein 3 (GATA3) pathway. However, the direct regulatory relationship between Oprm1 and miR-155 remains undetermined (57).
miR-155 is extensively involved in physiological and pathological processes, including immune regulation, inflammatory responses, tumorigenesis and metabolic homeostasis, by targeting the expression of downstream genes (36,38,53,54). miR-155 contributes to the development of CCVDs by regulating the expression of multiple molecules, some of which are shown in Fig. 3. miR-155 serves a critical role in both adaptive and innate immunity. One study has shown that in T cells, miR-155 targets and inhibits the protein expression of SOCS1, enhances STAT5 signaling, promotes the differentiation of T helper type 1 (Th1) and T helper type 17 (Th17) cells, and sustains the anti-infective immune response (36). In the regulation of inflammation, miR-155 is the only miRNA upregulated by macrophages in response to various inflammatory stimuli, including virus-related signals [such as the poly(I) synthetic analog of double-stranded RNA and interferons IFN-β/γ] and bacteria-related stimuli [for example, TLR ligands such as lipopolysaccharide, CpG DNA and Pam3Cys-Ser-(Lys) 4]. These stimuli activate miR-155 expression via myeloid differentiation primary response 88 (MYD88)- or Toll/IL-1 receptor domain-containing adaptor-inducing interferon-dependent TLR signaling pathways, while IFN-β/γ indirectly induces miR-155 expression through TNF-α autocrine signaling, involving TNFR1. miR-155 integrates the TLR, IFN and TNF-α signaling networks, activating transcription through the JNK/AP-1 pathway (38). miR-155 exhibits notable carcinogenic properties within the tumor microenvironment. The overexpression of miR-155 promotes tumor cell proliferation, invasion and angiogenesis by targeting and inhibiting tumor suppressor genes, such as tumor protein p53 inducible nuclear protein 1 (50,51) and SH2-containing inositol 5'-phosphatase 1 (58,59). miR-155 may also exert anticancer effects in certain solid tumors (for example, breast cancer, liver cancer and lymphoma) by inhibiting TGF-β signaling (60-63), highlighting its functional complexity.
As a non-coding RNA molecule, miR-155 serves as a multidimensional regulator in the pathological processes of CCVDs. miR-155 participates in key mechanisms such as inflammation, endothelial dysfunction, oxidative stress and apoptosis by targeting the expression of downstream genes (64-68). This section will delve into the specific molecular mechanisms through which miR-155 influences the progression of CCVDs, including AS, HF, MI, hypertension, IRI, stroke and aneurysm (Tables I-III).
Table IIISpecific molecular mechanisms of miR-155 regulating hypertension, stroke and arterial aneurysm. |
AS represents a pathological condition characterized by inflammatory responses and lipid accumulation, serving a role in the progression of CCVDs (69). The pathogenesis of AS involves a prolonged immunological inflammatory process, initiated by various pro-inflammatory mediators interacting with multiple cell types, including endothelial cells (ECs), vascular smooth muscle cells (VSMCs) and monocytes/macrophages (25,54,70). miR-155, a versatile miRNA, is highly expressed in atherosclerotic plaques of both murine and human subjects (71), emerging as a critical molecular factor in controlling the initiation and progression of AS (Fig. 4).
ECs serve as the interface between the blood vessel wall and the bloodstream, facilitating the exchange of oxygen and nutrients between the blood and tissues (72). ECs serve a pivotal role in inflammation responses, thrombosis regulation and vascular tone modulation, directly influencing CVD progression (72). eNOS generates nitric oxide (NO), a crucial molecule for maintaining cardiovascular homeostasis. Aberrant eNOS expression is often linked to endothelial dysfunction and CVD (73,74). Sun et al (75) demonstrated that miR-155 directly targeted eNOS, where elevated miR-155 levels reduced eNOS expression and NO production in HUVECs, impairing acetylcholine-mediated endothelium-dependent vasodilation in human mammary arteries. miR-155 is co-expressed with the angiotensin II type I receptor (AT1R) in ECs and VSMCs. The molecular mechanism involves miR-155 binding specifically to the 3'-UTR of AT1R mRNA, inhibiting its translation, and thus, mitigating the pathological effects of angiotensin II (Ang II) on HUVECs (76). This finding highlights the pivotal role of the miR-155-AT1R axis in vascular function regulation. EC apoptosis is a form of endothelial injury and is closely associated with the development of AS (73,74). Lee et al (77) demonstrated that G protein subunit α12 protected HUVECs from serum withdrawal-induced apoptosis by maintaining miR-155 expression. Additionally, another study demonstrated that overexpression of miR-155 inhibited palmitic acid-induced apoptosis, reactive oxygen species (ROS) production and inflammatory cytokine release in HUVECs by suppressing the Wnt signaling pathway (78). Endothelial autophagy, as a cytoprotective mechanism, maintains endothelial homeostasis by clearing damaged organelles and misfolded proteins, serving a critical role in preventing the onset and progression of AS (79,80). Under physiological conditions, ECs utilize autophagy to remove ox-LDL and ROS, thereby preventing lipid accumulation and inflammatory cytokine release (81). In HUVECs, ox-LDL induces autophagy and elevates miR-155 expression. Increased miR-155 levels promote autophagy, while reduced miR-155 expression suppresses autophagic activity in vascular ECs (82). Further research by Yin et al (83) indicated that elevated miR-155 levels enhanced ox-LDL-mediated autophagy in HUVECs by inhibiting the PI3K/AKT/mTOR signaling pathway. Metabolic abnormalities and genetic variations in homocysteine (Hcy) metabolism lead to hyperhomocysteinemia and endothelial dysfunction, which are both hallmark features of AS and key contributors to CVD (84). Research by Witucki and Jakubowski (85) demonstrated that metabolites of Hcy elevated the expression levels of miR-21, miR-155, miR-216 and miR-320c, which in turn reduced autophagy in human ECs, a process critical for maintaining vascular homeostasis. The stress-responsive enzyme heme oxygenase-1 (HO-1) protects cells under stress conditions and promotes endothelial anti-inflammatory and vasodilatory responses (86). Pulkkinen et al (87) showed that miR-155 exerted a protective effect on endothelial inflammation by reducing the translation of BTB domain and CNC homolog 1 (BACH1), which induced HO-1 expression in ECs. Another study demonstrated that both Abelmoschus esculentus and metformin ameliorated endothelial inflammation induced by a high-fat diet in rats through enhanced miR-155 expression, primarily by inhibiting TNF receptor associated factor 6 and NF-κB p65 activation (88). Yang et al (89) found that miR-155 not only regulated hypoxia-inducible factor 1α (HIF-1α) expression under hypoxic conditions but also enhanced the angiogenic potential of ECs by targeting E2F transcription factor 2. Promoting angiogenesis is a key feature of healthy ECs.
VSMCs serve a critical role in maintaining vascular function (90). A study revealed that TNF downregulated protein kinase G1 via NF-κB-dependent miR-155-5p biosynthesis, impairing the maintenance of the NO/cGMP-mediated VSMC contractile phenotype and vasodilation (91). The NF-κB/miR-155-5p/protein kinase G axis may contribute to atherosclerotic intimal hyperplasia (91). Another study found that miR-155, induced by NF-κB, disrupted the contractile phenotype of VSMCs and inhibited NO-mediated vasodilation by downregulating soluble guanylate cyclase β1 subunit (sGCβ1) expression, leading to functional impairment of VSMCs (26). Overexpression of miR-155-5p reduces AKT1 levels by inhibiting AKT1 phosphorylation, suppressing VSMC proliferation and migration (92), as well as reducing VSMC apoptosis (71). Furthermore, overexpression of miR-155-5p alleviates oxidative stress and migration of VSMCs by inhibiting BACH1 expression (93). Notably, miR-155 has been reported to downregulate the NADPH oxidase activator 1-phagocyte oxidase protein 47-related signaling pathway in apolipoprotein E (ApoE)-/- mice, inhibiting VSMC migration and excessive proliferation, thereby attenuating AS progression in these mice (94).
The inflammatory process driven by macrophages serves a critical role in AS (54). ox-LDL induces distinct miR-155 expression within atherosclerotic lesions and macrophages (41,54). Huang et al (41) demonstrated an increase in miR-155 expression under high ox-LDL level conditions, where elevated miR-155 levels suppressed TNF, IL-6 and IL-8 production by inhibiting MYD88-dependent NF-κB pathway activation. Further investigation demonstrated that miR-155 accumulated in ox-LDL-stimulated EC-derived extracellular vesicles (EVs), which were subsequently transferred to human monocyte THP1 cells (41). These vesicles promoted monocyte activation by shifting the monocyte/macrophage balance from the anti-inflammatory M2 phenotype to the pro-inflammatory M1 phenotype (95). These studies highlighted that miR-155 serves a role in negatively regulating the inflammatory response induced by ox-LDL across various signaling pathways (41,95). CTRP12 is a conserved homolog of adiponectin, which may participate in the development of CVDs by promoting endothelial injury or the inflammatory response (23). Wang et al (56) demonstrated that CTRP12 reduced miR-155-5p levels, leading to increased expression of its target gene LXRα. This elevation enhanced ATP binding cassette transporter A1 (ABCA1) and ATP binding cassette transporter G1-mediated cholesterol efflux, promoting macrophage differentiation towards the M2 phenotype, suppressing inflammation and alleviating AS (56). However, one study has suggested that miR-155 may also facilitate AS development (54). Specifically, during AS progression, miR-155 enhances NF-κB pathway activation by downregulating Bcl-6 expression, thereby promoting NLRP3 activation. Bcl-6 expression amplifies the pro-inflammatory transformation of monocytes/macrophages, accelerates lipid accumulation in plaques, and contributes to the formation of an inflammatory microenvironment (54). This illustrates the multifaceted role of miR-155.
The accumulation of foam cells is a key pathological feature of AS (96). One study has shown that miR-155 inhibited foam cell formation in macrophages by promoting cholesteryl ester hydrolase expression (97). Li et al (98) identified elevated miR-155 levels in both plasma and atherosclerotic plaques of patients with AS. In AS animal models, the specific molecular mechanism involved TNF-α inflammatory signaling triggering NF-κB pathway activation, thereby promoting upregulation of miR-155 expression. Elevated miR-155 levels reduced chronic inflammation via a negative feedback mechanism and operated through the miR-155-calcium regulated heat stable protein 1-TNF-α signaling cascade, exerting a protective effect during foam cell formation associated with AS (98).
Vascular inflammation is a key contributor to the development of AS (69). Wu et al (25) demonstrated that miR-155 overexpression suppressed TNF-α-induced endothelial inflammatory responses by reducing the expression of NF-κB p65 and adhesion molecules (such as intercellular adhesion molecule-1 and VCAM-1), thereby decreasing monocyte-EC adhesion. Conversely, miR-155 inhibition enhanced p65 levels and endothelial inflammation, while p65 knockdown using small interfering RNA reversed this effect. Their study further revealed that TNF-α induced miR-155 expression, and this induction was attenuated by endogenous p65 deficiency, suggesting miR-155 may act as a negative feedback regulator to mitigate excessive inflammation triggered by TNF-α (25). Zhu et al (99) found that miR-155 expression was elevated in both AS mouse models and patients with coronary artery disease (CAD) compared with healthy controls. Operating within a negative feedback loop, miR-155 suppresses inflammatory cytokine production, with its expression decreasing as the condition progresses (99). Additionally, miR-155 has been shown to mediate inflammatory responses and the MAPK pathway by targeting MAP3K10 (99). Ye et al (100) revealed that miR-155 served dual roles in AS mouse models: It directly inhibited SOCS1 expression, while increasing phosphorylated-STAT and programmed cell death 4 (PDCD4) levels, as well as increasing the production of inflammatory mediators such as IL-6 and TNF-α. Suppression of miR-155 reduced these molecular levels of PDCD4, STAT, IL-6 and TNF-α. Therefore, it is suggested that miR-155 mediates inflammatory cytokine production in AS through the SOCS1-STAT3-PDCD4 axis (100). Furthermore, miR-155 knockout reduces CD4+ T cell-induced EC apoptosis and promotes VSMC proliferation (101). By suppressing miR-155 expression, inflammation related to CD4+ T cells is diminished through decreased lymphocyte proliferation and altered differentiation patterns (101). Additionally, miR-155 has been shown to activate NLR family pyrin domain containing 3 (NLRP3) inflammasomes by regulating the MEK/ERK/NF-κB pathway, contributing to atherosclerotic plaque formation in ApoE-/- mice (102). These findings suggest that miR-155 holds promise as both a biomarker for AS and a potential target for therapeutic interventions.
miR-155 is a critical regulator involved in cardiac inflammation and hypertrophic responses (103-106). Bao and Lin (107) observed a marked increase in miR-155 levels in the myocardial tissue of mice with Coxsackievirus B3-induced myocarditis. Functional experiments in vitro showed that miR-155 mitigated myocardial damage by inhibiting the NF-κB pathway (107). Macrophage infiltration is a hallmark of viral myocarditis, and Zhang et al (108) found that silencing of miR-155 reduced viral myocarditis-induced cardiac damage and dysfunction by promoting macrophage M2 phenotype polarization. Additionally, inhibition of miR-155 improved experimental autoimmune myocarditis in mice by enhancing the Th17/regulatory T cell (Treg) immune response (109). These findings suggest that miR-155 suppression could serve as an effective treatment for autoimmune myocarditis. Zhang et al (110) demonstrated that Astragalus mongholicus (Fisch.) Bge alleviated immune imbalances in peripheral Tregs in children with viral myocarditis by reducing miR-155 levels. Collectively, these studies (107-109) position miR-155 as a promising diagnostic biomarker and potential therapeutic target for cardiomyopathy.
Research has also indicated a decrease in miR-155 levels in animal and patient models of myocardial hypertrophy (111-113). Overexpression of miR-155 induces hypertrophy in H9C2 cardiomyocytes in vitro (114). Conversely, inhibition of miR-155 alleviates myocardial hypertrophy in H9C2 rat cardiomyocytes by downregulating angiotensin II receptor subtype 1 (AGTR1) and inhibiting the calcium signaling pathway activated by AGTR1 (115). Seok et al (116) found that the absence of miR-155 protected the heart from pathological cardiac hypertrophy, primarily by reducing jumonji and AT-rich interaction domain containing 2 expression. Another study revealed that EVs derived from hypertrophic cardiomyocytes activated the miR-155-mediated MAPK (ERK, JNK and p38) pathway, inducing macrophage inflammation and exacerbating myocardial cell damage (117). Studies have demonstrated that miR-155-overexpressing macrophages directly targeted and suppressed FoxO3a expression in cardiomyocytes of uremic mice via EVs, leading to reduced FoxO3a levels, which promoted cardiomyocyte pyroptosis, and exacerbated hypertrophy and fibrosis (118,119). Additionally, Fan et al (120) demonstrated that resveratrol, a polyphenol compound, alleviated cardiac hypertrophy and improved cardiac function by activating BRCA1 in cardiomyocytes. This mechanism involved BRCA1 activation, which suppressed miR-155 expression, thereby enhancing FoxO3a expression and reducing cardiac hypertrophy (120). Furthermore, Yuan et al (121) demonstrated that cytoskeleton regulator RNA (CYTOR) deletion decreased IKKI protein expression, while IKKI deficiency triggered cardiac hypertrophy via the AKT and NF-κB pathways. miR-155 suppression partially attenuated the effects of CYTOR. The authors proposed that CYTOR functions as a ceRNA for miR-155, counteracting miR-155-mediated inhibitor of nuclear factor κB kinase subunit ε suppression, thus offering protection against cardiac hypertrophy (121). These studies highlight the critical role of miR-155 in cardiac hypertrophy (117-121) (Fig. 5).
MI is an acute cardiovascular event resulting from the rapid reduction or interruption of coronary blood flow, leading to ischemia, hypoxia and necrosis of myocardial cells. The primary cause is coronary atherosclerotic plaque rupture or thrombosis (122). Plaque rupture often results from an imbalance between macrophage-mediated degradation and fibroblast repair functions (123). Wang et al (124) found that upregulated miR-155 was predominantly present in macrophages and fibroblasts in the damaged heart, while pri-miR-155 was exclusively expressed in macrophages. Mice deficient in miR-155 exhibited increased proliferation of cardiac fibroblasts and collagen production, along with reduced inflammation in the damaged heart (124). Notably, suppression of miR-155 reduces nerve growth factor expression by decreasing the phagocytic activity of M1 macrophages and the inflammation mediated by the SOCS1/NF-κB pathway, thereby mitigating sympathetic remodeling and ventricular arrhythmias induced by MI in mice (125,126). Pro-inflammatory macrophage-mediated degradation of connexin 43 (Cx43) serves a pivotal role in arrhythmia following MI (127). A study has demonstrated that miR-155 inhibited the downregulation of macrophage-mediated IL-1β and MMP7 expression via the SOCS1/NF-κB pathway, reducing Cx43 degradation after MI (128).
Aging impairs the function of human mesenchymal stem cells (MSCs), reducing their therapeutic potential in MI (129). Hong et al (130) showed that inhibition of miR-155-5p suppressed MSC aging through the Cab39/AMPK signaling pathway. This suggests that miR-155-5p could be a novel target for restoring the vitality of bone marrow MSCs and enhancing their cardioprotective effects (130). Guo et al (21) also found that miR-155 levels dynamically increased in the hearts of mice with MI and in neonatal rat ventricular myocytes injured by hydrogen peroxide (H2O2). Downregulation of miR-155 promoted apoptosis induced by MI by targeting RNA binding protein Quaking (21). Hypoxia/reoxygenation (H/R)-induced cardiomyocyte apoptosis serves a critical role in MI pathogenesis. Inhibition of miR-155-5p can prevent NLRP3 inflammasome activation by targeting sirtuin 1 (SIRT1), thus reducing H/R-induced cardiomyocyte pyroptosis (131). lncRNA XIST promotes cardiac fibroblast proliferation and extracellular matrix (ECM) accumulation by acting as a sponge for miR-155-5p, thus facilitating MI formation (132). Another study demonstrated that in a mouse model of MI with dyslipidemia, the deletion of the miR-155 gene did not reduce infarct size or chronic HF but decreased the density of myofibroblasts in ischemic scars (133). An investigation into miR-155-based therapeutic strategies has revealed that rosuvastatin potentially reduces cardiovascular events and inflammatory markers (INF-γ, TNF-α and IL-6) in patients with acute coronary syndrome by suppressing the miR-155/Src homology 2-containing inositol 5-phosphatase 1 (SHIP-1) signaling cascade (134). Furthermore, combination of American ginseng with Danshen increased the serum levels of hepatocyte growth factor and basic fibroblast growth factor in acute MI rats, enhanced myocardial microvascular density and CD31 levels, and suppressed the miR-155-5p/HIF-1α/VEGF pathway, promoting angiogenesis, compared with those in the acute myocardial infarction model group (135). These findings highlight the potential of miR-155 as a target for the treatment of MI (Fig. 5).
IRI refers to the pathological phenomenon where tissue, such as cardiac or brain tissue, experiences restored blood flow after a period of ischemia, but this restoration exacerbates cell damage (136). This process is commonly observed in vascular recanalization therapies (such as thrombolysis and percutaneous coronary intervention surgery), cardiac or cerebrovascular surgeries, and organ transplantation following acute MI or IS (137,138). IRI poses a challenge in the field of CCVDs, highlighting the urgent need for the identification of novel therapeutic targets.
miR-155 expression increases in myocardial tissue following IRI, which is associated with elevated levels of TNF-α, IL-1β, CD105 and caspase-3, as well as enhanced leukocyte infiltration (139). Knockout of miR-155 reduces inflammatory cell recruitment and decreases ROS production in white blood cells (139). Notably, miR-155 exacerbates the inflammatory response, leukocyte infiltration and tissue damage in IRI by regulating SOCS-1-dependent ROS production (139). Chen et al (20) found that inhibition of miR-155 reduced the MI area by specifically regulating HIF-1α, inhibited IRI-induced cardiomyocyte apoptosis, maintained MMP levels and alleviated myocardial injury in rats. Furthermore, inhibition of miR-155 led to targeted regulation of Bcl-2-associated athanogene 5 and MAPK/JNK signaling, reducing myocardial IRI and the size of MI (140). Another study found that, in IRI mice, miR-155 was expressed at high levels, while SIRT1 expression was low. The expression levels of SIRT1, confirmed to be a target gene of miR-155, were increased following sevoflurane treatment, which reduced miR-155 levels, improved cardiac function, reduced infarct size and inhibited myocardial cell apoptosis (141). Greco et al (142) observed that myocardial IRI-induced EVs exhibited pro-inflammatory features, exacerbating cardiac injury. The specific molecular mechanism involved these EVs transferring miR-155-5p to macrophages, thereby enhancing the inflammatory response via activation of the JAK2/STAT1 pathway (142). This suggests that targeting EVs could be a potential therapeutic approach for the management of IRI. An investigation has demonstrated that miR-155 deletion improved cardiac ultrasound measurements in IRI mice, with reductions in MI areas, myocardial fibrosis and cellular apoptosis (including reducing the expression levels of caspase-3, caspase-4 and caspase-11) (143). Investigation of the underlying mechanism revealed that decreased miR-155 expression increased SH2 domain-containing protein tyrosine phosphatase 2 levels and alleviated IRI-induced necroptosis by suppressing ERK1/2 pathway activation (143). These findings highlight the potential of miR-155 as a key therapeutic target in the treatment of myocardial IRI (Fig. 5).
The elimination of the miR-155 gene protects against brain injury and hemorrhagic transformation induced by I/R (142). Multiple studies have corroborated these findings. Jiang et al (144) found that miR-155 deficiency reduced NO production and eNOS expression by activating the Notch pathway, thereby alleviating the damage caused by cerebral I/R in mice with middle cerebral artery occlusion (MCAO). Inhibition of miR-155 increases cell viability and reduces apoptosis by targeting the nuclear factor erythroid 2-related factor 2/HO-1 pathway, preventing neuronal damage induced by cerebral I/R (65). Furthermore, downregulation of miR-155 mitigates IRI by targeting V-Maf avian musculoaponeurotic fibrosarcoma oncogene homolog B, improving the neurological function and inhibiting inflammatory responses [IL-1β, IL-6, TNF-α, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2] (145). Another study also found that in MCAO/reperfusion mouse models and oxygen glucose deprivation/reoxygenation (OGD/R)-induced SH-SY5Y cells, miR-155-5p targeted dual specificity phosphatase 14 (DUSP14) by regulating the NF-κB and MAPK signaling pathways, thereby accelerating cerebral IRI (146). Suppression of miR-155-5p reduces cellular apoptosis and cerebral injury (146). Research by Shi et al (147) revealed that decreased miR-155-5p expression alleviated cerebral I/R-induced inflammation and cell death by modulating the DUSP14/thioredoxin interacting protein/NLRP3 signaling cascade. Notably, increased expression of lncRNA Oprm1 mitigates apoptosis following cerebral I/R damage via the Oprm1/miR-155/GATA3 regulatory axis (57). Furthermore, elevated Parkinson's disease protein 7 expression suppresses miR-155 levels, thereby enhancing SHP-1 expression and modulating astrocyte activation during cerebral IRI (148). These findings suggest potential therapeutic strategies for managing cerebral IRI (Fig. 6).
HF is a clinical syndrome characterized by a decline in the pumping function of the heart, leading to an inability to meet the metabolic needs of the body, with common symptoms including dyspnea, fatigue and fluid retention (149). The etiology of HF encompasses conditions such as hypertension, coronary heart disease and cardiomyopathy (150). The underlying pathological mechanisms involve myocardial remodeling, inflammation and fibrosis (151). Li et al (152) utilized bioinformatics and experimental validation to investigate the potential role of miR-155 in HF. The authors found that miR-155 may regulate the viability and apoptosis of H9c2 cardiomyocytes by targeting and modulating G protein-coupled receptor 18 (152). In the same year, another study reported that in established rat and cell models of HF, overexpression of Sirt1 upregulated NF-κB p65 and miR-155, promoting brain-derived neurotrophic factor (BDNF) expression and reducing cardiomyocyte apoptosis. The authors proposed that Sirt1 alleviated HF in rats via the NF-κB p65/miR-155/BDNF signaling cascade (153). Gao et al (154) found that schisandrin protected rat cardiomyocytes and prevented congestive HF by regulating miR-155 expression and mediating the AKT/cAMP response element-binding protein (CREB) signaling pathway. Another study revealed that miR-155 expression was downregulated in the myocardial tissue of mice with HF. Overexpression of miR-155 inhibited myocardial cell apoptosis (via inhibition of Bax and downstream caspase-3) through HIF-1α, reducing cardiac function damage in HF mice (155). These studies (152-154) highlight the critical role of miR-155 in myocardial protection and suggest its potential for improving cardiac function through the regulation of specific molecules (for example, HIF-1α, AKT and CREB) (Fig. 5).
Elevated miR-155 levels in the circulation of hypertensive patients have been found to be positively associated with inflammatory markers, indicating their involvement in the pathological process of hypertension through immune regulation (68,156). Animal models have confirmed that miR-155 deficiency alleviates perivascular inflammation and reduces blood pressure (157). Yang et al (68) demonstrated that levamlodipine improved vascular inflammation and endothelial dysfunction by regulating miR-155 in hypertensive rats, thereby modulating the receptor activator of nuclear factor κ-B/receptor activator of nuclear factor κ-B ligand/osteoprotegerin pathway. Additionally, studies have shown that miR-155 regulates vascular tone and endothelial-dependent vasodilation by targeting the eNOS and vascular endothelial growth factor signaling pathways (75,158). Overexpression of miR-155 can inhibit eNOS activity, reducing NO production, promoting endothelial dysfunction and contributing to hypertension development (75). VSMCs are essential for maintaining vascular function, and inflammation-induced VSMC dysfunction can also lead to hypertension (159). Park et al (26) found that miR-155, induced by NF-κB, downregulated sGCβ1 expression, impaired the VSMC contractile phenotype and disrupted NO induced vasodilation, leading to VSMC functional damage. Another study revealed that in the tunica media (VSMCs) of hypertensive rats, miR-155 expression was elevated. Inhibition of miR-155 reduced systolic and diastolic blood pressure, increased expression of p27 and α-smooth muscle actin (α-SMA) in the tunica media, and decreased the thickness of the tunica media. These findings suggest that miR-155 has therapeutic potential for hypertension, with its expression levels being positively associated with vascular wall thickness (160).
Ang II is a key peptide hormone in the renin-angiotensin system, serving a critical role in regulating vasoconstriction and renal sodium reabsorption (161). The physiological and pathophysiological effects of Ang II are primarily mediated through AT1R (162). Zheng et al (157) found that overexpression of miR-155 in cells reduced Ang II-induced α-SMA expression, suggesting that miR-155 may regulate the differentiation of rat aortic adventitia fibroblasts and inhibit AT1R expression. Additionally, research has shown that vascular mineralocorticoid receptors promote vasoconstriction and increase blood pressure with age by modulating miR-155 (163). Specifically restoring miR-155 in aged mineralocorticoid receptor-intact mice reduces calcium voltage-gated channel subunit α1 2 and AgtR1 mRNA levels, alleviating L-type calcium channel-mediated and Ang II-induced vasoconstriction and oxidative stress (163). Atrial natriuretic peptide (ANP), secreted by primary atrial myocytes, lowers blood pressure by increasing cGMP levels, inducing vasodilation, diuresis and sodium excretion (164). Vandenwijngaert et al (165) found that compared with individual use of miR-425 or miR-155, the combination of miR-425 and miR-155 demonstrated greater suppression of natriuretic peptide A expression and cGMP production in cardiomyocytes. These studies showed that promoting the expression of miR-155 may represent an effective strategy for regulating blood pressure in hypertensive disorders (163-165).
Stroke is the second leading cause of death worldwide, and IS is a major subtype (166). miR-155 serves a role in the progression of IS (167,168). With ongoing research, the molecular mechanisms of miR-155 in protecting against IS are becoming clearer (Fig. 6). Xing et al (169) used an in vivo rat model of MCAO and an in vitro oxygen-glucose deprivation cell model to simulate IS onset. The authors found that inhibition of miR-155 could protect against IS by promoting the phosphorylation of S6K through the Ras homolog enriched in brain/mTOR pathway (169). Ischemia induces autophagy via miR-155, contributing to nerve damage (170). Yang et al (170) revealed that miR-155-induced autophagy altered inflammatory responses and exacerbated ischemic brain injury by regulating the TLR4/NF-κB pathway in ischemic brain tissue. Another study indicated that miR-155 worsened cellular damage in IS by activating the TLR4/MYD88 signaling pathway (22). The study by Adly Sadik et al (171) demonstrated that miR-155 may promote inflammatory responses post-IS by activating the JAK2/STAT3 axis. A recent study found that miR-155 inhibited the activation of Wnt/β-catenin signaling, restored the Th17/Treg balance and prevented acute IS in mice (172). These findings highlight that miR-155 is involved in multiple signaling pathways and targets in IS. Knockdown of miR-155 expression and the use of specific inhibitors to block miR-155 targets may offer a novel approach for treating IS by interrupting signaling pathway transmission. Research has shown that geniposide and ginsenoside Rg1 protect against focal cerebral ischemia in MCAO model rats by inhibiting miR-155-5p in microglia following ischemic injury (173). Additionally, Opa-interacting protein 5-AS1 inhibits oxidative stress and inflammation by regulating the miR-155-5p/interferon regulatory factor 2 binding protein 2 axis, alleviating OGD/R-induced damage in HMC3 and SH-SY5Y cells, offering a novel targeted therapeutic molecule for IS treatment (174).
Arterial aneurysms are typically asymptomatic in their early stages, but as the aneurysm expands, the risk of rupture increases, which can lead to fatal bleeding (175). Based on anatomical location, aneurysms are classified into abdominal aortic aneurysms (AAAs), thoracic aortic aneurysms and intracranial aneurysms (IAs) (176). miR-155 promotes macrophage infiltration by upregulating pro-inflammatory factors such as TNF-α and IL-6, suggesting that miR-155 may contribute to the inflammatory process during aneurysm development (24,177). Zhang et al (24) found that inhibition of miR-155 prevented AAA formation by regulating macrophage inflammation. The development of AAA was linked to the proliferation and apoptosis of VSMCs. Their study also found that overexpression of miR-155 increased the levels of MMP-2, MMP-9, iNOS and monocyte chemoattractant protein-1 in ApoE-/- mouse models, stimulating VSMC proliferation and migration. Another study found that miR-155-5p expression was increased in VSMCs damaged by H2O2 or NaAsO2. Overexpression of miR-155-5p inhibited VSMC survival and promoted aneurysm formation by targeting FOS proto-oncogene (FOS) and Zic family member 3 (ZIC3) (178). These findings suggest that miR-155 could be a therapeutic target for AAA treatment. The degradation of the ECM in blood vessels is another key factor in aneurysm formation. miR-155 enhances the activity of MMP-2/9, accelerating the degradation of collagen and elastin (24). Inhibition of miR-155 can alleviate ECM damage and delay AAA progression (24). Yang et al (178) found that reduced miR-155 expression increased the incidence of IA rupture by upregulating MMP-2 expression, particularly in subjects with the SNP rs767649 genotype. This SNP in the miR-155 promoter reduces its transcriptional activity, suggesting a genetic predisposition to increased aneurysm risk (178). Additionally, another study found that miR-155-5p, derived from tumor-associated macrophages, can target IA formation and macrophage infiltration induced by Gremlin1 (179). These findings highlight miR-155-5p as a potential therapeutic target for IAs.
miR-155, a key regulatory miRNA, serves a pivotal role in the onset and progression of CCVDs by influencing pathological and physiological processes such as inflammation, oxidative stress and apoptosis (180-182). Research has shown that miR-155 is upregulated in the blood and tissues of patients with CCVDs such as MI (183) and stroke (171), indicating its potential as an effective marker for early diagnosis and prognosis evaluation.
Li et al (98) measured plasma miR-155 levels in 70 patients with AS and 55 normal controls. The results revealed higher miR-155 levels in patients with AS. Reverse transcription-quantitative PCR (RT-qPCR) analysis also showed markedly elevated miR-155 expression in 17 paired atherosclerotic lesions compared with normal veins from the same patients (98). Another study examined surface marker expression and miR-155 content in urinary EVs (uEVs) from patients diagnosed with unstable (n=12) and stable (n=12) CAD (184). Compared with uEVs from patients with stable CAD, uEVs from patients with unstable CAD exhibited increased miR-155 expression. Furthermore, miR-155 levels decreased during AS regression and increased during disease progression (184), suggesting its potential as an early diagnostic and prognostic biomarker.
In a clinical study of 89 patients with inflammatory cardiomyopathy (iCMP), Obradovic et al (185) observed elevated plasma miR-155 levels in patients with iCMP compared with those with dilated cardiomyopathy, suggesting miR-155 as a novel biomarker for iCMP diagnosis. Previous studies have indicated a marked increase in miR-155 expression in both myocardial tissue and blood after MI, which was strongly associated with inflammatory responses (IL-17A, IL-6 and TNF-α) and myocardial injury (183,186). Notably, Wang et al (187) found urinary miR-155 levels to be 30-fold higher in the MI group compared with healthy individuals, suggesting urinary miR-155 as a potential non-invasive diagnostic biomarker for MI.
miR-155 levels are elevated in the myocardium of patients with HF (188) and are positively associated with worsening heart function (32,105,189). However, Ikitimur et al (189) observed reduced serum miR-155 levels in 42 patients with systolic HF compared with 15 healthy controls, showing a notable positive association between miR-155 and left ventricular mass index. Conversely, a study by Ding et al (190), involving plasma samples from 62 healthy controls and 62 patients with HF, revealed higher miR-155-5p mRNA expression in patients with HF, suggesting miR-155-5p as a potential novel diagnostic biomarker for HF and related disorders. These contrasting findings highlight the functional complexity of miR-155 in cardiovascular pathophysiology.
In terms of cerebral stroke research, Zhang et al (31) recruited 93 patients with IS and 70 healthy controls, reporting elevated plasma endothelial microvesicle (EMV) and EMV-miR-155 levels during the acute and subacute phases of IS, with stable levels in the chronic phase. These elevations were positively associated with both infarct volume and National Institutes of Health Stroke Scale scores (31), suggesting plasma EMVs and their carried miRNA-155 as potential biomarkers for IS. Similarly, Adly Sadik et al (171) analyzed peripheral serum miR-155 expression in 46 patients with acute IS, revealing an 8.5-fold increase compared with healthy controls (171).
In terms of AAA research, Kin et al (191) collected tissue samples from AAA walls (n=13) during repair surgery and normal aortic walls (n=7) during valve replacement. Using high-throughput miRNA arrays for expression profiling, followed by qPCR validation of differentially expressed miRNAs, the authors revealed upregulation of miR-155 in AAA tissues (191). A complementary study comparing serum samples from 10 patients with AAA repair and 10 age- and sex-matched AAA-free controls revealed markedly elevated serum miR-155 levels in patients with AAA compared with controls (192). These collective findings highlight the need for further investigation into the pathogenic role of miR-155 in AAA development and its potential as a diagnostic biomarker.
miR-155 holds promise as a biomarker for CCVDs, serving a pivotal role in their pathogenesis and progression. Alterations in miR-155 expression are strongly associated with disease severity and clinical prognosis. Quantitative detection of miR-155 expression can facilitate earlier identification of disease risk, providing a foundation for timely clinical intervention and therapeutic management (171,184,185,189). Therefore, further exploration of the mechanistic pathways of miR-155, along with the development of miR-155-based diagnostic tools and therapeutic strategies targeting miR-155, presents substantial potential for breakthroughs in the prevention and treatment of these diseases.
In the field of precision medicine, innovative therapies based on miRNAs are gaining increasing attention, with miR-155 standing out due to its pivotal role in the pathological and physiological processes of CVDs (19,60,193). As a critical therapeutic candidate, miR-155 has garnered interest in biomedical research. While preclinical investigations of miR-155 extend across various disciplines beyond CCVDs, the mechanistic insights gained have considerable translational implications for the advancement of vascular medicine (13,46,58). Notably, cobomarsen, a synthetic oligonucleotide inhibitor specifically targeting miR-155, has shown therapeutic promise in oncology. Its action, validated in hematologic malignancies, demonstrates particular efficacy in managing non-Hodgkin lymphoma and cutaneous T-cell lymphoma (in a xenograft NSG mouse model of the activated B-cell subtype of diffuse large B-cell lymphoma), with ongoing clinical trials confirming favorable pharmacodynamic profiles (194,195). These studies indicate that cobomarsen can inhibit tumor growth and regulate critical signaling pathways, such as the JAK/STAT, MAPK/ERK and PI3K/AKT pathways, to exert antitumor effects (194,195). In fibrosis research, local injection of miR-155 antagonists has been shown to inhibit the Wnt/β-catenin and AKT signaling pathways, thereby reducing skin collagen deposition and improving fibrosis (196). Experiments have revealed that this antagomiR-155 targets the regulation of casein kinase 1α and SHIP-1, blocking key fibrotic pathways (196). Additionally, systemic delivery of miR-155-5p inhibitors (antigomiR-155) has been shown to reduce lipid accumulation in macrophages and reduce atherosclerotic plaque burden in ApoE-/- mice (197). Another study involving intravenous injection of miR-155 inhibitors starting 48 h post-distal MCAO in mice demonstrated a reduction in C-C motif chemokine ligand 3 and C-C motif chemokine ligand 12 cytokine expression after 7 days, with notable increases in IL-10, IL-4, IL-6, macrophage inflammatory protein-1α, IL-5 and IL-17 levels after 14 days (27). These findings suggest that miR-155 inhibition in stroke models alters the temporal progression of cytokine expression, potentially influencing inflammation and tissue repair following cerebral ischemia (27). Furthermore, magnetic resonance imaging examination revealed that the miR-155 inhibitor group exhibited a 34% reduction in infarct volume compared with the control group after 21 days (14.43% in controls vs. 9.5% in the inhibitor group (42). These findings highlight the potential of miRNA-based therapies in targeting key pathogenic pathways in diseases. miR-155-based therapies, in particular, hold great promise for the treatment of CVDs. With ongoing research and technological advancements, this novel approach may offer new options for the treatment of patients with CVD.
miR-155, a highly conserved non-coding RNA, regulates target gene expression and serves a critical role in the pathological processes of various CCVDs, including AS, MI, HF, hypertension and stroke. miR-155 is involved in mechanisms such as endothelial dysfunction, inflammation, oxidative stress, apoptosis and fibrosis. Notably, miR-155 exhibits a dual nature, exerting both pro-inflammatory and anti-inflammatory effects, depending on the specific disease context and microenvironment. Additionally, miR-155 holds promise as a diagnostic biomarker for CCVDs and represents a potential target for gene therapy. For example, cobomarsen, an anti-miR-155 oligonucleotide, has shown therapeutic efficacy in a xenograft NSG mouse model of the activated B-cell subtype of diffuse large B-cell lymphoma (195).
However, current research on miR-155 faces several challenges. Firstly, the mechanistic complexity of miR-155 remains incompletely understood. For instance, in AS, overexpression of miR-155 inhibits palmitic acid-induced apoptosis, ROS production and pro-inflammatory cytokine release in HUVECs by suppressing the Wnt signaling pathway (78), thereby mitigating the progression of AS. However, studies have also demonstrated that overexpression of miR-155-5p reduces AKT1 levels and its phosphorylation, thereby inhibiting VSMC proliferation and migration (92) while decreasing VSMC apoptosis (71), which conversely promotes AS development. Across different diseases, miR-155 exhibits functional complexity. In AS, elevated miR-155 levels enhance ox-LDL-mediated autophagy in HUVECs by inhibiting the PI3K/AKT/mTOR signaling pathway (83), thereby alleviating inflammation during AS progression. Conversely, researchers have found that overexpression of miR-155-5p promotes aneurysm formation by targeting FOS and ZIC3 to inhibit VSMC survival (178). These discrepancies may stem from the multifunctionality of its upstream and downstream genes. While multiple mRNAs have been identified as direct targets of miR-155-5p, these targets could also be regulated by other miRNAs. Furthermore, alterations in miR-155-5p may induce expression changes in associated upstream genes, such as lncRNAs and circular RNAs, potentially contributing to its differential expression patterns in the same disease. This remains a key area for future research. Secondly, validation of miR-155 target genes remains insufficient. Numerous studies have demonstrated that miR-155 can regulate the expression levels of various molecules (for example, Oprm1 and CTRP12) (23,57), but have not yet confirmed whether miR-155 directly targets and modulates their expression, with the intermediate molecules between them remaining unclear. Thirdly, animal models, predominantly murine models (such as ApoE- mice), have limitations due to species-specific differences from human diseases, and clinical data are still limited. Fourthly, most clinical studies related to miR-155 have small sample sizes, which compromises the stability and reliability of the findings and makes it difficult to comprehensively and accurately reflect the true role of miR-155 in relevant diseases or physiological processes. Additionally, small-scale studies may fail to adequately account for inter-individual variations, such as variations in age, sex, ethnicity, lifestyle and underlying medical conditions, which influence miR-155 expression and function, thereby limiting the generalizability and clinical applicability of the conclusions. Lastly, therapeutic strategies targeting miR-155 are in the early stages. While cobomarsen, a miR-155 inhibitor, shows efficacy in oncology, its safety, delivery efficiency and long-term effects in CCVDs require further validation.
The proposed solutions to address these challenges primarily include: First, utilizing single-cell sequencing and spatial transcriptomics to elucidate the cell type-specific functions of miR-155, complemented by the development of humanized animal models that better replicate human disease microenvironments. Second, employing multimodal experimental validation, such as dual-luciferase assays to confirm direct binding interactions, followed by RT-qPCR and western blot analyses of target genes (such as Oprm1 and CTRP12) after miR-155 overexpression or inhibition, with subsequent validation in both cellular and animal models. Third, implementing multicenter clinical studies incorporating organoid models to assess therapeutic efficacy and safety. Finally, phase I/II trials should prioritize high-risk CCVD populations (for example, patients with familial hypercholesterolemia) (198), integrating dynamic monitoring technologies such as fluorescent reporter genes for real-time tracking of miR-155 activity, with dosage adjustments based on circulating miR-155 levels.
Systemic regulation targeting miR-155 requires careful consideration of several key issues. First, off-target effects present significant risks: miR-155 modulates hundreds of genes (such as eNOS, SOCS1 and Bcl-6), and systemic inhibition or overexpression may disrupt physiological functions in non-target tissues, potentially causing immune dysregulation. As a key regulator of Th1/Th17 cell differentiation, miR-155 suppression could compromise anti-infective immunity and increase susceptibility to infections (172,199). Paradoxically, inhibition of miR-155 could enhance VSMC proliferation, leading to vascular stenosis or restenosis and disrupting vascular homeostasis (200). Second, delivery systems lack specificity: Current technologies, such as lipid nanoparticles or viral vectors, struggle to precisely target diseased tissues, risking hepatorenal toxicity (201,202). Third, dose-dependent toxicity may arise: miR-155 exhibits nonlinear disease associations; insufficient levels may impair anti-inflammatory effects (41), while excessive expression could promote fibrosis [for example, cardiac fibrosis (203)]. Future research should integrate single-cell sequencing and CRISPR screening to clarify cell-specific miR-155 functions and develop tissue-targeted delivery systems (such as exosomal vectors) (92,177,180,204,205). Conventional carriers, such as lipid or viral vectors, face issues such as hepatic sequestration (causing hepatotoxicity) and poor vascular barrier penetration (for example, of the blood-brain barrier) (206,207). Innovative solutions include: i) Bioinspired nanocarriers, including engineering exosomes to encapsulate miR-155 inhibitors (such as antagomirs) for degradation protection, with surface-conjugated VCAM-1 antibodies for precise binding to inflamed endothelium; ii) biomimetic membrane coatings, such as macrophage membrane-wrapped nanoparticles [for example, poly(lactide-co-glycolide)-polyethylene glycol cores loaded with inhibitors] that leverage innate chemotaxis to migrate toward lesions (such as plaques and infarcted myocardium); and iii) preclinical validation in large animal models with dynamic monitoring (such as fluorescent reporters) for real-time dose adjustments (208-210).
Despite being in the early stages of development, miRNA-based therapies demonstrate substantial potential. As of August 2025, no miRNA drugs have received global approval, although ~100 candidates are actively being investigated across therapeutic areas, including oncology, rare diseases, CVDs, metabolic diseases and inflammatory conditions (211-213). Clinical progress remains measured, with only a few candidates advancing to clinical trials. Notably, obefazimod (targeting miR-124) for ulcerative colitis achieved a 16.4% clinical remission rate in phase III trials (NCT05507216; July 2025), with the ABTECT-1 subgroup reaching a clinical remission rate of 19.3% (214). This indicates that it is a highly promising miRNA drug candidate for the future treatment of ulcerative colitis. In CVDs, CDR132L (a miR-132 inhibitor) completed a phase II trial (NCT05350969), demonstrating myocardial functional recovery in HF (215). Other notable candidates include miravirsen (miR-122 inhibitor for hepatitis C virus; phase II; NCT01200420) (216), CWT-001 (miR-29a mimic for tendinopathy; phase II; NCT06192927) and TTX-MC138 (miR-10b inhibitor for breast/pancreatic cancers; phase I; NCT06260774). However, safety concerns have stalled some pipelines. For instance, a trial investigating MRX34 (miR-34a mimic; NCT01829971) was halted due to severe immune-related adverse events and is currently under reevaluation (217), highlighting the translational challenges. No miR-155-targeted therapies have currently entered clinical trials. However, these clinical studies (214-217) strongly support the general feasibility of miRNA-based therapeutics, providing robust evidence for miR-155 as a potential therapeutic target. With deepening insights into miRNA mechanisms, future development of miR-155-specific therapies may yield breakthroughs in treating CCVDs.
In conclusion, the integration of preclinical research with emerging breakthroughs in miRNA therapeutics across diverse medical fields positions miR-155 as a promising therapeutic candidate for CCVDs. This strategic approach may pave the way for novel therapeutic interventions in cerebrovascular disease. The expanding understanding of the multifunctional regulation of miR-155 in CCVD pathophysiology has enhanced the prospects for precision-targeted therapies, potentially driving paradigm shifts in vascular medicine. As knowledge of the dual regulatory roles of this miRNA in both vascular homeostasis and disease progression deepens, rationally designed miR-155 modulators may ultimately redefine therapeutic standards for complex cerebrovascular conditions.
Not applicable.
PW contributed to the conception and design, and critically revised the manuscript. XZ was responsible for conceptualization, validation and writing the original draft of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
AAA |
abdominal aortic aneurysm |
|
ABCA1 |
ATP binding cassette transporter A1 |
|
AGTR1 |
angiotensin II receptor subtype 1 |
|
ANP |
atrial natriuretic peptide |
|
AS |
atherosclerosis |
|
AT1R |
angiotensin II type I receptor |
|
BACH1 |
BTB domain and CNC homolog 1 |
|
BDNF |
brain-derived neurotrophic factor |
|
BIC |
B-cell integration cluster |
|
CCVD |
cardiovascular and cerebrovascular disease |
|
ceRNA |
competitive endogenous RNA |
|
CVD |
cardiovascular disease |
|
CTRP12 |
C1q tumor necrosis factor related protein 12 |
|
Cx43 |
connexin 43 |
|
CYTOR |
cytoskeleton regulator RNA |
|
DUSP14 |
dual specificity phosphatase 14 |
|
EC |
endothelial cell |
|
EMV |
endothelial microvesicle |
|
eNOS |
endothelial nitric oxide synthase |
|
HIF-1α |
hypoxia-inducible factor 1α |
|
HF |
heart failure |
|
IA |
intracranial aneurysm |
|
iCMP |
inflammatory cardiomyopathy |
|
I/R |
ischemia-reperfusion |
|
IRI |
ischemia-reperfusion injury |
|
IS |
ischemic stroke |
|
JAK |
Janus kinase |
|
LIN28 |
lineage protein 28 |
|
LXRα |
liver X receptor α |
|
MCAO |
middle cerebral artery occlusion |
|
MI |
myocardial infarction |
|
miR-155 |
microRNA-155 |
|
MSC |
mesenchymal stem cell |
|
NLRP3 |
NLR family pyrin domain containing 3 |
|
NO |
nitric oxide |
|
OGD/R |
oxygen glucose deprivation/reoxygenation |
|
PDCD4 |
programmed cell death 4 |
|
ROS |
reactive oxygen species |
|
SIRT1 |
sirtuin 1 |
|
sGCβ1 |
soluble guanylate cyclase β1 subunit |
|
SOCS1 |
suppressor of cytokine signaling 1 |
|
TLR |
toll like receptor |
|
VSMC |
vascular smooth muscle cell |
Not applicable.
The present study was supported by the Liaoning Province Science and Technology Plan Joint Program (grant no. 2024-MSLH-305), the Basic Research Projects for Higher Education Institutions of Liaoning Province in 2022 (grant no. LJKMZ20221337), and the Joint Project of Shenyang Science and Technology Bureau (grant no. 21-174-9-13).
|
Magalhães JE and Sampaio Rocha-Filho PA: Migraine and cerebrovascular diseases: Epidemiology, pathophysiological, and clinical considerations. Headache. 58:1277–1286. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zanon Zotin MC, Sveikata L, Viswanathan A and Yilmaz P: Cerebral small vessel disease and vascular cognitive impairment: From diagnosis to management. Curr Opin Neurol. 34:246–257. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Gottesman RF and Seshadri S: Risk factors, lifestyle behaviors, and vascular brain health. Stroke. 53:394–403. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Nordestgaard LT, Christoffersen M and Frikke-Schmidt R: Shared risk factors between dementia and atherosclerotic cardiovascular disease. Int J Mol Sci. 23:97772022. View Article : Google Scholar : PubMed/NCBI | |
|
Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Beaton AZ, Boehme AK, Buxton AE, et al: Heart disease and stroke statistics-2023 update: A report from the American heart association. Circulation. 147:e93–e621. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Hilkens NA, Casolla B, Leung TW and de Leeuw FE: Stroke. Lancet. 403:2820–2836. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Goldsborough E III, Osuji N and Blaha MJ: Assessment of cardiovascular disease risk: A 2022 update. Endocrinol Metab Clin North Am. 51:483–509. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Z, Ma L, Liu M, Fan J and Hu S: Summary of the 2022 report on cardiovascular health and diseases in China. Chin Med J (Engl). 136:2899–2908. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Zhang H and Zou Z: Changing profiles of cardiovascular disease and risk factors in China: A secondary analysis for the Global Burden of Disease Study 2019. Chin Med J (Engl). 136:2431–2441. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W, Liu Y, Liu J, Yin P, Wang L, Qi J, You J, Lin L, Meng S, Wang F, et al: Mortality and years of life lost of cardiovascular diseases in China, 2005-2020: Empirical evidence from national mortality surveillance system. Int J Cardiol. 340:105–112. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yun CW and Lee SH: Enhancement of functionality and therapeutic efficacy of Cell-based therapy using mesenchymal stem cells for cardiovascular disease. Int J Mol Sci. 20:9822019. View Article : Google Scholar : PubMed/NCBI | |
|
Biasiolo M, Sales G, Lionetti M, Agnelli L, Todoerti K, Bisognin A, Coppe A, Romualdi C, Neri A and Bortoluzzi S: Impact of host genes and strand selection on miRNA and miRNA* expression. PLoS One. 6:e238542011. View Article : Google Scholar : PubMed/NCBI | |
|
Due H, Svendsen P, Bødker JS, Schmitz A, Bøgsted M, Johnsen HE, El-Galaly TC, Roug AS and Dybkær K: miR-155 as a biomarker in B-cell malignancies. Biomed Res Int. 2016:95130372016. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Gao D, Shao Z, Zheng Q and Yu Q: miR-155 indicates the fate of CD4+ T cells. Immunol Lett. 224:40–49. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang X, Zeng X, Shu J, Bao H and Liu X: MiR-155 enhances phagocytosis of alveolar macrophages through the mTORC2/RhoA pathway. Medicine (Baltimore). 102:e345922023. View Article : Google Scholar : PubMed/NCBI | |
|
Tili E, Croce CM and Michaille JJ: miR-155: On the crosstalk between inflammation and cancer. Int Rev Immunol. 28:264–284. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Suofu Y, Wang X, He Y, Li F, Zhang Y, Carlisle DL and Friedlander RM: Mir-155 knockout protects against Ischemia/reperfusion-induced brain injury and hemorrhagic transformation. Neuroreport. 31:235–239. 2020. View Article : Google Scholar | |
|
Wu L, Pu L and Zhuang Z: miR-155-5p/FOXO3a promotes pulmonary fibrosis in rats by mediating NLRP3 inflammasome activation. Immunopharmacol Immunotoxicol. 45:257–267. 2023. View Article : Google Scholar | |
|
Cao RY, Li Q, Miao Y, Zhang Y, Yuan W, Fan L, Liu G, Mi Q and Yang J: The emerging role of MicroRNA-155 in cardiovascular diseases. Biomed Res Int. 2016:98692082016. View Article : Google Scholar : PubMed/NCBI | |
|
Chen JG, Xu XM, Ji H and Sun B: Inhibiting miR-155 protects against myocardial ischemia/reperfusion injury via targeted regulation of HIF-1α in rats. Iran J Basic Med Sci. 22:1050–1058. 2019.PubMed/NCBI | |
|
Guo J, Liu HB, Sun C, Yan XQ, Hu J, Yu J, Yuan Y and Du ZM: MicroRNA-155 promotes myocardial infarction-induced apoptosis by targeting RNA-Binding protein QKI. Oxid Med Cell Longev. 2019:45798062019. View Article : Google Scholar : PubMed/NCBI | |
|
Chen W, Wang L and Liu Z: MicroRNA-155 influences cell damage in ischemic stroke via TLR4/MYD88 signaling pathway. Bioengineered. 12:2449–2458. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Bai B, Ji Z, Wang F, Qin C, Zhou H, Li D and Wu Y: CTRP12 ameliorates post-myocardial infarction heart failure through down-regulation of cardiac apoptosis, oxidative stress and inflammation by influencing the TAK1-p38 MAPK/JNK pathway. Inflamm Res. 72:1375–1390. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Liang K, Zou G, Chen X, Shi S, Wang G, Zhang K, Li K and Zhai S: Inhibition of miR-155 attenuates abdominal aortic aneurysm in mice by regulating macrophage-mediated inflammation. Biosci Rep. 38:BSR201714322018. View Article : Google Scholar : PubMed/NCBI | |
|
Wu XY, Fan WD, Fang R and Wu GF: Regulation of microRNA-155 in endothelial inflammation by targeting nuclear factor (NF)-κB P65. J Cell Biochem. 115:1928–1936. 2014.PubMed/NCBI | |
|
Park M, Choi S, Kim S, Kim J, Lee DK, Park W, Kim T, Jung J, Hwang JY, Won MH, et al: NF-κB-responsive miR-155 induces functional impairment of vascular smooth muscle cells by downregulating soluble guanylyl cyclase. Exp Mol Med. 51:1–12. 2019. | |
|
Pena-Philippides JC, Caballero-Garrido E, Lordkipanidze T and Roitbak T: In vivo inhibition of miR-155 significantly alters post-stroke inflammatory response. J Neuroinflammation. 13:2872016. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Han W, Zhang Y, Zong Y, Tan N, Zhang Y, Li L, Liu C and Liu L: Soluble epoxide hydrolase inhibitor t-AUCB ameliorates vascular endothelial dysfunction by influencing the NF-κB/miR-155-5p/eNOS/NO/IκB Cycle in hypertensive rats. Antioxidants (Basel). 11:13722022. View Article : Google Scholar | |
|
Faccini J, Ruidavets JB, Cordelier P, Martins F, Maoret JJ, Bongard V, Ferrières J, Roncalli J, Elbaz M and Vindis C: Circulating miR-155, miR-145 and let-7c as diagnostic biomarkers of the coronary artery disease. Sci Rep. 7:429162017. View Article : Google Scholar : PubMed/NCBI | |
|
Huang YQ, Huang C, Zhang B and Feng YQ: Association of circulating miR-155 expression level and inflammatory markers with white coat hypertension. J Hum Hypertens. 34:397–403. 2020. View Article : Google Scholar | |
|
Zhang H, Chen G, Qiu W, Pan Q, Chen Y, Chen Y and Ma X: Plasma endothelial microvesicles and their carrying miRNA-155 serve as biomarkers for ischemic stroke. J Neurosci Res. 98:2290–2301. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Jin ZQ: MicroRNA targets and biomarker validation for diabetes-associated cardiac fibrosis. Pharmacol Res. 174:1059412021. View Article : Google Scholar : PubMed/NCBI | |
|
Sun G, Yan J, Noltner K, Feng J, Li H, Sarkis DA, Sommer SS and Rossi JJ: SNPs in human miRNA genes affect biogenesis and function. RNA. 15:1640–1651. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Tam W: Identification and characterization of human BIC, a gene on chromosome 21 that encodes a noncoding RNA. Gene. 274:157–167. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Li C, He H, Zhu M, Zhao S and Li X: Molecular characterisation of porcine miR-155 and its regulatory roles in the TLR3/TLR4 pathways. Dev Comp Immunol. 39:110–116. 2013. View Article : Google Scholar | |
|
Rodriguez A, Vigorito E, Clare S, Warren MV, Couttet P, Soond DR, van Dongen S, Grocock RJ, Das PP, Miska EA, et al: Requirement of bic/microRNA-155 for normal immune function. Science. 316:608–611. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Ruggiero T, Trabucchi M, De Santa F, Zupo S, Harfe BD, McManus MT, Rosenfeld MG, Briata P and Gherzi R: LPS induces KH-type splicing regulatory protein-dependent processing of microRNA-155 precursors in macrophages. FASEB J. 23:2898–2908. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
O'Connell RM, Taganov KD, Boldin MP, Cheng G and Baltimore D: MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci USA. 104:1604–1609. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Guo Q, Zhang H, Zhang B, Zhang E and Wu Y: Tumor necrosis factor-alpha (TNF-α) enhances miR-155-Mediated endothelial senescence by targeting sirtuin1 (SIRT1). Med Sci Monit. 25:8820–8835. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Guo G, Zhong Z, Sun L, Liao L, Wang X, Cao Q and Chen H: Long non-coding RNA FLVCR1-AS1 sponges miR-155 to promote the tumorigenesis of gastric cancer by targeting c-Myc. Am J Transl Res. 11:793–805. 2019.PubMed/NCBI | |
|
Huang RS, Hu GQ, Lin B, Lin ZY and Sun CC: MicroRNA-155 silencing enhances inflammatory response and lipid uptake in oxidized Low-density Lipoprotein-stimulated human THP-1 macrophages. J Investig Med. 58:961–967. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Caballero-Garrido E, Pena-Philippides JC, Lordkipanidze T, Bragin D, Yang Y, Erhardt EB and Roitbak T: In Vivo inhibition of miR-155 promotes recovery after experimental mouse stroke. J Neurosci. 35:12446–12464. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Bruen R, Fitzsimons S and Belton O: miR-155 in the resolution of atherosclerosis. Front Pharmacol. 10:4632019. View Article : Google Scholar : PubMed/NCBI | |
|
Uva P, Da Sacco L, Del Cornò M, Baldassarre A, Sestili P, Orsini M, Palma A, Gessani S and Masotti A: Rat mir-155 generated from the lncRNA Bic is 'hidden' in the alternate genomic assembly and reveals the existence of novel mammalian miRNAs and clusters. RNA. 19:365–379. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Thai TH, Calado DP, Casola S, Ansel KM, Xiao C, Xue Y, Murphy A, Frendewey D, Valenzuela D, Kutok JL, et al: Regulation of the germinal center response by microRNA-155. Science. 316:604–608. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Schliesser MG, Claus R, Hielscher T, Grimm C, Weichenhan D, Blaes J, Wiestler B, Hau P, Schramm J, Sahm F, et al: Prognostic relevance of miRNA-155 methylation in anaplastic glioma. Oncotarget. 7:82028–82045. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Heo I, Joo C, Cho J, Ha M, Han J and Kim VN: Lin28 mediates the terminal uridylation of let-7 precursor MicroRNA. Mol Cell. 32:276–284. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Lu C, Chen B, Chen C, Li H, Wang D, Tan Y and Weng H: CircNr1h4 regulates the pathological process of renal injury in salt-sensitive hypertensive mice by targeting miR-155-5p. J Cell Mol Med. 24:1700–1712. 2020. View Article : Google Scholar | |
|
Pasca S, Jurj A, Petrushev B, Tomuleasa C and Matei D: MicroRNA-155 implication in M1 polarization and the impact in inflammatory diseases. Front Immunol. 11:6252020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Cheng C, Yuan X, He JT, Pan QH and Sun FY: microRNA-155 acts as an oncogene by targeting the tumor protein 53-induced nuclear protein 1 in esophageal squamous cell carcinoma. Int J Clin Exp Pathol. 7:602–610. 2014.PubMed/NCBI | |
|
Li Y, Zhang L, Dong Z, Xu H, Yan L, Wang W, Yang Q and Chen C: MicroRNA-155-5p promotes tumor progression and contributes to paclitaxel resistance via TP53INP1 in human breast cancer. Pathol Res Pract. 220:1534052021. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao L, Li X, Mu Z, Zhou J, Zhou P, Xie C and Jiang S: FTO inhibition enhances the antitumor effect of temozolomide by targeting MYC-miR-155/23a Cluster-MXI1 feedback circuit in glioma. Cancer Res. 80:3945–3958. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Costinean S, Zanesi N, Pekarsky Y, Tili E, Volinia S, Heerema N and Croce CM: Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice. Proc Natl Acad Sci USA. 103:7024–7029. 2006. 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. 122:4190–4202. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Kurowska-Stolarska M, Alivernini S, Ballantine LE, Asquith DL, Millar NL, Gilchrist DS, Reilly J, Ierna M, Fraser AR, Stolarski B, et al: MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis. Proc Natl Acad Sci USA. 108:11193–11198. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Wang G, Chen JJ, Deng WY, Ren K, Yin SH and Yu XH: CTRP12 ameliorates atherosclerosis by promoting cholesterol efflux and inhibiting inflammatory response via the miR-155-5p/LXRα pathway. Cell Death Dis. 12:2542021. View Article : Google Scholar | |
|
Jing H, Liu L, Jia Y, Yao H and Ma F: Overexpression of the long non-coding RNA Oprm1 alleviates apoptosis from cerebral Ischemia-reperfusion injury through the Oprm1/miR-155/GATA3 axis. Artif Cells Nanomed Biotechnol. 47:2431–2439. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Xue H, Hua LM, Guo M and Luo JM: SHIP1 is targeted by miR-155 in acute myeloid leukemia. Oncol Rep. 32:2253–2259. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Husain K, Villalobos-Ayala K, Laverde V, Vazquez OA, Miller B, Kazim S, Blanck G, Hibbs ML, Krystal G, Elhussin I, et al: Apigenin targets MicroRNA-155, enhances SHIP-1 expression, and augments anti-tumor responses in pancreatic cancer. Cancers (Basel). 14:36132022. View Article : Google Scholar : PubMed/NCBI | |
|
Faraoni I, Antonetti FR, Cardone J and Bonmassar E: miR-155 gene: A typical multifunctional microRNA. Biochim Biophys Acta. 1792:497–505. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Johansson J, Berg T, Kurzejamska E, Pang MF, Tabor V, Jansson M, Roswall P, Pietras K, Sund M, Religa P, et al: MiR-155-mediated loss of C/EBPβ shifts the TGF-β response from growth inhibition to epithelial-mesenchymal transition, invasion and metastasis in breast cancer. Oncogene. 32:5614–5624. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang D and Aguiar RC: MicroRNA-155 controls RB phosphorylation in normal and malignant B lymphocytes via the noncanonical TGF-β1/SMAD5 signaling module. Blood. 123:86–93. 2014. View Article : Google Scholar : | |
|
Li DP, Fan J, Wu YJ, Xie YF, Zha JM and Zhou XM: MiR-155 up-regulated by TGF-β promotes epithelial-mesenchymal transition, invasion and metastasis of human hepatocellular carcinoma cells in vitro. Am J Transl Res. 9:2956–2965. 2017. | |
|
Ke F, Wang H, Geng J, Jing X, Fang F, Fang C and Zhang BH: MiR-155 promotes inflammation and apoptosis via targeting SIRT1 in hypoxic-ischemic brain damage. Exp Neurol. 362:1143172023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhai Y, Liu B, Wu L, Zou M, Mei X and Mo X: Pachymic acid prevents neuronal cell damage induced by hypoxia/reoxygenation via miR-155/NRF2/HO-1 axis. Acta Neurobiol Exp (Wars). 82:197–206. 2022. | |
|
Zhang W, Wang L, Wang R, Duan Z and Wang H: A blockade of microRNA-155 signal pathway has a beneficial effect on neural injury after intracerebral haemorrhage via reduction in neuroinflammation and oxidative stress. Arch Physiol Biochem. 128:1235–1241. 2022. View Article : Google Scholar | |
|
Sun L, Ji S and Xing J: Inhibition of microRNA-155 alleviates neurological dysfunction following transient global ischemia and contribution of neuroinflammation and oxidative stress in the hippocampus. Curr Pharm Des. 25:4310–4317. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yang J, Si D, Zhao Y, He C and Yang P: S-amlodipine improves endothelial dysfunction via the RANK/RANKL/OPG system by regulating microRNA-155 in hypertension. Biomed Pharmacother. 114:1087992019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Y, Xian X, Wang Z, Bi Y, Chen Q, Han X, Tang D and Chen R: Research progress on the relationship between atherosclerosis and inflammation. Biomolecules. 8:802018. 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 | |
|
González-López P, Ares-Carral C, López-Pastor AR, Infante-Menéndez J, González Illaness T, Vega de Ceniga M, Esparza L, Beneit N, Martín-Ventura JL, Escribano Ó and Gómez-Hernández A: Implication of miR-155-5p and miR-143-3p in the vascular insulin resistance and instability of human and experimental atherosclerotic plaque. Int J Mol Sci. 23:102532022. View Article : Google Scholar : PubMed/NCBI | |
|
Gimbrone MA Jr and García-Cardeña G: Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res. 118:620–636. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Ota H, Eto M, Ogawa S, Iijima K, Akishita M and Ouchi Y: SIRT1/eNOS axis as a potential target against vascular senescence, dysfunction and atherosclerosis. J Atheroscler Thromb. 17:431–435. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Hong FF, Liang XY, Liu W, Lv S, He SJ, Kuang HB and Yang SL: Roles of eNOS in atherosclerosis treatment. Inflamm Res. 68:429–441. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Sun HX, Zeng DY, Li RT, Pang RP, Yang H, Hu YL, Zhang Q, Jiang Y, Huang LY, Tang YB, et al: Essential role of microRNA-155 in regulating Endothelium-dependent vasorelaxation by targeting endothelial nitric oxide synthase. Hypertension. 60:1407–1414. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu N, Zhang D, Chen S, Liu X, Lin L, Huang X, Guo Z, Liu J, Wang Y, Yuan W and Qin Y: Endothelial enriched microRNAs regulate angiotensin II-induced endothelial inflammation and migration. Atherosclerosis. 215:286–293. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lee HJ, Lee EJ and Seo M: Galpha12 protects vascular endothelial cells from serum Withdrawal-induced apoptosis through regulation of miR-155. Yonsei Med J. 57:247–253. 2016. View Article : Google Scholar : | |
|
Zhao Y, Rao W, Wan Y, Yang X, Wang G, Deng J, Dai M and Liu Q: Overexpression of microRNA-155 alleviates palmitate-induced vascular endothelial cell injury in human umbilical vein endothelial cells by negatively regulating the Wnt signaling pathway. Mol Med Rep. 20:3527–3534. 2019.PubMed/NCBI | |
|
Wei DH, Jia XY, Liu YH, Guo FX, Tang ZH, Li XH, Wang Z, Liu LS, Wang GX, Jian ZS and Ruan CG: Cathepsin L stimulates autophagy and inhibits apoptosis of ox-LDL-induced endothelial cells: Potential role in atherosclerosis. Int J Mol Med. 31:400–406. 2013. View Article : Google Scholar | |
|
Hu M, Ladowski JM and Xu H: The role of autophagy in vascular endothelial cell health and physiology. Cells. 13:8252024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang X, Ma C, Gao Y, Cui H, Zheng Y, Li J, Zong W and Zhang Q: Tongxinluo attenuates atherosclerosis by inhibiting ROS/NLRP3/caspase-1-mediated endothelial cell pyroptosis. J Ethnopharmacol. 304:1160112023. View Article : Google Scholar | |
|
Zhang Z, Pan X, Yang S, Ma A, Wang K, Wang Y, Li T and Liu S: miR-155 Promotes ox-LDL-Induced autophagy in human umbilical vein endothelial cells. Mediators Inflamm. 2017:91748012017. View Article : Google Scholar : PubMed/NCBI | |
|
Yin S, Yang S, Pan X, Ma A, Ma J, Pei H, Dong Y, Li S, Li W and Bi X: MicroRNA-155 promotes ox-LDL-induced autophagy in human umbilical vein endothelial cells by targeting the PI3K/Akt/mTOR pathway. Mol Med Rep. 18:2798–2806. 2018.PubMed/NCBI | |
|
McCully KS: Homocysteine and the pathogenesis of atherosclerosis. Expert Rev Clin Pharmacol. 8:211–219. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Witucki Ł and Jakubowski H: Homocysteine metabolites inhibit autophagy by upregulating miR-21-5p, miR-155-5p, miR-216-5p, and miR-320c-3p in human vascular endothelial cells. Sci Rep. 14:71512024. View Article : Google Scholar : PubMed/NCBI | |
|
Alonso-Piñeiro JA, Gonzalez-Rovira A, Sánchez-Gomar I, Moreno JA and Durán-Ruiz MC: Nrf2 and heme Oxygenase-1 involvement in atherosclerosis related oxidative stress. Antioxidants (Basel). 10:14632021. View Article : Google Scholar : PubMed/NCBI | |
|
Pulkkinen KH, Ylä-Herttuala S and Levonen AL: Heme oxygenase 1 is induced by miR-155 via reduced BACH1 translation in endothelial cells. Free Radic Biol Med. 51:2124–2131. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Gou L, Liu G, Ma R, Regmi A, Zeng T, Zheng J, Zhong X and Chen L: High fat-induced inflammation in vascular endothelium can be improved by Abelmoschus esculentus and metformin via increasing the expressions of miR-146a and miR-155. Nutr Metab (Lond). 17:352020. View Article : Google Scholar : PubMed/NCBI | |
|
Yang D, Wang J, Xiao M, Zhou T and Shi X: Role of Mir-155 in controlling HIF-1α level and promoting endothelial cell maturation. Sci Rep. 6:353162016. View Article : Google Scholar | |
|
Frismantiene A, Philippova M, Erne P and Resink TJ: Smooth muscle cell-driven vascular diseases and molecular mechanisms of VSMC plasticity. Cell Signal. 52:48–64. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Choi S, Park M, Kim J, Park W, Kim S, Lee DK, Hwang JY, Choe J, Won MH, Ryoo S, et al: TNF-α elicits phenotypic and functional alterations of vascular smooth muscle cells by miR-155-5p-dependent down-regulation of cGMP-dependent kinase 1. J Biol Chem. 293:14812–14822. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Zheng SY, Yang CQ, Ma BM and Jiang D: MiR-155-5p inhibits the proliferation and migration of VSMCs and HUVECs in atherosclerosis by targeting AKT1. Eur Rev Med Pharmacol Sci. 23:2223–2233. 2019.PubMed/NCBI | |
|
Tong Y, Zhou MH, Li SP, Zhao HM, Zhang YR, Chen D, Wu YX and Pang QF: MiR-155-5p attenuates vascular smooth muscle cell oxidative stress and migration via inhibiting BACH1 expression. Biomedicines. 11:16792023. View Article : Google Scholar : PubMed/NCBI | |
|
Tang Y, Song H, Shen Y, Yao Y, Yu Y, Wei G, Long B and Yan W: MiR-155 acts as an inhibitory factor in atherosclerosis-associated arterial pathogenesis by down-regulating NoxA1 related signaling pathway in ApoE(-/-) mouse. Cardiovasc Diagn Ther. 11:1–13. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
He S, Wu C, Xiao J, Li D, Sun Z and Li M: Endothelial extracellular vesicles modulate the macrophage phenotype: Potential implications in atherosclerosis. Scand J Immunol. 87:e126482018. View Article : Google Scholar : PubMed/NCBI | |
|
Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV and Orekhov AN: Mechanisms of foam cell formation in atherosclerosis. J Mol Med (Berl). 95:1153–1165. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang F, Zhao J, Sun D and Wei N: MiR-155 inhibits transformation of macrophages into foam cells via regulating CEH expression. Biomed Pharmacother. 104:645–651. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Kong D, Chen H, Liu S, Hu H, Wu T, Wang J, Chen W, Ning Y, Li Y and Lu Z: miR-155 acts as an anti-inflammatory factor in atherosclerosis-associated foam cell formation by repressing calcium-regulated heat stable protein 1. Sci Rep. 6:217892016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu J, Chen T, Yang L, Li Z, Wong MM, Zheng X, Pan X, Zhang L and Yan H: Regulation of microRNA-155 in atherosclerotic inflammatory responses by targeting MAP3K10. PLoS One. 7:e465512012. View Article : Google Scholar : PubMed/NCBI | |
|
Ye J, Guo R, Shi Y, Qi F, Guo C and Yang L: miR-155 Regulated inflammation response by the SOCS1-STAT3-PDCD4 axis in atherogenesis. Mediators Inflamm. 2016:80601822016. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng J, Wang W, Hong T, Yang S, Shen J and Liu C: Suppression of microRNA-155 exerts an anti-inflammatory effect on CD4+ T cell-mediated inflammatory response in the pathogenesis of atherosclerosis. Acta Biochim Biophys Sin (Shanghai). 52:654–664. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Peng Q, Yin R, Zhu X, Jin L, Wang J, Pan X and Ma A: miR-155 activates the NLRP3 inflammasome by regulating the MEK/ERK/NF-κB pathway in carotid atherosclerotic plaques in ApoE(-/-) mice. J Physiol Biochem. 78:365–375. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Li X, Wang D, Jiang X, Zhang M and Lv K: Serum exosome microRNA panel as a noninvasive biomarker for molecular diagnosis of fulminant myocarditis. Mol Ther Methods Clin Dev. 20:142–151. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Oh JH, Kim GB and Seok H: Implication of microRNA as a potential biomarker of myocarditis. Clin Exp Pediatr. 65:230–238. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lewandowski P, Goławski M, Baron M, Reichman-Warmusz E and Wojnicz R: A systematic review of miRNA and cfDNA as potential biomarkers for liquid biopsy in myocarditis and inflammatory dilated cardiomyopathy. Biomolecules. 12L:14762022. View Article : Google Scholar | |
|
Aleshcheva G, Baumeier C, Harms D, Bock CT, Escher F and Schultheiss HP: MicroRNAs as novel biomarkers and potential therapeutic options for inflammatory cardiomyopathy. ESC Heart Fail. 10:3410–3418. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Bao JL and Lin L: MiR-155 and miR-148a reduce cardiac injury by inhibiting NF-κB pathway during acute viral myocarditis. Eur Rev Med Pharmacol Sci. 18:2349–2356. 2014.PubMed/NCBI | |
|
Zhang Y, Zhang M, Li X, Tang Z, Wang X, Zhong M, Suo Q, Zhang Y and Lv K: Silencing MicroRNA-155 attenuates cardiac injury and dysfunction in viral myocarditis via promotion of M2 phenotype polarization of macrophages. Sci Rep. 6:226132016. View Article : Google Scholar : PubMed/NCBI | |
|
Yan L, Hu F, Yan X, Wei Y, Ma W, Wang Y, Lu S and Wang Z: Inhibition of microRNA-155 ameliorates experimental autoimmune myocarditis by modulating Th17/Treg immune response. J Mol Med (Berl). 94:1063–1079. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Z, Dai X, Qi J, Ao Y, Yang C and Li Y: Astragalus mongholicus (Fisch.) bge improves peripheral treg cell immunity imbalance in the children with viral myocarditis by reducing the levels of miR-146b and miR-155. Front Pediatr. 6:1392018. View Article : Google Scholar : PubMed/NCBI | |
|
Derda AA, Thum S, Lorenzen JM, Bavendiek U, Heineke J, Keyser B, Stuhrmann M, Givens RC, Kennel PJ, Schulze PC, et al: Blood-based microRNA signatures differentiate various forms of cardiac hypertrophy. Int J Cardiol. 196:115–122. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
He W, Huang H, Xie Q, Wang Z, Fan Y, Kong B, Huang D and Xiao Y: MiR-155 knockout in fibroblasts improves cardiac remodeling by targeting tumor protein p53-Inducible nuclear protein 1. J Cardiovasc Pharmacol Ther. 21:423–435. 2016. View Article : Google Scholar | |
|
Kelly M, Bagnall RD, Peverill RE, Donelan L, Corben L, Delatycki MB and Semsarian C: A polymorphic miR-155 binding site in AGTR1 is associated with cardiac hypertrophy in Friedreich ataxia. J Mol Cell Cardiol. 51:848–854. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
de Oliveira G, Freire PP, Omoto ACM, Cury SS, Fuziwara CS, Kimura ET, Dal-Pai-Silva M and Carvalho RF: Osteoglycin post-transcriptional regulation by miR-155 induces cellular architecture changes in H9c2 cardiomyoblasts. Gene. 676:9–15. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Zhou Y, Cao Z, Tong XZ, Xie HQ, Luo T, Hua XP and Wang HQ: miR-155 functions downstream of angiotensin II receptor subtype 1 and calcineurin to regulate cardiac hypertrophy. Exp Ther Med. 12:1556–1562. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Seok HY, Chen J, Kataoka M, Huang ZP, Ding J, Yan J, Hu X and Wang DZ: Loss of MicroRNA-155 protects the heart from pathological cardiac hypertrophy. Circ Res. 114:1585–1595. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yu H, Qin L, Peng Y, Bai W and Wang Z: Exosomes derived from hypertrophic cardiomyocytes induce inflammation in macrophages via miR-155 mediated MAPK Pathway. Front Immunol. 11:6060452020. View Article : Google Scholar | |
|
Wang B, Wang ZM, Ji JL, Gan W, Zhang A, Shi HJ, Wang H, Lv L, Li Z, Tang T, et al: Macrophage-derived exosomal Mir-155 regulating cardiomyocyte pyroptosis and hypertrophy in uremic cardiomyopathy. JACC Basic Transl Sci. 5:148–166. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Y, Feng T, Duan S, Shi Y, Li S, Zhang X and Zhang L: miR-155 promotes fibroblast-like synoviocyte proliferation and inflammatory cytokine secretion in rheumatoid arthritis by targeting FOXO3a. Exp Ther Med. 19:1288–1296. 2020.PubMed/NCBI | |
|
Fan Y, Liu L, Fang K, Huang T, Wan L, Liu Y, Zhang S, Yan D, Li G, Gao Y, et al: Resveratrol Ameliorates cardiac hypertrophy by Down-regulation of miR-155 through activation of breast cancer type 1 susceptibility protein. J Am Heart Assoc. 5:e0026482016. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan Y, Wang J, Chen Q, Wu Q, Deng W, Zhou H and Shen D: Long non-coding RNA cytoskeleton regulator RNA (CYTOR) modulates pathological cardiac hypertrophy through miR-155-mediated IKKi signaling. Biochim Biophys Acta Mol Basis Dis. 1865:1421–1427. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Murphy A and Goldberg S: Mechanical complications of myocardial infarction. Am J Med. 135:1401–1409. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Bentzon JF, Otsuka F, Virmani R and Falk E: Mechanisms of plaque formation and rupture. Circ Res. 114:1852–1866. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wang C, Zhang C, Liu L, A X, Chen B, Li Y and Du J: Macrophage-Derived mir-155-Containing exosomes suppress fibroblast proliferation and promote fibroblast inflammation during cardiac injury. Mol Ther. 25:192–204. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Hu J, Huang CX, Rao PP, Zhou JP, Wang X, Tang L, Liu MX and Zhang GG: Inhibition of microRNA-155 attenuates sympathetic neural remodeling following myocardial infarction via reducing M1 macrophage polarization and inflammatory responses in mice. Eur J Pharmacol. 851:122–132. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Hu J, Huang CX, Rao PP, Cao GQ, Zhang Y, Zhou JP, Zhu LY, Liu MX and Zhang GG: MicroRNA-155 inhibition attenuates endoplasmic reticulum stress-induced cardiomyocyte apoptosis following myocardial infarction via reducing macrophage inflammation. Eur J Pharmacol. 857:1724492019. View Article : Google Scholar : PubMed/NCBI | |
|
Palatinus JA, Valdez S, Taylor L, Whisenant C, Selzman CH, Drakos SG, Ranjan R, Hong T, Saffitz JE and Shaw RM: GJA1-20k rescues Cx43 localization and arrhythmias in arrhythmogenic cardiomyopathy. Circ Res. 132:744–746. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yang HT, Li LL, Li SN, Wu JT, Chen K, Song WF, Zhang GB, Ma JF, Fu HX, Cao S, et al: MicroRNA-155 inhibition attenuates myocardial infarction-induced connexin 43 degradation in cardiomyocytes by reducing pro-inflammatory macrophage activation. Cardiovasc Diagn Ther. 12:325–339. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu W, Du W, Duan R, Liu Y, Zong B, Jin X, Dong Z, Wang H, Shahab S, Wang H, et al: miR-873-5p suppression reinvigorates aging mesenchymal stem cells and improves cardiac repair after myocardial infarction. ACS Pharmacol Transl Sci. 7:743–756. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Hong Y, He H, Jiang G, Zhang H, Tao W, Ding Y, Yuan D, Liu J, Fan H, Lin F, et al: miR-155-5p inhibition rejuvenates aged mesenchymal stem cells and enhances cardioprotection following infarction. Aging Cell. 19:e131282020. View Article : Google Scholar : PubMed/NCBI | |
|
Lu Q, Shen Q, Su J, Li X, Xia B and Tang A: Inhibition of mir-155-5p alleviates cardiomyocyte pyroptosis induced by hypoxia/reoxygenation via targeting SIRT1-mediated activation of the NLRP3 inflammasome. J Cardiothorac Surg. 20:1352025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Ma J, Liu F and Zhang J: Long non-coding RNA XIST promotes the proliferation of cardiac fibroblasts and the accumulation of extracellular matrix by sponging microRNA-155-5p. Exp Ther Med. 21:4772021. View Article : Google Scholar : PubMed/NCBI | |
|
Schumacher D, Curaj A, Simsekyilmaz S, Schober A, Liehn EA and Mause SF: miR155 deficiency reduces myofibroblast density but fails to improve cardiac function after myocardial infarction in dyslipidemic mouse model. Int J Mol Sci. 22:54802021. View Article : Google Scholar : PubMed/NCBI | |
|
Xie W, Li P, Wang Z, Chen J, Lin Z, Liang X and Mo Y: Rosuvastatin may reduce the incidence of cardiovascular events in patients with acute coronary syndromes receiving percutaneous coronary intervention by suppressing miR-155/SHIP-1 signaling pathway. Cardiovasc Ther. 32:276–282. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Liu R, Liu W, Liu X, Fan Z, Cui J, Wu Y, Yin H and Lin Q: Panax quinquefolium L. and Salvia miltiorrhiza Bunge. enhances angiogenesis by regulating the miR-155-5p/HIF-1α/VEGF axis in acute myocardial infarction. Drug Des Devel Ther. 17:3249–3267. 2023. View Article : Google Scholar : | |
|
Zhang M, Liu Q, Meng H, Duan H, Liu X, Wu J, Gao F, Wang S, Tan R and Yuan J: Ischemia-reperfusion injury: Molecular mechanisms and therapeutic targets. Signal Transduct Target Ther. 9:122024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Q, Jia M, Wang Y, Wang Q and Wu J: Cell death mechanisms in cerebral ischemia-reperfusion injury. Neurochem Res. 47:3525–3542. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Y, Li L, Wang Z, Zhang J and Zhou Z: Myocardial ischemia-reperfusion injury; Molecular mechanisms and prevention. Microvasc Res. 149:1045652023. View Article : Google Scholar : PubMed/NCBI | |
|
Eisenhardt SU, Weiss JB, Smolka C, Maxeiner J, Pankratz F, Bemtgen X, Kustermann M, Thiele JR, Schmidt Y, Bjoern Stark G, et al: MicroRNA-155 aggravates ischemia-reperfusion injury by modulation of inflammatory cell recruitment and the respiratory oxidative burst. Basic Res Cardiol. 110:322015. View Article : Google Scholar : PubMed/NCBI | |
|
Xi J, Li QQ, Li BQ and Li N: miR-155 inhibition represents a potential valuable regulator in mitigating myocardial hypoxia/reoxygenation injury through targeting BAG5 and MAPK/JNK signaling. Mol Med Rep. 21:1011–1020. 2020.PubMed/NCBI | |
|
Huang G, Hao F and Hu X: Downregulation of microRNA-155 stimulates sevoflurane-mediated cardioprotection against myocardial ischemia/reperfusion injury by binding to SIRT1 in mice. J Cell Biochem. 120:15494–15505. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Greco R, Demartini C, Zanaboni AM, Blandini F, Amantea D and Tassorelli C: Endothelial nitric oxide synthase inhibition triggers inflammatory responses in the brain of male rats exposed to ischemia-reperfusion injury. J Neurosci Res. 96:151–159. 2018. View Article : Google Scholar | |
|
Liu M, Fu D, Gao T, Jiang H, Yang P and Li X: The low expression of miR-155 promotes the expression of SHP2 by inhibiting the activation of the ERK1/2 pathway and improves cell pyroptosis induced by I/R in mice. Aging (Albany NY). 16:4778–4788. 2024.PubMed/NCBI | |
|
Jiang T, Zhou S, Li X, Song J, An T, Huang X, Ping X and Wang L: MicroRNA-155 induces protection against cerebral ischemia/reperfusion injury through regulation of the Notch pathway in vivo. Exp Ther Med. 18:605–613. 2019.PubMed/NCBI | |
|
Zhang L, Liu C, Huang C, Xu X and Teng J: miR-155 knockdown protects against cerebral ischemia and reperfusion injury by targeting MafB. Biomed Res Int. 2020:64582042020. View Article : Google Scholar : PubMed/NCBI | |
|
Shi Y, Li K, Xu K and Liu QH: MiR-155-5p accelerates cerebral ischemia-reperfusion injury via targeting DUSP14 by regulating NF-κB and MAPKs signaling pathways. Eur Rev Med Pharmacol Sci. 24:1408–1419. 2020.PubMed/NCBI | |
|
Shi Y, Li Z, Li K and Xu K: miR-155-5p accelerates cerebral ischemia-reperfusion inflammation injury and cell pyroptosis via DUSP14/TXNIP/NLRP3 pathway. Acta Biochim Pol. 69:787–793. 2022.PubMed/NCBI | |
|
Xue Y, Wang Y, Chen T, Peng L, Wang C, Xue G and Yu S: DJ-1 regulates astrocyte activation through miR-155/SHP-1 signaling in cerebral ischemia/reperfusion injury. J Neurochem. 169:e162302025. View Article : Google Scholar | |
|
Tanai E and Frantz S: Pathophysiology of heart failure. Compr Physiol. 6:187–214. 2015. View Article : Google Scholar | |
|
McMurray JJ and Pfeffer MA: Heart failure. Lancet. 365:1877–1889. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Mosterd A and Hoes AW: Clinical epidemiology of heart failure. Heart. 93:1137–1146. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Li J, Su H, Zhu Y, Cao Y and Ma X: ETS2 and microRNA-155 regulate the pathogenesis of heart failure through targeting and regulating GPR18 expression. Exp Ther Med. 19:3469–3478. 2020.PubMed/NCBI | |
|
Lin B, Zhao H, Li L, Zhang Z, Jiang N, Yang X, Zhang T, Lian B, Liu Y, Zhang C, et al: Sirt1 improves heart failure through modulating the NF-κB p65/microRNA-155/BNDF signaling cascade. Aging (Albany NY). 13:14482–14498. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gao L, Li T, Li S, Song Z, Chang Y and Yuan L: Schisandrin A protects against isoproterenol-induced chronic heart failure via miR-155. Mol Med Rep. 25:242022. View Article : Google Scholar | |
|
Luo Y, Deng X, Chen Q, Cai Y, Bie M, Zhang Y, Peng L, Yao K, Chen X and Cai H: Up-regulation of miR-155 protects against chronic heart failure by inhibiting HIF-1α. Am J Transl Res. 15:6425–6436. 2023. | |
|
Heymans S, Corsten MF, Verhesen W, Carai P, van Leeuwen RE, Custers K, Peters T, Hazebroek M, Stöger L, Wijnands E, et al: Macrophage microRNA-155 promotes cardiac hypertrophy and failure. Circulation. 128:1420–1432. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng L, Xu CC, Chen WD, Shen WL, Ruan CC, Zhu LM, Zhu DL and Gao PJ: MicroRNA-155 regulates angiotensin II type 1 receptor expression and phenotypic differentiation in vascular adventitial fibroblasts. Biochem Biophys Res Commun. 400:483–488. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Shi L and Fleming I: One miR level of control: microRNA-155 directly regulates endothelial nitric oxide synthase mRNA and protein levels. Hypertension. 60:1381–1382. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Touyz RM, Alves-Lopes R, Rios FJ, Camargo LL, Anagnostopoulou A, Arner A and Montezano AC: Vascular smooth muscle contraction in hypertension. Cardiovasc Res. 114:529–539. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Xu D, Liao R, Wang XX and Cheng Z: Effects of miR-155 on hypertensive rats via regulating vascular mesangial hyperplasia. Eur Rev Med Pharmacol Sci. 22:7431–7438. 2018.PubMed/NCBI | |
|
Lai KN, Leung JCK and Tang SCW: The renin-angiotensin system. Contrib Nephrol. 170:135–144. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R and Eguchi S: Angiotensin II signal transduction: An update on mechanisms of physiology and pathophysiology. Physiol Rev. 98:1627–1738. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
DuPont JJ, McCurley A, Davel AP, McCarthy J, Bender SB, Hong K, Yang Y, Yoo JK, Aronovitz M, Baur WE, et al: Vascular mineralocorticoid receptor regulates microRNA-155 to promote vasoconstriction and rising blood pressure with aging. JCI Insight. 1:e889422016. View Article : Google Scholar : PubMed/NCBI | |
|
Theilig F and Wu Q: ANP-induced signaling cascade and its implications in renal pathophysiology. Am J Physiol Renal Physiol. 308:F1047–1055. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Vandenwijngaert S, Ledsky CD, Agha O, Wu C, Hu D, Bagchi A, Domian IJ, Buys ES, Newton-Cheh C and Bloch DB: MicroRNA-425 and microRNA-155 cooperatively regulate atrial natriuretic peptide expression and cGMP production. PLoS One. 13:e01966972018. View Article : Google Scholar : PubMed/NCBI | |
|
Stonesifer C, Corey S, Ghanekar S, Diamandis Z, Acosta SA and Borlongan CV: Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Prog Neurobiol. 158:94–131. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Putaala J: Ischemic stroke in young adults. Continuum (Minneap Minn). 26:386–414. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Choi GH, Ko KH, Kim JO, Kim J, Oh SH, Han IB, Cho KG, Kim OJ, Bae J and Kim NK: Association of miR-34a, miR-130a, miR-150 and miR-155 polymorphisms with the risk of ischemic stroke. Int J Mol Med. 38:345–356. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Xing G, Luo Z, Zhong C, Pan X and Xu X: Influence of miR-155 on cell apoptosis in rats with ischemic stroke: Role of the ras homolog enriched in brain (Rheb)/mTOR pathway. Med Sci Monit. 22:5141–5153. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Zhang N, Wang S and Wen Y: MicroRNA-155 regulates inflammatory response in ischemic cerebral tissues through autophagy. Curr Neurovasc Res. 15:103–110. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Adly Sadik N, Ahmed Rashed L and Ahmed Abd-El Mawla M: Circulating miR-155 and JAK2/STAT3 axis in acute ischemic stroke patients and its relation to Post-ischemic inflammation and associated ischemic stroke risk factors. Int J Gen Med. 14:1469–1484. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang W, Hong Q, Wang H, Zhu Z and Gong S: MicroRNA-155 inhibition activates Wnt/β-Catenin signaling to restore Th17/Treg cell balance and protect against acute ischemic stroke. eNeuro. 12:ENEURO.0347-24.2024. 2025. View Article : Google Scholar | |
|
Wang J, Li D, Hou J and Lei H: Protective effects of geniposide and ginsenoside Rg1 combination treatment on rats following cerebral ischemia are mediated via microglial microRNA-155-5p inhibition. Mol Med Rep. 17:3186–3193. 2018. | |
|
Zhang JK, Li Y, Yu ZT, Jiang JW, Tang H, Tu GL and Xia Y: OIP5-AS1 inhibits oxidative stress and inflammation in ischemic stroke through miR-155-5p/IRF2BP2 axis. Neurochem Res. 48:1382–1394. 2023. | |
|
Bossone E and Eagle KA: Epidemiology and management of aortic disease: Aortic aneurysms and acute aortic syndromes. Nat Rev Cardiol. 18:331–348. 2021. View Article : Google Scholar | |
|
Spin JM, Li DY, Maegdefessel L and Tsao PS: Non-coding RNAs in aneurysmal aortopathy. Vascul Pharmacol. 114:110–121. 2019. View Article : Google Scholar | |
|
Hu J, Huang S, Liu X, Zhang Y, Wei S and Hu X: miR-155: An important role in inflammation response. J Immunol Res. 2022:74372812022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang X, Peng J, Pang J, Wan W and Chen L: A functional polymorphism in the promoter region of miR-155 predicts the risk of intracranial hemorrhage caused by rupture intracranial aneurysm. J Cell Biochem. 120:18618–18628. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Z, Zhang X, Li L, Wang C, Feng M, Zhao K, Zhao R, Liu J and Fang Y: Tumor-associated macrophage-derived exosomal microRNA-155-5p stimulates intracranial aneurysm formation and macrophage infiltration. Clin Sci (Lond). 133:2265–2282. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang RL, Wang WM, Li JQ, Li RW, Zhang J, Wu Y and Liu Y: The role of miR-155 in cardiovascular diseases: Potential diagnostic and therapeutic targets. Int J Cardiol Cardiovasc Risk Prev. 24:2003552025.PubMed/NCBI | |
|
Yang WW, Li QX, Wang F, Zhang XR, Zhang XL, Wang M, Xue D, Zhao Y and Tang L: Exosomal miR-155-5p promote the occurrence of carotid atherosclerosis. J Cell Mol Med. 28:e701872024. View Article : Google Scholar : PubMed/NCBI | |
|
Eshraghi R, Rafiei M, Hadian Jazi Z, Shafie D, Raisi A and Mirzaei H: MicroRNA-155 and exosomal microRNA-155: Small pieces in the cardiovascular diseases puzzle. Pathol Res Pract. 257:1552742024. View Article : Google Scholar | |
|
Kazimierczyk E, Eljaszewicz A, Zembko P, Tarasiuk E, Rusak M, Kulczynska-Przybik A, Lukaszewicz-Zajac M, Kaminski K, Mroczko B, Szmitkowski M, et al: The relationships among monocyte subsets, miRNAs and inflammatory cytokines in patients with acute myocardial infarction. Pharmacol Rep. 71:73–81. 2019. View Article : Google Scholar | |
|
Fitzsimons S, Oggero S, Bruen R, McCarthy C, Strowitzki MJ, Mahon NG, Ryan N, Brennan EP, Barry M, Perretti M, et al: microRNA-155 is decreased during atherosclerosis regression and is increased in urinary extracellular vesicles during atherosclerosis progression. Front Immunol. 11:5765162020. View Article : Google Scholar | |
|
Obradovic D, Rommel KP, Blazek S, Klingel K, Gutberlet M, Lücke C, Büttner P, Thiele H, Adams V, Lurz P, et al: The potential role of plasma miR-155 and miR-206 as circulatory biomarkers in inflammatory cardiomyopathy. ESC Heart Fail. 8:1850–1860. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yao R, Ma Y, Du Y, Liao M, Li H, Liang W, Yuan J, Ma Z, Yu X, Xiao H and Liao Y: The altered expression of inflammation-related microRNAs with microRNA-155 expression correlates with Th17 differentiation in patients with acute coronary syndrome. Cell Mol Immunol. 8:486–495. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Liu L, Chen X, Wang T, Zhou H, Huang H, Qing L and Luo P: Noninvasive prognosis of postmyocardial infarction using urinary miRNA ultratrace detection based on Single-Target DNA-functionalized AuNPs. ACS Appl Mater Interfaces. 14:3633–3642. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Tijsen AJ, Pinto YM and Creemers EE: Non-cardiomyocyte microRNAs in heart failure. Cardiovasc Res. 93:573–582. 2012. View Article : Google Scholar | |
|
Ikitimur B, Cakmak HA, Coskunpinar E, Barman HA and Vural VA: The relationship between circulating microRNAs and left ventricular mass in symptomatic heart failure patients with systolic dysfunction. Kardiol Pol. 73:740–746. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Ding H, Wang Y, Hu L, Xue S, Wang Y, Zhang L, Zhang Y, Qi H, Yu H, Aung LHH, et al: Combined detection of miR-21-5p, miR-30a-3p, miR-30a-5p, miR-155-5p, miR-216a and miR-217 for screening of early heart failure diseases. Biosci Rep. 40:BSR201916532020. View Article : Google Scholar : PubMed/NCBI | |
|
Kin K, Miyagawa S, Fukushima S, Shirakawa Y, Torikai K, Shimamura K, Daimon T, Kawahara Y, Kuratani T and Sawa Y: Tissue- and plasma-specific MicroRNA signatures for atherosclerotic abdominal aortic aneurysm. J Am Heart Assoc. 1:e0007452012. View Article : Google Scholar | |
|
Biros E, Moran CS, Wang Y, Walker PJ, Cardinal J and Golledge J: microRNA profiling in patients with abdominal aortic aneurysms: The significance of miR-155. Clin Sci (Lond). 126:795–803. 2014. View Article : Google Scholar | |
|
Mashima R: Physiological roles of miR-155. Immunology. 145:323–333. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Seto AG, Beatty X, Lynch JM, Hermreck M, Tetzlaff M, Duvic M and Jackson AL: Cobomarsen, an oligonucleotide inhibitor of miR-155, co-ordinately regulates multiple survival pathways to reduce cellular proliferation and survival in cutaneous T-cell lymphoma. Br J Haematol. 183:428–444. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Anastasiadou E, Seto AG, Beatty X, Hermreck M, Gilles ME, Stroopinsky D, Pinter-Brown LC, Pestano L, Marchese C, Avigan D, et al: Cobomarsen, an oligonucleotide inhibitor of miR-155, slows DLBCL tumor cell growth in vitro and in vivo. Clin Cancer Res. 27:1139–1149. 2021. View Article : Google Scholar : | |
|
Yan Q, Chen J, Li W, Bao C and Fu Q: Targeting miR-155 to treat experimental scleroderma. Sci Rep. 6:203142016. View Article : Google Scholar : PubMed/NCBI | |
|
Tian FJ, An LN, Wang GK, Zhu JQ, Li Q, Zhang YY, Zeng A, Zou J, Zhu RF, Han XS, et al: Elevated microRNA-155 promotes foam cell formation by targeting HBP1 in atherogenesis. Cardiovasc Res. 103:100–110. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Tokgozoglu L and Kayikcioglu M: Familial hypercholesterolemia: Global burden and approaches. Curr Cardiol Rep. 23:1512021. View Article : Google Scholar : PubMed/NCBI | |
|
Yao R, Ma YL, Liang W, Li HH, Ma ZJ, Yu X and Liao YH: MicroRNA-155 modulates Treg and Th17 cells differentiation and Th17 cell function by targeting SOCS1. PLoS One. 7:e460822012. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Ouyang Y, Bai Y, Gong J and Liao H: miR-155-5p inhibits the viability of vascular smooth muscle cell via targeting FOS and ZIC3 to promote aneurysm formation. Eur J Pharmacol. 853:145–152. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Ertl HCJ: Immunogenicity and toxicity of AAV gene therapy. Front Immunol. 13:9758032022. View Article : Google Scholar : PubMed/NCBI | |
|
Kahil N, Abouzeinab NS, Hussein MAA and Khalil MI: Intraperitoneal hepatorenal toxicity of zinc oxide and nickel oxide nanoparticles in rats: A systematic review. Nanotoxicology. 18:583–598. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wei Y, Yan X, Yan L, Hu F, Ma W, Wang Y, Lu S, Zeng Q and Wang Z: Inhibition of microRNA-155 ameliorates cardiac fibrosis in the process of angiotensin II-induced cardiac remodeling. Mol Med Rep. 16:7287–7296. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Tang H, Mao J, Ye X, Zhang F, Kerr WG, Zheng T and Zhu Z: SHIP-1, a target of miR-155, regulates endothelial cell responses in lung fibrosis. FASEB J. 34:2011–2023. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kalkusova K, Taborska P, Stakheev D and Smrz D: The role of miR-155 in antitumor immunity. Cancers (Basel). 14:54142022. View Article : Google Scholar : PubMed/NCBI | |
|
Ghoumari AM, Rixe O, Yarovoi SV, Zerrouqi A, Mouawad R, Poynard T, Opolon P, Khayat D and Soubrane C: Gene transfer in hepatocarcinoma cell lines: In vitro optimization of a virus-free system. Gene Ther. 3:483–490. 1996.PubMed/NCBI | |
|
Wu J, Wu GY and Zern MA: The prospects of hepatic drug delivery and gene therapy. Expert Opin Investig Drugs. 7:1795–1817. 1998. View Article : Google Scholar | |
|
Golubovic A, Tsai S and Li B: Bioinspired lipid nanocarriers for RNA delivery. ACS Bio Med Chem Au. 3:114–136. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Xu F, Reiser M, Yu X, Gummuluru S, Wetzler L and Reinhard BM: Lipid-mediated targeting with membrane-wrapped nanoparticles in the presence of corona formation. ACS Nano. 10:1189–1200. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Streeter SS, Hebert KA, Bateman LM, Ray GS, Dean RE, Geffken KT, Resnick CT, Austin DC, Bell JE, Sparks MB, et al: Current and future applications of fluorescence guidance in orthopaedic surgery. Mol Imaging Biol. 25:46–57. 2023. View Article : Google Scholar : | |
|
Brillante S, Volpe M and Indrieri A: Advances in MicroRNA therapeutics: From preclinical to clinical studies. Hum Gene Ther. 35:628–648. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Miao Y, Fu C, Yu Z, Yu L, Tang Y and Wei M: Current status and trends in small nucleic acid drug development: Leading the future. Acta Pharm Sin B. 14:3802–3817. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Nappi F: Non-coding RNA-targeted therapy: A state-of-the-Art review. Int J Mol Sci. 25:36302024. View Article : Google Scholar : PubMed/NCBI | |
|
Vermeire S, Nitcheu J, Gineste P, Flatres A, Santo J, Scherrer D, Peyrin-Biroulet L, Dulai PS, Danese S, Dubinsky M, et al: Obefazimod in patients with moderate-to-severely active ulcerative colitis: Efficacy and safety analysis from the 96-week open-label maintenance phase 2b study. J Crohns Colitis. 19:jjaf0742025. View Article : Google Scholar : PubMed/NCBI | |
|
Bauersachs J, Solomon SD, Anker SD, Antorrena-Miranda I, Batkai S, Viereck J, Rump S, Filippatos G, Granzer U, Ponikowski P, et al: Efficacy and safety of CDR132L in patients with reduced left ventricular ejection fraction after myocardial infarction: Rationale and design of the HF-REVERT trial. Eur J Heart Fail. 26:674–682. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y, et al: Treatment of HCV infection by targeting microRNA. N Engl J Med. 368:1685–1694. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Hong DS, Kang YK, Borad M, Sachdev J, Ejadi S, Lim HY, Brenner AJ, Park K, Lee JL, Kim TY, et al: Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br J Cancer. 122:1630–1637. 2020. View Article : Google Scholar : PubMed/NCBI |