Next Article in Journal
Biomineral-Based Composite Materials in Regenerative Medicine
Next Article in Special Issue
Calcium Regulation of Connexin Hemichannels
Previous Article in Journal
NaV1.8 as Proarrhythmic Target in a Ventricular Cardiac Stem Cell Model
Previous Article in Special Issue
Gap Junction Channel Regulation: A Tale of Two Gates—Voltage Sensitivity of the Chemical Gate and Chemical Sensitivity of the Fast Voltage Gate
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Perspective and Therapeutic Potential of the Noncoding RNA–Connexin Axis

State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(11), 6146; https://doi.org/10.3390/ijms25116146
Submission received: 6 April 2024 / Revised: 27 May 2024 / Accepted: 30 May 2024 / Published: 2 June 2024

Abstract

:
Noncoding RNAs (ncRNAs) are a class of nucleotide sequences that cannot be translated into peptides. ncRNAs can function post-transcriptionally by splicing complementary sequences of mRNAs or other ncRNAs or by directly engaging in protein interactions. Over the past few decades, the pervasiveness of ncRNAs in cell physiology and their pivotal roles in various diseases have been identified. One target regulated by ncRNAs is connexin (Cx), a protein that forms gap junctions and hemichannels and facilitates intercellular molecule exchange. The aberrant expression and misdistribution of connexins have been implicated in central nervous system diseases, cardiovascular diseases, bone diseases, and cancer. Current databases and technologies have enabled researchers to identify the direct or indirect relationships between ncRNAs and connexins, thereby elucidating their correlation with diseases. In this review, we selected the literature published in the past five years concerning disorders regulated by ncRNAs via corresponding connexins. Among it, microRNAs that regulate the expression of Cx43 play a crucial role in disease development and are predominantly reviewed. The distinctive perspective of the ncRNA–Cx axis interprets pathology in an epigenetic manner and is expected to motivate research for the development of biomarkers and therapeutics.

1. Introduction

Noncoding RNAs (ncRNAs) consist of microRNAs (miRNAs), transfer RNA (tRNA)-derived small RNAs (tsRNAs), PIWI-interacting RNAs (piRNAs), and long noncoding RNAs (lncRNAs); among lncRNAs, distinct subtypes have been identified, such as pseudogene-derived lncRNAs and circular RNAs (circRNAs) [1]. Although ncRNAs do not encode proteins, they are involved in essential biological processes, and their dysregulation contributes to various diseases. The overexpression of miRNAs in cancers can function as oncogenes and promote cancer development by negatively regulating tumor suppressor genes and/or genes that control cell differentiation or apoptosis, whereas the underexpression of miRNAs in cancers can function as tumor suppressor genes that can inhibit cancers by regulating oncogenes and/or genes that control cell differentiation or apoptosis [1]. Several ncRNAs have been identified to participate in atherogenesis during the development of atherosclerosis [2]. Through epigenetic mechanisms, ncRNAs affect neurogenesis, synaptic plasticity, and neurotransmitter systems to regulate depression [3,4]. Owing to the close relationship between ncRNAs and diseases, therapeutic approaches targeting ncRNAs have received growing interest and are considered promising [5,6,7].
Human genes encode 21 kinds of connexin (Cx) proteins comprising four transmembrane domains, two extracellular loops, and one cytoplasmic loop [8]. Six connexins form a hemichannel termed a connexon [8]. Connexons that dock between the membranes of two adjacent cells form intercellular channels termed gap junctions, which aggregate into plaques [8]. Gap junctions allow the transfer of ions and small molecules across cell membranes and regulate intercellular communication, cell differentiation, and cell proliferation [9]. Hemichannels mediate the transfer of small molecules between the cytoplasm and extracellular environment and primarily function under pathological conditions [10]. Connexin gene mutations and connexin dysregulation are implicated in central nervous system (CNS) disorders and cardiovascular diseases [11,12,13]. Among all connexins, connexin 43 (Cx43) is the most studied, and potential therapeutic approaches based on Cx43 have been widely implicated [14,15,16]. (See Table S1 for a summary of connexin genes and corresponding proteins.)
In this review, we focus on ncRNA–Cx axis dysregulation in diseases and pathological processes, as well as ncRNA delivery through gap junctions. Diseases are stratified according to organ systems, and the ncRNA–Cx axis summarized in this review has been documented in scientific publications over the past five years. miRNAs directly bind to the mRNAs of connexins, inhibiting mRNA translation. In addition, lncRNAs or circRNAs can be spliced with miRNAs to interrupt the effects of miRNAs. Moreover, ncRNAs interact with proteins to regulate Cx function. The mechanisms involved in the ncRNA–Cx axis are shown in Figure 1.

2. Disorders Modulated by Noncoding RNAs through Relevant Connexins

2.1. Disorders in the CNS

The ncRNAs presented herein may regulate connexin expression or participate in the modification of connexins. Recent studies have implicated the expression and modification of connexins in depression, brain injury, neurodegenerative diseases, and numerous pathophysiological mechanisms, including apoptosis, neuroinflammation, and cellular proliferation and migration.
Gap junctions assembled from Cx43 play vital roles in maintaining astrocyte networks and consequently coordinate neuronal activity and synaptic transmission [17,18]. The downregulation of Cx43 reduces gap junction coupling, which alters the transmission of monoamines, glutamate, calcium, ATP, and cAMP and affects neuronal activity and synaptic function [19]. Decreased Cx43 expression and gap junction dysfunction in an animal model of depression were reversed by antidepressant treatment [20]. Moreover, astrocytic Cx43 is widely involved in CNS regulation, contributing to gap junction dysfunction and neuroinflammation in depression [16]. In a study using an unpredictable chronic mild stress (UCMS)-induced mouse model of depression, miR-221/222 directly bound to the 3′ untranslated regions (3′ UTR) of Cx43 mRNA and downregulated Cx43 protein levels; this could be reversed by genistein treatment, subsequently ameliorating depression-like behaviors in the mice [21]. miR-205 also targeted the Gja1 gene, which encodes the Cx43 protein [22]. The administration of Mahonia alkaloids reduced miR-205 content and consequently improved Cx43 levels, alleviating depression in reserpine-treated rats [22]. miR-106a was confirmed to bind to the 3′ UTR of Cx43 mRNA, and an miR-106a antagonist could repress apoptosis, alleviate oxidative stress, and enhance Cx43 protein content in glucose oxidase-treated rat cochlear marginal cells [23]. Furthermore, in gentamicin-induced rat models, the miR-106a antagonist exerted therapeutic effects on the pathological state of sensorineural hearing loss by regulating Cx43 [23].
Although not an immediate binding target, miR-182 inhibition tends to reverse Cx43 degradation in injured astrocyte cultures, indicating that miR-182 may decrease Cx43 expression [24]. Cx43 has been phosphorylated by extracellular-signal-regulated kinase (ERK)1/2 in rats subjected to cerebral ischemia [25]. In vitro models have shown that the overexpression of the miR-302/367 cluster has suppressed the phosphorylation of both ERK1/2 and Cx43, and in vivo studies revealed that elevated expression of the miR-302/367 cluster improved cognitive function and neuronal damage in rats after traumatic brain injury [26]. These findings suggest that the miR-302/367 cluster regulates ERK signaling to inhibit Cx43 phosphorylation and mitigates brain damage [26]. Jullienne et al. found that aquaporin 4 (AQP4) and Cx43 were significantly decreased after intracortical injection of siAQP4, and miR-224 and miR-19a were significantly increased [27]. They reported that this may be responsible for losses in astrocyte connectivity and tissue water mobility [27]. These off-target effects of siAQP4 suggest that miR-224 and miR-19a may be involved in amyotrophic lateral sclerosis, which shows concomitant and aberrant overexpression of AQP4 and Cx43 [27,28].
lncRNAs also function as the regulators of several CNS diseases [29,30]. lncRNA Pnky has a modulatory effect on Cx43 during neural stem cell migration [31]. lncRNA Pnky overexpression has enhanced the protein levels of some critical regulators of neural stem cell migration, including Cx43, whereas lncRNA Pnky depletion has decreased them in vitro. Consistently, cells overexpressing lncRNA Pnky transplanted into mice exhibited greater migration distances in the spinal canal than those in which lncRNA Pnky was depleted [31]. Knockdown of lncRNA NEAT1 downregulated Cx32 expression via the promotion of miR-1301-3p, a suppressor of Gjb1 (Cx32); suppressed the α-synuclein-induced inflammatory response; and reduced apoptosis in a cellular Parkinson’s disease (PD) model [32]. Abnormal α-synuclein aggregation contributes to neuroinflammation and is a key player in the pathology of PD [33]. This miR-1301-3p/Gjb1 signaling pathway, which is regulated by lncRNA NEAT1, offers potential therapeutic targets for PD treatment.
However, Cx43 enrichment does not always provide benefits. In cultured γ-radiation-induced astrocytes, miR-206 significantly lowered Cx43 expression and inflammatory cytokine release. In contrast, the Cx43-plasmid had the opposite effect, revealing that miR-206 relieves neuroinflammation by downregulating Cx43 [34]. Astrocytes transfected with miR-301a-3p, which can bind to Cx43 and downregulate its expression, have exhibited increased proliferative potential and decreased apoptosis; however, astrocytes overexpressing the Cx43 protein have exhibited high cell death rates [35].
Cx43 overexpression is widely observed in the inflammation, apoptosis, and proliferation associated with cancer cells. The detrimental role of Cx43 may originate from its channel-dependent and channel-independent functions. Cell death could be caused by gap junction-mediated bystander killing [36]. In addition, although hemichannel-mediated diseases have not yet been proven, Caruso et al. reviewed the molecular mechanisms of Cx43 hemichannel opening and their contribution to neuroinflammation and oxidative stress [37]. Activation of the inflammasome complex, NF-κB signaling pathways, and alterations in intracellular calcium levels can induce the opening of Cx43 hemichannels, thereby facilitating the release of ATP [37]. Dysregulated secretion of ATP and other gliotransmitters through Cx43 hemichannels disrupts CNS homeostasis [37]. The non-canonical functions of Cx43 include participation in gene transcription, development, and scaffolding complexes, as well as facilitating glioma invasion via a channel-independent pathway [38,39]. The conundrum of connexins or gap junctions serving as protectors or risk factors is determined by intercellular signaling [40].
Connexins have long been considered drug targets for neurological and neuropsychiatric disorders [17]. As shown in Table 1, recently established interactions between ncRNAs and connexins may augment the array of therapeutic strategies available for managing CNS diseases.

2.2. Disorders in the Cardiovascular System

Cx43 is the most abundant and extensively studied connexin in the cardiovascular system. Downregulated and altered distribution of Cx43 levels have been observed in the pathologies of several heart diseases [13]. Studies involving antisense peptides that knock down Cx43 expression or block Cx43 channels to alleviate ischemia/reperfusion injury have demonstrated the adverse effects of Cx43 [16,41]. Recently, ncRNAs were implicated in the regulation of connexins in the abovementioned phenomena. miR-1, miR-206, and lncRNAs are highly correlated with the regulation of Cx43, thereby participating in cardiovascular disorders [42,43,44].
For example, Cx43 may be a key target of miR-1 in the regulation of cardiovascular diseases. A recent study has identified the potential binding sites of miR-1 in the 3′ UTR of Cx43 [42]. The overexpression of miR-1 in mouse cardiomyocytes induced atrioventricular blocks accompanied by reduced Cx43 protein expression [45]. Under oxidative stress, miR-1 levels increase and Cx43 expression decreases [46,47]. The downregulation of Cx43 was also correlated with increased miR-1 levels in in vitro models of ischemia/hypoxia, and treatment with miR-1 inhibitors elevated Cx43 expression [48,49]. Furthermore, an endothelial dysfunction model induced by the transfection of miR-1 exhibited reduced Cx43 protein levels [50].
miR-206 is associated with arrhythmia and thrombosis by directly binding to Cx43 mRNAs, resulting in decreased Cx43 protein levels [43]. Conversely, inhibiting miR-206 has upregulated Cx43 levels and alleviated ischemia–reperfusion arrhythmia [43]. Consistent with these findings, miR-206 overexpression caused cardiac arrhythmia in mice by downregulating Cx43 [51]. miR-206 modulates cellular autophagy by targeting Cx43 mRNA [52]. The upregulation of miR-206 reduces Cx43 expression and increases autophagy-related proteins; Gja1 silencing exerts the same effects [52].
The involvement of other miRNAs and lncRNAs in cardiovascular disease is summarized in Table 2. Not only a direct target, Cx43 is a downstream factor modulated by the ncRNA–protein/miRNA axis. Signaling factors, including the platelet-derived growth factor [53] and Wnt3a [54], regulate Cx43 via miRNAs. Notably, ncRNA/protein complexes also have extensive functions. lncRNA CCRR binds to the Cx43-interacting protein CIP85 and prevents the endocytic trafficking of Cx43, thereby controlling the membrane distribution of Cx43 and gap junction functioning [44]. Similarly, lncRNA Gm2694 interacts with the chaperone protein GRP78 to trigger an unfolded protein response, which is associated with endoplasmic reticulum disruption [55]. miRNAs that interact with proteins are involved in the crucial mechanism of RNA splicing [56].

2.3. Disorders in Other Systems

Cx43 upregulation is associated with the inflammatory response and apoptosis observed during the development of osteoarthritis (OA), intervertebral disc degeneration, and ossification [64,65,66,67]. In vitro, chondrocytes induced by interleukin-1β have exhibited Cx43 overexpression, which has facilitated the upregulation of Toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), and nuclear factor κB (NF-κB) [65]. Simultaneously, miR-382-3p could suppress Cx43 and inhibit this TLR4/MyD88/NF-κB signaling pathway [65].
Ferroptosis is involved in the pathology of OA and differs from apoptosis and autophagy [68]. Cx43 overexpression aggravates ferroptosis in chondrocytes, a process rescued by miR-1 [69]. Excess Cx43 triggered by a knockdown of miR-1 could also form hemichannels that can promote the extracellular release of ATP [70]. Osteogenesis requires Cx43 expression, and upstream ncRNAs that regulate Cx43 have been identified [67,71].
Connexin26 (Cx26) and connexin30 (Cx30) form heteromeric and heterotypic gap junctions in the cochlea, and the mutations of Cx26 are responsible for congenital hearing loss [72,73]. miR-34c-5p directly binds to Cx26 mRNAs and downregulates their expression [74]. Instead of inhibiting upstream ncRNAs first, Gentile et al. knocked out Cx30 in a mouse model and investigated the miRNAs whose expressions were altered [74]. They identified the upregulated miRNAs and downstream targets linked to hearing loss [74]. This method shows that the dysregulation of connexins can cause abnormal ncRNA expression, leading to pathological processes. The ncRNAs and related connexins associated with pathological processes are summarized in Table 3.

2.4. Several Kinds of Cancer

Cell proliferation and migration in lung cancer [76], bladder cancer [77,78], and hypopharyngeal squamous cell carcinoma [79] can be suppressed by the overexpression of certain ncRNAs along with decreased Cx43 levels. This indicates the crucial role of Cx43 in the development of cancer and tumor metastasis [79,80,81]. In contrast, miRNAs are upregulated and connexins are downregulated in ovarian cancer [82], leukemia [83], gastric cancer [84], pancreatic cancer [85,86], and melanoma [87,88], suggesting an inhibitory effect of Cx43 on cancer progression.
In addition to the biochemical perspective of the ncRNA–Cx axis in cancer development, research on tumor drug resistance is worth investigating. Non-small-cell lung cancer (NSCLC) cells are more sensitive to cisplatin due to the overexpression of miR-613, and this enhanced chemosensitivity can be partially reversed by the upregulation of Gja1 [76]. This decreased drug resistance is partially owing to the lack of Cx43 and few hemichannels that may transport drug molecules out of cells [70]. However, the overexpression of miR-206 and subsequent downregulation of Cx43 enhance cisplatin resistance in ovarian cancer cells [82]. The conflicting reports on the effects of Cx43 on chemoresistance above are partly attributed to the multiple targets of miRNAs in addition to connexins. Furthermore, other molecules are implicated in this phenomenon [89,90], and the roles of connexins are tissue-specific [73]. The ncRNAs and related connexins that were identified during investigations of cancer development are summarized in Table 4.

3. The Transfer of Noncoding RNAs through Gap Junctions

In 2011, Lim et al. reported that miR-127, -197, -222, and -223 could be transported via gap junctions from the bone marrow stroma to breast cancer cells to modulate the cell cycle [93]. This phenomenon is considered one of the key discoveries showing that gap junctions directly regulate cancer [40]. Lemcke et al. summarized the co-culture systems that deliver miRNAs through gap junctions (composed of Cx43 or other connexins) in vitro [94]. The co-culture systems encompass mesenchymal stem cells, macrophages, glioma cells, cardiomyocytes, and astrocytes as donor or recipient cells for certain miRNAs [94,95]. Similar heterocellular Cx43-dependent pathways and miRNA transfer were observed in co-cultured melanoma cells and T lymphocytes [96]. Valiunas et al. transfected miR-16 and small interfering RNAs (siRNAs) into stem cells, which were then used to transfer these toxic oligonucleotides into three cancer cell lines via gap junctions to inhibit cell proliferation or induce cell death [97]. These facts have persuaded researchers to study the molecular mechanisms underlying these phenomena, particularly the molecular diameters of oligonucleotides and energy supply. Lemcke et al. proposed possible molecular mechanisms for miRNA shuttling via gap junctions based on experimental and computational data, suggesting that the transfer of miRNAs requires the assistance of the miRNA-binding protein Argonaute (AGO) and non-AGO proteins [98]. However, the present models of gap junction-dependent miRNA transport remain speculative, and novel miRNA-binding proteins must be identified to clarify the underlying mechanisms of gap junction-mediated miRNA transport [98].
In addition to mediating the transmission of miRNAs between adjacent cells, connexins also participate in extracellular vesicle (EV) functions. Cx43 has been detected in the EVs released from multiple human cell lines cultured in vitro [99]. Martins-Marques et al. isolated Cx43-containing EVs from human plasma and proposed that Cx43 participates in miRNA selection, forming functional channels at the EV surface that deliver miRNAs containing stable secondary structural elements to distant cells [100,101]. Similarly, Acuña et al. confirmed that Cx46-containing EVs could be released by breast cancer cells and proposed that Cx46 in the membranes of EVs facilitates the docking and internalization of the receptor cells [102]. Whether the miRNAs contained in EVs can be delivered to remote cells via hemichannel pairing requires further investigation.

4. Therapeutic Potential of the ncRNA–Cx Axis

ncRNA biomarkers and RNA interference (RNAi) technology have gained significant traction in clinical research. ncRNA profiles primarily serve as biomarkers to reveal predisposition to disease. During different stages of multiple sclerosis (MS), specifically clinically isolated syndrome (CIS) and relapsing–remitting MS (RRMS), serum EV miRNAs were significantly dysregulated [103]. Notably, CIS-specific serum EV miRNAs were downregulated, whereas RRMS-specific serum EV miRNAs were upregulated [103]. These MS stage-specific EV-derived miRNAs have potential predictive value [103]. Circulating miRNA profiles show features distinguishing transient ischemic attack (TIA) from acute ischemic stroke (AIS) [104]. Eleven miRNAs were differentially expressed in the sera of patients with TIA and AIS, which could help discriminate between these two diseases [104]. Clinical trials of ncRNA therapeutics have exploited RNAi technology to directly suppress target mRNA. Recently, siRNAs olpasiran [105] and lepodisiran [106], targeting lipoprotein synthesis, have emerged as effective treatments for cardiovascular disease. The siRNA agent lumasiran was the first FDA-approved prescription for primary hyperoxaluria type 1 that targets glycolate oxidase [107]. However, compared to the encouraging progress of ncRNAs and siRNAs in clinical practice, the clinical application of connexins is limited. Despite the association between connexin dysregulation and various diseases, no molecules targeting connexins are currently being tested in clinical trials.
To bridge the gap between the essential biological roles of connexins and their limited clinical utility, we suggest that connexins can be regulated by modulating the ncRNAs upstream or downstream of the connexins. Cx43 plays a key role in neuroinflammation [108,109]. Antagomirs to miR-221/222 [21], miR-205 [22], and miR-224 [27] can upregulate Cx43 protein levels in glial cells and have anti-neuroinflammatory potential. Carefully designed siRNAs and antisense oligonucleotides can suppress upstream ncRNAs, such as lncRNA ANRIL [61] to rescue Cx43 or to reduce Cx43 levels by suppressing lncRNA CCRR [44] and lncRNA MALAT1 [62]. More directly, CRISPR-Cas9 technology can be employed to edit ncRNAs, such as lncRNA Pnky, to regulate connexin gene expression. In addition, the use of EVs as drug delivery systems to target the ncRNA–Cx axis is an intriguing strategy. Cx43 can form channels on the surface of EVs and allow drug molecules to pass through [110]. Inspired by this phenomenon, we propose that Cx43 hemichannels can be embedded in artificial EV surfaces to facilitate the delivery of molecules targeting the ncRNA–Cx axis. The abovementioned oligonucleotides, ncRNA-targeted small molecules [111], and adeno-associated virus vectors carrying connexin genes are desirable cargos for these EVs. These EV drug carriers can enhance the uptake efficiency and infiltration of oligonucleotides into the blood–brain barrier, providing a promising approach for treating cancers and CNS diseases [97,112,113].

5. Conclusions and Perspectives

In the past few years, researchers have published exceptional reviews on ncRNAs [6,114] and connexins [115,116]; however, summaries on the ncRNA–connexin axis are inadequate. Herein, we have compiled examples published in the past five years that illustrate how connexins are modulated by relevant ncRNAs and how this interaction is associated with the development of many diseases.
Generally, miRNAs bind to the 3′ UTR of the connexin mRNA and interfere with mRNA translation, blocking connexin protein synthesis. This pathological process dysregulates the miRNA levels, leading to a negative correlation with connexin levels. lncRNAs and circRNAs act as sponges that inhibit miRNA functions. Therefore, downstream connexin levels are positively correlated with lncRNAs and circRNAs. Although less common, cases in which the connexin (Cx43) mRNA is located upstream of the miRNAs have been reported [117,118]. Indirectly, ncRNAs regulate or interact with the proteins upstream of connexins, thereby facilitating pathological processes. In particular, ncRNAs can be transported through gap junctions, particularly Cx43, which transfers ncRNAs between neighboring cells in vitro. Cx43 has been proposed to constitute EVs that deliver ncRNAs to remote tissues; however, this mechanism requires further investigation.
However, this study has some shortcomings. Owing to complex protein–protein interactions and tissue specificity, the underlying mechanisms through which connexins and ncRNAs contribute to disease development have not been clearly explained. Notably, developing therapies that regulate the ncRNA–Cx axis is challenging. First, researchers should be aware that, because of limited clinical samples and variations in research methodologies, the selected ncRNA profiles may exhibit limited repeatability, which is directly associated with the selection of ncRNAs to be further studied [119]. Second, in addition to their role as gap junctions, connexins engage in intricate downstream protein–protein interactions [39]. Therefore, the regulation of the ncRNA–Cx axis may potentially result in off-target effects. Third, although many in vitro studies on the intercellular transport of miRNAs through gap junctions have been reported, in vivo research is lacking. Therefore, it is important to acknowledge that utilizing EVs with Cx43 channels for the delivery of oligonucleotides or small molecules is still at a conceptual stage because transferring oligonucleotides through gap junctions requires a substantial energy supply. In the future, researchers should attempt to elucidate the molecular mechanisms before and after the ncRNA–connexin axis to pave the way for efficacious therapies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25116146/s1.

Author Contributions

Conceptualization, X.L. and Z.W.; resources, software and writing—original draft preparation, X.L. and Z.W.; writing—review and editing, X.L. and Z.W.; funding acquisition, Z.W. and N.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (82130109, 81773924, 81573636).

Data Availability Statement

The data used to support the findings of this study are included within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef] [PubMed]
  2. Fasolo, F.; Di Gregoli, K.; Maegdefessel, L.; Johnson, J.L. Non-coding RNAs in cardiovascular cell biology and atherosclerosis. Cardiovasc. Res. 2019, 115, 1732–1756. [Google Scholar] [CrossRef] [PubMed]
  3. Yuan, M.; Yang, B.; Rothschild, G.; Mann, J.J.; Sanford, L.D.; Tang, X.; Huang, C.; Wang, C.; Zhang, W. Epigenetic regulation in major depression and other stress-related disorders: Molecular mechanisms, clinical relevance and therapeutic potential. Signal Transduct. Target. Ther. 2023, 8, 309. [Google Scholar] [CrossRef] [PubMed]
  4. Liu, N.; Wang, Z.Z.; Zhao, M.; Zhang, Y.; Chen, N.H. Role of non-coding RNA in the pathogenesis of depression. Gene 2020, 735, 144276. [Google Scholar] [CrossRef] [PubMed]
  5. Di Martino, M.T.; Arbitrio, M.; Caracciolo, D.; Cordua, A.; Cuomo, O.; Grillone, K.; Riillo, C.; Caridà, G.; Scionti, F.; Labanca, C.; et al. miR-221/222 as biomarkers and targets for therapeutic intervention on cancer and other diseases: A systematic review. Mol. Ther. Nucleic Acids 2022, 27, 1191–1224. [Google Scholar] [CrossRef] [PubMed]
  6. Matsui, M.; Corey, D.R. Non-coding RNAs as drug targets. Nat. Rev. Drug Discov. 2017, 16, 167–179. [Google Scholar] [CrossRef]
  7. Weng, R.Y.; Jiang, Z.W.; Gu, Y.X. Noncoding RNA as Diagnostic and Prognostic Biomarkers in Cerebrovascular Disease. Oxidative Med. Cell. Longev. 2022, 2022, 8149701. [Google Scholar] [CrossRef]
  8. Lampe, P.D.; Laird, D.W. Recent advances in connexin gap junction biology. Fac. Rev. 2022, 11, 14. [Google Scholar] [CrossRef] [PubMed]
  9. Totland, M.Z.; Rasmussen, N.L.; Knudsen, L.M.; Leithe, E. Regulation of gap junction intercellular communication by connexin ubiquitination: Physiological and pathophysiological implications. Cell. Mol. Life Sci. 2020, 77, 573–591. [Google Scholar] [CrossRef]
  10. Van Campenhout, R.; Gomes, A.R.; De Groof, T.W.M.; Muyldermans, S.; Devoogdt, N.; Vinken, M. Mechanisms Underlying Connexin Hemichannel Activation in Disease. Int. J. Mol. Sci. 2021, 22, 3503. [Google Scholar] [CrossRef]
  11. Qiu, Y.; Zheng, J.L.; Chen, S.; Sun, Y. Connexin Mutations and Hereditary Diseases. Int. J. Mol. Sci. 2022, 23, 4255. [Google Scholar] [CrossRef] [PubMed]
  12. Delmar, M.; Laird, D.W.; Naus, C.C.; Nielsen, M.S.; Verselis, V.K.; White, T.W. Connexins and Disease. CSH Perspect. Biol. 2018, 10, a029348. [Google Scholar] [CrossRef] [PubMed]
  13. Rodríguez-Sinovas, A.; Sánchez, J.A.; Valls-Lacalle, L.; Consegal, M.; Ferreira-González, I. Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int. J. Mol. Sci. 2021, 22, 4413. [Google Scholar] [CrossRef] [PubMed]
  14. Bonacquisti, E.E.; Nguyen, J. Connexin 43 (Cx43) in cancer: Implications for therapeutic approaches via gap junctions. Cancer Lett. 2019, 442, 439–444. [Google Scholar] [CrossRef] [PubMed]
  15. Leybaert, L.; De Smet, M.A.; Lissoni, A.; Allewaert, R.; Roderick, H.L.; Bultynck, G.; Delmar, M.; Sipido, K.R.; Witschas, K. Connexin hemichannels as candidate targets for cardioprotective and anti-arrhythmic treatments. J. Clin. Investig. 2023, 133, e168117. [Google Scholar] [CrossRef] [PubMed]
  16. Zhang, N.N.; Zhang, Y.; Wang, Z.Z.; Chen, N.H. Connexin 43: Insights into candidate pathological mechanisms of depression and its implications in antidepressant therapy. Acta Pharmacol. Sin. 2022, 43, 2448–2461. [Google Scholar] [CrossRef] [PubMed]
  17. Charvériat, M.; Mouthon, F.; Rein, W.; Verkhratsky, A. Connexins as therapeutic targets in neurological and neuropsychiatric disorders. Biochim. Biophys. Acta-Mol. Basis Dis. 2021, 1867, 166098. [Google Scholar] [CrossRef] [PubMed]
  18. Xing, L.Y.; Yang, T.; Cui, S.S.; Chen, G. Connexin Hemichannels in Astrocytes: Role in CNS Disorders. Front. Mol. Neurosci. 2019, 12, 23. [Google Scholar] [CrossRef] [PubMed]
  19. Xia, C.Y.; Wang, Z.Z.; Yamakuni, T.; Chen, N.H. A novel mechanism of depression: Role for connexins. Eur. Neuropsychopharmacol. 2018, 28, 483–498. [Google Scholar] [CrossRef]
  20. Sun, J.D.; Liu, Y.; Yuan, Y.H.; Li, J.; Chen, N.H. Gap junction dysfunction in the prefrontal cortex induces depressive-like behaviors in rats. Neuropsychopharmacology 2012, 37, 1305–1320. [Google Scholar] [CrossRef]
  21. Shen, F.; Huang, W.L.; Xing, B.P.; Fang, X.; Feng, M.; Jiang, C.M. Genistein Improves the Major Depression through Suppressing the Expression of miR-221/222 by Targeting Connexin 43. Psychiatry Investig. 2018, 15, 919–925. [Google Scholar] [CrossRef] [PubMed]
  22. He, J.H.; Li, D.M.; Wei, J.; Wang, S.; Chu, S.F.; Zhang, Z.; He, F.; Wei, D.M.; Li, Y.; Xie, J.X.; et al. Mahonia Alkaloids (MA) Ameliorate Depression Induced Gap Junction Dysfunction by miR-205/Cx43 Axis. Neurochem. Res. 2022, 47, 3761–3776. [Google Scholar] [CrossRef] [PubMed]
  23. Ding, L.; Wang, J. MiR-106a facilitates the sensorineural hearing loss induced by oxidative stress by targeting connexin-43. Bioengineered 2022, 13, 14080–14093. [Google Scholar] [CrossRef] [PubMed]
  24. Alhadidi, Q.M.; Xu, L.J.; Sun, X.Y.; Althobaiti, Y.S.; Almalki, A.; Alsaab, H.O.; Stary, C.M. MiR-182 Inhibition Protects Against Experimental Stroke in vivo and Mitigates Astrocyte Injury and Inflammation in vitro via Modulation of Cortactin Activity. Neurochem. Res. 2022, 47, 3682–3696. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, W.; Feng, J.G.; Tong, W.S. Phosphorylation of astrocytic connexin43 by ERK1/2 impairs blood-brain barrier in acute cerebral ischemia. Cell Biosci. 2017, 7, 43. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, W.; Zhao, L.; Zhang, J.X.; Wang, B.B.; Xu, G.H.; Lin, C.; Liu, N. Elevated expression of miR-302 cluster improves traumatic brain injury by inhibiting phosphorylation of connexin43 via ERK signaling. J. Chem. Neuroanat. 2019, 99, 1–8. [Google Scholar] [CrossRef]
  27. Jullienne, A.; Fukuda, A.M.; Ichkova, A.; Nishiyama, N.; Aussudre, J.; Obenaus, A.; Badaut, J. Modulating the water channel AQP4 alters miRNA expression, astrocyte connectivity and water diffusion in the rodent brain. Sci. Rep. 2018, 8, 4186. [Google Scholar] [CrossRef] [PubMed]
  28. Almad, A.A.; Doreswamy, A.; Gross, S.K.; Richard, J.P.; Huo, Y.Q.; Haughey, N.; Maragakis, N.J. Connexin 43 in Astrocytes Contributes to Motor Neuron Toxicity in Amyotrophic Lateral Sclerosis. Glia 2016, 64, 1154–1169. [Google Scholar] [CrossRef] [PubMed]
  29. Wu, Y.Y.; Kuo, H.C. Functional roles and networks of non-coding RNAs in the pathogenesis of neurodegenerative diseases. J. Biomed. Sci. 2020, 27, 49. [Google Scholar] [CrossRef]
  30. Wu, X.Z.; Wei, H.C.; Wu, J.Q. Coding and long non-coding gene expression changes in the CNS traumatic injuries. Cell. Mol. Life Sci. 2022, 79, 123. [Google Scholar] [CrossRef]
  31. Du, J.N.; Li, Y.; Su, Y.T.; Zhi, W.Q.; Zhang, J.L.; Zhang, C.; Wang, J.; Deng, W.S.; Zhao, S.S. LncRNA Pnky Positively Regulates Neural Stem Cell Migration by Modulating mRNA Splicing and Export of Target Genes. Cell. Mol. Neurobiol. 2023, 43, 1199–1218. [Google Scholar] [CrossRef]
  32. Sun, Q.; Zhang, Y.; Wang, S.; Yang, F.; Cai, H.; Xing, Y.; Chen, Z.; Chen, J. NEAT1 Decreasing Suppresses Parkinson’s Disease Progression via Acting as miR-1301-3p Sponge. J. Mol. Neurosci. 2020, 71, 369–378. [Google Scholar] [CrossRef]
  33. Yao, J.; Wang, Z.; Song, W.; Zhang, Y. Targeting NLRP3 inflammasome for neurodegenerative disorders. Mol. Psychiatry 2023, 28, 4512–4527. [Google Scholar] [CrossRef] [PubMed]
  34. Zeng, W.; Fu, L.; Xu, H.F. MicroRNA-206 relieves irradiation-induced neuroinflammation by regulating connexin 43. Exp. Ther. Med. 2021, 22, 1186. [Google Scholar] [CrossRef]
  35. Dong, Y.; Wang, J.; Du, K.X.; Jia, T.M.; Zhang, Y.; Song, J.; Li, M.M.; Zhang, H.L. in the pathogenesis of bacterial meningitis by targeting. Neuroreport 2019, 30, 174–181. [Google Scholar] [CrossRef] [PubMed]
  36. Cusato, K.; Bosco, A.; Rozental, R.; Guimarães, C.A.; Reese, B.E.; Linden, R.; Spray, D.C. Gap junctions mediate bystander cell death in developing retina. J. Neurosci. 2003, 23, 6413–6422. [Google Scholar] [CrossRef] [PubMed]
  37. Caruso, G.; Di Pietro, L.; Caraci, F. Gap Junctions and Connexins in Microglia-Related Oxidative Stress and Neuroinflammation: Perspectives for Drug Discovery. Biomolecules 2023, 13, 505. [Google Scholar] [CrossRef]
  38. Sin, W.C.; Aftab, Q.; Bechberger, J.F.; Leung, J.H.; Chen, H.; Naus, C.C. Astrocytes promote glioma invasion via the gap junction protein connexin43. Oncogene 2016, 35, 1504–1516. [Google Scholar] [CrossRef]
  39. Martins-Marques, T.; Ribeiro-Rodrigues, T.; Batista-Almeida, D.; Aasen, T.; Kwak, B.R.; Girao, H. Biological Functions of Connexin43 Beyond Intercellular Communication. Trends Cell Biol. 2019, 29, 835–847. [Google Scholar] [CrossRef]
  40. Aasen, T.; Mesnil, M.; Naus, C.C.; Lampe, P.D.; Laird, D.W. Gap junctions and cancer: Communicating for 50 years. Nat. Rev. Cancer 2016, 16, 775–788. [Google Scholar] [CrossRef]
  41. Rusiecka, O.M.; Montgomery, J.; Morel, S.; Batista-Almeida, D.; Van Campenhout, R.; Vinken, M.; Girao, H.; Kwak, B.R. Canonical and Non-Canonical Roles of Connexin43 in Cardioprotection. Biomolecules 2020, 10, 1225. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, Y.; Li, J.; Xuan, L.Y.; Liu, Y.F.; Shao, L.Q.; Ge, H.Y.; Gu, J.Y.; Wei, C.X.; Zhao, M. Astragalus Root dry extract restores connexin43 expression by targeting miR-1 in viral myocarditis. Phytomedicine 2018, 46, 32–38. [Google Scholar] [CrossRef] [PubMed]
  43. Jin, Y.; Zhou, T.; Feng, Q.; Yang, J.; Cao, J.; Xu, X.; Yang, C. Inhibition of MicroRNA-206 Ameliorates Ischemia–Reperfusion Arrhythmia in a Mouse Model by Targeting Connexin43. J. Cardiovasc. Transl. Res. 2019, 13, 584–592. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, Y.; Sun, L.H.; Xuan, L.N.; Pan, Z.W.; Hu, X.L.; Liu, H.Y.; Bai, Y.L.; Jiao, L.; Li, Z.G.; Cui, L.N.; et al. Long non-coding RNA CCRR controls cardiac conduction via regulating intercellular coupling. Nat. Commun. 2018, 9, 4176. [Google Scholar] [CrossRef] [PubMed]
  45. Lv, L.F.; Zheng, N.; Zhang, L.J.; Li, R.T.; Li, Y.N.; Yang, R.; Li, C.; Fang, R.N.; Shabanova, A.; Li, X.L.; et al. Metformin ameliorates cardiac conduction delay by regulating microRNA-1 in mice. Eur. J. Pharmacol. 2020, 881, 173131. [Google Scholar] [CrossRef] [PubMed]
  46. Qin, W.; Zhang, L.; Li, Z.; Xiao, D.; Zhang, Y.; Yang, H.; Zhang, H.; Xu, C.; Zhang, Y. Metoprolol protects against myocardial infarction by inhibiting miR-1 expression in rats. J. Pharm. Pharmacol. 2020, 72, 76–83. [Google Scholar] [CrossRef] [PubMed]
  47. Wahl, C.M.; Schmidt, C.; Hecker, M.; Ullrich, N.D. Distress-Mediated Remodeling of Cardiac Connexin-43 in a Novel Cell Model for Arrhythmogenic Heart Diseases. Int. J. Mol. Sci. 2022, 23, 10174. [Google Scholar] [CrossRef] [PubMed]
  48. Trotta, M.C.; Ferraro, B.; Messina, A.; Panarese, I.; Gulotta, E.; Nicoletti, G.F.; D’Amico, M.; Pieretti, G. Telmisartan cardioprotects from the ischaemic/hypoxic damage through a miR-1-dependent pathway. J. Cell. Mol. Med. 2019, 23, 6635–6645. [Google Scholar] [CrossRef] [PubMed]
  49. Lei, Y.; Peng, X.Y.; Li, T.; Liu, L.M.; Yang, G.M. ERK and miRNA-1 target Cx43 expression and phosphorylation to modulate the vascular protective effect of angiotensin II. Life Sci. 2019, 216, 59–66. [Google Scholar] [CrossRef]
  50. Mondejar-Parreno, G.; Callejo, M.; Barreira, B.; Morales-Cano, D.; Esquivel-Ruiz, S.; Filice, M.; Moreno, L.; Cogolludo, A.; Perez-Vizcaino, F. miR-1 induces endothelial dysfunction in rat pulmonary arteries. J. Physiol. Biochem. 2019, 75, 519–529. [Google Scholar] [CrossRef]
  51. Jin, Y.; Zhou, T.Y.; Cao, J.N.; Feng, Q.T.; Fu, Y.J.; Xu, X.; Yang, C.J. MicroRNA-206 Downregulates Connexin43 in Cardiomyocytes to Induce Cardiac Arrhythmias in a Transgenic Mouse Model. Heart Lung Circ. 2019, 28, 1755–1761. [Google Scholar] [CrossRef] [PubMed]
  52. Li, Y.; Ge, J.P.; Yin, Y.Y.; Yang, R.W.; Kong, J.; Gu, J.P. Upregulated miR-206 Aggravates Deep Vein Thrombosis by Regulating GJA1-Mediated Autophagy of Endothelial Progenitor Cells. Cardiovasc. Ther. 2022, 2022, 9966306. [Google Scholar] [CrossRef] [PubMed]
  53. Lv, X.W.; Lu, P.; Hu, Y.S.; Xu, T.T. Overexpression of MiR-29b-3p Inhibits Atrial Remodeling in Rats by Targeting PDGF-B Signaling Pathway. Oxidative Med. Cell. Longev. 2021, 2021, 3763529. [Google Scholar] [CrossRef] [PubMed]
  54. Lv, X.W.; Li, J.Y.; Hu, Y.S.; Wang, S.R.; Yang, C.Y.; Li, C.X.; Zhong, G.Q. Overexpression of miR-27b-3p Targeting Wnt3a Regulates the Signaling Pathway of Wnt/β-Catenin and Attenuates Atrial Fibrosis in Rats with Atrial Fibrillation. Oxidative Med. Cell. Longev. 2019, 2019, 5703764. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, H.S.; Wang, J.; Li, H.H.; Wang, X.; Zhang, S.Q.; Deng, T.; Li, Y.K.; Zou, R.S.; Wang, H.J.; Zhu, R.; et al. Long noncoding RNA Gm2694 drives depressive-like behaviors in male mice by interacting with GRP78 to disrupt endoplasmic reticulum homeostasis. Sci. Adv. 2022, 8, eabn2496. [Google Scholar] [CrossRef] [PubMed]
  56. Gomes, C.P.D.; Schroen, B.; Kuster, G.M.; Robinson, E.L.; Ford, K.; Squire, I.B.; Heymans, S.; Martelli, F.; Emanueli, C.; Devaux, Y.; et al. Regulatory RNAs in Heart Failure. Circulation 2020, 141, 313–328. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, L.N.; Li, Q.; Diao, J.Y.; Lin, L.; Wei, J. MiR-23a Is Involved in Myocardial Ischemia/Reperfusion Injury by Directly Targeting CX43 and Regulating Mitophagy. Inflammation 2021, 44, 1581–1591. [Google Scholar] [CrossRef]
  58. Dufeys, C.; Daskalopoulos, E.P.; Castanares-Zapatero, D.; Conway, S.J.; Ginion, A.; Bouzin, C.; Ambroise, J.; Bearzatto, B.; Gala, J.L.; Heymans, S.; et al. AMPKα1 deletion in myofibroblasts exacerbates post-myocardial infarction fibrosis by a connexin 43 mechanism. Basic Res. Cardiol. 2021, 116, 10. [Google Scholar] [CrossRef] [PubMed]
  59. Sun, B.; Zhao, C.M.; Mao, Y. MiR-218-5p Mediates Myocardial Fibrosis after Myocardial Infarction by Targeting CX43. Curr. Pharm. Des. 2021, 27, 4504–4512. [Google Scholar] [CrossRef]
  60. Garcia-Elias, A.; Tajes, M.; Yañez-Bisbe, L.; Enjuanes, C.; Comín-Colet, J.; Serra, S.A.; Fernández-Fernández, J.M.; Aguilar-Agon, K.W.; Reilly, S.; Martí-Almor, J.; et al. Atrial Fibrillation in Heart Failure Is Associated with High Levels of Circulating microRNA-199a-5p and 22-5p and a Defective Regulation of Intracellular Calcium and Cell-to-Cell Communication. Int. J. Mol. Sci. 2021, 22, 10377. [Google Scholar] [CrossRef]
  61. Kumar, A.; Thomas, S.K.; Wong, K.C.; Lo Sardo, V.; Cheah, D.S.; Hou, Y.H.; Placone, J.K.; Tenerelli, K.P.; Ferguson, W.C.; Torkamani, A.; et al. Mechanical activation of noncoding-RNA-mediated regulation of disease-associated phenotypes in human cardiomyocytes. Nat. Biomed. Eng. 2019, 3, 137–146. [Google Scholar] [CrossRef] [PubMed]
  62. Yang, Z.J.; Liu, R.; Han, X.J.; Qiu, C.L.; Dong, G.L.; Liu, Z.Q.; Liu, L.H.; Luo, Y.; Jiang, L.P. Knockdown of the long non-coding RNA MALAT1 ameliorates TNF-α-mediated endothelial cell pyroptosis via the miR-30c-5p/Cx43 axis. Mol. Med. Rep. 2023, 27, 90. [Google Scholar] [CrossRef]
  63. Dai, W.R.; Chao, X.Y.; Li, S.S.; Zhou, S.; Zhong, G.Q.; Jiang, Z.Y.; Hirata, R.D.C. Long Noncoding RNA HOTAIR Functions as a Competitive Endogenous RNA to Regulate Connexin43 Remodeling in Atrial Fibrillation by Sponging MicroRNA-613. Cardiovasc. Ther. 2020, 2020, 5925342. [Google Scholar] [CrossRef]
  64. Costa, V.; De Fine, M.; Carina, V.; Conigliaro, A.; Raimondi, L.; De Luca, A.; Bellavia, D.; Salamanna, F.; Alessandro, R.; Pignatti, G.; et al. How miR-31-5p and miR-33a-5p Regulates SP1/CX43 Expression in Osteoarthritis Disease: Preliminary Insights. Int. J. Mol. Sci. 2021, 22, 2471. [Google Scholar] [CrossRef] [PubMed]
  65. Lei, J.L.; Fu, Y.H.; Zhuang, Y.; Zhang, K.; Lu, D.G. miR-382-3p suppressed IL-1β induced inflammatory response of chondrocytes via the TLR4/MyD88/NF-κB signaling pathway by directly targeting CX43. J. Cell. Physiol. 2019, 234, 23160–23168. [Google Scholar] [CrossRef] [PubMed]
  66. Zhou, P.; Xu, P.; Yu, W.T.; Li, H. MiR-206 improves intervertebral disk degeneration by targeting GJA1. J. Orthop. Surg. Res. 2022, 17, 157. [Google Scholar] [CrossRef]
  67. Yuan, X.Q.; Guo, Y.F.; Chen, D.C.; Luo, Y.B.; Chen, D.Y.; Miao, J.H.; Chen, Y. Long non-coding RNA MALAT1 functions as miR-1 sponge to regulate Connexin 43-mediated ossification of the posterior longitudinal ligament. Bone 2019, 127, 305–314. [Google Scholar] [CrossRef]
  68. Tong, L.P.; Yu, H.; Huang, X.Y.; Shen, J.; Xiao, G.Z.; Chen, L.; Wang, H.Y.; Xing, L.P.; Chen, D. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res. 2022, 10, 60. [Google Scholar] [CrossRef]
  69. Zhou, M.; Zhai, C.J.; Shen, K.; Liu, G.; Liu, L.; He, J.; Chen, J.; Xu, Y.Z. miR-1 Inhibits the Ferroptosis of Chondrocyte by Targeting CX43 and Alleviates Osteoarthritis Progression. J. Immunol. Res. 2023, 2023, 2061071. [Google Scholar] [CrossRef]
  70. Nakamura, T.; Iwamoto, T.; Nakamura, H.M.; Shindo, Y.; Saito, K.; Yamada, A.; Yamada, Y.; Fukumoto, S.; Nakamura, T. Regulation of miR-1-Mediated Connexin 43 Expression and Cell Proliferation in Dental Epithelial Cells. Front. Cell Dev. Biol. 2020, 8, 156. [Google Scholar] [CrossRef]
  71. Zhang, B.; Huo, S.B.; Cen, X.; Pan, X.F.; Huang, X.Q.; Zhao, Z.H. circAKT3 positively regulates osteogenic differentiation of human dental pulp stromal cells via miR-206/CX43 axis. Stem Cell Res. Ther. 2020, 11, 531. [Google Scholar] [CrossRef] [PubMed]
  72. Kamiya, K.; Yum, S.W.; Kurebayashi, N.; Muraki, M.; Ogawa, K.; Karasawa, K.; Miwa, A.; Guo, X.S.; Gotoh, S.; Sugitani, Y.; et al. Assembly of the cochlear gap junction macromolecular complex requires connexin 26. J. Clin. Investig. 2014, 124, 1598–1607. [Google Scholar] [CrossRef] [PubMed]
  73. Laird, D.W.; Lampe, P.D. Cellular mechanisms of connexin-based inherited diseases. Trends Cell Biol. 2022, 32, 58–69. [Google Scholar] [CrossRef] [PubMed]
  74. Gentile, G.; Paciello, F.; Zorzi, V.; Spampinato, A.G.; Guarnaccia, M.; Crispino, G.; Tettey-Matey, A.; Scavizzi, F.; Raspa, M.; Fetoni, A.R.; et al. miRNA and mRNA Profiling Links Connexin Deficiency to Deafness via Early Oxidative Damage in the Mouse Stria Vascularis. Front. Cell Dev. Biol. 2021, 8, 616878. [Google Scholar] [CrossRef] [PubMed]
  75. Tang, Y.; Ji, H.; Liu, H.; Liu, J.; Gu, W.; Peng, T.; Li, X. Pro-inflammatory cytokine-induced microRNA-212-3p expression promotes myocyte contraction via methyl-CpG-binding protein 2: A novel mechanism for infection-related preterm parturition. MHR Basic Sci. Reprod. Med. 2019, 25, 274–282. [Google Scholar] [CrossRef] [PubMed]
  76. Luo, J.H.; Jin, Y.; Li, M.Y.; Dong, L.Y. Tumor suppressor miR-613 induces cisplatin sensitivity in non-small cell lung cancer cells by targeting GJA1. Mol. Med. Rep. 2021, 23, 385. [Google Scholar] [CrossRef] [PubMed]
  77. Li, G.; Sun, L.F.; Mu, Z.Y.; Liu, S.B.; Qu, H.C.; Xie, Q.P.; Hu, B. MicroRNA-1298-5p inhibits cell proliferation and the invasiveness of bladder cancer cells via down-regulation of connexin 43. Biochem. Cell Biol. 2020, 98, 227–237. [Google Scholar] [CrossRef] [PubMed]
  78. Chi, Q.; Wang, Z.Y.; Li, H.Y.; Song, D.B.; Xu, H.; Ma, G.; Wang, Z.M.; Li, X.M. Tumor-suppressor microRNA-139-5p restrains bladder cancer cell line ECV-304 properties via targeting Connexin 43. Chin. Med. J. 2019, 132, 2354–2361. [Google Scholar] [CrossRef] [PubMed]
  79. Fan, J.D.; Wang, C.; Zhai, X.Y.; Li, J.H.; Ju, J.; Zhu, Y.Y.; Zheng, S.K.; Ren, N.; Huang, B.Q.; Jiang, X.Y.; et al. lncRNA LEF1-AS1 Acts as a Novel Biomarker and Promotes Hypopharyngeal Squamous Cell Carcinoma Progression and Metastasis by Targeting the miR-221-5p/GJA1 Axis. Dis. Markers 2022, 2022, 3881310. [Google Scholar] [CrossRef]
  80. Zheng, H.L.; Ding, B.S.; Xue, K.; Yu, J.X.; Lou, W.Y. Construction of a lncRNA/pseudogene-hsa-miR-30d-5p-GJA1 regulatory network related to metastasis of pancreatic cancer. Genomics 2021, 113, 1742–1753. [Google Scholar] [CrossRef]
  81. Li, D.H.; Li, L.D.; Chen, X.; Zhou, C.S.; Hao, B.; Cao, Y.Q. Dysregulation of lncRNA-CCRR contributes to brain metastasis of breast cancer by intercellular coupling via regulating connexin 43 expression. J. Cell. Mol. Med. 2021, 25, 4826–4834. [Google Scholar] [CrossRef]
  82. Yu, X.T.; Zhang, X.C.; Wang, G.; Wang, B.; Ding, Y.F.; Zhao, J.Y.; Liu, H.L.; Cui, S.Y. miR-206 as a prognostic and sensitivity biomarker for platinum chemotherapy in epithelial ovarian cancer. Cancer Cell Int. 2020, 20, 534. [Google Scholar] [CrossRef] [PubMed]
  83. Chai, C.; Sui, K.; Tang, J.; Yu, H.; Yang, C.; Zhang, H.; Li, S.C.; Zhong, J.F.; Wang, Z.; Zhang, X. BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution. Theranostics 2023, 13, 3943–3963. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, S.S.; Zhao, Y.; Liu, H.; Zhao, X.; Shen, X.B. miR-301-3p directly regulates Cx43 to mediate the development of gastric cancer. J. Int. Med. Res. 2021, 49, 3000605211033185. [Google Scholar] [CrossRef] [PubMed]
  85. Chen, K.; Wang, Q.; Liu, X.X.; Wang, F.; Yang, Y.M.; Tian, X.D. Hypoxic pancreatic cancer derived exosomal miR-30b-5p promotes tumor angiogenesis by inhibiting GJA1 expression. Int. J. Biol. Sci. 2022, 18, 1220–1237. [Google Scholar] [CrossRef] [PubMed]
  86. Georgikou, C.; Yin, L.B.; Gladkich, J.; Xiao, X.; Sticht, C.; de la Torre, C.; Gretz, N.; Gross, W.; Schäfer, M.; Karakhanova, S.; et al. Inhibition of miR30a-3p by sulforaphane enhances gap junction intercellular communication in pancreatic cancer. Cancer Lett. 2020, 469, 238–245. [Google Scholar] [CrossRef] [PubMed]
  87. Scatolini, M.; Patel, A.; Grosso, E.; Mello-Grand, M.; Ostano, P.; Coppo, R.; Vitiello, M.; Venesio, T.; Zaccagna, A.; Pisacane, A.; et al. GJB5 association with BRAF mutation and survival in cutaneous malignant melanoma. Br. J. Dermatol. 2022, 186, 117–128. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, J.L.; Li, H.; Zhang, J.B.; Zhang, C.H.; Hou, X.Q. Suppression of connexin 43 expression by miR-106a promotes melanoma cell proliferation. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 965–971. [Google Scholar] [CrossRef] [PubMed]
  89. Khalilian, S.; Imani, S.Z.H.; Ghafouri-Fard, S. Emerging roles and mechanisms of miR-206 in human disorders: A comprehensive review. Cancer Cell Int. 2022, 22, 412. [Google Scholar] [CrossRef]
  90. Mei, J.; Xu, R.; Hao, L.; Zhang, Y. MicroRNA-613: A novel tumor suppressor in human cancers. Biomed. Pharmacother. 2020, 123, 109799. [Google Scholar] [CrossRef]
  91. Li, Y.; Wang, L.; Tian, J.; Zu, Y.; Wang, F.; Yang, Y.; Ma, S.; Cao, J.; Huang, Q.; Ha, C. The role of Connexin26 regulated by MiR-2114-3p in the pathogenesis of ovarian cancer. Biochem. Biophys. Res. Commun. 2023, 640, 105–116. [Google Scholar] [CrossRef] [PubMed]
  92. Jiang, L.L.; Huang, H.; Qian, Y.F.; Li, Y.; Chen, X.L.; Di, N.; Yang, D.Z. miR-130b regulates gap junctional intercellular communication through connexin 43 in granulosa cells from patients with polycystic ovary syndrome. Mol. Hum. Reprod. 2020, 26, 576–584. [Google Scholar] [CrossRef] [PubMed]
  93. Lim, P.K.; Bliss, S.A.; Patel, S.A.; Taborga, M.; Dave, M.A.; Gregory, L.A.; Greco, S.J.; Bryan, M.; Patel, P.S.; Rameshwar, P. Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res. 2011, 71, 1550–1560. [Google Scholar] [CrossRef] [PubMed]
  94. Lemcke, H.; Steinhoff, G.; David, R. Gap junctional shuttling of miRNA—A novel pathway of intercellular gene regulation and its prospects in clinical application. Cell. Signal. 2015, 27, 2506–2514. [Google Scholar] [CrossRef] [PubMed]
  95. Fukuda, S.; Akiyama, M.; Niki, Y.; Kawatsura, R.; Harada, H.; Nakahama, K. Inhibitory effects of miRNAs in astrocytes on C6 glioma progression via connexin 43. Mol. Cell. Biochem. 2021, 476, 2623–2632. [Google Scholar] [CrossRef] [PubMed]
  96. Tittarelli, A.; Navarrete, M.; Lizana, M.; Hofmann-Vega, F.; Salazar-Onfray, F. Hypoxic Melanoma Cells Deliver microRNAs to Dendritic Cells and Cytotoxic T Lymphocytes through Connexin-43 Channels. Int. J. Mol. Sci. 2020, 21, 7567. [Google Scholar] [CrossRef] [PubMed]
  97. Valiunas, V.; Gordon, C.; Valiuniene, L.; Devine, D.; Lin, R.Z.; Cohen, I.S.; Brink, P.R. Intercellular delivery of therapeutic oligonucleotides. J. Drug Deliv. Sci. Technol. 2022, 72, 103404. [Google Scholar] [CrossRef] [PubMed]
  98. Lemcke, H.; David, R. Potential mechanisms of microRNA mobility. Traffic 2018, 19, 910–917. [Google Scholar] [CrossRef]
  99. Gemel, J.; Kilkus, J.; Dawson, G.; Beyer, E.C. Connecting Exosomes and Connexins. Cancers 2019, 11, 476. [Google Scholar] [CrossRef]
  100. Martins-Marques, T.; Costa, M.C.; Catarino, S.; Simoes, I.; Aasen, T.; Enguita, F.J.; Girao, H. Cx43-mediated sorting of miRNAs into extracellular vesicles. EMBO Rep. 2022, 23, e54312. [Google Scholar] [CrossRef]
  101. Soares, A.R.; Martins-Marques, T.; Ribeiro-Rodrigues, T.; Ferreira, J.V.; Catarino, S.; Pinho, M.J.; Zuzarte, M.; Isabel Anjo, S.; Manadas, B.; Sluijter, J.P.G.; et al. Gap junctional protein Cx43 is involved in the communication between extracellular vesicles and mammalian cells. Sci. Rep. 2015, 5, 13243. [Google Scholar] [CrossRef]
  102. Acuña, R.A.; Varas-Godoy, M.; Berthoud, V.M.; Alfaro, I.E.; Retamal, M.A. Connexin-46 Contained in Extracellular Vesicles Enhance Malignancy Features in Breast Cancer Cells. Biomolecules 2020, 10, 676. [Google Scholar] [CrossRef]
  103. Cuomo-Haymour, N.; Bergamini, G.; Russo, G.; Kulic, L.; Knuesel, I.; Martin, R.; Huss, A.; Tumani, H.; Otto, M.; Pryce, C.R. Differential Expression of Serum Extracellular Vesicle miRNAs in Multiple Sclerosis: Disease-Stage Specificity and Relevance to Pathophysiology. Int. J. Mol. Sci. 2022, 23, 1664. [Google Scholar] [CrossRef] [PubMed]
  104. Toor, S.M.; Aldous, E.K.; Parray, A.; Akhtar, N.; Al-Sarraj, Y.; Abdelalim, E.M.; Arredouani, A.; El-Agnaf, O.; Thornalley, P.J.; Pananchikkal, S.V.; et al. Circulating MicroRNA Profiling Identifies Distinct MicroRNA Signatures in Acute Ischemic Stroke and Transient Ischemic Attack Patients. Int. J. Mol. Sci. 2022, 24, 108. [Google Scholar] [CrossRef] [PubMed]
  105. O’Donoghue, M.L.; Rosenson, R.S.; Gencer, B.; López, J.A.G.; Lepor, N.E.; Baum, S.J.; Stout, E.; Gaudet, D.; Knusel, B.; Kuder, J.F.; et al. Small Interfering RNA to Reduce Lipoprotein(a) in Cardiovascular Disease. N. Engl. J. Med. 2022, 387, 1855–1864. [Google Scholar] [CrossRef] [PubMed]
  106. Nissen, S.E.; Linnebjerg, H.; Shen, X.; Wolski, K.; Ma, X.; Lim, S.; Michael, L.F.; Ruotolo, G.; Gribble, G.; Navar, A.M.; et al. Lepodisiran, an Extended-Duration Short Interfering RNA Targeting Lipoprotein(a): A Randomized Dose-Ascending Clinical Trial. JAMA 2023, 330, 2075–2083. [Google Scholar] [CrossRef] [PubMed]
  107. Garrelfs, S.F.; Frishberg, Y.; Hulton, S.A.; Koren, M.J.; O’Riordan, W.D.; Cochat, P.; Deschênes, G.; Shasha-Lavsky, H.; Saland, J.M.; Van’t Hoff, W.G.; et al. Lumasiran, an RNAi Therapeutic for Primary Hyperoxaluria Type 1. N. Engl. J. Med. 2021, 384, 1216–1226. [Google Scholar] [CrossRef] [PubMed]
  108. Jiang, H.; Zhang, Y.; Wang, Z.Z.; Chen, N.H. Connexin 43: An Interface Connecting Neuroinflammation to Depression. Molecules 2023, 28, 1820. [Google Scholar] [CrossRef] [PubMed]
  109. Guo, A.; Zhang, H.; Li, H.; Chiu, A.; García-Rodríguez, C.; Lagos, C.F.; Sáez, J.C.; Lau, C.G. Inhibition of connexin hemichannels alleviates neuroinflammation and hyperexcitability in temporal lobe epilepsy. Proc. Natl. Acad. Sci. USA 2022, 119, e2213162119. [Google Scholar] [CrossRef]
  110. Martins-Marques, T.; Pinho, M.J.; Zuzarte, M.; Oliveira, C.; Pereira, P.; Sluijter, J.P.; Gomes, C.; Girao, H. Presence of Cx43 in extracellular vesicles reduces the cardiotoxicity of the anti-tumour therapeutic approach with doxorubicin. J. Extracell. Vesicles 2016, 5, 32538. [Google Scholar] [CrossRef]
  111. Warner, K.D.; Hajdin, C.E.; Weeks, K.M. Principles for targeting RNA with drug-like small molecules. Nat. Rev. Drug Discov. 2018, 17, 547–558. [Google Scholar] [CrossRef] [PubMed]
  112. Usman, W.M.; Pham, T.C.; Kwok, Y.Y.; Vu, L.T.; Ma, V.; Peng, B.Y.; San Chan, Y.; Wei, L.K.; Chin, S.M.; Azad, A.; et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat. Commun. 2018, 9, 2359. [Google Scholar] [CrossRef] [PubMed]
  113. Nieland, L.; Mahjoum, S.; Grandell, E.; Breyne, K.; Breakefield, X.O. Engineered EVs designed to target diseases of the CNS. J. Control. Release 2023, 356, 493–506. [Google Scholar] [CrossRef] [PubMed]
  114. Toden, S.; Zumwalt, T.J.; Goel, A. Non-coding RNAs and potential therapeutic targeting in cancer. Biochim. Biophys. Acta-Rev. Cancer 2021, 1875, 188491. [Google Scholar] [CrossRef] [PubMed]
  115. Laird, D.W.; Lampe, P.D. Therapeutic strategies targeting connexins. Nat. Rev. Drug Discov. 2018, 17, 905–921. [Google Scholar] [CrossRef] [PubMed]
  116. Laird, D.W.; Naus, C.C.; Lampe, P.D. SnapShot: Connexins and Disease. Cell 2017, 170, 1260–1260.e1. [Google Scholar] [CrossRef] [PubMed]
  117. Naser Al Deen, N.; AbouHaidar, M.; Talhouk, R. Connexin43 as a Tumor Suppressor: Proposed Connexin43 mRNA-circularRNAs-microRNAs Axis Towards Prevention and Early Detection in Breast Cancer. Front. Med. 2019, 6, 192. [Google Scholar] [CrossRef]
  118. Naser Al Deen, N.; Atallah Lanman, N.; Chittiboyina, S.; Fostok, S.; Nasr, R.; Lelièvre, S.; Talhouk, R. Over-expression of miR-183-5p or miR-492 triggers invasion and proliferation and loss of polarity in non-neoplastic breast epithelium. Sci. Rep. 2022, 12, 21974. [Google Scholar] [CrossRef]
  119. 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. [Google Scholar] [CrossRef]
Figure 1. Diagram of the ncRNA–Cx axis. miRNAs act as translation suppressors by inhibiting transcription or inducing mRNA degradation. miRNAs bind to the 3′ untranslated regions of connexin mRNAs, preventing the Cx mRNAs from being translated, thereby contributing to disease development. lncRNAs, including circRNAs, act as sponges to abolish miRNA function, abrogating the miRNA-induced inhibition of Cx mRNAs. Moreover, miRNAs and lncRNAs interact with various proteins or serve as scaffolds to influence Cx function and distribution. tsRNAs and piRNAs regulate gene expression; however, how they regulate Cx remains unclear. Notably, (1) gap junctions particularly assembled from Cx43 provide pathways for the intercellular transport of miRNAs between cancer cells; (2) lncRNA CCRR’s reaction with CIP85 interrupts the binding of CIP85 and Cx43, thereby preventing the removal of Cx43 from the myocardial cell membrane; and (3) extracellular vesicles equipped with hemichannels have been implicated in the delivery of miRNAs to distant cells. However, supporting evidence for this mechanism is currently limited. The interaction between tsRNAs, piRNAs, and connexins needs to be further investigated. Cx, connexin; lncRNA, long noncoding RNA; circRNA, circular RNA; miRNA, microRNA; tsRNAs, transfer RNA (tRNA)-derived small RNAs; piRNAs, PIWI-interacting RNAs; nt, nucleotide; PPI, protein–protein interaction.
Figure 1. Diagram of the ncRNA–Cx axis. miRNAs act as translation suppressors by inhibiting transcription or inducing mRNA degradation. miRNAs bind to the 3′ untranslated regions of connexin mRNAs, preventing the Cx mRNAs from being translated, thereby contributing to disease development. lncRNAs, including circRNAs, act as sponges to abolish miRNA function, abrogating the miRNA-induced inhibition of Cx mRNAs. Moreover, miRNAs and lncRNAs interact with various proteins or serve as scaffolds to influence Cx function and distribution. tsRNAs and piRNAs regulate gene expression; however, how they regulate Cx remains unclear. Notably, (1) gap junctions particularly assembled from Cx43 provide pathways for the intercellular transport of miRNAs between cancer cells; (2) lncRNA CCRR’s reaction with CIP85 interrupts the binding of CIP85 and Cx43, thereby preventing the removal of Cx43 from the myocardial cell membrane; and (3) extracellular vesicles equipped with hemichannels have been implicated in the delivery of miRNAs to distant cells. However, supporting evidence for this mechanism is currently limited. The interaction between tsRNAs, piRNAs, and connexins needs to be further investigated. Cx, connexin; lncRNA, long noncoding RNA; circRNA, circular RNA; miRNA, microRNA; tsRNAs, transfer RNA (tRNA)-derived small RNAs; piRNAs, PIWI-interacting RNAs; nt, nucleotide; PPI, protein–protein interaction.
Ijms 25 06146 g001
Table 1. Noncoding RNAs and related connexins involved in CNS diseases.
Table 1. Noncoding RNAs and related connexins involved in CNS diseases.
Animal Tissue/Primary CellsCell LineNoncoding RNAsMolecular TargetRelated ConnexinDisease/PathologyRef
Prefrontal cortexes of miceHuman U87-MG astrocytesmiR-221/222 ↑Cx43 ↓Cx43 ↓Depression[21]
Prefrontal cortexes and hippocampuses of ratsHuman glioma cells U251miR-205 ↑Cx43 ↓Cx43 ↓Depression[22]
Rat cochlear marginal cells/miR-106a ↑Cx43 ↓Cx43 ↓Cell apoptosis[23]
Cortexes of mouse brains/mouse cortical astrocytes/miR-182 ↑Cortactin ↓Cx43 ↓Stroke[24]
/Human neuroblastoma SH-SY5Y cellsmiR-302/367 ↓p-ERK1/2 ↑p-Cx43 ↑Traumatic brain injury[26]
Cortexes of rat brains/mouse cortical astrocytes/miR-224 ↑Cx43 ↓Cx43 ↓Astrocyte connectivity decrease[27]
/HA-1800 normal astrocytesmiR-206 ↓Cx43 ↑Cx43 ↑Neuroinflammation[34]
Cortex of rat brain/rat cortical astrocytes/miR-301a-3p ↓Cx43 ↑Cx43 ↑Cell apoptosis[35]
Spinal cord of miceMurine neural stem cells C17.2 and NE4ClncRNA Pnky ↑U2AF1Cx43 ↑Cell migration[31]
/Human neuroblastoma SH-SY5Y cellslncRNA NEAT1 ↑miR-1301-3p ↓Cx32 ↑Cell apoptosis[32]
↑: upregulation; ↓: downregulation. U2AF1: U2 small nuclear RNA auxiliary factor 1.
Table 2. Noncoding RNAs and related connexins involved in cardiovascular disorders.
Table 2. Noncoding RNAs and related connexins involved in cardiovascular disorders.
Animal Tissue/Primary CellsCell LineNoncoding RNAsMolecular TargetRelated ConnexinDisease/PathologyRef
Cardiac tissue of miceHL-1 cellsmiR-1 ↑Cx43 ↓Cx43 ↓Viral myocarditis[42]
Cardiac tissues of mice/mouse ventricular cardiomyocytes/miR-1 ↑Cx43 ↓Cx43 ↓Cardiac conduction delay[45]
Rat cardiomyocytes/miR-1 ↑Cx43 ↓Cx43 ↓Myocardial infarction[46]
/iPSC-CMs, HL-1 cellsmiR-1 ↑Cx43 ↓Cx43 ↓Arrhythmia[47]
Cardiac tissue of ratsH9c2miR-1 ↑Cx43 ↓Cx43 ↓Ischemia/reperfusion injury[48]
Rat VSMCs/miR-1 ↑Cx43 ↓Cx43 ↓Hypoxia[49]
Rat PASMCs/miR-1 ↑Cx43 ↓Cx43 ↓Endothelial dysfunction[50]
Cardiac tissue of mice/miR-206 ↑Cx43 ↓Cx43 ↓Arrhythmia[43]
Cardiac tissue of mice/miR-206 ↑Cx43 ↓Cx43 ↓Arrhythmia[51]
Vascular tissues of miceEPCsmiR-206 ↑Cx43 ↓Cx43 ↓Autophagy[52]
Myocardial tissue of rats/rat myocardial cells293T cellsmiR-23a ↑Cx43 ↓Cx43 ↓Ischemia/reperfusion injury[57]
Human cardiac fibroblast cells/miR-125b-5p ↑Cx43 ↓Cx43 ↓Cardiac fibrosis[58]
Heart tissue of miceRat myocardial fibroblast cellsmiR-218-5p ↓Cx43 ↑Cx43 ↑Myocardial fibrosis[59]
/HL-1 cellsmiR-199a+miR-22 ↑Cx40 ↓Cx40 ↓Atrial fibrillation[60]
Atrial tissue of rats/miR-29b-3p ↓PDGF-B ↑Cx43 ↓Atrial fibrosis[53]
Atrial tissue of rats/miR-27b-3p ↓Wnt3a ↑Cx43 ↓Atrial fibrillation[54]
Mouse ventricular myocytesAC16 cellslncRNA CCRR ↓CIP85 ↑Cx43 ↓Cardiac conduction block[44]
Human cardiomyocytes/lncRNA ANRIL ↑CDKN2ACx43 ↓Cardiac fibrosis[61]
Rat aortic endothelial cell293T cellslncRNA MALAT1 ↑miR-30c-5p ↓Cx43 ↑Atherosclerosis[62]
Right atrial appendages of humansHL-1 cellslncRNA HOTAIR ↓miR-613 ↑Cx43 ↓Atrial fibrillation[63]
↑: upregulation; ↓: downregulation. iPSC-CMs, murine induced pluripotent stem cell-derived cardiomyocytes; VSMC, vascular smooth muscle cell; PASMC, pulmonary artery smooth muscle cell; EPC, endothelial progenitor cell; CDKN2A, cyclin-dependent kinase inhibitor 2A.
Table 3. Noncoding RNAs and related connexins involved in other systems.
Table 3. Noncoding RNAs and related connexins involved in other systems.
Animal Tissue/Primary CellsNoncoding RNAsMolecular TargetRelated ConnexinDisease/PathologyRef
Uteruses of mice/human myometrial smooth muscle cellsmiR-212-3p ↑MeCP2 ↓Cx43 ↑Myocyte contraction[75]
Human articular chondrocytesmiR-382-3p ↓Cx43 ↑Cx43 ↑Osteoarthritis[65]
Joints of mice/human articular chondrocytesmiR-1 ↓Cx43 ↑Cx43 ↑Osteoarthritis[69]
Human chondrocytesmiR-33a-5p ↑Sp1 ↓Cx43 ↑Osteoarthritis[64]
Human osteoblastsmiR-31-5p ↑Sp1 ↓Cx43 ↓Osteoarthritis[64]
Nucleus pulposus tissue of rats/human nucleus pulposus cellsmiR-206 ↓Cx43 ↑Cx43 ↑Intervertebral disk degeneration[66]
Ligament tissue of humans/human ligament fibroblast cellslncRNA MALAT1 ↑miR-1 ↓Cx43 ↑Ossification[67]
Molars of mice/rat dental epithelial cellsmiR-1 ↑Cx43 ↓Cx43 ↓Cell proliferation[70]
Human dental pulp stromal cellscircAKT3 ↑miR-206 ↓Cx43 ↑Osteogenesis[71]
Cochlea of micemiR-34c-5p ↑Cx26 ↓Cx26 ↓Hearing impairment[74]
↑: upregulation; ↓: downregulation. MeCP2: methyl-CpG-binding protein 2; Sp1: specific protein (Sp)-transcription factor 1.
Table 4. Noncoding RNAs and related connexins involved in cancers.
Table 4. Noncoding RNAs and related connexins involved in cancers.
Animal Tissue/Primary CellsCell LineNoncoding RNAsMolecular TargetRelated ConnexinDisease/PathologyRef
NSCLC tissue of patients/human lung cancer cells/miR-613 ↓Cx43 ↑Cx43 ↑Non-small cell lung cancer[76]
Human bladder cancer cells/miR-139-5p ↓Cx43 ↑Cx43 ↑Bladder cancer[78]
Bladder cancer tissue of patients/human bladder cancer cells/miR-1298-5p ↓Cx43 ↑Cx43 ↑Bladder cancer[77]
Human hypopharyngeal squamous carcinoma cells/lncRNA LEF1-AS1 ↑miR-221-5p ↓Cx43 ↑Hypopharyngeal squamous carcinoma[79]
Human metastatic breast cancer cells/lncRNA CCRR ↑CIP85 ↓Cx43 ↑Breast cancer metastasis[81]
/Metastasis pancreatic cancer cell M8 and its parental cell BxPC.3lnRNA HELLPAR ↑, lncRNA OIP-AS1 ↑hsa-miR-30d-5p ↓Cx43 ↑Pancreatic cancer metastasis[80]
Ovarian cancer tissue of humans/human ovarian cancer cells/miR-2114-3p ↓Cx26 ↑Cx26 ↑Ovarian cancer[91]
Tumors of mice/human ovarian cancer cells/miR-206 ↑Cx43 ↓Cx43 ↓Epithelial ovarian cancer[82]
Ovarian cortexes of patients/miR-130b ↑Cx43 ↓Cx43 ↓Polycystic ovary syndrome[92]
Bone marrow aspirates and peripheral blood of patientsLeukemic and HEK293T cellsmiR130a/b-3p ↑Cx43 ↓Cx43 ↓Leukemia[83]
Gastric cancer tissue of patients/human gastric cancer cells/miR-301-3p ↑Cx43 ↓Cx43 ↓Gastric cancer[84]
Pancreatic cancer tissue of patients/human pancreatic ductal adenocarcinoma cells/miR-30a-3p ↑Cx43 ↓Cx43 ↓Pancreatic cancer[86]
Pancreatic cancer tissue of patients/human pancreatic ductal adenocarcinoma cells/miR-30b-5p ↑Cx43 ↓Cx43 ↓Pancreatic cancer[85]
Human malignant melanoma cells, human epidermal melanocytes/miR-106a ↑Cx43 ↓Cx43 ↓Melanoma[88]
Human melanoma cells/miR-335-5p ↑Cx31 ↓Cx31 ↓Melanoma[87]
↑: upregulation; ↓: downregulation.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, X.; Wang, Z.; Chen, N. Perspective and Therapeutic Potential of the Noncoding RNA–Connexin Axis. Int. J. Mol. Sci. 2024, 25, 6146. https://doi.org/10.3390/ijms25116146

AMA Style

Li X, Wang Z, Chen N. Perspective and Therapeutic Potential of the Noncoding RNA–Connexin Axis. International Journal of Molecular Sciences. 2024; 25(11):6146. https://doi.org/10.3390/ijms25116146

Chicago/Turabian Style

Li, Xinmu, Zhenzhen Wang, and Naihong Chen. 2024. "Perspective and Therapeutic Potential of the Noncoding RNA–Connexin Axis" International Journal of Molecular Sciences 25, no. 11: 6146. https://doi.org/10.3390/ijms25116146

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop