- Review
- Open access
- Published:
Non-coding RNAs affecting NLRP3 inflammasome pathway in diabetic cardiomyopathy: a comprehensive review of potential therapeutic options
Journal of Translational Medicine volume 23, Article number: 249 (2025)
Abstract
Cardiomyopathies are a heterogeneous group of disorders that can lead to fulminant heart failure and sudden cardiac death. In recent years, the prevalence of all types of cardiomyopathies has shown an upward trend globally. Up to 40% of patients with cardiomyopathy-related heart failure have diabetes mellitus (DM). With the fast global spread of DM, the prevalence of DCM is increasing accordingly and it remains the leading cause of morbidity and mortality in chronic diabetic patients. NLRP3 inflammasome significantly contributes to the development and pathological progression of DCM. Targeting the inflammasome or any of the mediators along its activation pathway provides new potential therapeutic targets for developing specialized drugs to treat DCM.
In this comprehensive review, we sought to introduce and summarize the non-coding RNAs with potential therapeutic effects targeting NLRP3 inflammasome signaling in DCM. We hope this general overview can aid future research in developing new therapies for DCM.
Background
Cardiomyopathies are a heterogeneous group of disorders with various etiologies causing functional and structural abnormalities of the myocardium in the absence of valvular disease, hypertension, coronary artery disease (CAD), and congenital heart disease [1, 2]. Presenting signs and symptoms often fall into a spectrum from subtle clinical manifestations to fulminant heart failure [1]. Arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D) and hypertrophic cardiomyopathy (HCM) are known as important common causes of sudden cardiac death in young patients and athletes, respectively [1, 3]. Nearly 50% of children and adolescents with sudden cardiac death or waiting for heart transplantation, are affected by cardiomyopathies [4]. In 2020, about 60% of heart transplant recipients in the United States were primarily diagnosed with cardiomyopathy [5]. A study by Brownrigg et al. showed that the prevalence of all types of cardiomyopathies increased from 2010 to 2018 [6]. Globally, six million people were affected by cardiomyopathy and myocarditis in 2020 [5, 7].
To date, many classification systems have been proposed for cardiomyopathies. In 2006, the American Heart Association (AHA) classified cardiomyopathies as either primary or secondary based on the predominant organ involved, with primary cardiomyopathies further subdivided into genetic, mixed, and acquired cardiomyopathies [1]. In 2008, the European Society of Cardiology (ESC) categorized cardiomyopathies into specific functional and morphological phenotypes, with each phenotype subcategorized into familial and non-familial forms [2].
The most recent classification system endorsed by the World Heart Federation (WHF), the MOGE(S) nosology of cardiomyopathies, was adopted from the tumor, node, metastases (TNM) staging in oncology; this classification embodies and integrates five attributes: the morphofunctional phenotype (M), organ(s) involvement (O), genetic or familial inheritance pattern (G), etiological annotation (E), and the functional status (S) [8]. In recent years, the number of recognized cardiomyopathies has increased due to the advances in genetic attributes of cardiomyopathies [8,9,10].
Inflammasome-mediated inflammation plays a central role in the development and pathological progression of several cardiovascular diseases (CVDs), including cardiomyopathies [11,12,13,14,15,16,17,18,19,20]. Inflammasome is a high molecular weight multimeric protein complex [20,21,22] (generally composed of 3 proteins [17, 23,24,25]) in the cytosol of immune cells as well as the cardiovascular system cells. It plays a key role in response to cellular stress [23]. Inflammasome functions by triggering the activation of the proinflammatory cytokines interleukin (IL)-1β and IL-18 [20], resulting in a series of inflammatory responses, ultimately leading to the pathophysiology of CVDs [13, 26,27,28].
Many inflammasomes have been identified to date, but the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome is the most widely studied inflammasome. Growing evidence highlights the critical role of NLRP3 inflammasome activation in the pathogenesis of ischemic and non-ischemic cardiomyopathies [11, 13, 23]. NLRP3 inflammasome induces pyroptosis, a form of inflammatory programmed cell death [23], which contributes to the development of dilated cardiomyopathy [15, 20]. Understanding the role of inflammasomes in cardiomyopathies and identifying the underlying mechanisms, has led to the emergence of novel therapies targeting the inflammasome [20, 21, 23].
In this review, we aim to provide a comprehensive summary of the non-coding RNAs targeting NLRP3 inflammasome signaling in diabetic cardiomyopathy (DCM).
NLRP3 inflammasome
Inflammasomes are an important component of innate immunity [22, 29], generally composed of three parts, sensor receptors that are triggered and activated by pathogen-associated molecular patterns (PAMPs) and host-derived danger-related molecular patterns (DAMPs) [30, 31], adaptors that facilitate an inflammatory reaction, and effectors that are able to initiate the inflammatory cascade [17, 25] (Fig. 1). The assembly of the NLRP3 inflammasome includes the receptor protein NLRP3, the adaptor apoptosis-associated speck-like protein containing a CARD (ASC), and the effector protein pro-caspase-1 [12, 29, 30, 32, 33]. The NLRP3 component belongs to the nucleotide-binding domain and leucine‐rich repeat (NLR) protein family [12, 13], a group of pattern recognition receptors (PRRs) [17, 34, 35]. The NLR proteins share a mutual organization [12, 35]: a C-terminal leucine-rich repeats (LRR) domain [34, 36], a central nucleotide binding and oligomerization domain (NOD, also known as NACHT) [13, 37], and an N-terminal effector domain [17, 30, 35]. NLRP3 has the common structure of NLRs employing pyrin as its N-terminal effector (PYD) [12, 29, 33], which connects downstream bridging proteins to effector molecules [34, 36]. ASC is a speck-like protein consisting of two domains (a C-terminal CARD and an N-terminal PYD) that serves as the connection between NLRP3 and pro-caspase-1 [12, 38]. Pro-caspase-1, the effector protein of the inflammasome, is cleaved into caspase-1 after autocatalytic maturation on ASC filaments [12, 39]. Caspase-1 is a vital proteolytic enzyme in human homeostasis, resulting in the cleavage of pro-IL-1β and pro-IL-18 into mature cytokines [12, 40].
Structure of the NLRP3 inflammasome. The receptor protein NLRP3 belongs to the NLR family, a group of pattern recognition receptors (PRRs) that function as cytoplasmic receptors. The common structure of NLR proteins is as follows: [1] a C-terminal leucine-rich repeats (LRR) domain that recognizes and binds PAMP or DAMP stimuli [2], a central nucleotide binding and oligomerization domain (NOD, also known as NACHT) that upon ligand binding, undergoes conformational changes, triggering oligomerization and activating adenosine triphosphate (ATP)ase activity through its ATP-binding site. This process is essential for NLRP3 self-association and activation [3]. and an N-terminal effector domain, which is pyrin (PYD) in the NLRP3 inflammasome. The adaptor apoptosis-associated speck-like protein containing a CARD (ASC) connects NLRP3 and pro-caspase-1. Its N-terminal PYD interacts with the PYD of the NLRP3, providing a tight connection, and its C-terminal CARD domain subsequently connects with pro-caspase-1 via the homotypic CARD–CARD reaction. The oligomerization of pro-caspase-1 on ASC filaments facilitates its proximity-driven autocatalytic cleavage into mature caspase-1, which then forms an active heterotetramer, resulting in the cleavage of pro-IL-1β and pro-IL-18 into mature cytokines and inducing their release
NLRP3 pathways in cardiomyopathy
NLRP3 inflammasome contributes to the pathophysiology of cardiomyopathy by inducing IL-1β and IL-18 release [12, 17]. In the cardiac tissue, IL-1β triggers calcium leak from the sarcoplasmic reticulum, promotes cell death and tissue remodeling, and ultimately impairs contractility [18, 41].
NLRP3 also plays a critical role in cardiomyocyte pyroptosis (a newly defined programmed cell death) that contributes to dilated cardiomyopathy [1, 12]. Increased mRNA levels of NLRP3 inflammasome components and over-activation of related pathways were observed in the serum and ventricular tissues of mice models of cardiomyopathies [13,14,15,16, 42, 43]. During physiological conditions, NLRP3 is inactive [44, 45]. Complete activation of the NLRP3 inflammasome occurs in two stages. It is strictly regulated at the level of priming and activation [12, 30, 44, 45]. The priming step, also known as ‘the first signal’, is initiated by the recognition of PAMPs and DAMPs by Toll-like receptors (TLRs), which activate TLR-adaptor molecule myeloid differentiation primary response 88 (MyD88) signaling and/or cytokine receptors (TNFRs). This leads to stimulation of cytokines such as TNF-𝛼 which induce the release and nuclear translocation of active transcription factor nuclear factor kappa B (NF‑κB), which, then, primes the transcription of inflammasome components such as pro-caspase-1, pro-IL-1β, pro-IL-18, and NLRP3 resulting in their increased intracellular transcript levels [11,12,13, 30, 33, 34, 44,45,46,47,48,49,50,51,52,53]. After recognition of the stimuli, TLRs also promote the transcription of inflammasome-related components through post-translational modification of the NLRP3 inflammasome [29, 54].
In the non-canonical (alternative) activation pathway of NLRP3, the transcription step is mediated by caspase-8 and depends on Gram-negative bacteria [13, 29]. Lipopolysaccharide (LPS) promotes the transcription of the inflammasome components through the TLR4-MyD88-TRIF-RIPK1-FADD-CASP8 signaling pathway [13, 29, 55]. The activation of this pathway also induces the oligomerization of the canonical NLRP3 inflammasome and results in the transcription of the caspase-11 gene [13, 55, 56].
When the inflammasome components are not adequately expressed, activation of NLRP3 alone will be insufficient to induce caspase-1 activation and IL-1B or IL-18 production, causing priming to act as the limiting step [44].
Once primed, subsequent NLRP3 inflammasome activation termed ‘the second signal’ is triggered via multiple inter-related pathways downstream of various unrelated stimuli leading to final inflammasome assembly, which is crucial for signal transduction from upstream molecules to the downstream effector molecules [11, 30, 34, 36, 44]. These complex activation pathways are classified according to the source of the activation signal [33].
Stimuli
Stimuli are divided into three main groups: ionic flux, mitochondrial dysfunction (via reactive oxygen species (ROS)), and lysosomal damage [11, 12, 57]. In addition, the Golgi apparatus, endoplasmic reticulum (ER) stress, and noncoding RNAs are gradually being discovered to contribute to NLRP3 inflammasome activation [11, 33].
Ionic flux
Alterations in the intracellular concentrations of K+, Ca2+, and Cl- are different cellular signals regulating the activation of the NLRP3 inflammasome [29].
K+ efflux
The most common mechanism of NLRP3 inflammasome activation is K+ efflux [11, 29, 33] (Fig. 2A). Recognition of bacterial toxins, particulate matter, and ATP by PAMPs/DAMPs can converge on K+ efflux [29, 34, 58, 59]. ATP is a common trigger of NLRP3 inflammasome activation via the purinergic receptor (P2 × 7R), a characteristic non-selective ATP-gated cationic channel that partially regulates K+ currents [12, 29, 30, 60]. Cell membrane destruction and some microbial toxins are other triggers of K+ efflux, which leads to NLRP3 inflammasome activation [29, 61, 62].
Ionic flux. (A) K+ efflux: Detection of the elevated levels of ATP in the extracellular medium by P2 × 7R and activation of this receptor by caspase-4/5/11 results in the opening of the channel which induces K+ efflux. The decrease in intracellular potassium concentration activates the NLRP3 inflammasome. The binding of never-in-mitosis A-related kinase 7 (NEK7, a well‑conserved serine/threonine kinase also known as NIMA‑related kinase 7), to NLRP3 downstream to K+ efflux plays a key role in this process resulting in the molecular complex formation and further activating the NLRP3 inflammasome. Cell membrane destruction, some microbial toxins, and the non-canonical pathway also contribute to the NLRP3 inflammasome activation via K+ efflux. (B) Ca2+ mobilization: Calcium ion mobilization from the ER through Ca2+ channels or extracellular Ca2+ influx through the P2 × 7 receptor triggered by ATP results in increased intracellular Ca2+ levels. Ca2+ overload prompts the assembly of the NLRP3 inflammasome complex and plays an important role in the activation of the NLRP3 inflammasome. Ca2+ may facilitate the interaction between NLRP3 and ASC by inducing conformational changes
Ca2+ mobilization
Increased cytosolic Ca2+ concentration (Ca2+ overload) due to the mobilization of Ca2+ from ER or extracellular Ca2+ influx through the P2 × 7 receptor contributes to NLRP3 inflammasome activation [33, 34] (Fig. 2B). Also, it is proposed that Ca2+ may facilitate the interaction between NLRP3 and ASC by inducing conformational changes [12, 63].
Cl-efflux
Elevated extracellular Cl- concentrations promote the maturation of IL-1β induced by ATP [12, 29]. Comparably, Na+ influx also takes part in NLRP3 activation [29].
Mitochondrial dysfunction
Mitochondrial dysfunction and ROS production contribute to NLRP3 inflammasome activation [11] (Fig. 3). Once the respiratory chain reaction in mitochondria is inhibited, oxygen accumulates in the form of mitochondrial ROS (mtROS), which is essential for the response of NLRP3 to LPS and ATP [12, 64]. In addition to interacting with NLRP3, the mtDNA is also crucial for the activation process [12, 65, 66].
Various stimuli, including PAMPs, DAMPs, LPS, ATP, Hyperglycemia, and asbestos, inhibit the respiratory chain reaction in mitochondria, leading to oxygen accumulation in the form of mtROS. Detection of increased ROS concentrations by thioredoxin interacting protein (TXNIP) results in its dissociation from thioredoxin (TRX). TXNIP then binds to NLRP3, activating the NLRP3 inflammasome complex. ROS can also promote the interaction between Nek7 and NLRP3, which recruits the ASC domain subsequently, inducing NLRP3 assembly and oligomerization
PAMPs, DAMPs, LPS, ATP, and asbestos induce mitochondrial dysfunction, ROS production, and cardiolipin externalization [12, 13, 33, 34, 57, 64]. Hyperglycemia leads to an increase in mtROS generation which contributes significantly to cardiac fibrosis in DCM [33, 67, 68]. Altered ROS concentrations are detected by thioredoxin interacting protein (TXNIP) causing it to dissociate from thioredoxin (TRX) and to bind to NLRP3, activating the NLRP3 inflammasome complex [11, 30, 34, 69,70,71]. Similar to potassium efflux, ROS can promote the interaction between Nek7 and NLRP3, which recruits the ASC domain subsequently, inducing NLRP3 assembly and oligomerization [13, 72].
Lysosomal damage
Formation of intracellular crystalline or granular structures following phagocytosis of foreign and self-derived particles, including monosodium urate, silica, amyloid-β (Aβ), cholesterol, and uric acid crystals causes lysosomal damage and rupture, leading to the release of its contents such as tissue protease B [11, 33, 34, 73,74,75,76]. Such lysosomal rupture also releases cathepsins that can activate the NLRP3 inflammasome [11, 34]. Cathepsin B, a lysosomal enzyme, is required for IL-1β release; it is an important mediator of NLRP3 activation promoting CVB3-induced myocarditis through pyroptosis [11, 12, 77].
Miscellaneous stimuli
Golgi apparatus and ER stress also contribute to NLRP3 inflammasome activation [11, 33]. ER-Golgi vesicle trafficking is suggested to be involved in NLRP3 inflammasome activation as the disruption of the ER-Golgi trafficking inhibited NLRP3 inflammasome assembly and caspase-1 activation. Furthermore, mitochondrial clustering around the Golgi was observed to accompany NLRP3 activation; so, the dispersed trans-Golgi network (dTGN) also provides a scaffold for NLRP3 inflammasome aggregation and activation [11, 78, 79].
Noncoding RNAs, mainly microRNAs (miRNAs), such as miR-223, miR-495-3p, miR-145a-5p, miR-30c-5p, and miR-9-5p, influence NLRP3 activation either by modulating the transcription process or targeting the upstream regulators of its activation [11, 80,81,82,83,84]. In addition, long noncoding RNAs (lncRNAs), such as lncRNA MEG3, can enhance NLRP3 expression by sponging miR-223 [11, 85].
Intracellular pH, metabolic alterations, and autophagy dysfunction have also been identified to be involved in NLRP3 activation [29, 33, 34, 36].
Pyroptosis
Pyroptosis is a kind of programmed cell death, characterized by pore formation in the plasma membrane resulting in persistent cell swelling and osmotic rupture, and the discharge of pro-inflammatory factors [12, 33, 86, 87].
In pyroptosis, the inflammasome serves as a mediator [12]. Concomitant with the cleavage of IL‑1β and IL-18, the classical pyroptotic pathway also results in the cleavage of GSDMD into two parts (NT-GSDMD and CT-GSDMD) by the activated caspase-1. The cleaved lipophilic N-terminal fragment binds to membrane phospholipids and forms GSDMD micropores by oligomerizing into cell membranes, causing cell rupture and the release of cytokines, including IL-1β and IL-18, leading to an inflammatory cascade and cell death eventually [11, 12, 33, 88,89,90]. Cleaved GSDMD also facilitates potassium efflux and activation of canonical NLRP3 signaling [13, 56].
In the non-canonical activation pathway of the NLRP3 inflammasome, at the oligomerization step, LPS released by Gram-negative bacteria triggers the activation of caspase-11 that induces the nonclassical pyroptotic pathway by cleaving GSDMD [13, 29, 33, 91,92,93].
Diabetic cardiomyopathy
Diabetes mellitus (DM) is a serious public health issue worldwide and one of the fastest-growing health emergencies in the 21st century [94, 95]. Diabetic cardiomyopathy (DCM), the most common diabetic complication, remains the leading cause of morbidity and mortality in chronic diabetic patients [94,95,96,97,98]. About 10–40% of patients with cardiomyopathy-related heart failure have diabetes [97, 99].
DCM is a chronic low-grade inflammatory cardiovascular disorder characterized by functional, structural, and metabolic changes of the heart resulting from the switch of cardiac energy substrates’ supply to free fatty acids (FFAs) in the absence of CAD, hypertension, valvular and congenital heart disease, and other conventional risk factors for myocardial remodeling [94, 96, 100, 101].
DCM induces myocardial fibrosis and hypertrophy, followed by cardiac remodeling and early left ventricular diastolic dysfunction, leading to systolic dysfunction and ultimately to heart failure [94,95,96, 102]. With the fast global spread of DM, the prevalence of DCM is increasing accordingly; the incidence of DCM is about 1% and 17% in community-based populations and in diabetic patients, respectively [98, 103].
Myocardial inflammation is implicated as a key process in the multifactorial pathogenesis of DCM, with the cardiac inflammasome complexes being the key inducers of inflammation [94, 95, 104]. Insulin resistance in DM is closely associated with inflammation [95]. Major inducers of the pro-inflammatory state in the diabetic heart include hyperglycemia, hyperlipidemia (high levels of saturated FFAs), circulating cytokines, ROS, angiotensin II (AgII), endothelin-1, and insulin resistance [96, 104,105,106,107]. Several conserved mutual signal transduction pathways are abnormally regulated in cardiomyocytes due to dysregulation of glucose and lipid metabolism and DCM is closely associated with abnormal myocardial energy metabolism [94, 96, 108, 109].
NLRP3 inflammasome activation significantly contributes to the development of DCM [95, 98]. NLRP3 inflammasome-mediated pyroptosis of cardiomyocytes is a key participant in this process [98, 110]. Elevated expression of NLRP3 inflammasome components and upregulation of NLRP3 activation and cardiac pyroptosis have been shown in the heart tissues of diabetic patients compared with their nondiabetic counterparts [95, 98]. Through silencing the NLRP3 gene, Luo et al. showed for the first time that NLRP3 contributes to the development of DCM [111]. The NLRP3 inflammasome is expressed across a wide range of cells, including macrophages, cardiofibroblasts (CFs), and cardiomyocytes [18, 104, 112]. Under a hyperglycemic environment, the myocardium induces NLRP3 inflammasome activation, ultimately leading to cardiomyocyte pyroptosis [95, 98, 101].
In addition, peroxisome proliferator-activated receptors (PPARs) and sirtuins (Sirts) also have a role in the myocardial substrate switch and can limit the inflammatory response by targeting different components of the NLRP3 inflammasome-dependent signal transduction pathways [104].
Targeting the inflammasome itself or any of the mediators along its activation pathway can be beneficial in slowing down the progression of DCM [97, 101]. The massive amount of research identifying the mechanisms involved in DCM development provides new potential therapeutic targets for developing specialized drugs to treat DCM [96, 97, 101] (Table 1). Although several substances have been introduced in different studies as potential drugs that can be helpful in treating DCM, none of them have been actively explored in the human population or in practice and thus, no such drug has been employed or developed, so far [95, 104].
Non-coding RNAs: possible treatments and therapeutic targets for DCM
MicroRNAs
miR-223
Xu et al. carried out in vitro and in vivo experiments investigating the role of miRNA-223 (miR-223) and the effects of miR-223 inhibition in DCM. They found that the level of miR-223 was significantly increased in high-glucose (HG)-induced H9c2 cells and also in the streptozotocin (STZ)-induced rat models of DCM. They discovered that after the introduction of the miR-223 inhibitor, the increase in the expression of the NLRP3 inflammasome-related gene (NLRP3), fibrosis-related genes (collagen-1 and collagen-3), and apoptosis-related genes (caspase-3 and bax) and the decrease in the expression of the apoptosis-related gene (bcl2) were partially or completely reversed and the NLRP3 inflammasome activation, myocardial fibrosis, and apoptosis were markedly attenuated in both HG-induced H9c2 cells and the DCM rat model. They also showed that inhibition of miR-223 prominently relieved cardiac dysfunction and significantly reduced blood glucose in DCM rat model. Evaluating and comparing the myocardial tissues of DCM rat model with those of the miR-223 inhibitor-treated group, they found that the miR-223 inhibitor significantly alleviated cardiac fibrosis and could improve the morphological structure in myocardial tissues of the DCM model rats [113].
Zhao et al. discovered that along with the significantly increased expression level of miR-223–3p, SPI1 (a transcription factor) was significantly downregulated in HG-treated H9c2 cardiomyocytes. They found that miR-223–3p knockdown in HG-treated cardiomyocytes reversed the decrease in SPI1 levels. Also, the inflammasome/pyroptosis-associated proteins NLRP3, GSDMD-N, caspase-1, and IL-1β were downregulated following inhibition of miR-223–3p expression. Through further investigation, they confirmed that inhibiting the expression of miR-223–3p can relieve cardiomyocyte pyroptosis by up-regulating SPI1. Thus, they proposed a new signaling pathway based on their findings: miR-223–3p↑ → SPI1↓ → caspase-1↑ → IL-1β and other pyroptosis-associated proteins↑ → pyroptosis↑. These findings propose miR-223–3p inhibition as a potential therapeutic option for DCM [114].
miR-21-3p
In 2021, Shi et al. investigated the role of miR-21–3p and its target androgen receptor (AR) in diabetic cardiac fibrosis. They found that miR-21–3p was significantly upregulated in STZ-induced diabetic cardiac fibrotic tissue and HG-induced cardiac fibroblasts, while at the same time, the AR was downregulated. Overexpression of miR-21–3p resulted in reduced expression of AR in cardiac fibroblasts, whereas inhibition of miR-21–3p caused an elevation in the expression of AR in cardiac fibroblasts. Also, the levels of NLRP3 and caspase-1 and collagen I expression were reduced in cardiac fibroblasts transfected by miR-21–3p inhibitors. These findings demonstrated that AR is a direct target of miR-21–3p in modulating cardiac fibroblast pyroptosis in the diabetic heart. Furthermore, they discovered that AR up-regulation reduced fibrosis markers, collagen I expression, and NLRP3 and caspase-1 levels in HG-treated cardiac fibroblasts. Thus, considering that miR-21–3p aggravates diabetic cardiac pyroptosis and fibrosis by suppressing AR expression, they postulated that over-expression of AR or downregulation of miR-21–3p reduces pyroptosis via the AR/caspase-1 pathway [115].
miR-9
Through bioinformatics analysis and target validation assays, Jeyabal et al. discovered that miRNA-9 (miR-9) inhibits ELAVL1 expression in human cardiomyocytes. They found that ELAVL1 expression was augmented in human diabetic hearts and HG-treated human cardiomyocytes. Through further investigation, they showed that ELAVL1 knockdown nullified TNF-α induced canonical pyroptosis via NLRP3, caspase-1, and IL-1β suppression, while it did not affect LPS-induced noncanonical pyroptosis. Moreover, they found that inhibition of miR-9 upregulates ELAVL1 expression and activates caspase-1. Further evaluation showed that miR-9 expression was significantly downregulated in human diabetic heart tissues compared to their non-diabetic healthy counterparts. Also, blocking miR-9 expression in human cardiomyocytes resulted in an increase in the expression of caspase-1 and IL-1β, while transfecting cardiomyocytes with miR-9 mimics caused downregulation of these proteins and inhibited cardiac pyroptosis. These findings highlight targeting miR-9/ELAVL1 expression as a novel potential therapeutic option in preventing cardiac pyroptosis in DCM [116].
miR-200a-3p
In recent studies, a significant decrease in miR-200a-3p was observed in cardiomyocytes of myocardial injury models [117]. Additionally, lower expression of miR-200a-3p in the serum was associated with necrosis and inflammation in intestinal epithelial cells of necrotizing enterocolitis patients [118]. Based on these findings, You et al. investigated the role and mechanism of miR-200a-3p in the pathological process of DCM. They observed a significant decrease in miR-200a-3p expression in myocardial tissues of DCM-induced myocardial injury model mice. They also showed that overexpression of miR-200a-3p notably led to the alleviation of myocardial injury and cardiac dysfunction, reduced cardiac fibrosis, inflammation, and cardiomyocyte apoptosis, enhanced autophagy, and improved LV function in DCM mice. The researchers discovered that upregulation of miR-200a-3p ameliorates apoptosis by affecting various cellular processes. For instance, it downregulates FOXO3, inactivates Mst1 transcription, enhances SIRT3 expression and AMPK phosphorylation levels, and promotes autophagy. This ultimately slows down the progression of DCM. These findings suggest that overexpressing miR-200a-3p alleviates DCM in mice by regulating autophagy and inhibiting apoptosis through the FOXO3/Mst1/SIRT3/AMPK axis, which provides a new therapeutic approach for the prevention and treatment of DCM [119].
miR-18a‐3p
Yang et al. studied the effects of Berberine (BBR), a natural compound extracted from a Chinese herb with application in treating inflammatory disorders and DM-induced cardiovascular injury, on DCM. They found that BBR treatment effectively improved cardiac dysfunction and fibrosis in the rat model of DCM. BBR markedly alleviated DCM by limiting GSDMD‐driven pyroptosis via inhibiting IL‐1β secretion and GSDMD expression at the post‐transcriptional level in HG‐stimulated H9c2 cells and diabetic rats. They discovered that the inhibitory effects of BBR on GSDMD transcription are induced by the upregulation of miR‐18a‐3p expression in vivo and in vitro, confirming the direct negative regulatory effect of miR‐18a‐3p on GSDMD expression. They also indicated that miR‐18a‐3p reduced the ROS levels and rate of cell death by suppressing GSDMD production in vitro and in vivo. Moreover, it was confirmed that BBR regulates the expression levels of miR‐18a‐3p by enhancing the activation of the miR‐18a‐3p promoter. Collectively, these findings suggest that BBR alleviates DCM by inhibiting miR‐18a‐3p‐mediated GSDMD activation and pore formation through the NLRP3 inflammasome pathway. Thus, BBR can be considered a potential novel therapy for treating DCM [120].
LncRNAs
LncRNA-MALAT1
Shi et al. carried out an experiment investigating the role of metastasis-associated lung adenocarcinoma transcript 1 lncRNA (lncRNA-MALAT1) in cardiomyocyte pyroptosis in DCM. They demonstrated that silencing MALAT1, which was overexpressed in myocardial tissue of diabetic mice and HG-induced cardiomyocytes, improves cardiac function and morphology in HG-induced cardiomyocytes and STZ-induced diabetic mice. They also showed that inhibition of MALAT1 attenuates cardiomyocyte pyroptosis through the NLRP3 pathway by inducing a significant decrease in caspase-1, IL 1β, and IL-18 protein levels. Hence, they suggested that MALAT1 could be a new therapeutic target for DCM [121].
Melatonin
Che et al. found that along with the elevated levels of lncR-MALAT1, miRNA-141 (miR-141) was downregulated in the myocardium of DM mice and in HG-treated CFs. Investigating the role of melatonin in DCM, they reported that melatonin significantly restored cardiac hypertrophy, normalized the structural anomalies, ameliorated the damage to cellular organelles by improving sarcomere arrangement and mitochondria morphology, decreased interstitial collagen production, and finally alleviated cardiac dysfunction significantly in STZ-induced DM mice by suppressing the upregulation of lncR-MALAT1 and by upregulating miR-141 expression. These findings confirmed miR-141 as a downstream target of lncR-MALAT1, which is regulated by melatonin. Through further investigation, they signified lncR-MALAT1 as a competitive endogenous RNA (ceRNA) that can sponge miR-141 to limit its functional availability. Finally, they verified that melatonin exerts its antifibrotic effect by inhibiting lncR-MALAT1/miR-141-mediated NLRP3 inflammasome activation and TGF-β1/Smads signaling, making it a potentially effective agent for treating DCM [122].
Pomegranate Peel extract
Abo-Saif et al. conducted a study on the effects of pomegranate peel extract (PPE) on DCM in rats. They showed that prophylactic administration of PPE to diabetic rats significantly increased their survival rate, ameliorated cardiac hypertrophy, and improved their lipid profile. Additionally, it decreased serum cardiac troponin-1 levels and reduced lipid peroxidation in the rats’ myocardial tissue. They also found that the levels of lncRNA-MALAT1, IL-1β, and pyroptosis-related genes (NLRP3 and caspase-1) significantly decreased in the cardiac tissue of diabetic rats following treatment with PPE. In addition, histopathological examination of their cardiac tissues showed preservation of normal myocardial structures and a marked reduction in fibrosis. They concluded that PPE exhibits a cardioprotective effect against the development of DCM in diabetic rats. This effect is attributed to its exceptional antioxidant, anti-inflammatory, and anti-fibrotic properties, as well as its ability to improve the lipid profile. This protective effect is probably achieved via the inhibition of pyroptosis (the NLRP3/caspase-1/IL-1β signaling pathway) and downregulation of lncRNA-MALAT1. These data show that PPE could be a promising therapy to prevent the development of DCM [123].
LncRNA GAS5
Xu et al. observed a significant decrease in the lncRNA GAS5 levels in the heart tissues of mice with DCM. They also found that overexpression of GAS5 improved cardiac function and alleviated myocardial hypertrophy in DCM mice. Additionally, upregulation of GAS5 markedly repressed the increased expression of NLRP3, caspase- 1, pro-caspase-1, IL-1β, and IL-18 seen in cardiac tissues of DCM mice and HG-treated HL-1 cells (cardiac muscle cell line). Overexpressing GAS5 also suppressed caspase-1 activity and the release of lactate dehydrogenase (LDH) in the HG-treated HL-1 cells. Furthermore, they discovered that the expression of miR-34b-3p was increased in the heart tissues of DCM mice as well as in the HG-induced HL-1 cells, accompanied by a concomitant decrease in the expression of aryl hydrocarbon receptor (AHR). Through further investigation, they demonstrated that overexpression of GAS5 inhibits NLRP3 inflammasome activation-induced pyroptosis by regulating the miR-34b-3p/AHR axis via inhibition of miR-34b-3p expression. Collectively, these findings elucidate that lncRNA GAS5 acts as a ceRNA to enhance AHR expression by sponging miR-34b-3p, thus repressing NLRP3 inflammasome activation-mediated pyroptosis. This data provides lncRNA GAS5 as a novel valuable target for DCM treatment [124].
TINCR LncRNA
N6-methyladenosine (m6A) is an important epigenetic regulator of RNAs, including lncRNAs. Methyltransferase-like 14 (METTL14) is an m6A writer protein known to widely participate in the pathogenesis of major diseases, such as CVDs [125]. Since the underlying regulatory mechanism of m6A and METTL14 in DCM was not well established, Meng et al. aimed to uncover the role of METTL14-mediated m6A modification in pyroptosis and the progression of DCM throughout their study. They found that METTL14 was downregulated in HG-treated cardiomyocytes and myocardium tissues of STZ-induced DCM rat models. They revealed that METTL14 increased the m6A methylation level of the TINCR gene, suppressing its expression and reducing NLRP3 mRNA stability resulting in decreased expression of NLRP3. As a result, METTL14 prevented the occurrence of cardiomyocyte pyroptosis and remarkably suppressed DCM progression. This newly discovered regulatory mechanism not only enhances our understanding of the epigenetic regulation of pyroptosis in DCM progression but also holds promise for the development of novel therapeutic strategies to combat DCM [126].
LncRNA homeobox transcript antisense RNA (HOTAIR)
Since lncRNAs play important roles in the pathogenesis of DCM, Xiong et al. attempted to clarify the function of lncRNA homeobox transcript antisense RNA (HOTAIR) in the pyroptosis of cardiomyocytes and its underlying molecular mechanism in DCM. They reported that SIRT3 and HOTAIR were significantly downregulated in HG-induced H9C2 cells. Additionally, they found that HOTAIR overexpression alleviated pyroptosis and suppressed the inflammatory response in HG-treated cardiomyocytes. Upon further investigation, they unveiled that HOTAIR induced SIRT3 expression in HG-treated H9C2 cells by recruiting FUS (a multifunctional protein involved in multiple aspects of RNA metabolism, such as transcription regulation and alternative splicing), thereby attenuating pyroptosis. Moreover, they showed that SIRT3 knockdown notably reversed the suppressive effect of HOTAIR on HG-induced pyroptosis in cardiomyocytes, suggesting that SIRT3 acts as a downstream regulator of HOTAIR. Through these findings, researchers identified a vital and effective role for HOTAIR in DCM and strongly speculated that the HOTAIR/FUS/SIRT3 signaling pathway effectively influences the progression of DCM. They indicated that HOTAIR alleviates pyroptosis and the development of DCM, making it a potential marker for diagnosis and a novel therapeutic target for treating DCM [127].
LncRNA KCNQ1OT1
The lncRNA Kcnq1ot1, the full name of which is KCNQ1 overlapping transcript 1 [128], has been reported to play a significant role in the heart and participate in many CVDs, such as long QT syndrome and myocardial ischemia/reperfusion injury [129,130,131,132]. A recent study found that Kcnq1ot1 might influence cataract formation by acting as a ceRNA to regulate caspase-1 expression via sponging miR-214-3p [133]. Considering the still unknown expression and mechanisms of Kcnq1ot1 in DCM, Yang et al. conducted a study to clarify the effects of Kcnq1ot1 on cardiac pyroptosis in DCM and define its mechanisms. They found that Kcnq1ot1 and caspase-1 were overexpressed in the serum of diabetic patients, the cardiac tissue of diabetic mice, and HG-induced cardiomyocytes. They demonstrated that silencing Kcnq1ot1 reduced cell death, DNA fractionation, cytoskeletal structure abnormalities, and calcium overload in HG-induced cardiomyocytes, improved cardiac function and morphology in STZ-induced diabetic mice, and significantly inhibited pyroptosis of cardiomyocytes in vitro and in vivo. They discovered that silencing Kcnq1ot1 attenuates pyroptosis by targeting caspase-1 via miR-214-3p in cardiomyocytes. Thus, they elaborated for the first time on the downregulation of miR-214-3p in HG-treated cardiomyocytes and fully illuminated the effect of Kcnq1ot1 on pyroptosis in DCM via miR-214-3p and caspase-1. Therefore, their research suggests Kcnq1ot1 as a novel therapeutic target for DCM [134].
PIWI-interacting RNAs (piRNAs)
piR112710
PIWI-interacting RNAs (piRNAs) are a group of small non-coding RNAs (ncRNAs) composed of 26–32 nucleotides. They are termed piRNAs due to their interaction with PIWI family proteins. piRNAs are abundantly expressed in the myocardial tissue and have been reported to regulate cardiomyocyte apoptosis and hypertrophic cardiomyopathy and to improve heart failure. In their attempt to identify the functions of piRNAs in regulating pyroptosis in DCM, Jiao et al. performed a combination of piRNA microarray expression analysis and transcriptome sequencing (RNA-seq). They identified piR112710 to be involved in DCM. The researchers then investigated the effects of piR112710 in DCM in db/db mice and HG + Palmitate-treated neonatal mice cardiomyocytes. They found that piR112710 was downregulated in HG + Palmitate-treated cardiomyocytes. Also, piR112710 significantly ameliorated cardiac dysfunction in db/db mice, characterized by downregulation of inflammatory factors and pyroptosis-associated proteins (NLRP3, ASC, TXNIP, caspase-1, and GSDMD-N), reduced fibrosis, enhanced mitochondrial respiratory functions of the myocardium, and improved echocardiography. Moreover, supplementation with piR112710 significantly decreased oxidative stress and mitochondrial dysfunction and reduced the expression levels of pyroptosis-associated proteins and inflammatory factors in cultured HG + palmitate-induced neonatal mice cardiomyocytes. Through further investigation, the researchers discovered that piR112710 prevents pyroptosis by inhibiting the TXNIP/NLRP3 axis through the effective elimination of ROS and reducing the expression levels of the pyroptosis-associated proteins NLRP3, ASC, TXNIP, caspase-1, and GSDMD-N, which subsequently alleviate mitochondrial dysfunction and reduce IL-18 and IL-1β levels. Taken together, these findings revealed that the piR112710 protects against DCM by inhibiting the TXNIP/NLRP3-mediated pyroptosis, thereby providing a useful and novel therapeutic target for the treatment of DCM [135].
Conclusions
Cardiomyopathies are a heterogeneous group of disorders that can lead to fulminant heart failure and sudden cardiac death [1, 3]. From 2010 to 2018, the prevalence of all types of cardiomyopathies had an upward trend [6] and in 2020, six million people were affected by cardiomyopathy and myocarditis worldwide [5, 7]. It is noteworthy that up to 40% of patients with cardiomyopathy-related heart failure have diabetes [97, 99]. With the increasing global trend of DM, the prevalence of DCM is increasing accordingly [98, 103] and it remains the leading cause of morbidity and mortality in chronic diabetic patients [94,95,96,97,98]. NLRP3 inflammasome significantly contributes to the development and pathological progression of DCM [95, 98, 111].
Targeting the inflammasome itself or the mediators along its activation pathway provides new potential therapeutic targets for developing drugs for DCM treatment [96, 97, 101]. The massive amount of research identifying the underlying mechanisms of NLRP3 inflammasome in DCM has led to the emergence of novel therapies beneficial in slowing down the progression of DCM [20, 21, 23, 97, 101].
In this comprehensive review, we sought to provide a summary of the non-coding RNAs with potential therapeutic effects targeting NLRP3 inflammasome signaling in DCM. We hope that this general overview helps set a foundation for future research concerning the development of new therapies for DCM.
Data availability
Not applicable.
References
Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D, et al. Contemporary definitions and classification of the cardiomyopathies. Circulation. 2006;113(14):1807–16.
Elliott P, Andersson B, Arbustini E, Bilinska Z, Cecchi F, Charron P, et al. Classification of the cardiomyopathies: a position statement from the European society of cardiology working group on myocardial and pericardial diseases. Eur Heart J. 2007;29(2):270–6.
Maron BJ, Epstein SE, Roberts WC. Causes of sudden death in competitive athletes. J Am Coll Cardiol. 1986;7(1):204–14.
McKenna WJ, Maron BJ, Thiene G. Classification, epidemiology, and global burden of cardiomyopathies. Circ Res. 2017;121(7):722–30.
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS et al. Heart disease and stroke Statistics—2022 update: A report from the American heart association. Circulation. 2022;145(8).
Brownrigg JR, Leo V, Rose J, Low E, Richards S, Carr-White G, et al. Epidemiology of cardiomyopathies and incident heart failure in a population-based cohort study. Heart. 2022;108(17):1383–91.
Vos T, Lim SS, Abbafati C, Abbas KM, Abbasi M, Abbasifard M, et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the global burden of disease study 2019. Lancet. 2020;396(10258):1204–22.
Arbustini E, Narula N, Tavazzi L, Serio A, Grasso M, Favalli V, et al. The MOGE(S) classification of cardiomyopathy for clinicians. J Am Coll Cardiol. 2014;64(3):304–18.
Jacoby D, McKenna WJ. Genetics of inherited cardiomyopathy. Eur Heart J. 2012;33(3):296–304.
Watkins H, Ashrafian H, Redwood C. Inherited cardiomyopathies. N Engl J Med. 2011;364(17):1643–56.
Wang Y, Liu X, Shi H, Yu Y, Yu Y, Li M, et al. NLRP3 inflammasome, an immune-inflammatory target in pathogenesis and treatment of cardiovascular diseases. Clin Transl Med. 2020;10(1):91–106.
Zheng Y, Xu L, Dong N, Li F. NLRP3 inflammasome: the rising star in cardiovascular diseases. Front Cardiovasc Med. 2022;9.
Pellegrini C, Martelli A, Antonioli L, Fornai M, Blandizzi C, Calderone V. NLRP3 inflammasome in cardiovascular diseases: pathophysiological and Pharmacological implications. Med Res Rev. 2021;41(4):1890–926.
Pereira N, de Queiroga S, Nunes TBD, Andrade DF, de Nascimento C, Do-Valle-Matta MSL. Innate immune receptors over expression correlate with chronic Chagasic cardiomyopathy and digestive damage in patients. PLoS Negl Trop Dis. 2018;12(7):e0006589.
Zeng C, Duan F, Hu J, Luo B, Huang B, Lou X, et al. NLRP3 inflammasome-mediated pyroptosis contributes to the pathogenesis of non-ischemic dilated cardiomyopathy. Redox Biol. 2020;34:101523.
Miteva K, Pappritz K, Sosnowski M, El-Shafeey M, Müller I, Dong F, et al. Mesenchymal stromal cells inhibit NLRP3 inflammasome activation in a model of coxsackievirus B3-induced inflammatory cardiomyopathy. Sci Rep. 2018;8(1):2820.
Zhou W, Chen C, Chen Z, Liu L, Jiang J, Wu Z, et al. NLRP3: A novel mediator in cardiovascular disease. J Immunol Res. 2018;2018:1–8.
Bracey NA, Beck PL, Muruve DA, Hirota SA, Guo J, Jabagi H, et al. The Nlrp3 inflammasome promotes myocardial dysfunction in structural cardiomyopathy through interleukin-1β. Exp Physiol. 2013;98(2):462–72.
Abbate A. The heart on fire: inflammasome and cardiomyopathy. Exp Physiol. 2013;98(2):385–385.
Wu J, Dong E, Zhang Y, Xiao H. The role of the inflammasome in heart failure. Front Physiol. 2021;12.
Lara-Reyna S, Caseley EA, Topping J, Rodrigues F, Jimenez Macias J, Lawler SE et al. Inflammasome activation: from molecular mechanisms to autoinflammation. Clin Transl Immunology. 2022;11(7).
Martinon F, Burns K, Tschopp J. The inflammasome. Mol Cell. 2002;10(2):417–26.
Chen G, Chelu MG, Dobrev D, Li N. Cardiomyocyte inflammasome signaling in cardiomyopathies and atrial fibrillation: mechanisms and potential therapeutic implications. Front Physiol. 2018;9.
Kanneganti T. The inflammasome: firing up innate immunity. Immunol Rev. 2015;265(1):1–5.
Amin J, Boche D, Rakic S. What do we know about the inflammasome in humans? Brain Pathol. 2017;27(2):192–204.
Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/Reperfusion injury. Circulation. 2011;123(6):594–604.
Sandanger Ø, Ranheim T, Vinge LE, Bliksøen M, Alfsnes K, Finsen AV, et al. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia–reperfusion injury. Cardiovasc Res. 2013;99(1):164–74.
Doyle S, Ozaki E, Campbell M. Targeting the NLRP3 inflammasome in chronic inflammatory diseases: current perspectives. J Inflamm Res. 2015;15.
Xu X, Wu X, Yue G, An Q, Lou J, Yang X, et al. The role of Nod-like receptor protein 3 inflammasome activated by ion channels in multiple diseases. Mol Cell Biochem. 2023;478(6):1397–410.
An N, Gao Y, Si Z, Zhang H, Wang L, Tian C et al. Regulatory mechanisms of the NLRP3 inflammasome, a novel Immune-Inflammatory marker in cardiovascular diseases. Front Immunol. 2019;10.
Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–32.
Wang L, Hauenstein AV. The NLRP3 inflammasome: mechanism of action, role in disease and therapies. Mol Aspects Med. 2020;76:100889.
Fan J, Ren M, Adhikari BK, Wang H, He Y. The NLRP3 inflammasome as a novel therapeutic target for cardiac fibrosis. J Inflamm Res. 2022;15:3847–58.
Zhang X, Wang Z, Zheng Y, Yu Q, Zeng M, Bai L, et al. Inhibitors of the NLRP3 inflammasome pathway as promising therapeutic candidates for inflammatory diseases (Review). Int J Mol Med. 2023;51(4):35.
Kim YK, Shin JS, Nahm MH. NOD-Like receptors in infection, immunity, and diseases. Yonsei Med J. 2016;57(1):5.
Swanson KV, Deng M, Ting JPY. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–89.
Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20(13):3328.
Srinivasula SM, Poyet JL, Razmara M, Datta P, Zhang Z, Alnemri ES. The PYRIN-CARD protein ASC is an activating adaptor for Caspase-1. J Biol Chem. 2002;277(24):21119–22.
Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, et al. A novel heterodimeric cysteine protease is required for interleukin-1βprocessing in monocytes. Nature. 1992;356(6372):768–74.
Guo H, Callaway JB, Ting JPY. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21(7):677–87.
Duncan DJ, Yang Z, Hopkins PM, Steele DS, Harrison SM. TNF-α and IL-1β increase Ca2 + leak from the sarcoplasmic reticulum and susceptibility to arrhythmia in rat ventricular myocytes. Cell Calcium. 2010;47(4):378–86.
Luo B, Wang F, Li B, Dong Z, Liu X, Zhang C et al. Association of nucleotide-binding oligomerization domain-like receptor 3 inflammasome and adverse clinical outcomes in patients with idiopathic dilated cardiomyopathy. Clin Chem Lab Med. 2013;51(7).
Chin LH, Hsu YJ, Hsu SC, Chen YH, Chang YL, Huang SM, et al. The regulation of NLRP3 inflammasome expression during the development of cardiac contractile dysfunction in chronic kidney disease. Oncotarget. 2017;8(69):113303–17.
Toldo S, Mezzaroma E, McGeough MD, Peña CA, Marchetti C, Sonnino C, et al. Independent roles of the priming and the triggering of the NLRP3 inflammasome in the heart. Cardiovasc Res. 2015;105(2):203–12.
Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, et al. Cutting edge: NF-κB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183(2):787–91.
Sutterwala FS, Ogura Y, Zamboni DS, Roy CR, Flavell RA. NALP3: a key player in caspase-1 activation. J Endotoxin Res. 2006;12(4):251–6.
Guarda G, Zenger M, Yazdi AS, Schroder K, Ferrero I, Menu P, et al. Differential expression of NLRP3 among hematopoietic cells. J Immunol. 2011;186(4):2529–34.
Franchi L, Eigenbrod T, Núñez G. Cutting edge: TNF-α mediates sensitization to ATP and silica via the NLRP3 inflammasome in the absence of microbial stimulation. J Immunol. 2009;183(2):792–6.
Xing Y, Yao X, Li H, Xue G, Guo Q, Yang G, et al. Cutting edge: TRAF6 mediates TLR/IL-1R Signaling–Induced nontranscriptional priming of the NLRP3 inflammasome. J Immunol. 2017;199(5):1561–6.
Vallabhapurapu S, Karin M. Regulation and function of NF-κB transcription factors in the immune system. Annu Rev Immunol. 2009;27(1):693–733.
Cornut M, Bourdonnay E, Henry T. Transcriptional regulation of inflammasomes. Int J Mol Sci. 2020;21(21):8087.
Dowling JK, O’Neill LAJ. Biochemical regulation of the inflammasome. Crit Rev Biochem Mol Biol. 2012;47(5):424–43.
Ulland TK, Ferguson PJ, Sutterwala FS. Evasion of inflammasome activation by microbial pathogens. J Clin Invest. 2015;125(2):469–77.
Zhao N, Li C, Di cui, Xu B. li. Recent advances in the NEK7-licensed NLRP3 inflammasome activation: mechanisms, role in diseases and related inhibitors. J Autoimmun. 2020;113:102515.
Gurung P, Anand PK, Malireddi RKS, Vande Walle L, Van Opdenbosch N, Dillon CP, et al. FADD and Caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes. J Immunol. 2014;192(4):1835–46.
Pellegrini C, Antonioli L, Lopez-Castejon G, Blandizzi C, Fornai M. Canonical and Non-Canonical activation of NLRP3 inflammasome at the crossroad between immune tolerance and intestinal inflammation. Front Immunol. 2017;8.
Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469(7329):221–5.
Karmakar M, Katsnelson MA, Dubyak GR, Pearlman E. Neutrophil P2X7 receptors mediate NLRP3 inflammasome-dependent IL-1β secretion in response to ATP. Nat Commun. 2016;7(1):10555.
Muñoz-Planillo R, Kuffa P, Martínez-Colón G, Smith BL, Rajendiran TM, Núñez G. K + Efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity. 2013;38(6):1142–53.
Bartlett R, Stokes L, Sluyter R. The P2X7 Receptor Channel: Recent Developments and the Use of P2X7 Antagonists in Models of Disease. Pharmacol Rev. 2014;66(3):638–75.
Hamon MA, Cossart P. K + Efflux is required for histone H3 dephosphorylation by Listeria monocytogenes Listeriolysin O and other Pore-Forming toxins. Infect Immun. 2011;79(7):2839–46.
Suresh R, Chandrasekaran P, Sutterwala FS, Mosser DM. Complement-mediated ‘bystander’ damage initiates host NLRP3 inflammasome activation. J Cell Sci. 2016;129(9):1928–39.
Lee GS, Subramanian N, Kim AI, Aksentijevich I, Goldbach-Mansky R, Sacks DB, et al. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2 + and cAMP. Nature. 2012;492(7427):123–7.
Nakahira K, Haspel JA, Rathinam VAK, Lee SJ, Dolinay T, Lam HC, et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12(3):222–30.
Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 2012;36(3):401–14.
Zhong Z, Liang S, Sanchez-Lopez E, He F, Shalapour S, Lin X, jia, et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature. 2018;560(7717):198–203.
Yue Y, Meng K, Pu Y, Zhang X. Transforming growth factor beta (TGF-β) mediates cardiac fibrosis and induces diabetic cardiomyopathy. Diabetes Res Clin Pract. 2017;133:124–30.
Sharma A, Tate M, Mathew G, Vince JE, Ritchie RH, de Haan JB. Oxidative stress and NLRP3-Inflammasome activity as significant drivers of diabetic cardiovascular complications: therapeutic implications. Front Physiol. 2018;9.
Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11(2):136–40.
Gross O, Thomas CJ, Guarda G, Tschopp J. The inflammasome: an integrated view. Immunol Rev. 2011;243(1):136–51.
Abais JM, Xia M, Zhang Y, Boini KM, Li PL. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector?? Antioxid Redox Signal. 2015;22(13):1111–29.
He Y, Zeng MY, Yang D, Motro B, Núñez G. NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature. 2016;530(7590):354–7.
Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9(8):847–56.
Maejima I, Takahashi A, Omori H, Kimura T, Takabatake Y, Saitoh T, et al. Autophagy sequesters damaged lysosomes to control lysosomal biogenesis and kidney injury. EMBO J. 2013;32(17):2336–47.
Otsuki T, Holian A, Di Gioacchino M. Immunological effects of environmental factors: focus on the fibrous and particulated materials. J Immunol Res. 2014;2014:1–1.
Wang Y, Qin X, Paudel HK. Amyloid Β peptide promotes lysosomal degradation of clusterin via sortilin in hippocampal primary neurons. Neurobiol Dis. 2017;103:78–88.
Wang Y, Jia L, Shen J, Wang Y, Fu Z, Su S et al. an,. Cathepsin B aggravates coxsackievirus B3-induced myocarditis through activating the inflammasome and promoting pyroptosis. PLoS Pathog. 2018;14(1):e1006872.
Hong S, Hwang I, Gim E, Yang J, Park S, Yoon SH, et al. Brefeldin A–sensitive ER-Golgi vesicle trafficking contributes to NLRP3‐dependent caspase‐1 activation. FASEB J. 2019;33(3):4547–58.
Chen J, Chen ZJ. PtdIns4P on dispersed trans-Golgi network mediates NLRP3 inflammasome activation. Nature. 2018;564(7734):71–6.
Haneklaus M, Gerlic M, Kurowska-Stolarska M, Rainey AA, Pich D, McInnes IB, et al. Cutting edge: miR-223 and EBV miR-BART15 regulate the NLRP3 inflammasome and IL-1β production. J Immunol. 2012;189(8):3795–9.
Zhou T, Xiang DK, Li SN, Yang LH, Gao LF, Feng C. MicroRNA-495 ameliorates cardiac microvascular endothelial cell injury and inflammatory reaction by suppressing the NLRP3 inflammasome signaling pathway. Cell Physiol Biochem. 2018;49(2):798–815.
Zhong W, Li B, Xu Y, Yang P, Chen R, Wang Z, et al. Hypermethylation of the Micro-RNA 145 promoter is the key regulator for NLRP3 Inflammasome-Induced activation and plaque formation. JACC Basic Transl Sci. 2018;3(5):604–24.
Li P, Zhong X, Li J, Liu H, Ma X, He R, et al. MicroRNA-30c-5p inhibits NLRP3 inflammasome-mediated endothelial cell pyroptosis through FOXO3 down-regulation in atherosclerosis. Biochem Biophys Res Commun. 2018;503(4):2833–40.
Wang Y, Han Z, Fan Y, Zhang J, Chen K, Gao L, et al. MicroRNA-9 inhibits NLRP3 inflammasome activation in human atherosclerosis inflammation cell models through the JAK1/STAT signaling pathway. Cell Physiol Biochem. 2017;41(4):1555–71.
Zhang Y, Liu X, Bai X, Lin Y, Li Z, Fu J et al. Melatonin prevents endothelial cell pyroptosis via regulation of long noncoding RNA MEG3/miR-223/NLRP3 axis. J Pineal Res. 2018;64(2).
Kovacs SB, Miao EA, Gasdermins. Effectors of pyroptosis. Trends Cell Biol. 2017;27(9):673–84.
Yu P, Zhang X, Liu N, Tang L, Peng C, Chen X. Pyroptosis: mechanisms and diseases. Signal Transduct Target Ther. 2021;6(1):128.
Burdette BE, Esparza AN, Zhu H, Wang S. Gasdermin D in pyroptosis. Acta Pharm Sin B. 2021;11(9):2768–82.
Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature. 2016;535(7610):111–6.
Zuo Y, Chen L, Gu H, He X, Ye Z, Wang Z, et al. GSDMD-mediated pyroptosis: a critical mechanism of diabetic nephropathy. Expert Rev Mol Med. 2021;23:e23.
Baker PJ, Boucher D, Bierschenk D, Tebartz C, Whitney PG, D’Silva DB, et al. NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase‐5. Eur J Immunol. 2015;45(10):2918–26.
Kayagaki N, Stowe IB, Lee BL, O’Rourke K, Anderson K, Warming S, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015;526(7575):666–71.
Shi J, Zhao Y, Wang Y, Gao W, Ding J, Li P, et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature. 2014;514(7521):187–92.
Chen MY, Meng XF, Han YP, Yan JL, Xiao C, Qian LB. Profile of crosstalk between glucose and lipid metabolic disturbance and diabetic cardiomyopathy: inflammation and oxidative stress. Front Endocrinol (Lausanne). 2022;13.
Cai Z, Yuan S, Luan X, Feng J, Deng L, Zuo Y et al. Pyroptosis-Related inflammasome pathway: A new therapeutic target for diabetic cardiomyopathy. Front Pharmacol. 2022;13.
Wenzl FA, Ambrosini S, Mohammed SA, Kraler S, Lüscher TF, Costantino S et al. Inflammation in metabolic cardiomyopathy. Front Cardiovasc Med. 2021;8.
Jia Y, Li D, Yu J, Jiang W, Liao X, Zhao Q. Potential diabetic cardiomyopathy therapies targeting pyroptosis: A mini review. Front Cardiovasc Med. 2022;9.
Ding K, Song C, Hu H, Yin K, Huang H, Tang H. The role of NLRP3 inflammasome in diabetic cardiomyopathy and its therapeutic implications. Oxid Med Cell Longev. 2022;2022:1–19.
Paolillo S, Marsico F, Prastaro M, Renga F, Esposito L, De Martino F, et al. Diabet Cardiomyopathy Heart Fail Clin. 2019;15(3):341–7.
Dillmann WH. Diabetic cardiomyopathy. Circ Res. 2019;124(8):1160–2.
Zhang L, Ai C, Bai M, Niu J, Zhang Z. NLRP3 inflammasome/pyroptosis: A key driving force in diabetic cardiomyopathy. Int J Mol Sci. 2022;23(18):10632.
Braunwald E, Cardiomyopathies. Circ Res. 2017;121(7):711–21.
Dandamudi S, Slusser J, Mahoney DW, Redfield MM, Rodeheffer RJ, Chen HH. The prevalence of diabetic cardiomyopathy: A Population-Based study in olmsted County, Minnesota. J Card Fail. 2014;20(5):304–9.
Fuentes-Antrás J, Ioan AM, Tuñón J, Egido J, Lorenzo Ó. Activation of Toll-Like receptors and inflammasome complexes in the diabetic Cardiomyopathy-Associated inflammation. Int J Endocrinol. 2014;2014:1–10.
Palomer X, Salvadó L, Barroso E, Vázquez-Carrera M. An overview of the crosstalk between inflammatory processes and metabolic dysregulation during diabetic cardiomyopathy. Int J Cardiol. 2013;168(4):3160–72.
Lorenzo O, Ramírez E, Picatoste B, Egido J, Tuñón J. Alteration of energy substrates and ROS production in diabetic cardiomyopathy. Mediators Inflamm. 2013;2013:1–11.
Frati G, Schirone L, Chimenti I, Yee D, Biondi-Zoccai G, Volpe M, et al. An overview of the inflammatory signalling mechanisms in the myocardium underlying the development of diabetic cardiomyopathy. Cardiovasc Res. 2017;113(4):378–88.
Jia G, Whaley-Connell A, Sowers JR. Diabetic cardiomyopathy: a hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia. 2018;61(1):21–8.
Jia G, Hill MA, Sowers JR. Diabetic cardiomyopathy. Circ Res. 2018;122(4):624–38.
Xie Y, Huang Y, Ling X, Qin H, Wang M, Luo B. Chemerin/CMKLR1 Axis promotes inflammation and pyroptosis by activating NLRP3 inflammasome in diabetic cardiomyopathy rat. Front Physiol. 2020;11.
Luo B, Li B, Wang W, Liu X, Xia Y, Zhang C, et al. NLRP3 gene Silencing ameliorates diabetic cardiomyopathy in a type 2 diabetes rat model. PLoS ONE. 2014;9(8):e104771.
Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17(2):179–88.
Xu D, Zhang X, Chen X, Yang S, Chen H. Inhibition of miR-223 attenuates the NLRP3 inflammasome activation, fibrosis, and apoptosis in diabetic cardiomyopathy. Life Sci. 2020;256:117980.
Zhao S, Tan Y, Qin J, Xu H, Liu L, Wan H, et al. MicroRNA-223–3p promotes pyroptosis of cardiomyocyte and release of inflammasome factors via downregulating the expression level of SPI1 (PU.1). Toxicology. 2022;476:153252.
Shi P, Zhao XD, Shi KH, Ding XS, Tao H. MiR-21–3p triggers cardiac fibroblasts pyroptosis in diabetic cardiac fibrosis via inhibiting androgen receptor. Exp Cell Res. 2021;399(2):112464.
Jeyabal P, Thandavarayan RA, Joladarashi D, Suresh Babu S, Krishnamurthy S, Bhimaraj A, et al. MicroRNA-9 inhibits hyperglycemia-induced pyroptosis in human ventricular cardiomyocytes by targeting ELAVL1. Biochem Biophys Res Commun. 2016;471(4):423–9.
Chen ZQ, Zhou Y, Chen F, Huang JW, Li HL, Li T et al. miR-200a-3p attenuates coronary Microembolization-Induced myocardial injury in rats by inhibiting TXNIP/NLRP3-Mediated cardiomyocyte pyroptosis. Front Cardiovasc Med. 2021;8.
Liu Y, Wang Z, Huang H, Shou K. miR-200a-3p improves neonatal necrotizing Enterocolitis by regulating RIPK1. Am J Transl Res. 2021;13(11):12662–72.
You P, Chen H, Han W, Deng J. miR-200a-3p overexpression alleviates diabetic cardiomyopathy injury in mice by regulating autophagy through the FOXO3/Mst1/Sirt3/AMPK axis. PeerJ. 2023;11:e15840.
Yang L, Cheng CF, Li ZF, Huang XJ, Cai SQ, Ye SY, et al. Berberine blocks inflammasome activation and alleviates diabetic cardiomyopathy via the miR-18a-3p/Gsdmd pathway. Int J Mol Med. 2023;51(6):49.
Shi C, Wu L, Li L. LncRNA-MALAT 1 regulates cardiomyocyte scorching in diabetic cardiomyopathy by targeting NLRP3. Cell Mol Biol. 2022;67(6):213–9.
Che H, Wang Y, Li H, Li Y, Sahil A, Lv J, et al. Melatonin alleviates cardiac fibrosis via inhibiting LncRNA MALAT1/miR-141‐mediated NLRP3 inflammasome and TGF‐β1/Smads signaling in diabetic cardiomyopathy. FASEB J. 2020;34(4):5282–98.
Abo-Saif MA, Ragab AE, Ibrahim AO, Abdelzaher OF, Mehanyd ABM, Saber-Ayad M et al. Pomegranate Peel extract protects against the development of diabetic cardiomyopathy in rats by inhibiting pyroptosis and downregulating LncRNA-MALAT1. Front Pharmacol. 2023;14.
Xu Y, Fang H, Xu Q, Xu C, Yang L, Huang C. LncRNA GAS5 inhibits NLRP3 inflammasome activation-mediated pyroptosis in diabetic cardiomyopathy by targeting miR-34b-3p/AHR. Cell Cycle. 2020;19(22):3054–65.
Zhang BY, Han L, Tang YF, Zhang GX, Fan XL, Zhang JJ, et al. METTL14 regulates M6A methylation-modified primary miR-19a to promote cardiovascular endothelial cell proliferation and invasion. Eur Rev Med Pharmacol Sci. 2020;24(12):7015–23.
Meng L, Lin H, Huang X, Weng J, Peng F, Wu S. METTL14 suppresses pyroptosis and diabetic cardiomyopathy by downregulating TINCR LncRNA. Cell Death Dis. 2022;13(1):38.
Xiong J, Zhou Q. The LncRNA HOTAIR attenuates pyroptosis of diabetic cardiomyocytes by recruiting FUS to regulate SIRT3 expression. Kaohsiung J Med Sci. 2023;39(5):458–67.
Kanduri C. Kcnq1ot1: A chromatin regulatory RNA. Semin Cell Dev Biol. 2011;22(4):343–50.
Li X, Dai Y, Yan S, Shi Y, Han B, Li J, et al. Down-regulation of LncRNA KCNQ1OT1 protects against myocardial ischemia/reperfusion injury following acute myocardial infarction. Biochem Biophys Res Commun. 2017;491(4):1026–33.
Jiang Y, Du W, Chu Q, Qin Y, Tuguzbaeva G, Wang H, et al. Downregulation of long Non-Coding RNA Kcnq1ot1: an important mechanism of arsenic Trioxide-Induced long QT syndrome. Cell Physiol Biochem. 2018;45(1):192–202.
Coto E, Calvo D, Reguero JR, Morís C, Rubín JM, Díaz-Corte C, et al. Differential methylation of LncRNA KCNQ1OT1 promoter polymorphism was associated with symptomatic cardiac long QT. Epigenomics. 2017;9(8):1049–57.
Ren K, Xu R, Huang J, Zhao J, Shi W. Knockdown of long non-coding RNA KCNQ1OT1 depressed chemoresistance to Paclitaxel in lung adenocarcinoma. Cancer Chemother Pharmacol. 2017;80(2):243–50.
Jin X, Jin H, Shi Y, Guo Y, Zhang H. Long Non-Coding RNA KCNQ1OT1 promotes cataractogenesis via miR-214 and activation of the Caspase-1 pathway. Cell Physiol Biochem. 2017;42(1):295–305.
Yang F, Qin Y, Wang Y, Li A, Lv J, Sun X, et al. LncRNA KCNQ1OT1 mediates pyroptosis in diabetic cardiomyopathy. Cell Physiol Biochem. 2018;50(4):1230–44.
Jiao A, Liu H, Wang H, Yu J, Gong L, Zhang H, et al. piR112710 attenuates diabetic cardiomyopathy through inhibiting Txnip/NLRP3-mediated pyroptosis in Db/db mice. Cell Signal. 2024;122:111333.
Acknowledgements
Not applicable.
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
Not applicable.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Radmehr, E., Yazdanpanah, N. & Rezaei, N. Non-coding RNAs affecting NLRP3 inflammasome pathway in diabetic cardiomyopathy: a comprehensive review of potential therapeutic options. J Transl Med 23, 249 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06269-w
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06269-w