Skip to main content
Non-coding RNA Research logoLink to Non-coding RNA Research
. 2024 Jun 4;9(4):1111–1119. doi: 10.1016/j.ncrna.2024.06.005

Noncoding RNAs in chronic obstructive pulmonary disease: From pathogenesis to therapeutic targets

Bingbing Ren a,b,, Hua Su a, Chang Bao a, Hangdi Xu a, Ying Xiao b
PMCID: PMC11254503  PMID: 39022682

Abstract

Chronic obstructive pulmonary disease (COPD) is the most prevalent chronic respiratory disorder that is becoming the leading cause of morbidity and mortality on a global scale. There is an unmet need to investigate the underlying pathophysiological mechanisms and unlock novel therapeutic avenues for COPD. Recent research has shed light on the significant roles played by diverse noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), in orchestrating the development and progression of COPD. This review provides an overview of the regulatory roles of ncRNAs in COPD, elucidating their underlying mechanisms, and illuminating the potential prospects of RNA-based therapeutics in the management of COPD.

Keywords: COPD, miRNA, lncRNA, circRNA

1. Introduction

Chronic obstructive pulmonary disease (COPD) is defined as a heterogeneous lung condition characterized by chronic respiratory symptoms, including dyspnea, cough, and sputum production, that cause persistent airflow obstruction [1]. It is considered one of the most prevalent chronic respiratory disorders worldwide, with an estimated prevalence of 10.3 %, corresponding to approximately 391.9 million individuals [2]. Furthermore, as of 2019, COPD has become the third leading cause of death worldwide [2]. The primary pathological characteristic of COPD is a persistent and atypical inflammatory reaction occurring in the lungs, leading to airway and alveolar changes, as evidenced by the presence of small airway disease and emphysema [3]. In individuals diagnosed with COPD, the emergence of pulmonary hypertension is linked to a decline in clinical status, deterioration in gas exchange, and heightened mortality rates [4]. Although it is widely accepted that smoking, outdoor and indoor air pollution, childhood respiratory infections, and genetic abnormalities are major risk factors in the pathogenesis and development of COPD, the understanding of the pathophysiology of COPD remains incomplete [2]. Consequently, there is a pressing need to investigate the pathophysiological mechanisms of COPD and therefore identify novel therapeutic targets and strategies.

Recent studies have linked a variety of noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs) to the occurrence and development of COPD [5]. Some of these ncRNAs have undergone functional characterization, revealing their potential for early diagnosis and therapeutic intervention in COPD. This comprehensive review aims to present the latest advancements in the field of COPD-associated ncRNAs and explore their prospective clinical applications.

2. MicroRNAs in COPD

MicroRNAs are a subset of small noncoding RNAs, typically spanning 21–25 nucleotides, which primarily modulate gene expression at the post-transcriptional level. The dysregulation of a series of miRNAs has been linked to the pathogenesis of COPD and other respiratory disorders [6,7]. Moreover, much research efforts have been dedicated to investigating the functional mechanisms of specific miRNAs in the progression and development of COPD (Table 1, Fig. 1).

Table 1.

Functional microRNAs validated in COPD model.

miRNA Dysregulation in Patients Target Gene Targeted Pharmacological Actions COPD models Ref.
miR223 Increased in lung tissue HDAC2 Inflammation CS + mouse [22]
miR181 Decreased in lung tissue CCN1 Inflammation CSE + HBEC; CS + C57BL/6 mouse [21]
miR218 Increased in serum and lung tissue TNFR1 Inflammation CSE + HBEC [23]
miR27 PPARγ Inflammation CS + C57BL/6 mouse [24]
miR195 Increased in lung tissue PHLPP2 Inflammation CSE + BEAS-2B; CS + C57BL/6J mouse [27]
miR3202 Decreased in plasma by smoking FAIM2 Inflammation CSE + HBEC; CS + SD rat [25,26]
miR197 Decreased in lung tissue E2F1 Vascular remodeling Primary SMC [19]
miR21 Increased in serum and lung tissue pVHL; SATB1 Airway remodeling; inflammation CSE + HBEC + MRC-5; CS + BALB/c mouse [41,42]
miR206 Increased in plasma and lung tissue Notch3; VEGFA Vascular remodeling CSE + HPMECs [17]
miR145 Decreased in lung tissue by smoking CFTR; SLC26A9; KLF5 Mocociliary clearance; inflammation CSE + BEAS-2B; CS + non-CF A/J mouse; CSE + HBEC [36,37]
miR30 Decreased in blood by cadmium exposure SNAIL Epithelial-mesenchymal transition CdCl2+ BEAS-2B [10,11]
miR155 Increased in lung tissue by smoking Inflammation CS + C57BL/6 mouse [29]
let-7 Decreased in lung tissue IL-6 Myofibroblast differentiation CSE + HBEC + MRC-5; CS + C57BL/6 mouse [9]
miR-34 Increased in lung tissue SIRT1
SIRT6
Senescence Primary HBEC [31]
miR-570 Increased in lung tissue SIRT1 Senescence Primary SAEC [32]
miR126 Decreased in lung tissue ADAM9 Vascular remodeling Primary HLMVEC [18]

Abbreviations: CS, cigarette smoke; CSE, cigarette smoke extract; EMT, epithelial-to-mesenchymal transition; HBEC, human biliary epithelial cell; HLMVEC, human lung microvascular epithelial cell; HPMEC, human pulmonary microvascular endothelial cell; SAEC, small airway epithelial cell; SMC, smooth muscle cell.

Fig. 1.

Fig. 1

Noncoding RNAs participate in COPD pathogenesis. Functional miRNAs, lncRNAs, and circRNAs have been validated in COPD models to participate in inflammation, mucociliary clearance, airway and vascular remodeling, oxidative stress response, and cell apoptosis.

2.1. MicrRNAs target airway remodeling

Airway remodeling is a crucial factor contributing to progressive airway limitation in individuals with COPD [8]. Exposure to cigarette smoke (CS) has been shown to reduce levels of let-7 in bronchial epithelial cells, leading to increased secretion of interleukin 6 (IL-6) and subsequent differentiation of fibroblasts into myofibroblasts [9]. The suppression of miR-30 has been observed in cadmium-induced COPD [10]. This microRNA was found to inhibit the expression of SNAIL, a master regulator of epithelial-mesenchymal transition (EMT) in lung epithelial cells [11]. Notably, cadmium is a known lung carcinogen, and the cadmium-induced downregulation of miR-30 and consequential upregulation of SNAIL may contribute to the development of cadmium-associated lung cancer as well [11].

2.2. MicroRNAs target vascular remodeling

Pulmonary vascular remodeling is another prominent characteristic of COPD and is widely recognized as a key factor in the development of pulmonary hypertension [12]. Nevertheless, the precise molecular mechanisms responsible for these vascular alterations remain incompletely elucidated. Alternations in miRNA expression profiles in individuals with COPD were analyzed by several studies [13,14].

Due to the negative prognosis associated with skeletal muscle dysfunction in individuals with COPD, there has been a particular focus on muscle-specific microRNAs (myo-miRNAs), known to be involved in myogenesis and differentiation, in COPD research. Several preliminary studies have identified significant dysregulations of myo-miRNAs, such as miR-1, miR-133, and miR-206, in COPD patients [[14], [15], [16]]. Among them, miR-206 consistently exhibited upregulation in COPD patients’ plasma and lung tissue (Table 1). According to a recent investigation conducted on human pulmonary microvascular endothelial cells (HPMECs), it was found that miR-206 could directly target Notch receptor 3 (NOTCH3) and vascular endothelial growth factor A (VEGFA), both of which play crucial roles in vascular remodeling [17]. Correspondingly, the introduction of antagomir against miR-206 to HPMECs resulted in elevated expression levels of NOTCH3 and VEGFA, thereby impeding the HPMEC apoptosis induced by cigarette smoke extract (CSE) [17].

Significant decreases in the expression levels of both miR-126 and miR-197 were observed in the lung tissue of patients with COPD. MiR-126, previously identified as a key regulator of endothelial cell differentiation, is believed to play a role in vascular remodeling in small pulmonary arteries and lung microvasculature by modulating the expression of ADAM9 (a disintegrin and a metalloprotease 9) [18]. Additionally, the expression level of miR-197 exhibited a strong negative correlation with the severity of airflow obstruction and the extent of vascular remodeling. In pulmonary arteries, miR-197 appears to mitigate aberrant vascular remodeling, potentially through the inhibition of a transcription factor E2F1 [19].

2.3. MicroRNAs target inflammation

Pulmonary inflammation is a common feature of COPD and various other respiratory diseases. Consequently, dysregulation of numerous inflammation-associated microRNAs is frequently observed across different respiratory disorders [20].

The microRNA miR-181 plays a role in modulating inflammatory cytokine levels through its direct target CCN1. Decreased expression of miR-181 in bronchial epithelial cells has been linked to heightened inflammatory responses, increased reactive oxygen species production, and the progression of COPD [21]. Interestingly, inflammatory cytokines could induce the expression of various miRNAs in return. For instance, the upregulation of miR-223, which regulates HDAC2 expression and activity in pulmonary cells, has been observed in COPD mouse models following stimulation with interleukin-1β and tumor necrosis factor-α [22].

In individuals with COPD, miR-218 expression is decreased in both bronchial epithelial cells and serum samples. In non-COPD bronchial epithelial cells, miR-218 is responsible for binding to the 3′UTR region of tumor necrosis factor receptor 1 (TNFR1) mRNA to inhibit the unwanted activation of NF-κB activation and the upregulation of interleukins [23]. Similarly, miR-27 targets the 3′UTR sequence of peroxisome proliferator-activated receptor γ (PPARγ), leading to the suppression of its activation, thereby preventing the induction of pulmonary inflammation and activation of alveolar macrophages in the development of COPD [24].

Levels of miR-3202 in blood samples exhibited a reduction among individuals with COPD, and further displayed a negative association with COPD exacerbation, indicating a potential protective role of miR-3202 in COPD patients, particularly those who smoke [25]. Subsequent investigations suggest that miR-3202 may facilitate the activation of the FAS signaling pathway by inhibiting FAIM2 (Fas apoptotic inhibitory molecule 2) expression in T-lymphocytes [25,26].

Conversely, miR-195 was found to be significantly elevated in the lung tissue of COPD patients. Mechanistically, miR-195 enhanced AKT phosphorylation by suppressing PHLPP2 (PH Domain And Leucine Rich Repeat Protein Phosphatase 2) expression [27]. MiR-155, a well-known modulator of the immune response, was also upregulated in COPD patients [28,29]. More importantly, its expression level is strongly associated with lung function parameters of airflow limitation and diffusing capacity in patients with COPD, with the highest expression observed in end-stage COPD [28]. In a mouse model, removing miR-155 significantly reduces elastase-induced emphysema and changes in lung function, indicating its critical involvement in pulmonary inflammation and the development of COPD [29]. However, the specific molecular mechanisms underlying these effects remain unclear.

2.4. MicroRNAs target senescence

Cellular senescence is also recognized as a significant contributing factor to COPD [30]. The expression of miR-34 was found to be elevated in the lung tissues of COPD patients and was found to be correlated with the expression of senescence markers. Further investigation into the underlying mechanism revealed that miR-34 plays a crucial role in downregulating sirtuin-1 and -6 (SIRT1 and SIRT6) in bronchial epithelial cells, and inhibiting this microRNA could reverse cell cycle arrest and reduce markers of senescence [31]. The modulation of SIRT1 level by miR-570 has also been observed in individuals with COPD, with the upregulation of miR-570 being activated by p38 mitogen-activated protein kinase [32]. It is noteworthy that these miRNAs have been shown to regulate senescence in various cell types, hinting that the release and circulation of these miRNAs may provide insight into the pathogenesis of other chronic lung diseases and comorbidities associated with COPD.

Furthermore, several key miRNAs were found to participate in the progression of COPD through multiple pathways (Table 1). One example is miR-145, an established regulator in the TGF-β signaling pathway, which has been considered a promising miRNA-based therapeutic agent in breast cancer, colon cancer, and prostate cancer [[33], [34], [35]]. In the context of COPD models, on the one hand, miR-145 is essential for protecting airway epithelial cells against CSE-induced apoptosis and inflammation by targeting KLF5 (Krüppel-like factor 5) [36]. On the other hand, upregulation of miR-145 has been observed to suppress the expression of CFTR (cystic fibrosis transmembrane conductance regulator) and its modifier SLC26A9 (Solute carrier family 26 member 9), both of which play crucial roles in the primary innate defense of mucociliary clearance [37]. Another intriguing example is miR-21, a non-specific pro-metastatic factor in various cancers, exhibiting promising therapeutic potential in cholangiocarcinoma, colorectal cancer, and ovarian cancer [[38], [39], [40]]. In COPD patients, exosomal miR-21 derived from bronchial epithelial cells regulates myofibroblast differentiation via the pVHL (von Hippel-Lindau protein)/HIF1α (hypoxia-inducible factor 1α) axis. Simultaneously, miR-21 in lung tissue mediates the proliferation of airway macrophages, neutrophils, and lymphocytes via the SATB1 (special AT-rich sequence–binding protein 1)/S100A9 (S100 calcium binding protein A9)/NF-κB axis. In both scenarios, the utilization of small interfering RNA (siRNA) to target miR-21 demonstrated enhanced lung function, suggesting its potential as a promising therapeutic target for COPD [41,42].

3. Long noncoding RNAs in COPD

LncRNAs are defined as noncoding transcripts that exceed 200 nucleotides in length [43]. Initial investigations utilizing high-throughput RNA sequencing have revealed hundreds of lncRNAs to be associated with the development of COPD [44,45]. However, only recently have some of these lncRNAs begun to be functionally characterized [[46], [47], [48]](Table 2, Fig. 1).

Table 2.

Functional lncRNAs validated in COPD models.

lncRNA Dysregulation in Patients Target Gene Targeted Pharmacological Actions COPD models Ref.
COPDA1 Increased in lung tissue MS4A1 Airway remodeling HBSMC [55]
SNHG5 Decreased in lung tissue miR132/PTEN Inflammation CSE+16HBE [58]
NNT-AS1 Increased in lung tissue miR582-5p/FBXO11 Airway remodeling; inflammation CSE+16HBE [62]
CASC2 Decreased in serum miR18a-5p/IGF1 Inflammation CSE+16HBE [57]
Nqo1-AS1 Increased in lung tissue and PBMC Nqo1 Oxidative stress CSE + MLE12; CS + C57BL/6J mouse [52]
LUCAT1 Increased in serum miR181a-5p/Wnt/β-catenin Pulmonary cell proliferation and apoptosis CSE+16HBE [60]
IL6-AS1 Increased in lung tissue IL-6 Inflammation CSE/LPS/PM + HBF/HFL1 [49]
OIP5-AS1 Increased in serum miR410-3p/IL-13 Inflammation CSE+16HBE [59]
LASI Increased in lung tissue ICAM-1 Mucus hyperexpression; inflammation CSE + HBEC; CS + macaque [50]
CCAT1 Increased in lung tissue miR152-3p/ERK Inflammation CSE + HBE [61]
HOTAIR Increased in lung tissue DNMMT1/Bcl-2 Pulmonary cell apoptosis CSE + HPVEC; CS + C57BL/6J mouse [54]
MHC-R Immune response PM + SD rat [64]
Lnc-IL7R Decreased in serum EZH2/p21 Pulmonary cell apoptosis PM + HSAEpC [65]
HSALR1 Increased in lung tissue HSP90AB1/pAKT Airway remodeling CS + C57BL/6J mouse [47]

Abbreviations: CS, cigarette smoke; CSE, cigarette smoke extract; HBEC, human biliary epithelial cell; HBF, human bronchial fibroblast; HBSMC, human bronchial smooth muscle cell; HPMEC, human pulmonary microvascular endothelial cell; HPVEC, human pulmonary vascular endothelial cell; HSAEpC, human small airway epithelial cell; LPS, lipopolysaccharide; PM, particulate matter.

3.1. LncRNAs regulate RNA stability

The majority of these lncRNAs exert their effects by modulating local gene expression. For example, IL6-AS1, which is upregulated in the lung tissue of COPD patients, is the antisense transcript of interleukin 6. Through promoting IL-6 gene transcription and stabilizing IL-6 mRNA, IL6-AS1 enhances IL-6 production and inflammation in COPD [49].

Another lncRNA, LASI, is located on the antisense strand of the important inflammatory factor ICAM-1 (intercellular adhesion molecule 1) [50]. In COPD patients, the expression of LASI in airway epithelial cells is upregulated and positively correlated with the severity of the disease [48]. Furthermore, LASI expression induced by cigarette smoke in both human bronchial epithelial cells and the cynomolgus macaque model is positively correlated with the secretory mucin MUC5AC and airway the expression of inflammatory factors ICAM-1 and IL-6. Accordingly, the knockdown of LASI using siRNA resulted in a substantial decrease in the levels of MUC5AC (mucin 5AC), ICAM-1, and IL-6, indicating its modulatory functions in airway mucus hyperexpression and inflammatory responses [48].

NQO1-AS1 is the antisense transcript that is positioned in a tail-to-tail orientation with the NAD(P)H quinone oxidoreductase coding gene NQO1. As one of the most crucial quinone reductases, NQO1 has well-established roles in safeguarding cells against CS-induced oxidative stress and obstructive bronchitis [51]. By forming a pairing with the 3’ UTR of NQO1, NQO1-AS1 serves to stabilize NQO1 mRNA and subsequently enhance NQO1 expression, thereby mitigating the CS-induced oxidative stress [52].

3.2. LncRNAs regulate transcription

HOTAIR, a well-known regulator of epithelial cell plasticity, has been implicated in facilitating the EMT in various cancer types [46]. Recently, the upregulation of HOTAIR was also observed in the lung tissue of COPD patients [53,54]. In both CS-treated mouse models and human pulmonary vascular endothelial cells, HOTAIR targets DNMT1 (DNA methyltransferase enzyme 1) to mediate the hypermethylation of the BCL-2 (B-cell lymphoma-2) promoter, thereby promoting endothelial cell apoptosis [54]. Accordingly, knocking down HOTAIR can attenuate pulmonary cell apoptosis and emphysema in CS-treated mice [54]. Another lncRNA, COPDA1, was identified to coexpress with MS4A1 (membrane-spanning 4-domains A1) in bronchial smooth muscle cells. Their upregulation was proved to be associated with the undesirable proliferation of smooth cells and airway remodeling, although the specific signaling pathways implicated in the process have not been fully elucidated [55].

3.3. LncRNAs act as protein scaffolds

Additionally, HSALR1 lncRNA not only exhibits a substantial increase in lung tissue of COPD patients but also demonstrates a robust correlation with TGF-β1 expression, a known contributor to COPD progression [56]. Under the stimulation of TGF-β1, SMAD3 (SMAD family member 3) could directly bind to the promoter of HSALR1, thereby triggering its upregulation. As a result, HSALR1 serves as a scaffold to stabilize the HSP90AB1 (heat shock protein 90AB1)-AKT complex and promote AKT phosphorylation [47]. Experiment results obtained from both human lung fibroblast cells and mice models have demonstrated that the SMAD3/HSALR1/AKT axis plays a role in promoting cell proliferation and airway remodeling during the progression of COPD [47].

3.4. LncRNAs act as miRNA sponges

The expression levels of lncRNA CASC2 (cancer susceptibility candidate 2) were found to be significantly decreased in COPD patients, particularly those classified in the severe group (GOLD stage III). CASC2 acts as a competing endogenous RNA (ceRNA) for miR-18a-5p, influencing bronchial epithelial cell apoptosis and inflammation through the miR-18/IGF1 (insulin-like growth factor 1) axis [57]. Another lncRNA SNHG5 (small nucleolar RNA host gene 5) was also observed to be downregulated in COPD lung tissues and exhibited a positive correlation with FEV1 (forced expiratory volume in 1 s), a marker of COPD severity. LncRNA SNHG5 functions as a ceRNA for miR-132, leading to increased expression of PTEN and ultimately promoting COPD exacerbation [58].

Several other lncRNAs were found to be significantly upregulated in patients with COPD. Among them, OIP5-AS1 (Opa interacting protein 5-antisense RNA 1) was found to enhance cell apoptosis and pulmonary inflammation by targeting the miR-410-3p/IL-13 axis [59]. LncRNA LUCAT1 (lung cancer-associated transcript 1) was upregulated in serum samples from COPD patients and was shown to modulate COPD progression through the Wnt/β-catenin pathway by acting as a sponge for miR-181a-5p [60]. Whereas, CCAT1 was highly upregulated in lung tissues of COPD patients and was found to promote inflammation by sponging miR-152-3p to activate the ERK signaling pathway [61]. The upregulation of NNT-AS1 was CSE-dependent. Its upregulation leads to the positive regulation of FBXO11 (F-Box protein 11) through the sponging of miR-582-5p, which in turn contributes to the promotion of apoptosis, inflammation, and airway remodeling in the context of COPD progression [62].

It is worth noting that, in addition to cigarette smoking, the global initiative for chronic obstructive lung disease (GOLD) guideline has listed air pollution as one of the major risk factors for COPD as well [1]. It was reported that the prevalence of COPD is projected to increase by approximately 1.5 times for every 5 μg/m3 increase in exposure to fine particulate matter (PM2.5) [63]. Therefore, in addition to those CS-induced COPD models, recent research has begun to utilize PM-induced COPD models. In a rat model exposed to PM, the lncRNA MHC-R exhibited drastic upregulation and likely participated in the pathogenesis of COPD through the regulation of immune activities in dendritic cells [64]. Furthermore, the level of another lncRNA, lnc-IL7R, was found to be decreased in the serum of COPD patients and displayed a correlation with PM exposure. By recruiting EZH2 (enhancer of zeste homolog 2) to the p21 promoter, lnc-IL7R was able to effectively mitigate PM-induced cell apoptosis and senescence [65].

4. Circular RNAs in COPD

CircRNAs, characterized by their distinct circularized conformations, have emerged as pivotal regulators in a wide range of diseases [66,67]. While recent research advances revealed the dysregulation of certain circRNAs in COPD, only a handful of them have been subjected to functional characterization [5,68,69] (Fig. 2, Table 3).

Fig. 2.

Fig. 2

Functional mechanisms of circRNAs in COPD development.

Table 3.

Functional circRNAs validated in COPD models.

circRNA Dysregulation in Patients Target Gene/RNA Targeted Pharmacological Actions COPD models Ref.
circ_0026344 Decreased in lung tissue miR-21 Apoptosis CS + BALB/c mouse [70]
circBBS9 Increased in lung tissue miR-30 Inflammation PM2.5+ICR mouse [71,74]
miR-103 Apoptosis CSE + HPMEC
circ_0026466 Increased in blood miR-153 Apoptosis CSE+16HBE [72]
circ_0006827 Increased in lung tissue miR-145 Apoptosis CSE + HPMEC [73]
CSE + BEAS-2B
circFOXO3 Increased in lung tissue miR-214 Inflammation CS + C57BL/6 mouse [75]
CSE + MLE12
circXPO1 Increased in lung tissue miR-23 Inflammation CSE + MLE12 [76]
CS + C57BL/6
circSAV1 Increased in lung tissue IREB2 Ferroptosis CSE + BEAS-2B [77]
CS + BALB/c mouse
circ_0005045 Increased in lung tissue PRDX2 Inflammation PM2.5+C57BL/6 mouse [78]

Abbreviations: CS, cigarette smoke; CSE, cigarette smoke extract; HPMEC, human pulmonary microvascular endothelial cell; PM, particulate matter.

4.1. CircRNAs act as miRNA sponges

MicroRNA sponging is a widely recognized functional mechanism of circRNAs, in which circRNAs competitively bind to microRNAs to modulate the expression of target genes. An illustrative example of this mechanism is observed in the case of circRNA_0026344, which functions as an anti-apoptosis factor in alveolar epithelial cells by sponging miR-21. This action inhibits miR-21's binding to PTEN. The overexpression of circ_0026344 has been found to partially block the CSE-induced autophagy and apoptosis, and certain blockages can be reversed by the introduction of a miR-21 mimic [70]. Whereas circBBS9, circ_0026466, and circ_0006872 have been identified as pro-apoptotic miRNA sponges against miR103a-3p/BCL2L13 (BCL2-like 13) axis, miR153-3p/TRAF6 (NF receptor associated factor 6) axis, and miR145/NF-κB axis, respectively [[71], [72], [73]]. It is worth noting that circRNA molecules have the potential to interact with multiple miRNAs. For example, circBBS9 has been found to also bind to miR-30e-5p to regulate the expression of ADAR, and exacerbate pulmonary inflammation in PM2.5-induced COPD mice [74]. Similarly, recent studies have identified the involvement of circFOXO3 and circXPO1 in CS-induced inflammation in COPD. Specifically, circFOXO3 has been shown to enhance pulmonary inflammation by targeting miR-214 to upregulate IKK-β (inhibitor kappa B kinase β) expression, silencing of which would disrupt the NF-κB signaling pathway [75]; while circXPO1 has been found to interact with miR-23b-3p to upregulate TAB3 (MAP3K7 binding protein 3) in the TGF-β signaling pathway [76].

4.2. CircRNAs act as protein scaffolds

CircRNAs also possess the ability to interact with RNA bind proteins (RBPs) and serve as protein scaffolds to regulate gene expression [67]. A recent study showed that m6A-modified circSAV1 forms a complex with YTHDF1 (YTH N6-methyladenosine RNA binding protein F1) and IREB2 (iron responsive element binding protein 2) mRNA, thereby facilitating the translation of IREB2. The upregulation of IREB2 disrupts iron homeostasis and ultimately leads to ferroptosis. In lung tissue of COPD patients, the level of circSAV1 level was found to be associated with the progression of COPD progression, whereas the silencing of circSAV1 in a mouse model effectively prevented CS-induced ferroptosis in lung epithelial cells [77]. Another circRNA, hsa_circ_0005045, was discovered to be upregulated in a specific subgroup of non-smoking PM2.5-sensitive COPD patients. This circRNA was found to interact with PRDX2 (oxidized peroxiredoxin-2), a proinflammatory factor [78]. Interestingly, knocking down the corresponding circRNA, mmu_circ_0002950, in mice effectively rescued the PM2.5-induced COPD-like lesions. These findings suggest that this circRNA plays a conserved role in protecting COPD patients against air pollution-associated acute disease exacerbation [79].

5. Therapeutic potentials of noncoding RNAs in COPD

Given the limited understanding of the underlying pathophysiological mechanisms of COPD, earlier therapeutics for COPD are largely adapted from the symptomatic treatments of asthma. The treatment options for COPD have significantly expanded in recent years, encompassing new oral and inhaled medications, as well as innovative surgical and bronchoscopic procedures. Nevertheless, the escalating global prevalence and mortality rates associated with COPD necessitate the development of safer and more effective pharmacological treatments for COPD.

Investigations into ncRNAs associated with COPD have not only identified novel therapeutic targets but have also offered alternative strategies for treating this disease [80]. RNA-based therapeutics, with their high specificity compared to small molecules and proteins and lower safety concerns compared to DNA therapies, have emerged as promising therapeutic options for various diseases. The approval of Fomivirsen, the first RNA-based therapy, by the FDA in 1998 for the treatment of cytomegalovirus retinitis in immunocompromised patients highlights the potential of RNA drugs in clinical usage. Currently, over 10 RNA-based therapeutics have received FDA approval, and several comprehensive reviews have documented these achievements [[81], [82], [83]]. Furthermore, an array of RNA drugs, such as Vutrisiran (Phase 3 trial NCT04153149), Tivanisiran (Phase 3 trial NCT04819269), and Olparsiran (Phase 2 trial NCT04270760), are presently under clinical development. The expanded applications of these RNA-based drugs have demonstrated the feasibility of utilizing RNA-based therapeutics for the treatment of human diseases [84].

RNA-based therapies are also receiving growing attention in treating respiratory diseases with high specificity and potency [85]. In 2000, Stenton and colleagues conducted an initial investigation into the utilization of antisense oligodeoxynucleotide (ASO) in the treatment of respiratory diseases. Specifically, ASO targeting spleen tyrosine kinase (Syk) was administered via aerosol to the lungs of mice to assess its efficacy in managing asthma [86]. However, due to issues of non-specificity and potential allergic reactions, further exploration of Syk ASO was halted. The challenge of achieving targeted delivery of RNA molecules to specific cells emerged as a significant barrier to the implementation of RNA therapies for pulmonary conditions. Attempts have been undertaken over the past two decades to enhance the pulmonary delivery of therapeutic RNAs. In 2008, Xu et al. developed a poly(ester amine)-mediated complex that effectively delivered AKT siRNAs via aerosol, leading to the inhibition of lung cancer progression [87]. Subsequently, in 2014, nanoparticles were utilized for the delivery of miR-34 to lung cancer cells, resulting in the successful inhibition of tumor growth in vitro [88]. And TargomiR, utilizing both a nanoparticle drug delivery system and an anti-epidermal growth factor receptor antibody to target miR-16 mimics to cancer cells, has completed phase I clinical trials for the treatment of malignant pleural mesothelioma and non-small cell lung cancer (NCT02369198) [89]. Noticeably, in addition to its efficacy in treating respiratory diseases, recent advancements in nanotechnology-based miRNA therapies have also demonstrated potential for early diagnosis and overcoming drug resistance in lung cancer treatment [90].

Efforts have also been undertaken to address critical pathways involved in the pathogenesis of COPD using RNA interference (RNAi) approaches (Table 4). This approach has been favored due to the ease of synthesizing small interfering RNAs (siRNAs) and their successful application in various other diseases [91]. Specifically, to mitigate inflammation in the airway epithelium and alveolar macrophages of COPD patients, siRNAs specific to the receptor-interacting serine/threonine-protein kinase 2 (RIP2) and ribosome protein S3 (RPS3) have been designed. Both of them are crucial factors involved in the NF-κB signaling pathway, a master proinflammatory regulator in COPD progression [92,93]. To mitigate airway remodeling and associated lung fibrosis, CHST3 (carbohydrate sulfotransferase 3) -specific siRNA and TROP2 (trophoblast cell surface antigen 2) -specific siRNA were employed to reduce emphysema and basal cell hyperplasia [94,95]. Additionally, to counteract pulmonary vascular remodeling, which may further lead to pulmonary arterial hypertension, siRNAs targeting HIF-1α and HIF-2α were utilized to inhibit smooth muscle cell proliferation and EMT [96].

Table 4.

Current studies for RNA-based therapeutics in COPD.

Therapeutic target RNA type Delivery system Target cell Ref.
RPS3 siRNA Naked siRNA Macrophage [92]
RIP2 siRNA Lipofectamine Macrophage Epithelial cell [93]
CHST3 siRNA Naked siRNA Macrophage [94]
TROP2 siRNA Lipofectamine Basal cell [95]
HIF1/2 siRNA Lipofectamine Pulmonary artery smooth muscle cells [96]

Presently, many siRNAs have demonstrated promising outcomes in mitigating inflammation, airway and vascular remodeling, and disease exacerbation in COPD models [70]. However, breakthroughs in the development of RNA-based drugs for COPD treatment have not emerged yet. Similar to other respiratory conditions, the primary obstacle in utilizing RNA therapies for COPD lies in achieving effective and specific delivery. Pulmonary delivery of inhaled RNA drugs is preferred and rapidly advancing due to its minimal patient discomfort [97]. Inhaled drugs may encounter interference from endogenous lung substances and various lung cell types prior to reaching their intended therapeutic targets. A thorough comprehension of the pathological microenvironment is imperative for the successful design and precise targeting of inhaled RNA drugs. Presently, the insufficient understanding of the diverse pathophysiological mechanisms contributing to the initiation and advancement of COPD appears to impede the advancement of RNA-based therapies for COPD treatments. Extensive endeavors are still required to elucidate the intricate networks involved in COPD pathogenesis and to identify pivotal regulatory factors for therapeutics. Meanwhile, the mitigation of undesirable off-target effects poses another significant obstacle, particularly for those genes and ncRNAs that exhibit multifunctionality across various pathways.

In summary, a multitude of ncRNAs have been unearthed as pivotal contributors to the prevalence of COPD. The functional investigations conducted on ncRNAs have substantially augmented our comprehension of the intricated network underpinning COPD pathogenesis, thereby presenting a novel avenue for the identification of therapeutic targets and drugs for COPD management.

CRediT authorship contribution statement

Bingbing Ren: Writing – original draft, Funding acquisition. Hua Su: Writing – review & editing, Funding acquisition. Chang Bao: Writing – review & editing. Hangdi Xu: Writing – review & editing. Ying Xiao: Writing – review & editing, Funding acquisition.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

This work was supported by the National Science Foundation of China (32000437), the Medical Science and Technology Project of Zhejiang Province (2023RC188, 2022RC037, 2024KY105), and the Opening Project of Zhejiang Provincial Preponderant and Characteristic Subject of Key University (Chinese Traditional Medicine), Zhejiang Chinese Medical University (ZYXZD2019003). Figures were created by FigDraw (FigDraw.com).

References

  • 1.Venkatesan P. GOLD report: 2022 update. Lancet Res. Med. 2022;10:e20. doi: 10.1016/S2213-2600(21)00561-0. [DOI] [PubMed] [Google Scholar]
  • 2.Adeloye D., Song P., Zhu Y., Campbell H., Sheikh A., Rudan I. Global, regional, and national prevalence of, and risk factors for, chronic obstructive pulmonary disease (COPD) in 2019: a systematic review and modelling analysis. Lancet Res. Med. 2022;10:447–458. doi: 10.1016/S2213-2600(21)00511-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Brandsma C.A., Van den Berge M., Hackett T.L., Brusselle G., Timens W. Recent advances in chronic obstructive pulmonary disease pathogenesis: from disease mechanisms to precision medicine. J. Pathol. 2020;250:624–635. doi: 10.1002/path.5364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Olsson K.M., Corte T.J., Kamp J.C., Montani D., Nathan S.D., Neubert L., Price L.C., Kiely D.G. Pulmonary hypertension associated with lung disease: new insights into pathomechanisms, diagnosis, and management. Lancet Res. Med. 2023;11:820–835. doi: 10.1016/S2213-2600(23)00259-X. [DOI] [PubMed] [Google Scholar]
  • 5.Li B., Zhang J., Dong H., Feng X., Yu L., Zhu J., Zhang J. Systematic analysis of various RNA transcripts and construction of biological regulatory networks at the post-transcriptional level for chronic obstructive pulmonary disease. J. Transl. Med. 2023;21:790. doi: 10.1186/s12967-023-04674-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zhou G., Chen T., Raj J.U. MicroRNAs in pulmonary arterial hypertension. AM J Resp. Cell Mol. 2015;52:139–151. doi: 10.1165/rcmb.2014-0166TR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zoulikha M., Xiao Q., Boafo G.F., Sallam M.A., Chen Z., He W. Pulmonary delivery of siRNA against acute lung injury/acute respiratory distress syndrome. Acta Pharm. Sin. B. 2022;12:600–620. doi: 10.1016/j.apsb.2021.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Liu G., Philp A.M., Corte T., Travis M.A., Schilter H., Hansbro N.G., Burns C.J., Eapen M.S., Sohal S.S., Burgess J.K., Hansbro P.M. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol. Therapeut. 2021;225 doi: 10.1016/j.pharmthera.2021.107839. [DOI] [PubMed] [Google Scholar]
  • 9.Di T., Yang Y., Fu C., Zhang Z., Qin C., Sai X., Liu J., Hu C., Zheng M., Wu Y., Bian T. Let-7 mediated airway remodelling in chronic obstructive pulmonary disease via the regulation of IL-6. Eur. J. Clin. Invest. 2021;51 doi: 10.1111/eci.13425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zheng L., Jiang Y.L., Fei J., Cao P., Zhang C., Xie G.F., Wang L.X., Cao W., Fu L., Zhao H. Circulatory cadmium positively correlates with epithelial-mesenchymal transition in patients with chronic obstructive pulmonary disease. Ecotox. Environ. Safe. 2021;215 doi: 10.1016/j.ecoenv.2021.112164. [DOI] [PubMed] [Google Scholar]
  • 11.Tanwar V.S., Zhang X., Jagannathan L., Jose C.C., Cuddapah S. Cadmium exposure upregulates SNAIL through miR-30 repression in human lung epithelial cells. Toxicol. Appl. Pharm. 2019;373:1–9. doi: 10.1016/j.taap.2019.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Blanco I., Piccari L., Barbera J.A. Pulmonary vasculature in COPD: the silent component. Respirology. 2016;21:984–994. doi: 10.1111/resp.12772. [DOI] [PubMed] [Google Scholar]
  • 13.Garcia-Lucio J., Argemi G., Tura-Ceide O., Diez M., Paul T., Bonjoch C., Coll-Bonfill N., Blanco I., Barbera J.A., Musri M.M., Peinado V.I. Gene expression profile of angiogenic factors in pulmonary arteries in COPD: relationship with vascular remodeling. AM J. Physiol.-Lung C. 2016;310:L583–L592. doi: 10.1152/ajplung.00261.2015. [DOI] [PubMed] [Google Scholar]
  • 14.Donaldson A., Natanek S.A., Lewis A., Man W.D., Hopkinson N.S., Polkey M.I., Kemp P.R. Increased skeletal muscle-specific microRNA in the blood of patients with COPD. Thorax. 2013;68:1140–1149. doi: 10.1136/thoraxjnl-2012-203129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lewis A., Riddoch-Contreras J., Natanek S.A., Donaldson A., Man W.D., Moxham J., Hopkinson N.S., Polkey M.I., Kemp P.R. Downregulation of the serum response factor/miR-1 axis in the quadriceps of patients with COPD. Thorax. 2012;67:26–34. doi: 10.1136/thoraxjnl-2011-200309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Puig-Vilanova E., Martinez-Llorens J., Ausin P., Roca J., Gea J., Barreiro E. Quadriceps muscle weakness and atrophy are associated with a differential epigenetic profile in advanced COPD. Clin. Sci. 2015;128:905–921. doi: 10.1042/CS20140428. [DOI] [PubMed] [Google Scholar]
  • 17.Sun Y., An N., Li J., Xia J., Tian Y., Zhao P., Liu X., Huang H., Gao J., Zhang X. miRNA-206 regulates human pulmonary microvascular endothelial cell apoptosis via targeting in chronic obstructive pulmonary disease. J. Cell. Biochem. 2019;120:6223–6236. doi: 10.1002/jcb.27910. [DOI] [PubMed] [Google Scholar]
  • 18.Goel K., Egersdorf N., Gill A., Cao D., Collum S.D., Jyothula S.S., Huang H.J., Sauler M., Lee P.J., Majka S., Karmouty-Quintana H., Petrache I. Characterization of pulmonary vascular remodeling and MicroRNA-126-targets in COPD-pulmonary hypertension. Resp. Res. 2022;23:349. doi: 10.1186/s12931-022-02267-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Musri M.M., Coll-Bonfill N., Maron B.A., Peinado V.I., Wang R.S., Altirriba J., Blanco I., Oldham W.M., Tura-Ceide O., Garcia-Lucio J., de la Cruz-Thea B., Meister G., Loscalzo J., Barbera J.A. MicroRNA dysregulation in pulmonary arteries from chronic obstructive pulmonary disease. Relationships with vascular remodeling. AM J Resp. Cell Mol. 2018;59:490–499. doi: 10.1165/rcmb.2017-0040OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Aghasafari P., George U., Pidaparti R. A review of inflammatory mechanism in airway diseases. Inflamm. Res. 2019;68:59–74. doi: 10.1007/s00011-018-1191-2. [DOI] [PubMed] [Google Scholar]
  • 21.Du Y., Ding Y., Chen X., Mei Z., Ding H., Wu Y., Jie Z. MicroRNA-181c inhibits cigarette smoke-induced chronic obstructive pulmonary disease by regulating CCN1 expression. Resp. Res. 2017;18:155. doi: 10.1186/s12931-017-0639-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Leuenberger C., Schuoler C., Bye H., Mignan C., Rechsteiner T., Hillinger S., Opitz I., Marsland B., Faiz A., Hiemstra P.S., Timens W., Camici G.G., Kohler M., Huber L.C., Brock M. MicroRNA-223 controls the expression of histone deacetylase 2: a novel axis in COPD. J. Mol. Med. 2016;94:725–734. doi: 10.1007/s00109-016-1388-1. [DOI] [PubMed] [Google Scholar]
  • 23.Xu H., Sun Q., Lu L., Luo F., Zhou L., Liu J., Cao L., Wang Q., Xue J., Yang Q., Yang P., Lu J., Xiang Q., Liu Q. MicroRNA-218 acts by repressing TNFR1-mediated activation of NF-kappaB, which is involved in MUC5AC hyper-production and inflammation in smoking-induced bronchiolitis of COPD. Toxicol. Lett. 2017;280:171–180. doi: 10.1016/j.toxlet.2017.08.079. [DOI] [PubMed] [Google Scholar]
  • 24.Wang D., He S., Liu B., Liu C. MiR-27-3p regulates TLR2/4-dependent mouse alveolar macrophage activation by targetting PPARgamma. Clin. Sci. 2018;132:943–958. doi: 10.1042/CS20180083. [DOI] [PubMed] [Google Scholar]
  • 25.Shen W., Liu J., Fan M., Wang S., Zhang Y., Wen L., Wang R., Wei W., Li N., Zhang Y., Zhao G. MiR-3202 protects smokers from chronic obstructive pulmonary disease through inhibiting FAIM2: an in vivo and in vitro study. Exp. Cell Res. 2018;362:370–377. doi: 10.1016/j.yexcr.2017.11.038. [DOI] [PubMed] [Google Scholar]
  • 26.Soliman H., Toso E.A., Darwish I.E., Ali S.M., Kyba M. Antiapoptotic Protein FAIM2 is targeted by miR-3202, and DUX4 via TRIM21, leading to cell death and defective myogenesis. Cell Death Dis. 2022;13:405. doi: 10.1038/s41419-022-04804-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gu W., Yuan Y., Yang H., Wu H., Wang L., Tang Z., Li Q. Role of miR-195 in cigarette smoke-induced chronic obstructive pulmonary disease. Int. Immunopharmacol. 2018;55:49–54. doi: 10.1016/j.intimp.2017.11.030. [DOI] [PubMed] [Google Scholar]
  • 28.Jafarzadeh A., Naseri A., Shojaie L., Nemati M., Jafarzadeh S., Bannazadeh B.H., Hamblin M.R., Akhlagh S.A., Mirzaei H. MicroRNA-155 and antiviral immune responses. Int. Immunopharmacol. 2021;101 doi: 10.1016/j.intimp.2021.108188. [DOI] [PubMed] [Google Scholar]
  • 29.De Smet E.G., Van Eeckhoutte H.P., Avila C.F., Blomme E., Verhamme F.M., Provoost S., Verleden S.E., Venken K., Maes T., Joos G.F., Mestdagh P., Brusselle G.G., Bracke K.R. The role of miR-155 in cigarette smoke-induced pulmonary inflammation and COPD. Mucosal Immunol. 2020;13:423–436. doi: 10.1038/s41385-019-0241-6. [DOI] [PubMed] [Google Scholar]
  • 30.Barnes P.J., Baker J., Donnelly L.E. Cellular senescence as a mechanism and target in chronic lung diseases. Am J. Resp. Crit. Care. 2019;200:556–564. doi: 10.1164/rccm.201810-1975TR. [DOI] [PubMed] [Google Scholar]
  • 31.Baker J.R., Vuppusetty C., Colley T., Papaioannou A.I., Fenwick P., Donnelly L., Ito K., Barnes P.J. Oxidative stress dependent microRNA-34a activation via PI3Kalpha reduces the expression of sirtuin-1 and sirtuin-6 in epithelial cells. Sci. Rep.-UK. 2016;6 doi: 10.1038/srep35871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Baker J.R., Vuppusetty C., Colley T., Hassibi S., Fenwick P.S., Donnelly L.E., Ito K., Barnes P.J. MicroRNA-570 is a novel regulator of cellular senescence and inflammaging. Faseb. J. 2019;33:1605–1616. doi: 10.1096/fj.201800965R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bertoli G., Cava C., Castiglioni I. MicroRNAs: new biomarkers for diagnosis, prognosis, therapy prediction and therapeutic tools for breast cancer. Theranostics. 2015;5:1122–1143. doi: 10.7150/thno.11543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yu Y., Nangia-Makker P., Farhana L., G R.S., Levi E., Majumdar A.P. miR-21 and miR-145 cooperation in regulation of colon cancer stem cells. Mol. Cancer. 2015;14:98. doi: 10.1186/s12943-015-0372-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang W.F., Liu S.Y., Qi Z.F., Lv Z.H., Ding H.R., Zhou W.J. MiR-145 targeting BNIP3 reduces apoptosis of chondrocytes in osteoarthritis through Notch signaling pathway. Eur. Rev. Med. Pharmaco. 2020;24:8263–8272. doi: 10.26355/eurrev_202008_22622. [DOI] [PubMed] [Google Scholar]
  • 36.Dang X., Yang L., Guo J., Hu H., Li F., Liu Y., Pang Y. miR-145-5p is associated with smoke-related chronic obstructive pulmonary disease via targeting KLF5. Chem. Biol. Interact. 2019;300:82–90. doi: 10.1016/j.cbi.2019.01.011. [DOI] [PubMed] [Google Scholar]
  • 37.Dutta R.K., Chinnapaiyan S., Rasmussen L., Raju S.V., Unwalla H.J. A neutralizing aptamer to TGFBR2 and miR-145 antagonism rescue cigarette smoke- and TGF-beta-mediated CFTR expression. Mol. Ther. 2019;27:442–455. doi: 10.1016/j.ymthe.2018.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Au Y.C., Co N.N., Tsuruga T., Yeung T.L., Kwan S.Y., Leung C.S., Li Y., Lu E.S., Kwan K., Wong K.K., Schmandt R., Lu K.H., Mok S.C. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 2016;7 doi: 10.1038/ncomms11150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lampis A., Carotenuto P., Vlachogiannis G., Cascione L., Hedayat S., Burke R., Clarke P., Bosma E., Simbolo M., Scarpa A., Yu S., Cole R., Smyth E., Mateos J.F., Begum R., Hezelova B., Eltahir Z., Wotherspoon A., Fotiadis N., Bali M.A., Nepal C., Khan K., Stubbs M., Hahne J.C., Gasparini P., Guzzardo V., Croce C.M., Eccles S., Fassan M., Cunningham D., Andersen J.B., Workman P., Valeri N., Braconi C. MIR21 drives resistance to heat shock protein 90 inhibition in cholangiocarcinoma. Gastroenterology. 2018;154:1066–1079.e5. doi: 10.1053/j.gastro.2017.10.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wang C., Peng R., Zeng M., Zhang Z., Liu S., Jiang D., Lu Y., Zou F. An autoregulatory feedback loop of miR-21/VMP1 is responsible for the abnormal expression of miR-21 in colorectal cancer cells. Cell Death Dis. 2020;11:1067. doi: 10.1038/s41419-020-03265-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Xu H., Ling M., Xue J., Dai X., Sun Q., Chen C., Liu Y., Zhou L., Liu J., Luo F., Bian Q., Liu Q. Exosomal microRNA-21 derived from bronchial epithelial cells is involved in aberrant epithelium-fibroblast cross-talk in COPD induced by cigarette smoking. Theranostics. 2018;8:5419–5433. doi: 10.7150/thno.27876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kim R.Y., Sunkara K.P., Bracke K.R., Jarnicki A.G., Donovan C., Hsu A.C., Ieni A., Beckett E.L., Galvao I., Wijnant S., Ricciardolo F.L., Di Stefano A., Haw T.J., Liu G., Ferguson A.L., Palendira U., Wark P.A., Conickx G., Mestdagh P., Brusselle G.G., Caramori G., Foster P.S., Horvat J.C., Hansbro P.M. A microRNA-21-mediated SATB1/S100A9/NF-kappaB axis promotes chronic obstructive pulmonary disease pathogenesis. Sci. Transl. Med. 2021;13 doi: 10.1126/scitranslmed.aav7223. [DOI] [PubMed] [Google Scholar]
  • 43.Quinn J.J., Chang H.Y. Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 2016;17:47–62. doi: 10.1038/nrg.2015.10. [DOI] [PubMed] [Google Scholar]
  • 44.Bi H., Zhou J., Wu D., Gao W., Li L., Yu L., Liu F., Huang M., Adcock I.M., Barnes P.J., Yao X. Microarray analysis of long non-coding RNAs in COPD lung tissue. Inflamm. Res. 2015;64:119–126. doi: 10.1007/s00011-014-0790-9. [DOI] [PubMed] [Google Scholar]
  • 45.Zhang H., Sun D., Li D., Zheng Z., Xu J., Liang X., Zhang C., Wang S., Wang J., Lu W. Long non-coding RNA expression patterns in lung tissues of chronic cigarette smoke induced COPD mouse model. Sci. Rep.-UK. 2018;8:7609. doi: 10.1038/s41598-018-25702-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Amicone L., Marchetti A., Cicchini C. The lncRNA HOTAIR: a pleiotropic regulator of epithelial cell plasticity. J. Exp. Clin. Canc. Res. 2023;42:147. doi: 10.1186/s13046-023-02725-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Yi E., Lin B., Zhang Y., Wang X., Zhang J., Liu Y., Jin J., Hong W., Lin Z., Cao W., Mei X., Bai G., Li B., Zhou Y., Ran P. Smad3-mediated lncRNA HSALR1 enhances the non-classic signalling pathway of TGF-beta1 in human bronchial fibroblasts by binding to HSP90AB1. Clin. Transl. Med. 2023;13 doi: 10.1002/ctm2.1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Manevski M., Devadoss D., Long C., Singh S.P., Nasser M.W., Borchert G.M., Nair M.N., Rahman I., Sopori M., Chand H.S. Increased expression of LASI lncRNA regulates the cigarette smoke and COPD associated airway inflammation and mucous cell hyperplasia. Front. Immunol. 2022;13 doi: 10.3389/fimmu.2022.803362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yi E., Zhang J., Zheng M., Zhang Y., Liang C., Hao B., Hong W., Lin B., Pu J., Lin Z., Huang P., Li B., Zhou Y., Ran P. Long noncoding RNA IL6-AS1 is highly expressed in chronic obstructive pulmonary disease and is associated with interleukin 6 by targeting miR-149-5p and early B-cell factor 1. Clin. Transl. Med. 2021;11:e479. doi: 10.1002/ctm2.479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Devadoss D., Daly G., Manevski M., Houserova D., Hussain S.S., Baumlin N., Salathe M., Borchert G.M., Langley R.J., Chand H.S. A long noncoding RNA antisense to ICAM-1 is involved in allergic asthma associated hyperreactive response of airway epithelial cells. Mucosal Immunol. 2021;14:630–639. doi: 10.1038/s41385-020-00352-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ross D., Siegel D. Functions of NQO1 in cellular protection and CoQ(10) metabolism and its potential role as a redox sensitive molecular switch. Front. Physiol. 2017;8:595. doi: 10.3389/fphys.2017.00595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhang H., Guan R., Zhang Z., Li D., Xu J., Gong Y., Chen X., Lu W. LncRNA Nqo1-AS1 attenuates cigarette smoke-induced oxidative stress by upregulating its natural antisense transcript Nqo1. Front. Pharmacol. 2021;12 doi: 10.3389/fphar.2021.729062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Xia H., Xue J., Xu H., Lin M., Shi M., Sun Q., Xiao T., Dai X., Wu L., Li J., Xiang Q., Tang H., Bian Q., Liu Q. Andrographolide antagonizes the cigarette smoke-induced epithelial-mesenchymal transition and pulmonary dysfunction through anti-inflammatory inhibiting HOTAIR. Toxicology. 2019;422:84–94. doi: 10.1016/j.tox.2019.05.009. [DOI] [PubMed] [Google Scholar]
  • 54.Dai Z., Liu X., Zeng H., Chen Y. Long noncoding RNA HOTAIR facilitates pulmonary vascular endothelial cell apoptosis via DNMT1 mediated hypermethylation of Bcl-2 promoter in COPD. Resp. Res. 2022;23:356. doi: 10.1186/s12931-022-02234-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zheng M., Hong W., Gao M., Yi E., Zhang J., Hao B., Liang C., Li X., Li C., Ye X., Liao B., He F., Zhou Y., Li B., Ran P. Long noncoding RNA COPDA1 promotes airway smooth muscle cell proliferation in chronic obstructive pulmonary disease. AM J Resp. Cell Mol. 2019;61:584–596. doi: 10.1165/rcmb.2018-0269OC. [DOI] [PubMed] [Google Scholar]
  • 56.Liu G., Philp A.M., Corte T., Travis M.A., Schilter H., Hansbro N.G., Burns C.J., Eapen M.S., Sohal S.S., Burgess J.K., Hansbro P.M. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol. Therapeut. 2021;225 doi: 10.1016/j.pharmthera.2021.107839. [DOI] [PubMed] [Google Scholar]
  • 57.Liu P., Zhang H., Zeng H., Meng Y., Gao H., Zhang M., Zhao L. LncRNA CASC2 is involved in the development of chronic obstructive pulmonary disease via targeting miR-18a-5p/IGF1 axis. Ther. Adv. Respir. Dis. 2021;15 doi: 10.1177/17534666211028072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Shen Q., Zheng J., Wang X., Hu W., Jiang Y., Jiang Y. LncRNA SNHG5 regulates cell apoptosis and inflammation by miR-132/PTEN axis in COPD. Biomed. Pharmacother. 2020;126 doi: 10.1016/j.biopha.2020.110016. [DOI] [PubMed] [Google Scholar]
  • 59.Hao W., Lin F., Shi H., Guan Z., Jiang Y. Long non-coding RNA OIP5-AS1 regulates smoke-related chronic obstructive pulmonary disease via targeting micro RNA -410-3p/IL-13. Bioengineered. 2021;12:11664–11676. doi: 10.1080/21655979.2021.2000199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zhao S., Lin C., Yang T., Qian X., Lu J., Cheng J. Expression of long non-coding RNA LUCAT1 in patients with chronic obstructive pulmonary disease and its potential functions in regulating cigarette smoke extract-induced 16HBE cell proliferation and apoptosis. J. Clin. Lab. Anal. 2021;35 doi: 10.1002/jcla.23823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zong D., Liu X., Li J., Long Y., Ouyang R., Chen Y. LncRNA-CCAT1/miR-152-3p is involved in CSE-induced inflammation in HBE cells via regulating ERK signaling pathway. Int. Immunopharmacol. 2022;109 doi: 10.1016/j.intimp.2022.108818. [DOI] [PubMed] [Google Scholar]
  • 62.Mei J., Zhang Y., Lu S., Wang J. Long non-coding RNA NNT-AS1 regulates proliferation, apoptosis, inflammation and airway remodeling of chronic obstructive pulmonary disease via targeting miR-582-5p/FBXO11 axis. Biomed. Pharmacother. 2020;129 doi: 10.1016/j.biopha.2020.110326. [DOI] [PubMed] [Google Scholar]
  • 63.Doiron D., de Hoogh K., Probst-Hensch N., Fortier I., Cai Y., De Matteis S., Hansell A.L. Air pollution, lung function and COPD: results from the population-based UK Biobank study. Eur. Respir. J. 2019;54 doi: 10.1183/13993003.02140-2018. [DOI] [PubMed] [Google Scholar]
  • 64.He F., Wang N., Yu X., Zheng Y., Liu Q., Chen Q., Pu J., Li N., Zou W., Li B., Ran P. GATA3/long noncoding RNA MHC-R regulates the immune activity of dendritic cells in chronic obstructive pulmonary disease induced by air pollution particulate matter. J. Hazard Mater. 2022;438 doi: 10.1016/j.jhazmat.2022.129459. [DOI] [PubMed] [Google Scholar]
  • 65.Lee K.Y., Ho S.C., Sun W.L., Feng P.H., Lin C.W., Chen K.Y., Chuang H.C., Tseng C.H., Chen T.T., Wu S.M. Lnc-IL7R alleviates PM(2.5)-mediated cellular senescence and apoptosis through EZH2 recruitment in chronic obstructive pulmonary disease. Cell Biol. Toxicol. 2022;38:1097–1120. doi: 10.1007/s10565-022-09709-1. [DOI] [PubMed] [Google Scholar]
  • 66.Chen L., Shan G. CircRNA in cancer: fundamental mechanism and clinical potential. Cancer Lett. 2021;505:49–57. doi: 10.1016/j.canlet.2021.02.004. [DOI] [PubMed] [Google Scholar]
  • 67.Chen L., Huang C., Shan G. Circular RNAs in physiology and non-immunological diseases. Trends Biochem. Sci. 2022;47:250–264. doi: 10.1016/j.tibs.2021.11.004. [DOI] [PubMed] [Google Scholar]
  • 68.Liu P., Wang Y., Zhang N., Zhao X., Li R., Wang Y., Chen C., Wang D., Zhang X., Chen L., Zhao D. Comprehensive identification of RNA transcripts and construction of RNA network in chronic obstructive pulmonary disease. Resp. Res. 2022;23:154. doi: 10.1186/s12931-022-02069-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Vallejo J., Saigusa R., Gulati R., Armstrong S.S., Suryawanshi V., Alimadadi A., Durant C.P., Ghosheh Y., Roy P., Ehinger E., Pattarabanjird T., Hanna D.B., Landay A.L., Tracy R.P., Lazar J.M., Mack W.J., Weber K.M., Adimora A.A., Hodis H.N., Tien P.C., Ofotokun I., Heath S.L., Shemesh A., McNamara C.A., Lanier L.L., Hedrick C.C., Kaplan R.C., Ley K. Combined protein and transcript single-cell RNA sequencing in human peripheral blood mononuclear cells. BMC Biol. 2022;20:193. doi: 10.1186/s12915-022-01382-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Li Q., Li S., Xu C., Zhao J., Hou L., Jiang F., Zhu Z., Wang Y., Tian L. microRNA-149-5p mediates the PM(2.5)-induced inflammatory response by targeting TAB2 via MAPK and NF-kappaB signaling pathways in vivo and in vitro. Cell Biol. Toxicol. 2023;39:703–717. doi: 10.1007/s10565-021-09638-5. [DOI] [PubMed] [Google Scholar]
  • 71.Guo P., Lu J., Lei Y. Significant role of circRNA BBS9 in chronic obstructive pulmonary disease via miRNA-103a-3p/BCL2L13. BMC Pulm. Med. 2023;23:257. doi: 10.1186/s12890-023-02540-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wang C., Tang Y., Hou H., Su C., Gao Y., Yang X. CIRC_0026466 knockdown protects human bronchial epithelial cells from cigarette smoke extract-induced injury by promoting the MIR-153-3P/TRAF6/NF-KappaB pathway. Shock. 2023;60:121–129. doi: 10.1097/SHK.0000000000002141. [DOI] [PubMed] [Google Scholar]
  • 73.Xue M., Peng N., Zhu X., Zhang H. Hsa_circ_0006872 promotes cigarette smoke-induced apoptosis, inflammation and oxidative stress in HPMECs and BEAS-2B cells through the miR-145-5p/NF-kappaB axis. Biochem. Bioph. Res. Co. 2021;534:553–560. doi: 10.1016/j.bbrc.2020.11.044. [DOI] [PubMed] [Google Scholar]
  • 74.Li M., Hua Q., Shao Y., Zeng H., Liu Y., Diao Q., Zhang H., Qiu M., Zhu J., Li X., Ling Y., Zhang R., Jiang Y. Circular RNA circBbs9 promotes PM(2.5)-induced lung inflammation in mice via NLRP3 inflammasome activation. Environ. Int. 2020;143 doi: 10.1016/j.envint.2020.105976. [DOI] [PubMed] [Google Scholar]
  • 75.Zhou L., Wu B., Yang J., Wang B., Pan J., Xu D., Du C. Knockdown of circFOXO3 ameliorates cigarette smoke-induced lung injury in mice. Resp. Res. 2021;22:294. doi: 10.1186/s12931-021-01883-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Du Y., Ding Y., Shi T., He W., Mei Z., Feng X., Zhang X., Jie Z. Suppression of circXPO1 attenuates cigarette smoke-induced inflammation and cellular senescence of alveolar epithelial cells in chronic obstructive pulmonary disease. Int. Immunopharmacol. 2022;111 doi: 10.1016/j.intimp.2022.109086. [DOI] [PubMed] [Google Scholar]
  • 77.Xia H., Wu Y., Zhao J., Cheng C., Lin J., Yang Y., Lu L., Xiang Q., Bian T., Liu Q. N6-Methyladenosine-modified circSAV1 triggers ferroptosis in COPD through recruiting YTHDF1 to facilitate the translation of IREB2. Cell Death Differ. 2023;30:1293–1304. doi: 10.1038/s41418-023-01138-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Salzano S., Checconi P., Hanschmann E.M., Lillig C.H., Bowler L.D., Chan P., Vaudry D., Mengozzi M., Coppo L., Sacre S., Atkuri K.R., Sahaf B., Herzenberg L.A., Herzenberg L.A., Mullen L., Ghezzi P. Linkage of inflammation and oxidative stress via release of glutathionylated peroxiredoxin-2, which acts as a danger signal. P Natl. Acad. Sci. USA. 2014;111:12157–12162. doi: 10.1073/pnas.1401712111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Meng Q., Wang J., Cui J., Li B., Wu S., Yun J., Aschner M., Wang C., Zhang L., Li X., Chen R. Prediction of COPD acute exacerbation in response to air pollution using exosomal circRNA profile and Machine learning. Environ. Int. 2022;168 doi: 10.1016/j.envint.2022.107469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ruger J., Ioannou S., Castanotto D., Stein C.A. Oligonucleotides to the (gene) rescue: FDA approvals 2017-2019. Trends Pharmacol. Sci. 2020;41:27–41. doi: 10.1016/j.tips.2019.10.009. [DOI] [PubMed] [Google Scholar]
  • 81.Winkle M., El-Daly S.M., Fabbri M., Calin G.A. Noncoding RNA therapeutics - challenges and potential solutions. Nat. Rev. Drug Discov. 2021;20:629–651. doi: 10.1038/s41573-021-00219-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Nemeth K., Bayraktar R., Ferracin M., Calin G.A. Non-coding RNAs in disease: from mechanisms to therapeutics. Nat. Rev. Genet. 2024;25:211–232. doi: 10.1038/s41576-023-00662-1. [DOI] [PubMed] [Google Scholar]
  • 83.Jadhav V., Vaishnaw A., Fitzgerald K., Maier M.A. RNA interference in the era of nucleic acid therapeutics. Nat. Biotechnol. 2024;42:394–405. doi: 10.1038/s41587-023-02105-y. [DOI] [PubMed] [Google Scholar]
  • 84.Winkle M., El-Daly S.M., Fabbri M., Calin G.A. Noncoding RNA therapeutics - challenges and potential solutions. Nat. Rev. Drug Discov. 2021;20:629–651. doi: 10.1038/s41573-021-00219-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zhang M., Lu H., Xie L., Liu X., Cun D., Yang M. Inhaled RNA drugs to treat lung diseases: disease-related cells and nano-bio interactions. Adv. Drug Deliv. Rev. 2023;203 doi: 10.1016/j.addr.2023.115144. [DOI] [PubMed] [Google Scholar]
  • 86.Stenton G.R., Kim M.K., Nohara O., Chen C.F., Hirji N., Wills F.L., Gilchrist M., Hwang P.H., Park J.G., Finlay W., Jones R.L., Befus A.D., Schreiber A.D. Aerosolized Syk antisense suppresses Syk expression, mediator release from macrophages, and pulmonary inflammation. J. Immunol. 2000;164:3790–3797. doi: 10.4049/jimmunol.164.7.3790. [DOI] [PubMed] [Google Scholar]
  • 87.Xu C.X., Jere D., Jin H., Chang S.H., Chung Y.S., Shin J.Y., Kim J.E., Park S.J., Lee Y.H., Chae C.H., Lee K.H., Beck G.J., Cho C.S., Cho M.H. Poly(ester amine)-mediated, aerosol-delivered Akt1 small interfering RNA suppresses lung tumorigenesis. Am J. Resp. Crit. Care. 2008;178:60–73. doi: 10.1164/rccm.200707-1022OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Xue W., Dahlman J.E., Tammela T., Khan O.F., Sood S., Dave A., Cai W., Chirino L.M., Yang G.R., Bronson R., Crowley D.G., Sahay G., Schroeder A., Langer R., Anderson D.G., Jacks T. Small RNA combination therapy for lung cancer. P Natl. Acad. Sci. USA. 2014;111:E3553–E3561. doi: 10.1073/pnas.1412686111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.van Zandwijk N., Pavlakis N., Kao S.C., Linton A., Boyer M.J., Clarke S., Huynh Y., Chrzanowska A., Fulham M.J., Bailey D.L., Cooper W.A., Kritharides L., Ridley L., Pattison S.T., MacDiarmid J., Brahmbhatt H., Reid G. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017;18:1386–1396. doi: 10.1016/S1470-2045(17)30621-6. [DOI] [PubMed] [Google Scholar]
  • 90.Li J., Zhu L., Kwok H.F. Nanotechnology-based approaches overcome lung cancer drug resistance through diagnosis and treatment. Drug Resist. Updates. 2023;66 doi: 10.1016/j.drup.2022.100904. [DOI] [PubMed] [Google Scholar]
  • 91.Dua K., Wadhwa R., Singhvi G., Rapalli V., Shukla S.D., Shastri M.D., Gupta G., Satija S., Mehta M., Khurana N., Awasthi R., Maurya P.K., Thangavelu L., S R., Tambuwala M.M., Collet T., Hansbro P.M., Chellappan D.K. The potential of siRNA based drug delivery in respiratory disorders: recent advances and progress. DRUG DEVELOP RES. 2019;80:714–730. doi: 10.1002/ddr.21571. [DOI] [PubMed] [Google Scholar]
  • 92.Dong J., Liao W., Peh H.Y., Chan T.K., Tan W.S., Li L., Yong A., Wong W.S. Ribosomal protein S3 gene silencing protects against experimental allergic asthma. Brit. J. Pharmacol. 2017;174:540–552. doi: 10.1111/bph.13717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Dong J., Liao W., Tan L.H., Yong A., Peh W.Y., Wong W. Gene silencing of receptor-interacting protein 2 protects against cigarette smoke-induced acute lung injury. Pharmacol. Res. 2019;139:560–568. doi: 10.1016/j.phrs.2018.10.016. [DOI] [PubMed] [Google Scholar]
  • 94.Kai Y., Tomoda K., Yoneyama H., Yoshikawa M., Kimura H. RNA interference targeting carbohydrate sulfotransferase 3 diminishes macrophage accumulation, inhibits MMP-9 expression and promotes lung recovery in murine pulmonary emphysema. Resp. Res. 2015;16:146. doi: 10.1186/s12931-015-0310-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Liu Q., Li H., Wang Q., Zhang Y., Wang W., Dou S., Xiao W. Increased expression of TROP2 in airway basal cells potentially contributes to airway remodeling in chronic obstructive pulmonary disease. Resp. Res. 2016;17:159. doi: 10.1186/s12931-016-0463-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Reimann S., Fink L., Wilhelm J., Hoffmann J., Bednorz M., Seimetz M., Dessureault I., Troesser R., Ghanim B., Klepetko W., Seeger W., Weissmann N., Kwapiszewska G. Increased S100A4 expression in the vasculature of human COPD lungs and murine model of smoke-induced emphysema. Resp. Res. 2015;16:127. doi: 10.1186/s12931-015-0284-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mei D., Tan W., Tay Y., Mukhopadhyay A., Wong W. Therapeutic RNA strategies for chronic obstructive pulmonary disease. Trends Pharmacol. Sci. 2020;41:475–486. doi: 10.1016/j.tips.2020.04.007. [DOI] [PubMed] [Google Scholar]

Articles from Non-coding RNA Research are provided here courtesy of KeAi Publishing

RESOURCES