v
Search
Advanced

Publications > Journals > Gene Expression> Article Full Text

  • OPEN ACCESS

MicroRNAs in the Regulation of Immune Response in Cardiovascular Diseases: New Diagnostic and Therapeutic Tools

  • Ilgiz Gareev1,* ,
  • Ozal Beylerli2,
  • Albert Sufianov2,
  • Leili Gulieva1,
  • Valentin Pavlov1 and
  • Huaizhang Shi3
 Author information 

Abstract

Cardiovascular diseases (CVDs) remain the leading cause of global morbidity and mortality, highlighting the urgent need for innovative diagnostic and prognostic approaches to address their complex pathophysiology. Recent advances in molecular cardiology have unveiled immune-derived microRNAs (miRNAs), or immuno-miRs, as pivotal regulators in the interplay between immune responses and cardiovascular pathology. Secreted by immune cells such as T lymphocytes, macrophages, and neutrophils, these small non-coding RNAs modulate critical signaling pathways by regulating gene expression. Immuno-miRs influence essential processes, including inflammation, endothelial dysfunction, and fibrotic remodeling—core mechanisms underlying conditions such as atherosclerosis, myocardial infarction, and heart failure. Moreover, their presence in systemic circulation within extracellular vesicles underscores their role in intercellular communication, impacting both immune and non-immune cardiovascular cells, such as cardiomyocytes and endothelial cells. This dual functionality renders immuno-miRs promising candidates as diagnostic biomarkers for early disease detection and as prognostic tools for assessing disease progression and therapeutic efficacy. Furthermore, emerging miRNA-based interventions—such as miRNA mimics and inhibitors—show considerable promise in modulating immune dysregulation in CVDs, although clinical translation remains a significant challenge. In this review, we comprehensively examine the regulatory roles of immuno-miRs in both innate and adaptive immune responses and explore recent advancements in miRNA-based therapies. By consolidating current knowledge and identifying existing gaps, we provide a comprehensive overview of the transformative potential of immuno-miRs in CVD management. Integrating these molecules into personalized medicine may pave the way for more effective, targeted, and minimally invasive strategies to combat one of the world’s most pressing health challenges.

Keywords

Cardiovascular diseases, miRNAs, miR-155, Immuno-miRs, Immune system, Therapy, Diagnosis, Inflammation

Introduction

Cardiovascular diseases (CVDs) are a leading cause of global mortality, accounting for approximately 18.6 million deaths annually—a number projected to rise due to increasing life expectancy, urbanization, and the growing prevalence of risk factors such as obesity, diabetes, and hypertension.1 Despite advancements in pharmacological and interventional treatments, the complex pathophysiology of CVD remains a significant challenge. Central to this complexity is the interplay between the immune system and cardiovascular tissues, which plays a pivotal role in the initiation, progression, and resolution of these diseases.2 The immune system, comprising innate and adaptive components, acts as a double-edged sword in cardiovascular health. While immune responses are essential for maintaining homeostasis and initiating tissue repair, their dysregulation can lead to chronic inflammation, fibrosis, and adverse remodeling—hallmark features of CVDs.3 For instance, macrophages contribute both to the clearance of necrotic debris and the exacerbation of inflammation in myocardial infarction, whereas T cells are implicated in the destabilization of atherosclerotic plaques.4 Understanding the mechanisms that govern these immune responses is critical for developing novel diagnostic and therapeutic strategies. In recent years, microRNAs (miRNAs) have emerged as key regulators of the crosstalk between the immune and cardiovascular systems. miRNAs are small, non-coding RNA molecules, typically 18–22 nucleotides in length, that regulate gene expression by binding to complementary sequences in the 3′-untranslated regions of target messenger RNAs (mRNAs) (Fig. 1).5

Biogenesis of microRNA (miRNA).
Fig. 1  Biogenesis of microRNA (miRNA).

DGCR8, DiGeorge syndrome critical region 8; mRNA, messenger RNA; RISC, RNA-induced silencing complex.

These molecules orchestrate a wide range of cellular processes, including apoptosis, differentiation, and immune modulation.6 Immune-derived miRNAs, often referred to as “immuno-miRs”, are a subset predominantly expressed in immune cells such as macrophages, T lymphocytes, B cells, and natural killer (NK) cells.7 Their roles in CVD have garnered increasing attention due to their capacity to modulate inflammatory pathways and immune cell phenotypes. For example, miR-223—highly expressed in neutrophils and macrophages—acts as a critical regulator of myeloid cell differentiation and inflammatory responses, influencing atherosclerotic plaque formation and stability.8 Similarly, miR-181a, a key player in T cell development and activation, has been shown to modulate immune aging, a factor contributing to the heightened risk of CVD in older adults.9 One of the most promising aspects of immune-derived miRNAs lies in their potential as biomarkers. Circulating miRNAs, found either freely or encapsulated within extracellular vesicles such as exosomes, offer a stable and non-invasive means of assessing disease states. Their expression profiles can reflect underlying immune activity, providing insights into disease progression, severity, and treatment response.10 For example, elevated levels of miR-21 and miR-155 in plasma have been correlated with adverse outcomes in heart failure and myocardial infarction, respectively, highlighting their diagnostic and prognostic value.11 Moreover, immune-derived miRNAs represent a novel therapeutic frontier in CVD. Advances in RNA-based therapeutics have facilitated the development of miRNA mimics and inhibitors (antagomiRs) designed to restore normal gene expression patterns. Preclinical studies have demonstrated the efficacy of these approaches in reducing inflammation, promoting tissue repair, and improving cardiac function following myocardial infarction.12 For instance, nanoparticle-based delivery systems targeting miR-21 in cardiac macrophages have shown promise in mitigating pathological remodeling and fibrosis.13 Despite these advances, challenges such as off-target effects, delivery efficiency, and long-term safety remain significant barriers to clinical translation.14 This review aimed to provide a comprehensive overview of the current understanding of immune-derived miRNAs in CVD. We will explore their roles in immune modulation, their potential as biomarkers for early diagnosis and risk stratification, and their therapeutic applications. By consolidating existing evidence and identifying gaps in knowledge, this work sought to support the integration of miRNAs into the field of precision cardiovascular medicine. Harnessing the potential of these molecules could revolutionize CVD management by offering more targeted, effective, and minimally invasive therapeutic strategies to address this global health burden.

miRNAs in immune cell development and function

miRNAs are essential non-coding RNAs that play pivotal roles in regulating the development, differentiation, activation, and function of immune cells. By fine-tuning gene expression through mRNA degradation or translational inhibition, miRNAs are critical for maintaining immune homeostasis and mounting appropriate responses to pathological stimuli.15 Immune-derived miRNAs, often termed “immuno-miRs”, regulate both innate and adaptive immune responses, influencing processes such as inflammation, tissue repair, and immune tolerance. Dysregulation of these miRNAs has been increasingly linked to the pathogenesis of CVDs, emphasizing their dual role as both contributors to disease progression and potential therapeutic targets (Fig. 2).16

Overview of immune cells and their regulatory microRNAs (miRNAs).
Fig. 2  Overview of immune cells and their regulatory microRNAs (miRNAs).

Th1, type 1 T helper.

miRNAs in innate immune cells

Innate immune cells—including macrophages, neutrophils, and NK cells—are the body’s first responders to tissue damage or infection. miRNAs regulate their development, polarization, and activation, shaping the balance between pro-inflammatory and anti-inflammatory responses. Macrophages, known for their plasticity, can polarize into pro-inflammatory M1 or anti-inflammatory M2 states depending on environmental cues. miR-223 is a key regulator of macrophage differentiation, preventing excessive inflammatory responses by targeting transcription factors such as nuclear factor I-A and other signal transducers.17 Studies have demonstrated that miR-223 deficiency leads to unrestrained M1 macrophage activation, exacerbating inflammation in conditions such as atherosclerosis and myocardial infarction.18 Another important miRNA, miR-155, is a well-characterized pro-inflammatory molecule that promotes M1 polarization by enhancing cytokine production—particularly tumor necrosis factor-alpha and interleukin-6—via activation of the nuclear factor kappa B (NF-κB) pathway.19 Overexpression of miR-155 has been associated with chronic inflammatory diseases including heart failure and hypertension, where persistent macrophage activation contributes to tissue damage.20 Neutrophils, another vital component of the innate immune response, also rely on miRNAs for proper function. miR-146a acts as a negative regulator of neutrophil-mediated inflammation by targeting key elements of Toll-like receptor (TLRs) and NF-κB signaling pathways, such as interleukin-1 receptor-associated kinase 1 (IRAK1) and tumor necrosis factor receptor-associated factor 6 (TRAF6).21 Dysregulation of miR-146a can lead to exaggerated neutrophilic responses, frequently observed in acute myocardial infarction and other inflammatory conditions.22 NK cells, which are essential for antiviral defense and tumor surveillance, are also regulated by miRNAs. miR-27a has been shown to modulate NK cell cytotoxicity by targeting genes involved in degranulation and cytokine release. Impaired miRNA expression in NK cells can compromise their function, contributing to the chronic inflammation seen in CVD.23

miRNAs in adaptive immune cells

In the adaptive immune system, miRNAs are indispensable for the regulation of T and B lymphocytes, ensuring proper immune activation and tolerance. T cells depend on miRNAs for both thymic development and peripheral activation. miR-181a is a master regulator of T cell receptor (TCR) sensitivity, fine-tuning the signaling threshold during positive and negative selection in the thymus. Reduced miR-181a expression in aged individuals has been associated with diminished T cell responsiveness, contributing to immune senescence and the increased risk of CVD in elderly populations.24 The miR-17-92 cluster, a polycistronic group of miRNAs, is crucial for T cell survival and proliferation. Dysregulation of this cluster impairs effector and memory T cell responses, weakening immunity during chronic inflammation, as seen in atherosclerosis and myocardial infarction.25 Regulatory T cells (Tregs) are critical for maintaining immune tolerance and preventing excessive inflammation. miR-142-3p and miR-142-5p regulate Treg function by modulating cyclic adenosine monophosphate signaling and transforming growth factor-beta (TGF-β) receptor expression, both of which are essential for Treg-mediated suppression of pro-inflammatory effector T cells.26 Deletion of miR-142 disrupts Treg homeostasis, leading to widespread immune activation and tissue damage in models of hypertension and atherosclerosis.27 B cells also rely on miRNAs for proper differentiation and antibody production. miR-150 is critical for the transition from precursor to mature B cells, while miR-34a regulates class-switch recombination and somatic hypermutation. Dysregulation of these miRNAs can impair humoral immunity, contributing to chronic inflammation and the production of autoantibodies often observed in autoimmune-associated CVD.28

miRNAs in immune cell crosstalk and extracellular vesicle communication

Beyond their intracellular functions, miRNAs are instrumental in intercellular communication via extracellular vesicles (EVs), such as exosomes. Immune cells, including macrophages and T cells, release miRNA-enriched EVs into circulation, influencing the activity of distant target cells (Fig. 3). For instance, EVs enriched with miR-223, secreted by macrophages, can downregulate inflammatory signaling in endothelial cells, offering protection against vascular damage.29 Conversely, EVs containing miR-155 released during chronic inflammation can amplify pro-inflammatory responses in recipient cells, thereby accelerating atherosclerosis progression.30 These findings highlight the dual role of miRNAs as both mediators and messengers of immune responses, making them attractive therapeutic targets for modulating immune cell communication in CVD.31

miRNA-loaded extracellular vesicles (EVs) can be produced directly by T cells upon engagement with vascular cells.
Fig. 3  miRNA-loaded extracellular vesicles (EVs) can be produced directly by T cells upon engagement with vascular cells.

CARs, chimeric antigen receptors; CD, cluster of differentiation; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; IL, interleukin; NK, natural killer; PD1, programmed cell death 1; PDL1, programmed death-ligand 1.

The regulatory functions of miRNAs in immune cells have profound implications for understanding and treating CVD. Their dysregulation contributes to disease pathophysiology while offering novel opportunities for therapeutic intervention. miRNA-based therapies—such as antagomiRs and miRNA mimics—have shown promise in restoring normal immune cell function in preclinical studies. For example, nanoparticle-mediated delivery of miR-21 to cardiac macrophages post-myocardial infarction has been shown to reduce fibrosis and improve cardiac function.32 Furthermore, the stability of circulating miRNAs makes them valuable diagnostic and prognostic biomarkers, enabling minimally invasive monitoring of immune activity and disease progression.33

Immunity and CVD risk factors

CVDs are influenced by genetic and environmental factors, but also by complex interactions within the immune system. Chronic low-grade inflammation, driven by dysregulated immune responses, has emerged as a hallmark of major CVD risk factors, including obesity, hypertension, diabetes, and dyslipidemia.34 The immune system modulates these risk factors through a network of innate and adaptive immune cells, cytokines, and miRNAs, which regulate inflammatory pathways and cellular communication. Understanding these interactions is crucial for identifying novel biomarkers and therapeutic targets for CVD prevention and management.

Obesity and adipose tissue inflammation

Obesity is a major risk factor for CVD and is often accompanied by chronic inflammation in adipose tissue. Once considered a passive fat reservoir, adipose tissue is now recognized as an active endocrine organ that produces pro-inflammatory cytokines and adipokines. In obesity, immune cells infiltrate adipose tissue, contributing to systemic inflammation and metabolic dysregulation.35 Epicardial adipose tissue (EAT), a visceral fat depot surrounding the myocardium, has been implicated in the development of coronary artery disease and atrial fibrillation. Increased EAT volume correlates with heightened expression of inflammatory miRNAs, such as miR-34a and miR-103, which modulate macrophage polarization and Th2 chemokine signaling, respectively.36 miR-34a promotes pro-inflammatory M1 macrophage activity, exacerbating local inflammation, while reduced miR-103 levels in EAT have been linked to impaired regulation of anti-inflammatory pathways.37 Furthermore, miRNAs in exosomes secreted by adipocytes, such as miR-223 and miR-155, influence immune cell behavior in distant tissues. These miRNAs enhance inflammatory responses in macrophages and endothelial cells, contributing to vascular dysfunction and insulin resistance.38

Hypertension and immune dysregulation

Hypertension, a major CVD risk factor, is increasingly recognized as an immune-mediated disorder. Early research demonstrated that Rag1 knockout mice, which are deficient in T and B lymphocytes, are protected against angiotensin II-induced hypertension, implicating adaptive immune responses in the disease pathogenesis.39 Subsequent studies have revealed specific miRNAs that regulate immune cells in hypertension. For instance, miR-214, which is upregulated in perivascular adipose tissue during angiotensin II-induced hypertension, amplifies vascular stiffening and fibrosis by modulating T cell activation and cytokine production.40 Deletion of miR-214 reduces inflammation and vascular damage, highlighting its therapeutic potential. Similarly, miR-31 influences the balance between Treg and T helper 17 cells, with its deletion reducing hypertension, cardiac hypertrophy, and renal fibrosis in experimental models.41 In addition to adaptive immune responses, innate immune cells such as monocytes and macrophages play critical roles in hypertension. miR-155, widely expressed in these cells, exacerbates vascular inflammation and remodeling by targeting transcriptional repressors of pro-inflammatory pathways.42

Diabetes and immune-driven inflammation

Diabetes is a significant CVD risk factor characterized by chronic systemic inflammation. Hyperglycemia and insulin resistance activate the immune system, leading to endothelial dysfunction and accelerated atherosclerosis. Immune cells, particularly macrophages, infiltrate pancreatic islets and vascular tissues, contributing to inflammation and tissue damage.43 miRNAs regulate immune responses in diabetes-associated CVD. For instance, miR-146a mitigates inflammation by targeting IRAK1 and TRAF6 in macrophages, reducing cytokine production.44 However, its downregulation in diabetic patients leads to unrestrained inflammation and vascular complications. Conversely, miR-21 is upregulated in diabetes, promoting fibrosis and endothelial dysfunction by enhancing TGF-β signaling pathways.45

Dyslipidemia and atherosclerosis

Dyslipidemia, characterized by elevated low-density lipoprotein levels and reduced high-density lipoprotein, is a major driver of atherosclerosis. Immune cells, particularly macrophages, play a central role in plaque formation and progression by engulfing oxidized low-density lipoprotein and forming foam cells. Dysregulated miRNAs contribute to this process by modulating lipid metabolism and inflammatory signaling. miR-33, a well-studied miRNA in dyslipidemia, suppresses cholesterol efflux from macrophages by targeting ATP-binding cassette transporter A1, leading to lipid accumulation and foam cell formation.46 Inhibition of miR-33 enhances cholesterol efflux and reduces plaque burden in experimental models of atherosclerosis. Similarly, miR-155 promotes foam cell formation by enhancing macrophage inflammatory responses and reducing cholesterol efflux.47 miRNAs also influence endothelial dysfunction, a key early event in atherosclerosis. miR-92a, enriched in endothelial cells exposed to disturbed flow, promotes endothelial activation and monocyte adhesion by targeting Krüppel-like Factor 2 (KLF2) and other atheroprotective genes. However, focusing solely on miR-92a may provide an incomplete understanding.48 Other endothelial miRNAs play critical roles in early atherogenesis. For instance, miR-126, an endothelial-specific miRNA, is essential for maintaining vascular integrity and promoting reparative angiogenesis; its dysregulation is closely associated with endothelial dysfunction and atherosclerotic development.49–51 Similarly, miR-103 has been implicated in modulating endothelial inflammation and permeability, further contributing to atherosclerosis.52 Emerging evidence also points to miR-10a, which can suppress endothelial activation by inhibiting NF-κB signaling pathways, thereby reducing inflammatory responses. Moreover, the miR-221/222 cluster regulates endothelial nitric oxide synthase expression and vascular tone, underscoring its role in modulating endothelial function.43 Collectively, these findings illustrate a multifaceted network of endothelial miRNAs that orchestrate vascular homeostasis and early atherogenic processes. This broader view emphasizes that a combination of miRNAs, rather than a single candidate, may better capture the complexity of endothelial regulation in atherosclerosis.

Immune crosstalk and risk factor amplification

CVD risk factors rarely act in isolation. The immune system plays a critical role in amplifying the effects of these risk factors through interconnected pathways. For example, obesity-induced inflammation exacerbates hypertension by promoting renal infiltration of immune cells, while diabetes-induced hyperglycemia accelerates atherosclerosis by enhancing monocyte activation and foam cell formation.53 miRNAs serve as molecular mediators in these interactions, with dysregulated miRNA profiles observed across multiple risk factors. Circulating miRNA signatures have emerged as potential biomarkers for assessing cumulative CVD risk. For instance, elevated plasma levels of miR-122 and miR-126 have been linked to obesity-related dyslipidemia, while miR-21 and miR-155 correlate with hypertension and diabetes-induced vascular damage.54 Targeting immune-mediated pathways using miRNA-based therapies offers promising strategies for mitigating CVD risk factors. For example, miRNA mimics and inhibitors, delivered via nanoparticles, are being explored to modulate inflammation, improve lipid metabolism, and restore endothelial function.55 The development of miRNA-based biomarkers also holds potential for early detection and personalized management of individuals at high risk for CVD.55

Regulation of immune cells by miRNAs in the context of CVDs and related complications

The progression of CVDs is intrinsically linked to the dysregulation of immune cell function. Immune cells, including macrophages, neutrophils, T lymphocytes, and regulatory Tregs, play critical roles in modulating inflammation, tissue remodeling, and repair. miRNAs have emerged as key regulators of these processes, influencing immune cell activation, polarization, and crosstalk. The intricate interplay between miRNAs and immune cells determines the balance between protective and pathological responses in CVDs such as myocardial infarction, atherosclerosis, and heart failure.56

Macrophage regulation by miRNAs in CVDs

Macrophages are central to the inflammatory response in CVDs, contributing to both tissue damage and repair. miRNAs regulate macrophage polarization into pro-inflammatory M1 or anti-inflammatory M2 phenotypes, influencing disease progression or resolution. miR-21, highly expressed in macrophages, plays a dual role in CVDs. It promotes M2 polarization by targeting the suppressor of cytokine signaling 1 (SOCS1), which enhances the production of anti-inflammatory cytokines such as interleukin-10.57 This is particularly beneficial during the later stages of myocardial infarction when tissue repair and remodeling are critical. However, overexpression of miR-21 during the early inflammatory phase may exacerbate fibrosis and pathological remodeling by activating the TGF-β/mothers against decapentaplegic homolog (SMAD) signaling pathway.58 miR-155, a hallmark of M1 macrophages, amplifies inflammation by targeting negative regulators of the NF-κB pathway, such as SHIP1 and SOCS1.59 Elevated miR-155 levels have been associated with increased pro-inflammatory cytokine production and plaque destabilization in atherosclerosis, emphasizing its pathological role in advanced CVDs.60 miR-223, predominantly expressed in myeloid cells, serves as a critical modulator of macrophage homeostasis. It suppresses excessive activation of inflammatory pathways by targeting transcription factors such as signal transducer and activator of transcription 3 and nuclear factor I-A, preventing chronic inflammation and foam cell formation in atherosclerosis.61 Mice deficient in miR-223 exhibit aggravated inflammation and plaque instability, highlighting its protective role.62

Neutrophil regulation by miRNAs in acute and chronic inflammation

Neutrophils are the first responders to tissue injury, and their dysregulation can lead to excessive inflammation and tissue damage. miRNAs modulate neutrophil recruitment, activation, and lifespan in CVDs. miR-146a is a key regulator of neutrophilic inflammation, targeting IRAK1 and TRAF6 to suppress TLRs and NF-κB signaling pathways. Reduced miR-146a expression has been observed in patients with acute myocardial infarction, correlating with heightened neutrophil-driven inflammation.63 Therapeutic restoration of miR-146a levels has been shown to mitigate neutrophil recruitment and improve outcomes in animal models of ischemic injury.64 miR-223 also plays a role in neutrophil regulation, suppressing pro-inflammatory cytokine production and promoting neutrophil apoptosis. Dysregulation of miR-223 leads to prolonged neutrophil activation, exacerbating tissue damage during acute inflammation in myocardial infarction and chronic inflammation in heart failure.65

T lymphocyte regulation by miRNAs in CVDs

T lymphocytes, particularly CD4+ subsets, significantly contribute to the immune landscape in CVDs. miRNAs regulate T cell activation, differentiation, and effector functions, influencing disease outcomes. miR-181a modulates TCR signaling thresholds, ensuring appropriate activation during immune responses. Age-related declines in miR-181a expression impair T cell activation, contributing to the immune senescence observed in elderly CVD patients.66 Restoring miR-181a levels in preclinical models has shown promise in rejuvenating T cell function and improving immune responses.67 The miR-17-92 cluster is crucial for the survival and proliferation of effector T cells. Dysregulation of this cluster impairs the generation of cytotoxic and memory T cells, weakening immune responses in chronic inflammation associated with heart failure and atherosclerosis.68

Regulatory T cells and miRNA regulation in CVDs

Tregs are essential for maintaining immune tolerance and preventing excessive inflammation (Fig. 4). miRNAs play a vital role in shaping Treg development, stability, and function. miR-142-3p and miR-142-5p regulate cyclic adenosine monophosphate signaling and TGF-β receptor expression, both of which are critical for Treg-mediated suppression of effector T cells. Dysregulation of these miRNAs impairs Treg function, exacerbating inflammatory responses in conditions like atherosclerosis and hypertension.69 Although miR-155 is pro-inflammatory in macrophages, it plays a different role in Tregs, enhancing their proliferation and suppressive capacity by targeting the SOCS1 pathway. This dual function highlights the context-dependent nature of miRNA activity in immune regulation.70

Aspects of T cell recruitment by vascular cells may differ between species.
Fig. 4  Aspects of T cell recruitment by vascular cells may differ between species.

Vascular cells, such as endothelial cells (ECs), modulate inflammation by regulating immune cell migration, activation status, and function. ALK, anaplastic lymphoma kinase; CCL22, C-C motif chemokine ligand 22; CD, cluster of differentiation; CTLA-4, cytotoxic T-lymphocyte associated protein 4; EGFR, epidermal growth factor receptor; GITR, glucocorticoid induced TNF receptor family-related protein; GITRL, glucocorticoid-induced TNF-related ligand; HER2, human epidermal growth factor receptor 2; ICOS, inducible T-cell costimulatory; IFNγ, interferon gamma; IL, interleukin; LAG-3, lymphocyte-activation gene 3; MAPK, mitogen-activated protein kinase; MHC, major histocompatibility complex; MMP, matrix metalloproteinase; MSC, mesenchymal stem cell; NF-kB, nuclear factor kappa-light-chain-enhancer of activated B cells; NK, natural killer; PD-1, programmed cell death 1; PD-L1, programmed death ligand 1; ROS, reactive oxygen species; RTK, receptor tyrosine kinase; TCR, T-cell receptor; TGF, transforming growth factor; Th1, type 1 T helper; TIM-3, hepatitis A virus cellular receptor 2; TNF, tumor necrosis factor.

The regulation of immune cells by miRNAs has profound implications for understanding and treating CVDs. miRNA-based therapies, including mimics and inhibitors, are being explored to restore immune balance. For example, nanoparticle-mediated delivery of miR-21 mimics has shown promise in enhancing macrophage-mediated tissue repair in myocardial infarction models.32 Similarly, antagomiRs targeting miR-155 have demonstrated efficacy in reducing inflammation and plaque burden in atherosclerosis.71 Circulating miRNAs also serve as potential biomarkers for monitoring immune cell activity in CVDs. Elevated plasma levels of miR-146a and miR-223, for instance, correlate with disease severity and outcomes in myocardial infarction and heart failure, offering minimally invasive tools for diagnosis and prognosis.72 While these findings highlight the therapeutic potential of miRNA modulation, challenges such as off-target effects, delivery efficiency, and long-term safety must be addressed before clinical translation. Continued research into miRNAs’ roles in immune cell regulation will pave the way for novel, targeted approaches to managing CVDs (Table 1).73–87

Table 1

MicroRNAs and their key targets in modulating regulatory T cell (Treg) function

miRNARegulationTargetsBiological effectsReference
miR-155-5pDownSOCS1FOXP3 induces the expression of miR-155-5p in Tregs. Deficiency in miR-155 disrupts STAT5 signaling, leading to a decrease in Treg numbers and imbalance in immune regulation73
miR-142-3pDownTGFBR1, TET2, KDM6AReduced levels of miR-142-3p enhance FOXP3 expression and suppressive Treg activity by increasing KDM6A and BCL-2 levels through the demethylation of H3K27me3. It also boosts TGFBR1 expression, helping prevent the rejection of mismatched allografts7476
miR-340-5pDownIL-4In allergic rhinitis, increased expression of miR-340-5p hinders the formation and functionality of Tregs, contributing to impaired immune tolerance77
miR-125a-5pDownSTAT3, IFNG, IL-13The absence of miR-125a-5p reduces Treg populations, worsening conditions like colitis and experimental autoimmune encephalomyelitis (EAE) by promoting inflammatory responses. This miRNA plays a crucial role in maintaining the stability and balance of Tregs78
miR-4281-3pUpFOXP3Through interaction with the TATA-box in the FOXP3 promoter, miR-4281-3p enhances FOXP3 expression, improving Treg differentiation, functionality, and persistence79
miR-202-5pUpMATN2Elevated miR-202-5p levels in allergic rhinitis negatively affect the development and suppressive capabilities of Tregs, leading to reduced immune regulation80
miR-15a-5p/16-5pUpFOXP3Overexpression of miR-15a-5p/16-5p reduces FOXP3 levels, impairing the suppressive capabilities of Tregs and potentially affecting immune tolerance81
miR-146a-5pDownSTAT1The lack of miR-146a-5p in Tregs leads to a failure in tolerogenic mechanisms and promotes a shift toward Th1 immune responses, disrupting immune balance82
miR-181a/b-5pDownCTLA-4Deficiency in miR-181a/b-5p affects thymic Treg formation but enhances the suppressive activity of peripheral Tregs, contributing to a complex regulatory balance83
miR-24-3pDownFOXP3Although naturally low in Tregs, artificial overexpression of miR-24-3p decreases FOXP3 expression, impairing Treg function and stability84
miR-1224-5pDownFOXP3AhR signaling suppresses miR-1224-5p, leading to increased FOXP3 expression and Treg maturation, helping mitigate systemic inflammation caused by pertussis toxin85
miR-146b-5pDownTRAF6Using antagomirs to reduce miR-146b-5p expression enhances thymic Treg effectiveness and their inhibitory impact on graft-versus-host disease (GvHD)86
miR-142-5pDownPDE3BThe suppression of miR-142-5p in Tregs reduces cAMP levels, impairing Treg activation and peripheral tolerance. This results in systemic autoimmune inflammation87

Recent research has highlighted the critical role of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) in fine-tuning T cell responses, particularly in CVDs such as atherosclerosis and post-infarction cardiac remodeling.88 Originally characterized for its role in immune tolerance and cancer immunotherapy, emerging data increasingly link CTLA-4 to vascular inflammation and myocardial injury.88 Below is an updated overview of how CTLA-4 signaling, alongside relevant miRNA-mediated regulation, influences T cell biology and cardiovascular health.89 T cell activation requires two main signals: (1) recognition of antigenic peptides presented on MHC class I or II molecules by the TCR and (2) co-stimulation via CD28 interaction with its ligands CD80 (B7-1) or CD86 (B7-2). Once activated, T cells initiate signaling cascades such as the phosphoinositide 3-kinases/Akt pathway, promoting proliferation, survival, and cytokine production. CTLA-4 competes with CD28 for binding to CD80/CD86, exerting an inhibitory effect that dampens T cell activation. When CTLA-4 is absent or blocked, T cells become hyperresponsive, potentially exacerbating inflammatory pathologies (Fig. 5).90–92

Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) in regulatory T cells (Tregs) and dendritic cells.
Fig. 5  Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) in regulatory T cells (Tregs) and dendritic cells.

CD, cluster of differentiation; MHC, major histocompatibility complex; PD1, programmed cell death 1; PDL1, programmed death ligand 1; TCR, T-cell receptor.

Preclinical studies underscore CTLA-4’s importance in restraining pathological inflammation within the vascular endothelium. Mice deficient in CTLA-4 or treated with anti-CTLA-4 blocking antibodies exhibit larger atherosclerotic lesions, indicating that unchecked T cell activity accelerates plaque formation.93 Conversely, overexpression of CTLA-4 or administration of abatacept (a CTLA-4 analog that competes with CD28 for CD80/CD86 binding) can reduce atherosclerotic lesion size, decrease T cell proliferation, and lower pro-inflammatory cytokine levels.94 These findings suggest the therapeutic potential of CTLA-4 agonists or fusion proteins in mitigating arterial inflammation. In myocardial infarction models, CTLA-4 signaling has garnered attention. Hypoxia-induced stress on cardiomyocytes, particularly in post-infarction border zones, elevates the expression of costimulatory molecules like CD80/CD86, fueling an inflammatory environment. Administration of abatacept in this context has been shown to protect against excessive cardiac damage and improve survival in mouse models, aligning with observations in some patient cohorts where outcomes after MI are more favorable when T cell overactivation is restrained.95,96

Within the immune system, Tregs are a specialized subset that helps maintain self-tolerance and immune homeostasis. Tregs require the transcription factor forkhead box P3 (FOXP3) to retain their suppressive phenotype, and CTLA-4 is one of the key molecules driving their regulatory function.97 By binding and depleting CD80/CD86 on antigen-presenting cells, Tregs limit co-stimulation to conventional T cells, reducing overall immune activation. This mechanism is especially relevant in pathologies where unchecked inflammation leads to tissue injury, such as advanced atherosclerosis and post-infarction remodeling. In recent years, attention has turned to miRNAs as crucial post-transcriptional regulators of CTLA-4 expression. Multiple miRNAs, including miR-9, miR-105, miR-155, and miR-487a-3p, directly target CTLA-4 transcripts, influencing T cell proliferation and effector functions.89 Additional miRNAs, such as miR-24 and miR-210, can indirectly suppress CTLA-4 by downregulating FOXP3, thereby impacting Treg stability and immunosuppressive capacity.89 Among these, miR-155 has garnered particular attention. Studies in cancer and inflammatory models suggest that miR-155 can reduce CTLA-4 levels by binding to the 3′UTR of its mRNA in both Treg and conventional T cells, lowering the activation threshold for T cells.98 However, recent findings indicate that miR-155 may require cooperation with other RNA-binding proteins or miRNAs to achieve full repression of CTLA-4 (Fig. 6).99,100 This interplay likely varies in different disease contexts, such as myocardial ischemia and atherosclerosis, where cytokine milieus and hypoxic signals modulate miRNA expression. Building on these insights, novel therapeutic strategies are exploring ways to modulate CTLA-4 and its regulatory miRNAs to achieve better outcomes in CVDs. Agents like abatacept reduce detrimental inflammation by blocking costimulatory signals, thus protecting arterial walls and the ischemic myocardium. Targeting miR-155 or other miRNAs that downregulate CTLA-4 could be a future approach to reinforce Treg function. However, off-target effects remain a concern, and additional co-factors may be needed for a robust clinical response. Combining CTLA-4–focused immunotherapy with established cardiovascular interventions (e.g., lipid-lowering agents, anti-hypertensives) may yield synergistic benefits, particularly for patients with high inflammatory burdens post-MI or advanced atherosclerosis.

Post-transcriptional silencing of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) by miR-155 in Treg cells may contribute to reducing CTLA-4 mRNA expression observed in cardiovascular diseases.
Fig. 6  Post-transcriptional silencing of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) by miR-155 in Treg cells may contribute to reducing CTLA-4 mRNA expression observed in cardiovascular diseases.

Overall, miR-155 can activate CTLA-4 in T cells, suggesting a role for miR-155 as a temporal filter regulating the onset of inflammation by activating CTLA-4 expression. miRNA, microRNA; mRNA, messenger RNA; RISC, RNA-induced silencing complex.

Despite these promising leads, further investigation is essential to clarify how miRNAs, RNA-binding proteins, and inflammatory mediators orchestrate CTLA-4 expression in vivo. Mapping these regulatory networks can guide the development of next-generation immunomodulatory treatments that preserve the delicate balance between pro- and anti-inflammatory pathways in CVD. Understanding the underlying molecular details will also facilitate personalized strategies, ensuring that interventions targeting CTLA-4 and associated miRNAs are both effective and safe for diverse patient populations. In summary, recent evidence underscores CTLA-4’s pivotal role in regulating T cell activation and preventing excessive immune damage in cardiovascular settings. Ongoing exploration of CTLA-4 and its post-transcriptional control by miRNAs promises to expand our therapeutic toolkit, offering new hope for individuals with advanced atherosclerosis and ischemic heart disease (Table 2).62,101–112

Table 2

Summary of key immune-derived miRNAs in cardiovascular diseases: mechanisms, functions, and therapeutic implications

miRNAsKey findingsImpact/ApplicationsMechanism of actionFunctionsReference
miR-21, miR-155, and miR-146aExplores how immune-miRs regulate gene expression in immune cells and their contribution to cardiovascular disease progressionHighlights their potential as biomarkers for early diagnosis and disease progressionmiR-21 targets PTEN, activating the PI3K/Akt pathway, promoting cell survivalInflammatory response regulation, vascular remodeling, apoptosis inhibition101
miR-146a, miR-155, and miR-223Investigates the role of immune-miRs in modulating immune signaling pathways affecting innate and adaptive immune responses in CVDSuggests potential therapeutic targets for treating inflammation and CVDmiR-146a inhibits NF-κB signaling by targeting TRAF6 and IRAK1Modulates inflammation, immune cell function, fibrosis in vascular tissues102
miR-146a and miR-155Discusses the role of inflamma-miRs in regulating inflammation in cardiovascular disease and their association with atherogenesis.Offers insights into using inflamma-miRs as biomarkers for atherosclerosis and CVDmiR-155 enhances macrophage activation via JAK/STAT pathwayPromotes atherosclerosis, endothelial dysfunction, macrophage polarization103
miR-1, miR-133a, and miR-21Focuses on specific miRNAs as diagnostic biomarkers for heart failure and their expression profiles in patientsDemonstrates the feasibility of using miRNAs as biomarkers for heart failure diagnosismiR-1 and miR-133a target GATA4 and Hand2, influencing cardiac developmentCardiac muscle function regulation, response to stress, cell differentiation104
miR-21 and miR-155Reviews the involvement of miRNAs in immune activation and inflammation during heart failure, emphasizing their therapeutic potentialSuggests miRNAs as targets for modulating immune responses in heart failure treatmentmiR-21 promotes fibrosis by targeting SMAD7 and TGF-β signaling pathwayFibrosis regulation, cardiac remodeling, inflammation in myocardial infarction105
miR-155Highlights miR-155’s involvement in cardiovascular inflammation, especially in atherosclerosis, and its potential as a therapeutic targetFocuses on miR-155 as a crucial mediator in inflammatory pathways in CVDmiR-155 targets SOCS1, enhancing inflammation by promoting NF-κB activationPro-inflammatory response, vascular injury, endothelial cell dysfunction106
miR-223miR-223 is involved in myeloid cell differentiation and modulation of immune responses, influencing atherosclerosis progressionSuggests miR-223 as a therapeutic target for modulating inflammation in CVDmiR-223 targets NFI-A and regulates granulocyte differentiation and inflammatory responseMyeloid differentiation, immune cell modulation, suppression of inflammation62
miR-146aExamines miR-146a’s role in modulating inflammation by targeting key inflammatory pathways in CVD, such as the TLR and NF-κB pathwaysProposes miR-146a as a potential therapeutic target for controlling inflammation in CVDmiR-146a inhibits TLR/IL-1R signaling by targeting IRAK1 and TRAF6Anti-inflammatory function, modulation of immune response, vascular health107
miR-21Reviews the dual role of miR-21 in promoting fibrosis and inflammation during myocardial infarction and heart failureIdentifies miR-21 as a biomarker for fibrosis and as a therapeutic target in CVDmiR-21 activates the PI3K/Akt pathway by inhibiting PTEN, promoting fibrosisFibrosis promotion, tissue remodeling, anti-apoptotic function.108
miR-223Explores how exosomal miR-223 can modulate inflammatory responses and prevent endothelial damage, highlighting its role as a non-invasive biomarkerSuggests the potential of exosomal miR-223 in therapeutic strategies for CVDmiR-223 is secreted via exosomes and regulates endothelial function by targeting NF-κBModulation of endothelial function, immune cell response, reduction of vascular damage109
miR-92a and miR-126Investigates the role of miRNAs in regulating endothelial function and promoting inflammation in atherosclerosisIdentifies miR-92a and miR-126 as key regulators of vascular inflammation in CVDmiR-92a regulates endothelial cell junctions and angiogenesis via integrinsEndothelial barrier function, inflammatory responses in atherosclerosis110
miR-21 and miR-92aFocuses on circulating miRNAs that serve as biomarkers for early detection and prognosis in acute myocardial infarctionHighlights the use of circulating miRNAs in early diagnosis of myocardial infarctionmiR-21 and miR-92a are released from injured myocardial cells, influencing inflammationCardiovascular injury markers, inflammation resolution, early diagnosis111
miR-34aReviews the involvement of miR-34a in endothelial aging and its contribution to cardiovascular diseases such as hypertension and atherosclerosisDiscusses the potential of miR-34a as a target for anti-aging therapies in CVDmiR-34a inhibits SIRT1, leading to increased cellular senescence and agingEndothelial aging, cellular senescence, inflammation regulation112

Future directions and limitations

The growing body of evidence surrounding miRNA-based interventions in CVDs underscores both the promise of these therapies and the formidable challenges ahead. Recent preclinical and early clinical findings highlight the potential of manipulating specific miRNAs to curb pathological processes such as chronic inflammation, fibrosis, and endothelial dysfunction. However, translating these insights into robust, safe, and effective clinical interventions will require systematic refinement of current strategies, innovative delivery methods, and integration of knowledge from immunology, materials science, and regulatory biology.

A key milestone in miRNA therapeutics has been the transition from preclinical studies to initial clinical investigations. Two early clinical reports evaluated the safety and preliminary efficacy of miRNA modulation in patients with heart failure.113,114 These trials demonstrated encouraging safety profiles for miRNA modulators, providing a foundation for larger, multicenter trials that will be critical for defining optimal dosing regimens, long-term effectiveness, and potential off-target effects. Such studies should include detailed patient stratification—using biomarkers, comorbidities, and genetic background—to identify which subgroups derive the greatest benefit from miRNA therapies. Overcoming barriers such as variable miRNA expression across patient populations and disease stages will likely require multiparametric analyses that account for environmental factors such as diet and exercise that influence cardiovascular risk.

One of the most pressing challenges in miRNA-based therapy is achieving precise and efficient delivery to disease-relevant tissues and cell types. Recent work has demonstrated the potential of nanoparticle-mediated delivery systems for miR-21 inhibitors in post-myocardial infarction models, showcasing improvements in cardiac function through the reduction of pathological remodeling.115 However, this and other preclinical approaches rely on carefully engineered platforms—such as biodegradable polymers, liposomes, or viral vectors—that must meet various criteria, including biocompatibility, immunogenicity, stability in circulation, and tissue specificity. Moving forward, advances in nanotechnology and polymer science will likely enhance targeted delivery. Cell-specific targeting ligands, for instance, could direct miRNA mimics or inhibitors to pathological immune cell subsets infiltrating atherosclerotic plaques or infarcted myocardium, reducing systemic exposure and off-target effects—critical considerations given that miRNAs regulate many biological pathways.95,96 Innovative exosome engineering is another promising direction, leveraging the inherent biocompatibility of these naturally occurring vesicles and their ability to deliver cargo to specific cell types. This approach could help mitigate immune responses triggered by artificially manufactured carriers.

Chronic inflammation is central to atherogenesis, plaque instability, and adverse cardiac remodeling. Multiple studies have illustrated how miRNAs modulate chemokine and chemokine receptor expression, influencing immune cell trafficking and activation in CVDs.116–119 By fine-tuning these chemokine axes, miRNA-based strategies could offer unprecedented control over immune cell recruitment, potentially stabilizing plaques and reducing inflammatory burden without globally suppressing the immune system. Future studies must explore the best ways to harness this knowledge. For instance, miR-155 antagomiRs can dampen pro-inflammatory macrophage activity, but this same miRNA can also have protective roles in certain immune contexts. Likewise, modulating miRNAs that target chemokine receptors must account for feedback loops, such as compensatory upregulation of other inflammatory pathways. Detailed mapping of miRNA-chemokine networks at the cellular and tissue level will be crucial for optimizing therapeutic regimens that safely achieve immune homeostasis.

While conventional approaches rely on miRNA mimics or antagomiRs to alter miRNA levels, clustered regularly interspaced short palindromic repeats (CRISPR)-based technologies have opened the door to more permanent and precise modifications.83 CRISPR/Cas tools can directly edit miRNA genes or their regulatory sequences, potentially correcting dysregulated miRNAs implicated in CVDs. Nevertheless, issues such as efficiency, mosaicism, and off-target editing must be addressed before gene editing becomes a clinically viable strategy. Simultaneously, artificial intelligence (AI)-driven methods are playing an increasingly important role in CVD research. By integrating large-scale multi-omics data—encompassing genomics, proteomics, metabolomics, and transcriptomics—AI can identify robust miRNA signatures that correlate with disease progression or therapeutic response. These computational models could guide personalized treatment strategies, predicting which miRNA targets would have the highest likelihood of success in specific patient subpopulations. Moreover, AI could refine delivery approaches by modeling the interactions of various carrier systems within the complex cardiovascular environment. Current single cell sequencing platforms are not optimized to detect mature miRNAs, which limits the ability to directly measure miRNA expression in individual cells.52 However, new specialized protocols and indirect inference methods are emerging, allowing researchers to correlate single-cell gene expression profiles with known miRNA-target interactions. Improvements in sample preparation and library construction may soon allow precise identification of miRNA roles in distinct immune cell subsets (e.g., M1 vs. M2 macrophages, T effector vs. T regulatory cells) within an atherosclerotic lesion or infarct area.

An intriguing frontier in CVD research is the interplay between the gut microbiome and miRNA expression. Early evidence suggests that microbiota composition influences systemic inflammation and immune cell phenotypes by altering the expression of regulatory miRNAs, such as miR-146a and miR-155.85 Future studies could explore whether modulating the gut microbiome—through diet, probiotics, or antibiotics—synergizes with miRNA-based therapies to yield superior cardiovascular outcomes. This dual-target approach could be especially beneficial for individuals whose microbiome profile contributes to a heightened inflammatory state.

Finally, drawing parallels with more advanced fields like cancer immunotherapy could further accelerate progress. In oncology, miRNAs have been used to enhance immune checkpoint inhibition and fine-tune T cell activity. A similar strategy could be adapted for CVDs, where immune checkpoints like CTLA-4 mediate vascular inflammation and plaque development. Identifying miRNAs that regulate these pathways in T cells could lead to combination therapies that pair miRNA modulators with traditional or experimental immunomodulatory agents, offering personalized treatment regimens that target pathological immune activation without compromising systemic immune competence.

In summary, research over the past decade has firmly established miRNAs as pivotal regulators of immune-driven cardiovascular pathologies. Early clinical trials have begun to address the feasibility and preliminary safety of miRNA-based therapies, laying the groundwork for more comprehensive investigations.113,114 However, critical questions remain regarding optimal delivery systems, off-target risks, tissue-specific expression, and the long-term sustainability of miRNA interventions. As more refined technologies—from CRISPR/Cas gene editing to AI-driven biomarker discovery—converge with an evolving understanding of chemokine networks and gut microbiome interactions, the field stands on the brink of transformative breakthroughs. Addressing current limitations and rigorously validating novel approaches will ultimately determine whether miRNAs can fulfill their potential as cornerstone therapies in personalized cardiovascular medicine (Table 3).8,17,18,21,49–52,59,61,62,98–100,106,107,109,120–127

Table 3

Immune-derived miRNAs (immuno-miRs) as diagnostic and therapeutic tools in CVDs

miRNAPrimary cellular source/contextRole in CVD pathogenesisDiagnostic/Prognostic utilityTherapeutic potentialReference
miR-21Macrophages, cardiac fibroblasts; also secreted in extracellular vesiclesModulates inflammation, fibrosis, and tissue remodeling; dual role in early (pro-inflammatory) and later (repair) phasesElevated plasma levels correlate with adverse outcomes in myocardial infarction (MI) and heart failure; potential biomarker for fibrosisInhibitors (antagomiRs) may reduce pathological remodeling and fibrosis120122
miR-146aInnate immune cells (e.g., neutrophils, macrophages)Acts as a negative regulator of inflammation by targeting IRAK1 and TRAF6; modulates Toll-like receptor signalingLower or dysregulated levels associated with exacerbated inflammation in atherosclerosis and heart failure; candidate for risk stratificationmiRNA mimics could restore regulatory functions to suppress excessive inflammation21,52,59,107
miR-223Myeloid cells (macrophages and neutrophils)Regulates macrophage polarization and limits excessive inflammatory activation; contributes to plaque stabilityCirculating levels reflect immune cell activation and plaque composition in atherosclerosis; potential prognostic markermiR-223 mimics might be used to balance macrophage responses and stabilize plaques8,17,18,61,62,109
miR-155Expressed in both macrophages and neutrophils (and other immune cells)Promotes pro-inflammatory responses via NF-κB signaling; involved in atherogenesis and myocardial injury; context-dependent roles (can be anti-inflammatory in certain stages)Elevated in inflammatory conditions such as atherosclerosis and MI; useful in evaluating immune activation statusAntagomiRs targeting miR-155 may reduce excessive inflammation and improve outcomes98100,106
miR-122Although primarily liver-derived, its circulating levels reflect systemic lipid metabolismContributes to dyslipidemia and atherosclerosis through regulation of cholesterol and lipid homeostasisServes as a biomarker for lipid dysregulation and associated atherosclerotic riskInhibiting miR-122 may improve cholesterol efflux and reduce plaque formation123
miR-126Endothelial cells (EC-specific)Critical for maintaining endothelial integrity, angiogenesis, and vascular homeostasis; influences endothelial activation in atherosclerosisAltered levels are associated with endothelial dysfunction and vascular injury; potential marker for early CVD detectionmiR-126 mimics could enhance endothelial repair and promote vascular health4951
miR-33Lesional macrophages and liver (species-specific: miR-33a in mice; miR-33a/b in humans/primates)Regulates cholesterol efflux by targeting ATP-binding cassette transporters (e.g., ABCA1); contributes to foam cell formation and atherosclerotic plaque progressionCirculating levels may reflect lipid metabolism disturbances and atherosclerotic burdenInhibitors of miR-33 can enhance cholesterol efflux and reduce foam cell formation124127

Conclusions

Immune-derived miRNAs have emerged as critical regulators in CVDs, influencing both innate and adaptive immune responses. Our review demonstrates that specific miRNAs—such as miR-21, miR-146a, miR-223, miR-122, miR-126, and miR-155—play pivotal roles in mediating inflammation, modulating tissue repair, and maintaining immune homeostasis. Their ability to circulate within extracellular vesicles highlights their potential as minimally invasive biomarkers for early diagnosis, risk stratification, and monitoring disease progression in conditions such as myocardial infarction, atherosclerosis, and heart failure. Moreover, preclinical studies on miRNA-based therapeutics, using mimics and inhibitors, indicate promising strategies for targeting dysregulated immune responses in CVDs. These approaches, by directly modulating specific miRNAs, have shown potential to reduce pathological inflammation and promote tissue repair. However, challenges remain in translating these findings into clinical practice. Issues such as the complexity of miRNA regulatory networks, potential off-target effects, and difficulties in achieving efficient, cell-specific delivery must be carefully addressed. Overall, the evidence supports the transformative potential of immune-derived miRNAs as diagnostic, prognostic, and therapeutic tools in cardiovascular medicine. Future research should focus on refining these strategies through advanced technologies, including high-throughput sequencing, gene editing, and artificial intelligence. Overcoming current translation barriers will be crucial for unlocking the full potential of immune-derived miRNAs, paving the way for more personalized and effective cardiovascular care, ultimately improving patient outcomes and reducing the global burden of CVDs.

Declarations

Acknowledgement

The authors utilized the highly esteemed software BioRender and FigDraw 2.0. to create the figures for this manuscript. BioRender.com (2025) and FigDraw (2025). Retrieved from https://www.biorender.com and https://www.figdraw.com.

Funding

This work was supported by the Bashkir State Medical University Strategic Academic Leadership Program (PRIORITY-2030).

Conflict of interest

IG has been an editorial board member of Gene Expression since February 2025. The authors have no other conflict of interest to note.

Authors’ contributions

Conceptualization, writing—original draft preparation, writing—review and editing (IG, OB), validation, investigation, resources, and visualization (VP, LG, AS, HS), project administration, and funding acquisition (IG). All authors have read and agreed to the published version of the manuscript.

References

  1. Lavie CJ. Progress in Cardiovascular Diseases Statistics 2022. Prog Cardiovasc Dis 2022;73:94-95 View Article PubMed/NCBI
  2. Fukumoto Y. Lifestyle intervention for primary prevention of cardiovascular diseases. Eur J Prev Cardiol 2022;29(17):2250-2251 View Article PubMed/NCBI
  3. Goldsborough E, Osuji N, Blaha MJ. Assessment of Cardiovascular Disease Risk: A 2022 Update. Endocrinol Metab Clin North Am 2022;51(3):483-509 View Article PubMed/NCBI
  4. Schiattarella GG, Wang Y, Tian R, Hill JA. Metabolism and Inflammation in Cardiovascular Health and Diseases: Mechanisms to Therapies. J Mol Cell Cardiol 2021;157:113-114 View Article PubMed/NCBI
  5. Gareev I, Beylerli O, Yang G, Sun J, Pavlov V, Izmailov A, et al. The current state of MiRNAs as biomarkers and therapeutic tools. Clin Exp Med 2020;20(3):349-359 View Article PubMed/NCBI
  6. Domínguez-Andrés J, Dos Santos JC, Bekkering S, Mulder WJM, van der Meer JWM, Riksen NP, et al. Trained immunity: adaptation within innate immune mechanisms. Physiol Rev 2023;103(1):313-346 View Article PubMed/NCBI
  7. Gareev I, de Jesus Encarnacion Ramirez M, Goncharov E, Ivliev D, Shumadalova A, Ilyasova T, et al. MiRNAs and lncRNAs in the regulation of innate immune signaling. Noncoding RNA Res 2023;8(4):534-541 View Article PubMed/NCBI
  8. Fu Y, Mackowiak B, Feng D, Lu H, Guan Y, Lehner T, et al. MicroRNA-223 attenuates hepatocarcinogenesis by blocking hypoxia-driven angiogenesis and immunosuppression. Gut 2023;72(10):1942-1958 View Article PubMed/NCBI
  9. Kotewitsch M, Heimer M, Schmitz B, Mooren FC. Non-coding RNAs in exercise immunology: A systematic review. J Sport Health Sci 2024;13(3):311-338 View Article PubMed/NCBI
  10. Sharma Y, Saini AK, Kashyap S, Chandan G, Kaur N, Gupta VK, et al. Host miRNA and immune cell interactions: relevance in nano-therapeutics for human health. Immunol Res 2022;70(1):1-18 View Article PubMed/NCBI
  11. Ding H, Wang Y, Hu L, Xue S, Wang Y, Zhang L, 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 2020;40(3):BSR20191653 View Article PubMed/NCBI
  12. Mansouri F, Seyed Mohammadzad MH. Molecular miR-19a in Acute Myocardial Infarction: Novel Potential Indicators of Prognosis and Early Diagnosis. Asian Pac J Cancer Prev 2020;21(4):975-982 View Article PubMed/NCBI
  13. Ramanujam D, Schön AP, Beck C, Vaccarello P, Felician G, Dueck A, et al. MicroRNA-21-Dependent Macrophage-to-Fibroblast Signaling Determines the Cardiac Response to Pressure Overload. Circulation 2021;143(15):1513-1525 View Article PubMed/NCBI
  14. Labonia MCI, Estapé Senti M, van der Kraak PH, Brans MAD, Dokter I, Streef TJ, et al. Cardiac delivery of modified mRNA using lipid nanoparticles: Cellular targets and biodistribution after intramyocardial administration. J Control Release 2024;369:734-745 View Article PubMed/NCBI
  15. Cho S, Dong J, Lu LF. Cell-intrinsic and -extrinsic roles of miRNAs in regulating T cell immunity. Immunol Rev 2021;304(1):126-140 View Article PubMed/NCBI
  16. Cui Y, Qi Y, Ding L, Ding S, Han Z, Wang Y, et al. miRNA dosage control in development and human disease. Trends Cell Biol 2024;34(1):31-47 View Article PubMed/NCBI
  17. Fazi F, Rosa A, Fatica A, Gelmetti V, De Marchis ML, Nervi C, et al. A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPalpha regulates human granulopoiesis. Cell 2005;123(5):819-831 View Article PubMed/NCBI
  18. Haneklaus M, Gerlic M, O’Neill LA, Masters SL. miR-223: infection, inflammation and cancer. J Intern Med 2013;274(3):215-226 View Article PubMed/NCBI
  19. Rastegar-Moghaddam SH, Ebrahimzadeh-Bideskan A, Shahba S, Malvandi AM, Mohammadipour A. Roles of the miR-155 in Neuroinflammation and Neurological Disorders: A Potent Biological and Therapeutic Target. Cell Mol Neurobiol 2023;43(2):455-467 View Article PubMed/NCBI
  20. Mahesh G, Biswas R. MicroRNA-155: A Master Regulator of Inflammation. J Interferon Cytokine Res 2019;39(6):321-330 View Article PubMed/NCBI
  21. Liu GJ, Zhang QR, Gao X, Wang H, Tao T, Gao YY, et al. MiR-146a Ameliorates Hemoglobin-Induced Microglial Inflammatory Response via TLR4/IRAK1/TRAF6 Associated Pathways. Front Neurosci 2020;14:311 View Article PubMed/NCBI
  22. Ardinal AP, Wiyono AV, Estiko RI. Unveiling the therapeutic potential of miR-146a: Targeting innate inflammation in atherosclerosis. J Cell Mol Med 2024;28(19):e70121 View Article PubMed/NCBI
  23. Gilyazova I, Timasheva Y, Karunas A, Kazantseva A, Sufianov A, Mashkin A, et al. COVID-19: Mechanisms, risk factors, genetics, non-coding RNAs and neurologic impairments. Noncoding RNA Res 2023;8(2):240-254 View Article PubMed/NCBI
  24. McIntyre G, Jackson Z, Colina J, Sekhar S, DiFeo A. miR-181a: regulatory roles, cancer-associated signaling pathway disruptions, and therapeutic potential. Expert Opin Ther Targets 2024;28(12):1061-1091 View Article PubMed/NCBI
  25. Baumjohann D. Diverse functions of miR-17-92 cluster microRNAs in T helper cells. Cancer Lett 2018;423:147-152 View Article PubMed/NCBI
  26. Ding S, Liang Y, Zhao M, Liang G, Long H, Zhao S, et al. Decreased microRNA-142-3p/5p expression causes CD4+ T cell activation and B cell hyperstimulation in systemic lupus erythematosus. Arthritis Rheum 2012;64(9):2953-2963 View Article PubMed/NCBI
  27. Dolff S, Abdulahad WH, Wilde B. Intrinsic T-cell regulator miR-142-3p/5p - a novel therapeutic target?. Cell Mol Immunol 2021;18(2):508-509 View Article PubMed/NCBI
  28. Ménoret A, Agliano F, Karginov TA, Karlinsey KS, Zhou B, Vella AT. Antigen-specific downregulation of miR-150 in CD4 T cells promotes cell survival. Front Immunol 2023;14:1102403 View Article PubMed/NCBI
  29. Gareev I, Yang G, Sun J, Beylerli O, Chen X, Zhang D, et al. Circulating MicroRNAs as Potential Noninvasive Biomarkers of Spontaneous Intracerebral Hemorrhage. World Neurosurg 2020;133:e369-e375 View Article PubMed/NCBI
  30. Gareev I, Beylerli O, Zhao B. MiRNAs as potential therapeutic targets and biomarkers for non-traumatic intracerebral hemorrhage. Biomark Res 2024;12(1):17 View Article PubMed/NCBI
  31. Zarzour A, Kim HW, Weintraub NL. Epigenetic Regulation of Vascular Diseases. Arterioscler Thromb Vasc Biol 2019;39(6):984-990 View Article PubMed/NCBI
  32. Li M, Tang X, Liu X, Cui X, Lian M, Zhao M, et al. Targeted miR-21 loaded liposomes for acute myocardial infarction. J Mater Chem B 2020;8(45):10384-10391 View Article PubMed/NCBI
  33. Drees EEE, Pegtel DM. Circulating miRNAs as Biomarkers in Aggressive B Cell Lymphomas. Trends Cancer 2020;6(11):910-923 View Article PubMed/NCBI
  34. Nicholls M. Optimizing Cardiovascular Risk Factors. Eur Heart J 2021;42(35):3420-3421 View Article PubMed/NCBI
  35. Kawai T, Autieri MV, Scalia R. Adipose tissue inflammation and metabolic dysfunction in obesity. Am J Physiol Cell Physiol 2021;320(3):C375-C391 View Article PubMed/NCBI
  36. Marí-Alexandre J, Barceló-Molina M, Sanz-Sánchez J, Molina P, Sancho J, Abellán Y, et al. Thickness and an Altered miRNA Expression in the Epicardial Adipose Tissue Is Associated With Coronary Heart Disease in Sudden Death Victims. Rev Esp Cardiol (Engl Ed) 2019;72(1):30-39 View Article PubMed/NCBI
  37. Vacca M, Di Eusanio M, Cariello M, Graziano G, D’Amore S, Petridis FD, et al. Integrative miRNA and whole-genome analyses of epicardial adipose tissue in patients with coronary atherosclerosis. Cardiovasc Res 2016;109(2):228-239 View Article PubMed/NCBI
  38. Engin AB. MicroRNA and Adipogenesis. Adv Exp Med Biol 2017;960:489-509 View Article PubMed/NCBI
  39. Trott DW, Harrison DG. The immune system in hypertension. Adv Physiol Educ 2014;38(1):20-24 View Article PubMed/NCBI
  40. Nosalski R, Siedlinski M, Denby L, McGinnigle E, Nowak M, Cat AND, et al. T-Cell-Derived miRNA-214 Mediates Perivascular Fibrosis in Hypertension. Circ Res 2020;126(8):988-1003 View Article PubMed/NCBI
  41. Ahmadian-Elmi M, Bidmeshki Pour A, Naghavian R, Ghaedi K, Tanhaei S, Izadi T, et al. miR-27a and miR-214 exert opposite regulatory roles in Th17 differentiation via mediating different signaling pathways in peripheral blood CD4+ T lymphocytes of patients with relapsing-remitting multiple sclerosis. Immunogenetics 2016;68(1):43-54 View Article PubMed/NCBI
  42. Tian K, Xu W, Chen M, Deng F. miR-155 promotes Th17 differentiation by targeting FOXP3 to aggravate inflammation in MRSA pneumonia. Cytokine 2024;180:156662 View Article PubMed/NCBI
  43. Mansouri F, Seyed Mohammadzad MH. Corrigendum to “Effects of metformin on changes of miR-19a and miR-221 expression associated with myocardial infarction in patients with type 2 diabetes” [Diabetes Metabol Syndr: Clin Res Rev 16 (2022) 102602] https://doi.org/10.1016/j.dsx.2022.102602. Diabetes Metab Syndr 2023;17(9):102833 View Article PubMed/NCBI
  44. Naidu C, Cox AJ, Lewohl JM. Influence of sex and liver cirrhosis on the expression of miR-146a-5p and its target genes, IRAK1 and TRAF6. Brain Res 2024;1827:148763 View Article PubMed/NCBI
  45. Song X, Liu F, Chen M, Zhu M, Zheng H, Wang W, et al. MiR-21 regulates skeletal muscle atrophy and fibrosis by targeting TGF-beta/SMAD7-SMAD2/3 signaling pathway. Heliyon 2024;10(12):e33062 View Article PubMed/NCBI
  46. Tate M, Wijeratne HRS, Kim B, Philtjens S, You Y, Lee DH, et al. Deletion of miR-33, a regulator of the ABCA1-APOE pathway, ameliorates neuropathological phenotypes in APP/PS1 mice. Alzheimers Dement 2024;20(11):7805-7818 View Article PubMed/NCBI
  47. Hu J, Huang S, Liu X, Zhang Y, Wei S, Hu X. miR-155: An Important Role in Inflammation Response. J Immunol Res 2022;2022:7437281 View Article PubMed/NCBI
  48. Liu H, Wu HY, Wang WY, Zhao ZL, Liu XY, Wang LY. Regulation of miR-92a on vascular endothelial aging via mediating Nrf2-KEAP1-ARE signal pathway. Eur Rev Med Pharmacol Sci 2017;21(11):2734-2742 PubMed/NCBI
  49. Schober A, Nazari-Jahantigh M, Wei Y, Bidzhekov K, Gremse F, Grommes J, et al. MicroRNA-126-5p promotes endothelial proliferation and limits atherosclerosis by suppressing Dlk1. Nat Med 2014;20(4):368-376 View Article PubMed/NCBI
  50. Santovito D, Egea V, Bidzhekov K, Natarelli L, Mourão A, Blanchet X, et al. Noncanonical inhibition of caspase-3 by a nuclear microRNA confers endothelial protection by autophagy in atherosclerosis. Sci Transl Med 2020;12(546):eaaz2294 View Article PubMed/NCBI
  51. Jia W, Liu J, Tian X, Jiang P, Cheng Z, Meng C. Correction to: MircoRNA-126-5p inhibits apoptosis of endothelial cell in vascular arterial walls via NF-κB/PI3K/AKT/mTOR signaling pathway in atherosclerosis. J Mol Histol 2022;53(5):869 View Article PubMed/NCBI
  52. Santovito D, Weber C. Zooming in on microRNAs for refining cardiovascular risk prediction in secondary prevention. Eur Heart J 2017;38(7):524-528 View Article PubMed/NCBI
  53. Riksen NP, Bekkering S, Mulder WJM, Netea MG. Trained immunity in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2023;20(12):799-811 View Article PubMed/NCBI
  54. Nosalski R, McGinnigle E, Siedlinski M, Guzik TJ. Novel Immune Mechanisms in Hypertension and Cardiovascular Risk. Curr Cardiovasc Risk Rep 2017;11(4):12 View Article PubMed/NCBI
  55. Gareev I, Pavlov V, Du W, Yang B. MiRNAs and Their Role in Venous Thromboembolic Complications. Diagnostics (Basel) 2023;13(21):3383 View Article PubMed/NCBI
  56. Zhou L, Seo KH, Wong HK, Mi QS. MicroRNAs and immune regulatory T cells. Int Immunopharmacol 2009;9(5):524-527 View Article PubMed/NCBI
  57. Wang Z, Brandt S, Medeiros A, Wang S, Wu H, Dent A, et al. MicroRNA 21 is a homeostatic regulator of macrophage polarization and prevents prostaglandin E2-mediated M2 generation. PLoS One 2015;10(2):e0115855 View Article PubMed/NCBI
  58. Zhang Y, Yuan B, Xu Y, Zhou N, Zhang R, Lu L, et al. MiR-208b/miR-21 Promotes the Progression of Cardiac Fibrosis Through the Activation of the TGF-β1/Smad-3 Signaling Pathway: An in vitro and in vivo Study. Front Cardiovasc Med 2022;9:924629 View Article PubMed/NCBI
  59. Renrick AN, Thounaojam MC, de Aquino MTP, Chaudhuri E, Pandhare J, Dash C, et al. Bortezomib Sustains T Cell Function by Inducing miR-155-Mediated Downregulation of SOCS1 and SHIP1. Front Immunol 2021;12:607044 View Article PubMed/NCBI
  60. Xue X, Wang J, Fu K, Dai S, Wu R, Peng C, et al. The role of miR-155 on liver diseases by modulating immunity, inflammation and tumorigenesis. Int Immunopharmacol 2023;116:109775 View Article PubMed/NCBI
  61. Flynn CL, Markey GE, Neudecker V, Farrelly C, Furuta GT, Eltzschig HK, et al. The MicroRNA miR-223 Constrains Colitis-associated Tumorigenesis by Limiting Myeloid Cell Infiltration and Chemokine Expression. J Immunol 2024;213(12):1869-1883 View Article PubMed/NCBI
  62. Jiao P, Wang XP, Luoreng ZM, Yang J, Jia L, Ma Y, et al. miR-223: An Effective Regulator of Immune Cell Differentiation and Inflammation. Int J Biol Sci 2021;17(9):2308-2322 View Article PubMed/NCBI
  63. Bukauskas T, Mickus R, Cereskevicius D, Macas A. Value of Serum miR-23a, miR-30d, and miR-146a Biomarkers in ST-Elevation Myocardial Infarction. Med Sci Monit 2019;25:3925-3932 View Article PubMed/NCBI
  64. Cecconi A, Navarrete G, Garcia-Guimaraes M, Vera A, Blanco-Dominguez R, Sanz-Garcia A, et al. Influence of air pollutants on circulating inflammatory cells and microRNA expression in acute myocardial infarction. Sci Rep 2022;12(1):5350 View Article PubMed/NCBI
  65. Zidar N, Boštjančič E, Glavač D, Stajer D. MicroRNAs, innate immunity and ventricular rupture in human myocardial infarction. Dis Markers 2011;31(5):259-265 View Article PubMed/NCBI
  66. Ye Z, Li G, Kim C, Hu B, Jadhav RR, Weyand CM, et al. Regulation of miR-181a expression in T cell aging. Nat Commun 2018;9(1):3060 View Article PubMed/NCBI
  67. Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G, et al. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 2007;129(1):147-161 View Article PubMed/NCBI
  68. Wu T, Wieland A, Araki K, Davis CW, Ye L, Hale JS, et al. Temporal expression of microRNA cluster miR-17-92 regulates effector and memory CD8+ T-cell differentiation. Proc Natl Acad Sci U S A 2012;109(25):9965-9970 View Article PubMed/NCBI
  69. Sun P, Wang N, Zhao P, Wang C, Li H, Chen Q, et al. Circulating Exosomes Control CD4(+) T Cell Immunometabolic Functions via the Transfer of miR-142 as a Novel Mediator in Myocarditis. Mol Ther 2020;28(12):2605-2620 View Article PubMed/NCBI
  70. Yu J, Mei J, Zuo D, Zhang M, Yu S, Li F, et al. Inflammatory factor-mediated miR-155/SOCS1 signaling axis leads to Treg impairment in systemic lupus erythematosus. Int Immunopharmacol 2024;141:113013 View Article PubMed/NCBI
  71. Rachmawati E, Sargowo D, Rohman MS, Widodo N, Kalsum U. miR-155-5p predictive role to decelerate foam cell atherosclerosis through CD36, VAV3, and SOCS1 pathway. Noncoding RNA Res 2021;6(2):59-69 View Article PubMed/NCBI
  72. Gangwar RS, Rajagopalan S, Natarajan R, Deiuliis JA. Noncoding RNAs in Cardiovascular Disease: Pathological Relevance and Emerging Role as Biomarkers and Therapeutics. Am J Hypertens 2018;31(2):150-165 View Article PubMed/NCBI
  73. Elton TS, Selemon H, Elton SM, Parinandi NL. Regulation of the MIR155 host gene in physiological and pathological processes. Gene 2013;532(1):1-12 View Article PubMed/NCBI
  74. Lu Y, Gao J, Zhang S, Gu J, Lu H, Xia Y, et al. miR-142-3p regulates autophagy by targeting ATG16L1 in thymic-derived regulatory T cell (tTreg). Cell Death Dis 2018;9(3):290 View Article PubMed/NCBI
  75. Gao J, Gu J, Pan X, Gan X, Ju Z, Zhang S, et al. Blockade of miR-142-3p promotes anti-apoptotic and suppressive function by inducing KDM6A-mediated H3K27me3 demethylation in induced regulatory T cells. Cell Death Dis 2019;10(5):332 View Article PubMed/NCBI
  76. Wang WL, Ouyang C, Graham NM, Zhang Y, Cassady K, Reyes EY, et al. microRNA-142 guards against autoimmunity by controlling Treg cell homeostasis and function. PLoS Biol 2022;20(2):e3001552 View Article PubMed/NCBI
  77. Rouas R, Merimi M, Najar M, El Zein N, Fayyad-Kazan M, Berehab M, et al. Human CD8(+) CD25 (+) CD127 (low) regulatory T cells: microRNA signature and impact on TGF-β and IL-10 expression. J Cell Physiol 2019;234(10):17459-17472 View Article PubMed/NCBI
  78. Li D, Kong C, Tsun A, Chen C, Song H, Shi G, et al. MiR-125a-5p Decreases the Sensitivity of Treg cells Toward IL-6-Mediated Conversion by Inhibiting IL-6R and STAT3 Expression. Sci Rep 2015;5:14615 View Article PubMed/NCBI
  79. Zhang Y, Liu W, Chen Y, Liu J, Wu K, Su L, et al. A Cellular MicroRNA Facilitates Regulatory T Lymphocyte Development by Targeting the FOXP3 Promoter TATA-Box Motif. J Immunol 2018;200(3):1053-1063 View Article PubMed/NCBI
  80. Wang L, Yang X, Li W, Song X, Kang S. MiR-202-5p/MATN2 are associated with regulatory T-cells differentiation and function in allergic rhinitis. Hum Cell 2019;32(4):411-417 View Article PubMed/NCBI
  81. Haralambieva IH, Kennedy RB, Simon WL, Goergen KM, Grill DE, Ovsyannikova IG, et al. Differential miRNA expression in B cells is associated with inter-individual differences in humoral immune response to measles vaccination. PLoS One 2018;13(1):e0191812 View Article PubMed/NCBI
  82. Garbin A, Contarini G, Damanti CC, Tosato A, Bortoluzzi S, Gaffo E, et al. MiR-146a-5p enrichment in small-extracellular vesicles of relapsed pediatric ALCL patients promotes macrophages infiltration and differentiation. Biochem Pharmacol 2023;215:115747 View Article PubMed/NCBI
  83. Li G, Yu M, Lee WW, Tsang M, Krishnan E, Weyand CM, et al. Decline in miR-181a expression with age impairs T cell receptor sensitivity by increasing DUSP6 activity. Nat Med 2012;18(10):1518-1524 View Article PubMed/NCBI
  84. Zhou YF, Song SS, Tian MX, Tang Z, Wang H, Fang Y, et al. Cystathionine β-synthase mediated PRRX2/IL-6/STAT3 inactivation suppresses Tregs infiltration and induces apoptosis to inhibit HCC carcinogenesis. J Immunother Cancer 2021;9(8):e003031 View Article PubMed/NCBI
  85. Al-Ghezi ZZ, Singh N, Mehrpouya-Bahrami P, Busbee PB, Nagarkatti M, Nagarkatti PS. AhR Activation by TCDD (2,3,7,8-Tetrachlorodibenzo-p-dioxin) Attenuates Pertussis Toxin-Induced Inflammatory Responses by Differential Regulation of Tregs and Th17 Cells Through Specific Targeting by microRNA. Front Microbiol 2019;10:2349 View Article PubMed/NCBI
  86. Tu Z, Xiong J, Xiao R, Shao L, Yang X, Zhou L, et al. Loss of miR-146b-5p promotes T cell acute lymphoblastic leukemia migration and invasion via the IL-17A pathway. J Cell Biochem 2019;120(4):5936-5948 View Article PubMed/NCBI
  87. Anandagoda N, Willis JC, Hertweck A, Roberts LB, Jackson I, Gökmen MR, et al. microRNA-142-mediated repression of phosphodiesterase 3B critically regulates peripheral immune tolerance. J Clin Invest 2019;129(3):1257-1271 View Article PubMed/NCBI
  88. Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood 2018;131(1):58-67 View Article PubMed/NCBI
  89. Ebrahimi A, Barati T, Mirzaei Z, Fattahi F, Mansoori Derakhshan S, Shekari Khaniani M. An overview on the interaction between non-coding RNAs and CTLA-4 gene in human diseases. Med Oncol 2024;42(1):13 View Article PubMed/NCBI
  90. Gergely TG, Drobni ZD, Sayour NV, Ferdinandy P, Varga ZV. Molecular fingerprints of cardiovascular toxicities of immune checkpoint inhibitors. Basic Res Cardiol 2025;120(1):187-205 View Article PubMed/NCBI
  91. Yousif LI, Tanja AA, de Boer RA, Teske AJ, Meijers WC. The role of immune checkpoints in cardiovascular disease. Front Pharmacol 2022;13:989431 View Article PubMed/NCBI
  92. Xiao Q, Li X, Li Y, Wu Z, Xu C, Chen Z, et al. Biological drug and drug delivery-mediated immunotherapy. Acta Pharm Sin B 2021;11(4):941-960 View Article PubMed/NCBI
  93. Callahan MK, Wolchok JD. At the bedside: CTLA-4- and PD-1-blocking antibodies in cancer immunotherapy. J Leukoc Biol 2013;94(1):41-53 View Article PubMed/NCBI
  94. Ewing MM, Karper JC, Abdul S, de Jong RC, Peters HA, de Vries MR, et al. T-cell co-stimulation by CD28-CD80/86 and its negative regulator CTLA-4 strongly influence accelerated atherosclerosis development. Int J Cardiol 2013;168(3):1965-1974 View Article PubMed/NCBI
  95. Mansouri F, Seyed Mohammadzad MH. Decreased Expression of Cytotoxic T Lymphocyte-associated Protein 4: A Risk Factor of Myocardial Infarction. Iran J Allergy Asthma Immunol 2022;21(1):86-91 View Article PubMed/NCBI
  96. Buehning F, Lerchner T, Vogel J, Hendgen-Cotta UB, Totzeck M, Rassaf T, et al. Preclinical models of cardiotoxicity from immune checkpoint inhibitor therapy. Basic Res Cardiol 2025;120(1):171-185 View Article PubMed/NCBI
  97. Mansouri F, Seyed Mohammadzad MH. Up-Regulation of Cell-Free MicroRNA-1 and MicroRNA-221-3p Levels in Patients with Myocardial Infarction Undergoing Coronary Angiography. Adv Pharm Bull 2021;11(4):719-727 View Article PubMed/NCBI
  98. Braile M, Luciano N, Carlomagno D, Salvatore G, Orlandella FM. Insight into the Role of the miR-584 Family in Human Cancers. Int J Mol Sci 2024;25(13):7448 View Article PubMed/NCBI
  99. Braile M, Marcella S, Cristinziano L, Galdiero MR, Modestino L, Ferrara AL, et al. VEGF-A in Cardiomyocytes and Heart Diseases. Int J Mol Sci 2020;21(15):5294 View Article PubMed/NCBI
  100. Xu D, Gareev I, Beylerli O, Pavlov V, Le H, Shi H. Integrative bioinformatics analysis of miRNA and mRNA expression profiles and identification of associated miRNA-mRNA network in intracranial aneurysms. Noncoding RNA Res 2024;9(2):471-485 View Article PubMed/NCBI
  101. Roberts LB, Kapoor P, Howard JK, Shah AM, Lord GM. An update on the roles of immune system-derived microRNAs in cardiovascular diseases. Cardiovasc Res 2021;117(12):2434-2449 View Article PubMed/NCBI
  102. Yibulayin K, Abulizi M. The function of miRNAs in the immune system’s inflammatory reaction to heart failure. Front Cardiovasc Med 2024;11:1506836 View Article PubMed/NCBI
  103. Zapata-Martínez L, Águila S, de Los Reyes-García AM, Carrillo-Tornel S, Lozano ML, González-Conejero R, et al. Inflammatory microRNAs in cardiovascular pathology: another brick in the wall. Front Immunol 2023;14:1196104 View Article PubMed/NCBI
  104. Fayez SS, Mishlish SM, Saied HM, Shaban SA, Suleiman AA, Hassan F, et al. Role of Different Types of miRNAs in Some Cardiovascular Diseases and Therapy-Based miRNA Strategies: A Mini Review. Biomed Res Int 2022;2022:2738119 View Article PubMed/NCBI
  105. van de Vrie M, Heymans S, Schroen B. MicroRNA involvement in immune activation during heart failure. Cardiovasc Drugs Ther 2011;25(2):161-170 View Article PubMed/NCBI
  106. Cao RY, Li Q, Miao Y, Zhang Y, Yuan W, Fan L, et al. The Emerging Role of MicroRNA-155 in Cardiovascular Diseases. Biomed Res Int 2016;2016:9869208 View Article PubMed/NCBI
  107. Arroyo AB, Águila S, Fernández-Pérez MP, Reyes-García AML, Reguilón-Gallego L, Zapata-Martínez L, et al. miR-146a in Cardiovascular Diseases and Sepsis: An Additional Burden in the Inflammatory Balance?. Thromb Haemost 2021;121(9):1138-1150 View Article PubMed/NCBI
  108. Khalaji A, Mehrtabar S, Jabraeilipour A, Doustar N, Rahmani Youshanlouei H, Tahavvori A, et al. Inhibitory effect of microRNA-21 on pathways and mechanisms involved in cardiac fibrosis development. Ther Adv Cardiovasc Dis 2024;18:17539447241253134 View Article PubMed/NCBI
  109. Zhang MW, Shen YJ, Shi J, Yu JG. MiR-223-3p in Cardiovascular Diseases: A Biomarker and Potential Therapeutic Target. Front Cardiovasc Med 2020;7:610561 View Article PubMed/NCBI
  110. Feinberg MW, Moore KJ. MicroRNA Regulation of Atherosclerosis. Circ Res 2016;118(4):703-720 View Article PubMed/NCBI
  111. Zhang L, Ding H, Zhang Y, Wang Y, Zhu W, Li P. Circulating MicroRNAs: Biogenesis and Clinical Significance in Acute Myocardial Infarction. Front Physiol 2020;11:1088 View Article PubMed/NCBI
  112. Hua CC, Liu XM, Liang LR, Wang LF, Zhong JC. Targeting the microRNA-34a as a Novel Therapeutic Strategy for Cardiovascular Diseases. Front Cardiovasc Med 2021;8:784044 View Article PubMed/NCBI
  113. Täubel J, Hauke W, Rump S, Viereck J, Batkai S, Poetzsch J, et al. Novel antisense therapy targeting microRNA-132 in patients with heart failure: results of a first-in-human Phase 1b randomized, double-blind, placebo-controlled study. Eur Heart J 2021;42(2):178-188 View Article PubMed/NCBI
  114. Abplanalp WT, Fischer A, John D, Zeiher AM, Gosgnach W, Darville H, et al. Efficiency and Target Derepression of Anti-miR-92a: Results of a First in Human Study. Nucleic Acid Ther 2020;30(6):335-345 View Article PubMed/NCBI
  115. Chipont A, Esposito B, Challier I, Montabord M, Tedgui A, Mallat Z, et al. MicroRNA-21 Deficiency Alters the Survival of Ly-6C(lo) Monocytes in ApoE(-/-) Mice and Reduces Early-Stage Atherosclerosis-Brief Report. Arterioscler Thromb Vasc Biol 2019;39(2):170-177 View Article PubMed/NCBI
  116. Xie Y, Wang Y, Li J, Hang Y, Oupický D. Promise of chemokine network-targeted nanoparticles in combination nucleic acid therapies of metastatic cancer. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2019;11(2):e1528 View Article PubMed/NCBI
  117. Cimen I, Natarelli L, Abedi Kichi Z, Henderson JM, Farina FM, Briem E, et al. Targeting a cell-specific microRNA repressor of CXCR4 ameliorates atherosclerosis in mice. Sci Transl Med 2023;15(720):eadf3357 View Article PubMed/NCBI
  118. Ge Q, Brichard S, Yi X, Li Q. microRNAs as a new mechanism regulating adipose tissue inflammation in obesity and as a novel therapeutic strategy in the metabolic syndrome. J Immunol Res 2014;2014:987285 View Article PubMed/NCBI
  119. Döring Y, Noels H, van der Vorst EPC, Neideck C, Egea V, Drechsler M, et al. Vascular CXCR4 Limits Atherosclerosis by Maintaining Arterial Integrity: Evidence From Mouse and Human Studies. Circulation 2017;136(4):388-403 View Article PubMed/NCBI
  120. Karakas M, Schulte C, Appelbaum S, Ojeda F, Lackner KJ, Münzel T, et al. Circulating microRNAs strongly predict cardiovascular death in patients with coronary artery disease-results from the large AtheroGene study. Eur Heart J 2017;38(7):516-523 View Article PubMed/NCBI
  121. Usuelli V, Ben Nasr M, D’Addio F, Liu K, Vergani A, El Essawy B, et al. miR-21 antagonism reprograms macrophage metabolism and abrogates chronic allograft vasculopathy. Am J Transplant 2021;21(10):3280-3295 View Article PubMed/NCBI
  122. Schober A, Blay RM, Saboor Maleki S, Zahedi F, Winklmaier AE, Kakar MY, et al. MicroRNA-21 Controls Circadian Regulation of Apoptosis in Atherosclerotic Lesions. Circulation 2021;144(13):1059-1073 View Article PubMed/NCBI
  123. Kostiniuk D, Marttila S, Raitoharju E. Circulatory miRNAs in essential hypertension. Atherosclerosis 2025;401:119069 View Article PubMed/NCBI
  124. Zhang X, Rotllan N, Canfrán-Duque A, Sun J, Toczek J, Moshnikova A, et al. Targeted Suppression of miRNA-33 Using pHLIP Improves Atherosclerosis Regression. Circ Res 2022;131(1):77-90 View Article PubMed/NCBI
  125. Price NL, Zhang X, Fernández-Tussy P, Singh AK, Burnap SA, Rotllan N, et al. Loss of hepatic miR-33 improves metabolic homeostasis and liver function without altering body weight or atherosclerosis. Proc Natl Acad Sci U S A 2021;118(5):e2006478118 View Article PubMed/NCBI
  126. Mandolini C, Santovito D, Marcantonio P, Buttitta F, Bucci M, Ucchino S, et al. Identification of microRNAs 758 and 33b as potential modulators of ABCA1 expression in human atherosclerotic plaques. Nutr Metab Cardiovasc Dis 2015;25(2):202-209 View Article PubMed/NCBI
  127. Santovito D, Marcantonio P, Mastroiacovo D, Natarelli L, Mandolini C, De Nardis V, et al. High dose rosuvastatin increases ABCA1 transporter in human atherosclerotic plaques in a cholesterol-independent fashion. Int J Cardiol 2020;299:249-253 View Article PubMed/NCBI

About this Article

Cite this article
Gareev I, Beylerli O, Sufianov A, Gulieva L, Pavlov V, Shi H. MicroRNAs in the Regulation of Immune Response in Cardiovascular Diseases: New Diagnostic and Therapeutic Tools. Gene Expr. Published online: Apr 23, 2025. doi: 10.14218/GE.2025.00010.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
January 23, 2025 March 7, 2025 March 26, 2025 April 23, 2025
DOI http://dx.doi.org/10.14218/GE.2025.00010
  • Gene Expression
  • eISSN 1555-3884
Back to Top

MicroRNAs in the Regulation of Immune Response in Cardiovascular Diseases: New Diagnostic and Therapeutic Tools

Ilgiz Gareev, Ozal Beylerli, Albert Sufianov, Leili Gulieva, Valentin Pavlov, Huaizhang Shi
  • Reset Zoom
  • Download TIFF