Open Access

Mechanisms and application strategies of miRNA‑146a regulating inflammation and fibrosis at molecular and cellular levels (Review)

  • Authors:
    • Zufang Liao
    • Rongjiong Zheng
    • Guofeng Shao
  • View Affiliations

  • Published online on: December 5, 2022     https://doi.org/10.3892/ijmm.2022.5210
  • Article Number: 7
  • Copyright: © Liao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In the progression of various diseases, inflammation has a critical role. Chronic persistent inflammation is a pivotal trigger of fibrosis. Several microRNAs (miRNAs) participate in inflammation and fibrosis. In recent years, it has been proved that miRNAs are a critical link in physiological and pathological processes. Among them, the miRNA miR‑146a has a pivotal role in the immune system and acquired immunity, making it one of the most studied miRNAs. Due to its essential roles at the molecular and cellular levels, it has broad application prospects in precision medicine. The present comprehensive review focused on the mechanisms of miR‑146a and its application strategies in inflammation and fibrosis, discussing its therapeutic potential. The main signaling pathways through which miR‑146a regulates inflammation and fibrosis and their relationships were covered. Furthermore, the functions and effects of miR‑146a in specific cells, which may join in the process of inflammation and fibrosis, were outlined. Application strategies were also summarized according to recent studies based on these mechanisms.

1. Introduction

Inflammation and fibrosis are inseparable from disease. Organ fibrosis is frequently induced by persistent chronic inflammation triggered by several stimuli, although it may have multiple causes (1). Type 2 T helper (Th2) cells have strong profibrotic capacities and induce M2-type expression of macrophages. Macrophages have essential effects on inflammation and fibrosis via several pathways (2,3). Chronic inflammation and epithelial-mesenchymal transition (EMT) create a profibrotic environment that promotes the production of collagen and smooth muscle α-actin (α-SMA) by infiltrating hematopoietic cells and resident fibroblasts. Several interactions among fibroblasts, fibrocytes and inflammatory cells also attenuate or exacerbate fibrosis (4).

MicroRNAs (miRNAs) mediate several functions in cells. They are pivotal regulators in cellular pathways and numerous pathologies, although they do not code for proteins. With RNA-polymerase II/III catalyst, miRNAs encoding genes are transcribed into primary miRNAs. The primary miRNAs are cleaved with Drosha (the first RNase III enzyme) into precursor miRNAs of ~60-70 nucleotides in the nucleus, and exportin-5 transports these precursor miRNAs into the cytoplasm. Subsequently, the second RNase III enzyme, Dicer, cleaves them into ~22-nucleotide double-stranded miRNA molecules (5). The RNA-induced silencing complex (RISC) incorporates the mature miRNAs, in which they further perform their functions. Finally, these miRNAs bind to the 3′-untranslated regions (3′UTRs) of the target genes and degenerate or reduce mRNA expression (6). miRNAs mediate mRNA degradation mainly by deadenylation, subsequent decapping and 5′-3′ exonucleolytic digestion (7).

In a physiological environment, the expression of miR-146a is confined to immune cells, and it negatively inhibits the innate and adaptive immune responses by regulating certain adapters or transcription factors, including signal transducer and activator of transcription 1 (STAT1) (8,9), tumor necrosis factor receptor-associated factor 6 (TRAF6) and interleukin-1 receptor-associated kinase 1 (IRAK1) (10-13). Fig. 1 illustrates the biological functions and top 30 enriched pathways of miR-146a-5p from the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis [the results were obtained from the CancerMIRNome database (http://bioinfo.jialab-ucr.org/CancerMIRNome/)]. The nuclear factor-κB (NF-κB) signaling pathway and Toll-like receptor (TLR) are related to inflammation. Binding cytokine receptor is also an inflammation-related biological function. The present review focused on describing the mechanisms and application strategies of miRNA-146a in regulating inflammation and fibrosis at molecular and cellular levels.

2. Location and transcription regulation of miR-146a

The miR-146 family (miR-146a and miR-146b) are homologous, with only two nucleotides differing in their 3′ region. Their coding genes are located on 10 (10q24.32) and chromosomes 5 (5q33.3) in humans (14). These isoforms have similarities and differences in function and target molecules. They have unique regulatory functions for the signaling pathway (15,16). The miR-146a precursor contains a 5′ arm sequence (miR-146a-5p) and a 3′ arm sequence (miR-146a-3p). miR-146a is the main topic of the present review. The gene motif's binding promoter of miR-146a reported in studies on inflammation or fibrosis will be discussed. Liu et al (17) found two pairs of NF-κB binding motifs (GGG ATT TCC and GGG ACT TTC C in humans) and forkhead box protein 3 (FOXP3) binding motifs (GCC AAC A and GCA AAT A in humans) in the miR-146a promoter. NF-κB is a pivotal factor in inflammation and fibrosis, while FOXP3 is the transcription factor of T-regulatory (Treg) cells and participants in immune responses. IL-1 receptors (IL-1R)/TLRs stimuli, such as lipopolysaccharide (LPS) and NF-κB signaling, lead to its expression. The expression of miR-146a downregulates the activity of these pathways (14,15,18). In rats with acute spinal cord injury, Ni et al (19) found that tri-methylation recruitment at lysine 27 of histone H3 (H3K27me3) around the upstream region in the promoter of miR-146a reduced miR-146 expression, suggesting that H3K27me3 participates in the epigenetic regulation of miR-146a. By interfering with the maturation process of miRNA, genetic variants in miRNA precursors affect miRNA expression levels and thereby cause disease susceptibility. These variants may occur in the miRNA promoter and precursor regions. Single nucleotide polymorphisms (SNPs) are essential. As a result of the rs2910164 polymorphism, the pre-miR-146a's stem region is mismatched, which indirectly influences the expression of mature miR-146a (20). G:U in rs2910164 changes to C:U, causing mispairing in miR-146a's hairpin and decreasing the efficiency of processing miRNA precursors into mature miRNA, thus suppressing the expression of miR-146a (21). V-Ets oncogene homolog 1 (Ets-1) is a member of the Ets family of transcription factors that activates the miR-146a promoter and directly regulates miR-146a expression. The inflammation risk-associated G allele of rs57095329 inhibits the binding affinity for Ets-1 and leads to an inappropriate compositional change, and is thus another genetic factor suppressing miR-146a expression (22). These two SNPs were in a strong linkage disequilibrium. Cui et al (23) found that the miRNA-146a rs57095329 A allele carried a lower risk and seizure frequency of drug-resistant epilepsy (Fig. 2).

3. miR-146a and inflammation

Inhibition of inflammation via the IL-1R/TLRs-NF-κB axis

It is thought that downstream of these receptors are similar signaling cascades, as TLRs and IL-1R both express Toll/IL-1 receptor (TIR) domains (24). The current understanding of TLR signaling is primarily based on studies focusing on downstream IL-1R signaling. TLR2 is involved in peptidoglycan, lipoteichoic acid (LTA) and bacterial lipoprotein metabolism. The stimuli of TLR4 contain LPS, LTA and oxidized low-density lipoprotein (25,26). TLRs transduce downstream signals by mediating the myeloid differentiation factor 88 (MyD88) or TIR domain-containing adapter molecule (TRIF)-related pathways. The binding of IRAK4 and IRAK1/2 are induced to form the myddosome when MyD88 recruits to the TIR domain of TLRs. TRAF6 further activates downstream of MyD88 and triggers NF-κB signaling, inducing pro-inflammatory responses. TRAF6 also induces endosomal TLR4, which is related to TRIF and contributes to pro-inflammatory responses (27). Activated inhibitor κ kinase β induces the phosphorylation of inhibitor of κBα (IκBα), leading to the ubiquitination and subsequent degradation of IκBα, which promotes the release of NF-κB dimers from the cytoplasm to the nucleus (28-30). The canonical NF κB pathway regulates the production of pro inflammatory cytokines, growth factors, chemokines, matrix metalloproteinase, anti apoptotic proteins, pro inflammatory enzymes, intercellular adhesion molecule 1 and inhibitors of NFκ B signaling (such as IκBα) (31,32).

The IL-1R/TLRs-NF-κB signaling pathway functions mainly by targeting IRAK1 and TRAF6. Numerous studies have reported that miR-146a reduced gene expression of IRAK1 and TRAF6 (33-36). In a study of kidney ischemia/reperfusion injury, Li et al (37) found that miR-146a downregulated the expression of IRAK1 through binding to its 3′UTR and ultimately inactivated NF-κB, inhibiting the infiltration of inflammatory cells and protecting kidney functions. Another study reported that miR-146a targeted the 3′UTR of TRAF6 and induced LPS-TLR4-NF-κB pathway inhibition, reducing inflammatory mediators such as IL6 and TNF-α (38). Zhang et al (39) reported that the 3′UTR of TRAF6 and IRAK1 had binding domains for miR-146a. The release of inflammatory cytokines was decreased by the upregulation of miR-146a, blunting IL-1R1/TLR4 (40). When TLR2 of THP-1 cells was stimulated by bacterial lipoprotein, Quinn et al (41) found that overexpression of miR-146a reduced TNF-α expression. When they used LPS to stimulate TLR4 of the human hepatic stellate cell (HSC) line LX2, Chen et al (42) found that inflammatory responses and fibrogenesis were inhibited by the overexpression of miR-146a. During inflammation, with the overactivation of the NF-κB pathway, the expression and activity of miR-146a in immune cells led to the downregulation of IRAK1 and TRAF6 genes, finally decreasing the expression of NF-κB transcription factors, particularly inflammatory cytokines. In mice, acute and chronic inflammatory autoimmune responses were overactive in miR-146a-deficient T cells (43). Certain studies reported worsening inflammation in several diseases associated with deficiency of miR-146a (44-46). miR-146a treatment of experimental autoimmune anterior uveitis resulted in significant variations in cytokine production. Pro-inflammatory cytokines were inhibited, such as IL-1β, IL-6 and IFN-γ, while anti-inflammatory cytokines such as IL-10 increased (47). Lv et al (48) found that overexpression of miR-146a inhibited TRAF6/NF-κB activation in LPS-induced nucleus pulposus cells. Furthermore, when miR-146a was deficient in injured mice, inflammation at the wound site deteriorated due to dysregulation of pro-inflammatory cytokines, IRAK1 and TRAF6 (49). These findings suggest that miR-146a attenuates inflammatory responses by targeting the upstream IRAK1 and TRAF6 of NF-κB, the center of the IL-1R/TLRs signaling pathway.

Inhibition of inflammation via the Janus kinase (JAK)-STAT axis

Tang et al (50) identified that upregulation of miR-146a decreased the expression of the STAT1 genes, thereby controlling human lupus erythematosus through the IFN pathway. He et al (51) demonstrated that hepatic schistosomiasis increased the expression of miR-146a/b in macrophages. IFN-γ inducing macrophages to differentiate into M1 macrophages was reduced by upregulated miR-146a targeting STAT1, presumably as a mechanism of it inhibiting the pro-inflammatory function of M1 macrophages (51). miR-146a regulated the IFN-γ-dependent Th1 cell-mediated spontaneous immunopathology and reduced Treg cell trans-differentiation into Th1 cells by controlling the overactivation of STAT1 in Treg cells (8).

STAT3 is involved in retinal endothelial inflammation in type 1 diabetes and high glucose-induced endoplasmic reticulum stress (52). Elevated STAT3 phosphorylation led to increased production of inflammatory cytokines from macrophages (53). Ocular JAK/STAT3 signaling is activated by IL-6 (54-56), while miR-146a inhibits IL-6. Ye and Steinle (57) reported that miR-146a inhibited inflammation and apoptosis as a potential molecular treatment in the diabetic retina by participating in inhibiting the IL-6-related JAK/STAT3 pathway. Furthermore, miR-146a was reported to inhibit the pro-inflammatory function of STAT3 by targeting homeodomain-interacting protein kinases 3 (58). Downregulation of miR-146a increased the levels of phosphorylated STAT3 protein (59), consistent with the result that STAT3 had significantly higher activity in miR-146a-deficient mice (60). A western blot study by Sun et al (61) also demonstrated that miRNA-146a-5p inhibited the activation of STAT3. miR-146a is also involved in inflammation-related Treg/Th17 differentiation by targeting STAT1/STAT3, which will be introduced in the next section.

miR-146a is involved in inflammation by regulating immune cells Regulation of macrophages

To be consistent regarding Th1 and Th2 differentiation paradigms for T cells, macrophage differentiation involved in the production of various phenotypes was divided into canonical M1 polarization and alternative M2 polarization. M1 macrophages are predominantly pro-inflammatory (62,63). The enhanced expressions of cell-surface activation factors, such as major histocompatibility complex class II (MHCII), CD80 and CD86, and the production of pro-inflammatory factors are the main characteristics involved in antigen presentation functions for M1 activation. M2 macrophages have numerous functions, including inhibiting inflammation and promoting tissue repair (64-67). Peng et al (68) demonstrated miR-146a-induced M2 polarization through TLR4/NF-κB to attenuate inflammation and repair wounds in mice with diabetic ulcers. miR-146a has also been found to be involved in inhibiting M1 macrophage polarization (69,70). Huang et al (70) demonstrated that the overexpression of miR-146a in M1 macrophages significantly reduced the expression of pro-inflammatory cytokines; on the contrary, miR-146a inhibition promoted the polarization of M1 macrophages. miR-146a inhibits M1 pro-inflammatory responses and miR-146a-deficient diabetic mice exhibited increased M1 and weakened M2 responses, aggravating renal injury (45). Overexpression of miR-146a improved LPS-induced inflammatory responses in macrophages of older mice (71). Macrophage maturation was downregulated by miR-146a through decreased expression of CD80 and CD86, attenuating inflammation (72). miR-146a inhibited M1 polarization and induced M2 polarization by regulating NLRP3 (73) and NOTCH1 (74). The inflammatory inhibition of miR-146a by regulating macrophage polarization was also identified in other studies. By sponging miR-146a, long non-coding RNAs (lncRNAs) such as lncRNA CHRF and lncRNA HCG18 induced M1 polarization and inflammatory responses (75,76).

Regulation of the Th17/Treg balance

Studies indicated that the expression of miR-146a increased in Treg cells (77,78). miR-146a-deficient mice had an elevated percentage of FOXP3-CD4+ T cells and IFN-γ-dependent Th1 pro-inflammatory responses increased in miR-146a-deficient mice through activating STAT1 (target of miR-146) (8). However, one study reported that in patients with allergic rhinitis, miR-146a elevated Treg functions and differentiation via targeting STAT5b (79). Guo et al (58) indicated that miR-146a mediated Treg-cell differentiation via upregulating FOXP3 expression. IL-6-driven STAT3 signaling is required for the nuclear retinoic acid receptor-related orphan receptor-γ-t function involved in Th17-cell differentiation. Li et al (80) reported that experimental autoimmune encephalomyelitis deteriorated and the differentiation into Th17 cells increased in miR-146a knock-out 2D2 T cells. miR-146a reduced the production of IL-6 and IL-21 in 2D2 T cells, reducing their Th17 differentiation via the STAT3 signaling pathway (80). One study further reported that the inhibition of miR-146a elevated the expression of the NF-κB, STAT3 and TRAF6 genes, leading to the production of pro-inflammatory cytokines such as IL-6, IL-17A and IL-21, which mediated Th17-cell differentiation (59). In oral lichen planus, one group reported that FOXP3 reduced the expression of downstream TRAF6 and promoted CD4+ T-cell differentiation into Treg cells by regulating miR-146a, thereby alleviating chronic inflammation (81). They further indicated that lncRNA DQ786243 overexpression significantly enhanced the expression of miR-146a by targeting FOXP3, thereby suppressing the NF-κB pathway (82).

Regulation of dendritic cell (DC) functions

DC maturation is characterized by the upregulation of the surface antigens CD80 and CD86 (83). It has been reported that miR-146a mimics contributed to immature DC, inducing Treg differentiation and reducing the expression of CD80 and CD86 in the DC surface via the Notch-1 signal pathway. By contrast, miR-146a inhibition had the opposite effect (84). miR-146a appears to be involved in DC tolerogenic properties based on these findings. A study reported that miR-146a overexpression inhibited DC maturation and antigen presentation (85). Through targeting the JAK/STAT1 signaling pathway, miR-146a was indicated to downregulate the expression of MHCII and activation of DCs and decrease the production of inflammatory factors (86). These findings are consistent with another study (87). miR-146a expression in human plasmacytoid DCs reduced their ability to mediate the proliferation of CD4+ T-cell proliferation by downregulating costimulatory molecules and MHCII on its surface (88). Park et al (89) demonstrated that miR-146a overexpression in mature DCs decreased cytokine production and enhanced DC apoptosis by targeting IRAK1 and TRAF6. These studies indicate that upregulation of miR-146a suppresses the activation, maturation and antigen presentation function of DCs via several signaling pathways, and miR-146a may mediate Treg differentiation via DCs.

Regulation of natural killer (NK)-cell functions

Xu et al (90) indicated that overexpression of miR-146a reduced the cytotoxic effect induced by NK cells and produced pro-inflammatory cytokines by targeting STAT1. They reported that anti-inflammatory factors transforming growth factor-β (TGF-β) and IL-10 induced the expression of miR-146a (90). Pesce et al (91) demonstrated that miR-146a regulated NK cell-mediated cytotoxicity via targeting killer Ig-like receptors. They reported that miR-146a downregulated the levels of IFN-γ in human NK cells and prevented NK cells from becoming over-activated and overproducing IFN-γ by targeting IRAK1 and TRAF6 (92). There are relatively limited studies on the relationship of miR-146a with NK cells, particularly in inflammation. These studies suggested that miR-146a may negatively regulate inflammation through participating in NK-cell functions (Fig. 3).

Figure 3

MiR-146a regulates inflammation. MiR-146a is critical in producing inflammatory cytokines and differentiating immune cells, such as CD+ T cells, DCs and macrophages. In DCs, miR-146a inhibits the expression of MHCII through the JAK/STAT1 signaling pathway, and the expression of CD80 and CD86 is also reduced, which impacts the maturation and antigen presentation function of DCs. By contrast, miR-146a suppresses the production of inflammatory cytokines of macrophages by promoting its M2 polarization and targeting the NF-κB signaling pathway. Finally, for the decrease of stimuli from antigen-presenting cells and the functions of miR-146a through different signaling pathways (such as the inhibition of JAK-STAT1/3 and the promotion of STAT5), CD4+ T cells increase anti-inflammatory Treg cell differentiation and decrease pro-inflammatory Th17 and Th1 differentiation. The figure was drawn with Figdraw (https://www.figdraw.com). miR, microRNA; Th1, type 1 T-helper cell; MHC, major histocompatibility complex; DC, dendritic cell; Treg cell, T-regulatory cell; TLR, Toll-like receptor; FOXP, forkhead box protein; LPS, lipopolysaccharide; PGN; peptidoglycan; LTA, lipoteichoic acid; IRAK1, interleukin-1 receptor-associated kinase 1; TRAF6, tumor necrosis factor receptor-associated factor 6; STAT, signal transducers and activators of transcription; NF-κB, nuclear factor-κB; JAK, Janus kinase; MYD88, myeloid differentiation factor 88; NLRP3, NACHT, LRR and PYD domain-containing protein 3; RORγ, retinoic acid receptor-related orphan receptor γ; CIITA, MHCII transactivator; TAK1, TGF-β-activated kinase 1; TAB, TAK-binding proteins; TCR, T-cell receptor.

4. miR-146a and fibrosis

Approximately 45-50% of deaths may be ascribed to fibrosis in developed countries (1,93). Excessive extracellular matrix (ECM) production is a feature of fibrosis and excessive matrix shrinkage in various organs (94). Fibrosis is primarily caused by chronic inflammation mediated by several stimuli (1). In fibrosis models, epithelial cells trans-differentiate into myofibroblasts via EMT activated by chronic inflammation (95). Myofibroblasts produce ECM in the progression of physiological tissue repair and organ fibrosis. Myofibroblasts are derived from endothelial cells, epithelial cells, fixed fibroblasts, smooth muscle cells, pericytes, bone marrow-derived progenitor cell lines and bone marrow-derived fibroblasts (96). The deposition of connective tissue components leads to increased stiffness in the affected tissue and impairs the diffusion of oxygen and nutrients, further inducing sustained myofibroblast activation and cell damage (97-99). Epigenetic modifications induced by profibrotic environments shield myofibroblasts from external stimuli, forming a malignant expansion loop (100,101). Due to the profibrotic effect of excessive fibrosis, TGF-β and other signaling pathways trigger further fibrosis, culminating in organ failure and dysfunction (102).

miR-146a is a pivotal antifibrotic miRNA in numerous tissues (103,104). Studies highlighted the possibility of treating fibrosis by interfering with miR-146 (105,106). Downregulation of miR-146a-5p was found in cirrhotic livers of CCl4-treated rats and upregulation of miR-146a suppressed the profibrotic function of TGF-β by targeting downstream TGF-β signaling (107). Src kinase inhibitor significantly attenuated the progression of skin and lung fibrosis in mice (108,109). miR-146a overexpression reduced the production of α-SMA and collagen 1 (Col-1) in TGF-β-treated human HSCs by inhibiting Src synthesis (110). It was found that miR-146 expression was downregulated in a fibrotic mouse model after acute muscle injury and overexpression of miR-146a attenuated the secretion of ECM in vitro and in vivo (111). Liu et al (112) reported that miR-146a reduced the production of Col-1 and hyaluronic acid of ECM in orbital fibroblasts. After induction of ischemia/reperfusion injury, tubular injury, inflammatory infiltrates and fibrosis worsened in miR-146a-deficient mice as compared to wild-type mice (113). miR-146a suppressed myocardial fibrosis by targeting the TRAF6 and IRAK1 genes by targeting the TLR-4-NF-κB pathway (114).

These studies indicate that miR-146a regulates the fibrosis process inside cells through signaling pathways, which are classified into two types in the present review (Fig. 4). In addition, it is also necessary to explore the functions of miR-146a at the level of fibrosis-related cells in the future.

Main signaling pathways of miR-146a
Small mothers against decapentaplegic (Smad)-dependent TGF-β signaling pathway

The TGF-β signaling pathway regulates adaptive and innate immunity, fibrosis and inflammation (115). TGF-β switching accelerated the development or progression of fibrosis in chronic inflammatory autoimmune conditions (116,117). The activation of fibrotic factors produced by inflammatory and epithelial cells was observed during the progression of chronic autoimmune diseases. Organ fibrosis is characterized by sustained fibroblast proliferation and inflammation (118). TGF-β1 treatment of HSCs led to elevated expression of α-SMA and Col-1 and decreased expression of miR-146a-5p (119). Overexpression of miR-146a reduced the levels of α-SMA and Col-1 in TGF-β-treated HSCs (110).

The primary signaling pathway mediated by TGF-β involves the phosphorylation of Smad2/3 protein to form receptor-mediated Smad/co-regulated Smad4 complex (120). This Smad complex accumulates in the nucleus and triggers the transcription of target genes (e.g., SNAIL) by activating transcription factors (121) and mediating signaling events associated with EMT activation. SNAIL is involved in the downregulation of E-cadherin and claudins, upregulating vimentin (VIM) and fibronectin (122). The induction of EMT markers and SNAIL activation led to upregulation of the ECM markers Col-1 and VIM and the suppression of the epithelial marker E-cadherin. Several studies indicated that Smad4 is the target gene of miR-146a (119,123,124). After miR-146a was transferred, the expression of α-SMA, VIM and Col-1 in injured mice was significantly reduced; however, after simultaneous treatment with Smad4 and miR-146a, the expression of these fibrosis markers was significantly enhanced, suggesting that miR-146 ameliorates skeletal muscle fibrosis by downregulating Smad4 (111). The inflammatory microenvironment, including IL-17, IL-22, IL-6 and other pro-inflammatory cytokines, may induce EMT through TGF-β/Smad or non-Smad signaling pathways (125). miR-146a may be crucial in the process of EMT-related fibrosis mediated by TGF-β.

NF-κB/BMP and endogenous bait receptors for activin membrane-binding inhibitors (Bambi) signaling pathway

In quiescent HSCs, TGF-β1 signaling was suppressed by Bambi, an endogenous decoy receptor for TGF-β (126). In addition to serving as bait, Bambi directly interfered with TGF-β1 signaling by targeting Smad7 (127). Jiang et al (128) found that Enhancer of zeste homolog 2 (EZH2) inhibition led to transcriptional block of the TGF-β1 pathway, the cell cycle pathway and numerous ECM components in primary HSCs. EZH2 inhibition reduced the recruitment of H3K27me3 to target genes encoding Bambi and increased Bambi expression. Ni et al (19) observed that LPS upregulated H3K27me3 and EZH2 inhibitors increased miR-146a expression by regulating H3K27me3 methylation. This finding suggests that suppressing the recruitment of H3K27me3 around the promoter may increase the expression of miR-146a and Bambi. Under profibrotic stimulation, Bambi expression was downregulated, activating the TGF-β1 pathway of hematopoietic stem cells and activating hematopoietic stem cells (129). LPS is an essential factor in the downregulation of Bambi during cirrhosis progression (130). Bambi is a functional inhibitor of the TGF-β receptor that is downregulated by NF-κB. With the progression of cirrhosis, bacterial translocation and LPS levels increased (131). LPS binds to TLR4 and recruits TRAF6, IRAK1 and TGF-β-activated kinase 1, inducing phosphorylation, ubiquitination and degradation of IκBα. Subsequently, NF-κB dissociates from IκBα and enters the nucleus, inducing downregulation of Bambi, which elevates the response of HSCs to TGF-β1 stimulation (132,133). A gene expression array of total RNA from miR-146a-overexpressed HSC-2 cells and control cells indicated that Bambi mRNA was upregulated in clones overexpressing miR-146a-5p (134). Zou et al (107) found that Bambi protein was highly expressed after mi-146a-5p overexpression in TGF-β1-stimulated cells pretreated by LPS.

During fibrosis, miR-146a suppresses the activation of NF-κB by targeting its upstream genes IRAK1 and TRAF6. The expression of Bambi then increases due to the inactivated NF-κB and induces the inhibition of the TGF-β/SAMD signaling pathway. While miR-146a regulates fibrosis, H3K27me3-mediated epigenetic regulation has a considerable role. The effect of miR-146a on fibrosis is summarized in Table I. These findings indicate that miR-146a regulates EMT and fibrotic factors such as α-SMA, COL-1 and COL-4 to exert its antifibrotic functions via these signaling pathways (Fig. 5).

Table I

Expression patterns and functions of microRNA-146a in different fibrosis models.

Table I

Expression patterns and functions of microRNA-146a in different fibrosis models.

Research subjectTarget or mediationSourceExpressionEffect(Refs.)
Liver fibrosis ratsTRAF6; IRAK1; Smad4; BambiHSCsDownAbrogation of hepatic fibrosis by suppressing both TGFβ/Smad and NF-κB/Bambi signaling pathways in HSCs(107)
Radiation-induced liver fibrosisSRCHSC LX2DownSuppression of α-SMA and Col-1 expression(110)
Hepatic fibrosis in ratsSmad4Liver fibrotic tissuesDownInhibition of TGF-β1-induced α-SMA expression(119)
Mice after acute muscle contusionSmad4Muscle cellsDownAttenuation of the effect of TGF-β on the expression of fibrosis markers(111)
LPS-induced HSC LX2TRAF6HSC LX2DownMediates downregulation of TRAF6; suppresses the phosphorylation of Smad2 and attenuates the expression of α-SMA(135)
Myocardial IRI miceNOX4Heart tissues and cardiomyocytesDownRegulates the transcription of NOX4, which affects P38 signaling in cardiac fibrosis(136)
A549 cells treated with TGF-β1L1CAMA549 cells-Inhibits EMT process via targeting L1CAM(137)
STZ-induced diabetes in miceTRAF6; IRAK1Heart endothelial cellsDownReduces the expression of Col-1 α1 and Col-4 α1(138)
Irradiated HSC LX2RAC1HSC LX2UpDecreases the expression of α-SMA and Col-1(139)
Nonalcoholic fibrosing steatohepatitis in miceWnt1; Wnt5Activated HSCsDownDecreases the expression of α-SMA and Col-1(140)
TGF-β1-induced dermal fibroblastSmad4MyofibroblastUpReduces the expression of α-SMA(106)
Isoproterenol-induced cardiac fibrosis in ratsEGF2Cardiac fibroblastsDownUpregulates the expression of basic FGF2, Col-1 and α-SMA(141)
Constrictive pericarditis-induced MF in ratsTRAF6; IRAK1Myocardial tissue8 weeks: Up; 16 weeks: DownSuppresses myocardial fibrosis by inhibiting TRAF6 and IRAK1 via the TLR-4 signaling pathway(142)
Human LFHSmad4Human LF tissuesDownAttenuates the progression of TGF-β1- induced fibrosis(143)

[i] FGF2, fibroblast growth factor 2; Col-1, collagen I; α-SMA, smooth muscle α-actin; MF, myocardial fibrosis; HSCs, hepatic stellate cells; LPS, lipopolysaccharide; IRI, ischemia/reperfusion injury; NOX4, NADPH oxidase 4; L1CAM, L1 cell adhesion molecule; LFH, ligamentum flavum hypertrophy; STZ, streptozotocin; Col-4, collagen 4; SRC, proto-oncogene tyrosine-protein kinase; RAC1, Ras-related C3 botulinum toxin substrate 1; EMT, endothelial to mesenchymal transition; IRAK1, interleukin-1 receptor-associated kinase 1; TRAF6, tumor necrosis factor receptor-associated factor 6; Bambi, BMP and endogenous bait receptors for activin membrane-binding inhibitors.

miR-146a regulates fibrosis through fibrosis-related cells Regulation of fibroblasts

Liu et al (106) reported that miR-146a downregulated α-SMA expression and myofibroblast transdifferentiation induced by TGF-β1 via inhibiting Smad4. In an isoproterenol-induced cardiac fibrosis model, Zhang et al (141) observed that the expression of miR-146a was reduced in cardiac fibroblasts, while α-SMA, Col-1 and fibroblast growth factor 2 were increased. These trends were reversed when miR-146a was upregulated by miR-146a mimics (141). The absence of miR-146a led to the proliferation of synovial fibroblasts due to increased expression of TRAF6 (144). miR-146a mimics decreased the production of α-SMA, Col-1α and fibronectin induced by TGF-β in orbital fibroblasts by targeting Smad4 and TRAF6 (145). These findings suggest that miR-146a participates in fibrocyte differentiation and regulates fibroblast functions via different pathways.

Regulation of macrophages

Macrophages derived from monocytes produce numerous factors, which modulate fibrosis and tissue repair. TGF-β derived from macrophages induces fibroblast migration and promotes fibroblast growth, activation and collagen synthesis (146,147). Due to the essential role of macrophages in regulating fibrosis, researchers adopted several approaches to transfer, reduce and restrain macrophage migration into tissues (148,149). Bhatt et al (45) found that the expression of M1 polarization antigens was enhanced, while the expression of M2 polarization antigens was inhibited in miR-146a-deficient macrophages, inducing the infiltration of macrophages and kidney fibrosis. Another study reported that delivery of miR-146a using polyethyleneimine nanoparticles inhibited macrophage infiltration and renal fibrosis in vivo (105). Another study found that miR-146a attenuated fibrosis in hepatic schistosomiasis by regulating macrophage differentiation into M2 cells (51). The differentiation of M1 macrophages to M2 macrophages reduced the stimulation of inflammation to fibrosis and promoted matrix remodeling and injury healing (3). There is substantial opportunity for studies on miR-146a regulating macrophages in fibrosis.

5. Application strategies

miRNAs are crucial regulators of gene expression, suggesting that they have the potential to serve as biomarkers for diagnosis and prediction of prognosis, as well as treatment targets for diseases (150).

miR-146a as a biomarker in inflammation and fibrosis
Diagnosis

Due to its significant relationship with inflammation and fibrosis, numerous scientists suggest that miR-146a may be essential for diagnosing related diseases. Shumnalieva et al (151) found that miR-146a was overexpressed in peripheral blood compared with a healthy control group. miR-146a was also downregulated in cases with lupus nephritis compared to negative controls, and based on its expression levels, it was possible to identify lupus nephritis and assess its activity (152). Abou-Zeid et al (153) found that miR-146a levels were elevated in cases with rheumatoid arthritis and may act as a potentially effective marker of disease activity. Li et al (154) reported that miR-146a regulated the inflammatory response during Helicobacter pylori infection via regulating the NF-κB pathway. miR-146a and IL-17A expression were positively correlated in H. pylori-infected human gastric mucosa (154). As inflammation has a crucial role in the pathogenesis of acute coronary syndrome, serum exome miR-146a levels were significantly higher in patients than in normal coronary arteries (155). Yang et al (156) reported that miR-146a inhibited the accumulation of low-density lipoprotein cholesterol and inflammatory responses by targeting TLR-4 signaling. They also demonstrated that expression levels of miR-146a increased initially and then decreased in atherosclerotic endothelial cells (156). In most fibrosis models, miR-146a expression was downregulated, while certain studies indicated that expression may be upregulated in the early stages of fibrosis (142). These findings suggest that increased miR-146a expression may be a protective factor in early disease stages.

Prognosis

Wu et al (157) found that the plasma miR-146a expression and inflammatory factors were positively associated with the severities of osteoarthritis, suggesting that miR-146a levels were able to act as a biomarker of osteoarthritis. Another study on generalized aggressive periodontitis demonstrated higher miR-146a expression in patients than in healthy controls that were directly consistent with disease severity and a reduction in the levels of pro-inflammatory cytokines, suggesting that miR-146a might indicate periodontal disease severity (158). miR-146a reduction was reported by Zhu et al (152) to increase the possibility of progression to end-stage renal disease and recurrence within one year. A study on tocilizumab treating multifocal interstitial pneumonia induced by COVID-19 found that in patients with the lowest peripheral expression of miR-146a-5p, the condition was the worst, linking inflammation and COVID-19 evolution (159). Serum exosomal miR-146a-5p levels had the potential to identify mild fibrosis from severe fibrosis and fibrosis (grades I-III) from no fibrosis (160). miR-146a-5p distinguished patients with mild vs. severe fibrosis (161). These findings suggest that miR-146a may serve as a prognostic marker due to its tight relationship with inflammation and fibrosis severity.

Therapy
Direct delivery

The primary challenge for delivering miRNAs is their limited ability to penetrate cell membranes due to their negative charge and rapid degradation in vivo. Effective internalization of therapeutic agents into target cells without cytotoxic or immunogenic effects is critical for obtaining the desired therapeutic effect, particularly when the aim is to target unwanted inflammation. To date, several substances have been used to develop delivery systems for miR-146a and its mimics, including viral vectors such as adenovirus (162) and nano-carriers consisting of polyethylene glycol-poly (lactic acid) nanoparticles (163), cerium oxide nanoparticles (164) and polyethyleneimine nanoparticles (105). To promote absorption, the nanoparticles are frequently modified with lectins or penetrating peptides (165) or doped with chitosan (166). Although viral vectors possess good stability and transfection efficiency, they may induce adverse inflammatory responses. Therefore, nano-carriers appear to be the primary focus. Studies indicated anti-fibrotic effects after delivering extracellular vesicles containing miR-146a (37,143,167,168). Studies on the delivery of miR-146a to treat inflammation and fibrosis are summarized in Table II. Their treatment effects appear to be dose-dependent.

Table II

Applications of delivering microRNA-146a in fibrosis and inflammation models.

Table II

Applications of delivering microRNA-146a in fibrosis and inflammation models.

DiseaseDelivery vectorsTargetEffect(Refs.)
Microglial-mediated neuroinflammationhUMSC-ExosIRAK1; TRAF6Ameliorates neuroinflammation(35)
IRIhUSC-ExoIRAK1Inhibits the activation of NF-κB and decreases the infiltration of inflammatory cells(37)
Renal fibrosisPolyethylenimine nanoparticlesTRAF6; Smad4Attenuates renal fibrosis(105)
NECAdenovirusNLRP3 inflammasome; CLIC4Attenuates inflammation and intestinal injury in the NEC-affected intestine(162)
Acute lung injuryCerium oxide nanoparticlesCol-1α2Reduces inflammation and decreases collagen deposition(164)
Allergic rhinitisChitosan hydrogel doped with PEG-PLA nanoparticlesNF-κBInduces less inflammation(166)
LFHhUCMSC-EVsSmad4Attenuates the progression of TGF-β1- induced fibrosis(143)
Liver fibrosisHLSCs-EVs-Decreases the expression of α-SMA and Col-1α1(167)
Myocardial infarctionExosomes within alginate derivative hydrogelTRAF6 IRAK1Induces lower degrees of fibrosis(169)
Contact dermatitisNickFect type of cell-penetrating peptidesIRAK1Reduces ear swelling response and downregulates pro-inflammatory cytokines (IL-6, IL-1β, IL-33 and TNF-α)(170)
SCCIICell permeable peptideIRAKI; TRAF6Regulates the neuroinflammatory response(171)
Injury during mechanical ventilationPolyethylenimine- lipid nanoparticle-Attenuates inflammatory response during mechanical ventilation(172)

[i] LFH, ligamentum flavum hypertrophy; hUCMSC-EVs, human umbilical cord mesenchymal stromal cell-derived extracellular vesicles; HLSCs-EVs, human liver stem cell-derived extracellular vesicles; PEG-PLA, polyethylene glycol-poly(lactic acid); SCCII, severe controlled cortical impact injury; NLRP3, NACHT, LRR and PYD domains-containing protein 3; NEC, necrotizing enterocolitis; hUMSC-Exos, human umbilical cord mesenchymal stem cell-derived exosomes; IRI, ischemia/reperfusion injury; hUSC-Exo, human urine-derived stem cell-derived exosomes; IRAK1, interleukin-1 receptor-associated kinase 1; TRAF6, tumor necrosis factor receptor-associated factor 6; SMA, smooth muscle actin; Col-1, collagen I.

miR-146a functions as a target

Drugs such as vildagliptin (173) and GSKJ4 (174) attenuated inflammation by regulating miR-146a expression. Other studies reported that inhibition of lncRNAs such as XIST (175) and SNHG16 (176) suppressed inflammatory responses by sponging miR-146a. Knockdown of the lncRNA MALAT1 affected inflammation by increasing miR-146a expression in an LPS-induced acute lung injury model (177). The recombinant Schistosoma japonicum protein P40 increased the levels of miR-146a in LX-2 cells and attenuated hepatic fibrosis by targeting Smad4 (178). Most drugs are designed according to miR-146a function by targeting the promoter of miR-146a, while lncRNAs act via their sponging effect.

6. Conclusions and perspectives

The present study focused on the functions of miR-146a in inflammation and fibrosis. The former may involve the IL-1R/TLRs-NF-κB axis and the JAK-STAT signaling pathway. By regulating microglial M1 and M2 polarization, miR-146a demonstrated critical roles in inflammation and fibrosis. The effects of miR-146a on fibrosis development were discussed in the present study. However, substantial knowledge gaps remain. There was frequent disagreement among the results obtained by different studies, possibly due to preanalytical factors such as acquiring samples, storage and small sample sizes (179). For instance, studies reported that they controlled inflammatory responses via the inhibition of miR-146a expression (173,180), while for inflammation therapy, miR-146a mimics were delivered in vivo to upregulate miR-146a levels (176). These principles appear mutually contradictory. In addition, M1 macrophages have a pro-inflammatory effect, while M2 tends to confine inflammation and remodel the matrix or promote fibrosis when inflammation cannot be resolved. Therefore, miR-146a inhibiting M1 polarization of macrophages and promoting M2 polarization may lead to increased fibrosis, while simultaneously serving anti-inflammatory functions, which appears contradictory. A reasonable explanation is that miR-146a promotes a proportional balance between them (51). It should be noted that off-target effects complicate the therapeutic journey, as a single miRNA possesses various targets controlled by cellular substances and environments.

Furthermore, exogenous miRNAs may alter the balance of miRISC-endogenous miRNAs by interacting with miRISC unnecessarily. Treatments based on oligonucleotides, such as miRNA mimics and miRNA inhibitors, should be specific to certain cell types. Synthetic double-stranded miRNAs, which regulate the expression of endogenous miRNA, must be constituted in a manner that allows them to be freely adsorbed by cells, as well as keeping their effectiveness for the intended time (181). A system that is able to deliver the exogenous miRNAs in a standardized and efficient manner should be established. Furthermore, it is necessary to determine mRNA target-specific activity based on a profound comprehension of cellular complexity.

A substantial body of evidence demonstrated that various miRNAs, particularly miR-146a, are essential regulators of inflammation and fibrosis, focusing on the potential of miR-146a as a biomarker and target with a clinical application value. A deeper understanding of the miR-146a-related signaling pathway and its functions will facilitate the early diagnosis and treatment of disease.

Availability of data and materials

Not applicable.

Authors' contributions

ZFL and GFS made all contributions to the design and conception for the study; RJZ performed the literature search; ZFL and GFS wrote and revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

EMT

epithelial-mesenchymal transition

α-SMA

smooth muscle α-actin

miRNA

microRNA

RISC

RNA-induced silencing complex

3′UTR

3′-untranslated region

IRAK1

interleukin-1 receptor-associated kinase 1

TRAF6

tumor necrosis factor receptor-associated factor 6

STAT

signal transducers and activators of transcription

NF-κB

nuclear factor-κB

JAK

Janus kinase

KEGG

Kyoto Encyclopedia of Genes and Genomes

GO

Gene Ontology

FOXP3

forkhead box protein 3

IL-1R

IL-1 receptor

TLR

Toll-like receptor

LPS

lipopolysaccharide

H3K27me3

tri-methylation at lysine 27 of histone H3

ETS-1

V-Ets oncogene homolog 1

TIR

Toll/IL-1 receptor

MYD88

myeloid differentiation factor 88

TRIF

TIR domain-containing adaptor molecule

IκBα

inhibitor of kappa Bα

HSC

hepatic stellate cell

Th1

type 1 T helper cell

Treg

regulatory T cell

MHCII

major histocompatibility complex class II

lncRNA

long non-coding RNA

DC

dendritic cell

NK

natural killer

TGF-β

transforming growth factor-β

ECM

extracellular matrix

Col-1

collagen 1

Smad

Small mothers against decapentaplegic

R-smad

receptor-mediated Smad

VIM

vimentin

Bambi

BMP and endogenous bait receptors for activin membrane-binding inhibitors

EZH2

enhancer of zeste homolog 2

Acknowledgments

Not applicable.

Funding

This work was supported by the Natural Science Foundation of Ningbo Municipality (grant nos. 202003N4269 and 2019C50069), the grants of basic public welfare projects in Zhejiang province (grant no. LGF19H020004), Zhejiang Province Medical and Health Project (grant nos. 2017ZD026 and 2020KY273), Ningbo Health Branding Subject Fund (grant no. PPXK2018-01) and Ningbo 'Technology Innovation 2025' Major Special Project (grant no. 2022Z150).

References

1 

Wynn TA: Cellular and molecular mechanisms of fibrosis. J Pathol. 214:199–210. 2008. View Article : Google Scholar

2 

Mack M: Inflammation and fibrosis. Matrix Biol. 68-69:106–121. 2018. View Article : Google Scholar

3 

Tang PM, Nikolic-Paterson DJ and Lan HY: Macrophages: Versatile players in renal inflammation and fibrosis. Nat Rev Nephrol. 15:144–158. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Kleaveland KR, Moore BB and Kim KK: Paracrine functions of fibrocytes to promote lung fibrosis. Expert Rev Respir Med. 8:163–172. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Gregory RI, Chendrimada TP, Cooch N and Shiekhattar R: Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell. 123:631–640. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Huang Y, Shen XJ, Zou Q, Wang SP, Tang SM and Zhang GZ: Biological functions of microRNAs: A review. J Physiol Biochem. 67:129–139. 2011. View Article : Google Scholar

7 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Lu LF, Boldin MP, Chaudhry A, Lin LL, Taganov KD, Hanada T, Yoshimura A, Baltimore D and Rudensky AY: Function of miR-146a in controlling Treg cell-mediated regulation of Th1 responses. Cell. 142:914–929. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Wang S, Zhang X, Ju Y, Zhao B, Yan X, Hu J, Shi L, Yang L, Ma Z, Chen L, et al: MicroRNA-146a feedback suppresses T cell immune function by targeting Stat1 in patients with chronic hepatitis B. J Immunol. 191:293–301. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Boldin MP, Taganov KD, Rao DS, Yang L, Zhao JL, Kalwani M, Garcia-Flores Y, Luong M, Devrekanli A, Xu J, et al: miR-146a is a significant brake on autoimmunity, myeloproliferation, and cancer in mice. J Exp Med. 208:1189–1201. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Zhao JL, Rao DS, Boldin MP, Taganov KD, O'Connell RM and Baltimore D: NF-kappaB dysregulation in microRNA-146a-deficient mice drives the development of myeloid malignancies. Proc Natl Acad Sci USA. 108:9184–9189. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Ho BC, Yu IS, Lu LF, Rudensky A, Chen HY, Tsai CW, Chang YL, Wu CT, Chang LY, Shih SR, et al: Inhibition of miR-146a prevents enterovirus-induced death by restoring the production of type I interferon. Nat Commun. 5:33442014. View Article : Google Scholar : PubMed/NCBI

13 

Alexander M, Hu R, Runtsch MC, Kagele DA, Mosbruger TL, Tolmachova T, Seabra MC, Round JL, Ward DM and O'Connell RM: Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Nat Commun. 6:73212015. View Article : Google Scholar : PubMed/NCBI

14 

Paterson MR and Kriegel AJ: MiR-146a/b: A family with shared seeds and different roots. Physiol Genomics. 49:243–252. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Curtale G, Mirolo M, Renzi TA, Rossato M, Bazzoni F and Locati M: Negative regulation of Toll-like receptor 4 signaling by IL-10-dependent microRNA-146b. Proc Natl Acad Sci USA. 110:11499–11504. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Kutty RK, Nagineni CN, Samuel W, Vijayasarathy C, Jaworski C, Duncan T, Cameron JE, Flemington EK, Hooks JJ and Redmond TM: Differential regulation of microRNA-146a and microRNA-146b-5p in human retinal pigment epithelial cells by interleukin-1β, tumor necrosis factor-α, and interferon-γ. Mol Vis. 19:737–750. 2013.

17 

Liu R, Liu C, Chen D, Yang WH, Liu X, Liu CG, Dugas CM, Tang F, Zheng P, Liu Y and Wang L: FOXP3 controls an miR-146/NF-κB negative feedback loop that inhibits apoptosis in breast cancer cells. Cancer Res. 75:1703–1713. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Taganov KD, Boldin MP, Chang KJ and Baltimore D: NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA. 103:12481–12486. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Ni S, Yang B, Xia L and Zhang H: EZH2 mediates miR-146a-5p/HIF-1 α to alleviate inflammation and glycolysis after acute spinal cord injury. Mediators Inflamm. 2021:55915822021. View Article : Google Scholar

20 

Damodaran M, Paul SFD and Venkatesan V: Genetic polymorphisms in miR-146a, miR-196a2 and miR-125a genes and its association in prostate cancer. Pathol Oncol Res. 26:193–200. 2020. View Article : Google Scholar

21 

Chae YS, Kim JG, Lee SJ, Kang BW, Lee YJ, Park JY, Jeon HS, Park JS and Choi GS: A miR-146a polymorphism (rs2910164) predicts risk of and survival from colorectal cancer. Anticancer Res. 33:3233–3239. 2013.PubMed/NCBI

22 

Luo X, Yang W, Ye DQ, Cui H, Zhang Y, Hirankarn N, Qian X, Tang Y, Lau YL, de Vries N, et al: A functional variant in microRNA-146a promoter modulates its expression and confers disease risk for systemic lupus erythematosus. PLoS Genet. 7:e10021282011. View Article : Google Scholar : PubMed/NCBI

23 

Cui L, Tao H, Wang Y, Liu Z, Xu Z, Zhou H, Cai Y, Yao L, Chen B, Liang W, et al: A functional polymorphism of the microRNA-146a gene is associated with susceptibility to drug-resistant epilepsy and seizures frequency. Seizure. 27:60–65. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Kang JY and Lee JO: Structural biology of the Toll-like receptor family. Annu Rev Biochem. 80:917–941. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Rowe DC, McGettrick AF, Latz E, Monks BG, Gay NJ, Yamamoto M, Akira S, O'Neill LA, Fitzgerald KA and Golenbock DT: The myristoylation of TRIF-related adaptor molecule is essential for Toll-like receptor 4 signal transduction. Proc Natl Acad Sci USA. 103:6299–6304. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Tanimura N, Saitoh S, Matsumoto F, Akashi-Takamura S and Miyake K: Roles for LPS-dependent interaction and relocation of TLR4 and TRAM in TRIF-signaling. Biochem Biophys Res Commun. 368:94–99. 2008. View Article : Google Scholar : PubMed/NCBI

27 

De Nardo D: Toll-like receptors: Activation, signalling and transcriptional modulation. Cytokine. 74:181–189. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Hayden MS and Ghosh S: Shared principles in NF-kappaB signaling. Cell. 132:344–362. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Chen LF and Greene WC: Shaping the nuclear action of NF-kappaB. Nat Rev Mol Cell Biol. 5:392–401. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Hoffmann A, Natoli G and Ghosh G: Transcriptional regulation via the NF-kappaB signaling module. Oncogene. 25:6706–6716. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Karin M: Nuclear factor-kappaB in cancer development and progression. Nature. 441:431–436. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Luo JL, Kamata H and Karin M: IKK/NF-kappaB signaling: Balancing life and death-a new approach to cancer therapy. J Clin Invest. 115:2625–2632. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Hou J, Wang P, Lin L, Liu X, Ma F, An H, Wang Z and Cao X: MicroRNA-146a feedback inhibits RIG-I-dependent type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2. J Immunol. 183:2150–2158. 2006. View Article : Google Scholar

34 

He L, Wang Z, Zhou R, Xiong W, Yang Y, Song N and Qian J: Dexmedetomidine exerts cardioprotective effect through miR-146a-3p targeting IRAK1 and TRAF6 via inhibition of the NF-κB pathway. Biomed Pharmacother. 133:1109932021. View Article : Google Scholar

35 

Zhang Z, Zou X, Zhang R, Xie Y, Feng Z, Li F, Han J, Sun H, Ouyang Q, Hua S, et al: Human umbilical cord mesenchymal stem cell-derived exosomal miR-146a-5p reduces microglial-mediated neuroinflammation via suppression of the IRAK1/TRAF6 signaling pathway after ischemic stroke. Aging (Albany NY). 13:3060–3079. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Hou J, Deng Q, Deng X, Zhong W, Liu S and Zhong Z: MicroRNA-146a-5p alleviates lipopolysaccharide-induced NLRP3 inflammasome injury and pro-inflammatory cytokine production via the regulation of TRAF6 and IRAK1 in human umbilical vein endothelial cells (HUVECs). Ann Transl Med. 9:14332021. View Article : Google Scholar : PubMed/NCBI

37 

Li X, Liao J, Su X, Li W, Bi Z, Wang J, Su Q, Huang H, Wei Y, Gao Y, et al: Human urine-derived stem cells protect against renal ischemia/reperfusion injury in a rat model via exosomal miR-146a-5p which targets IRAK1. Theranostics. 10:9561–9578. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Wang XP, Luoreng ZM, Zan LS, Li F and Li N: Bovine miR-146a regulates inflammatory cytokines of bovine mammary epithelial cells via targeting the TRAF6 gene. J Dairy Sci. 100:7648–7658. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Zhang X, Guo Y, Xu X, Tang T, Sun L, Wang H, Zhou W, Fang L, Li Q and Xie P: miR-146a promotes Borna disease virus 1 replication through IRAK1/TRAF6/NF-κB signaling pathway. Virus Res. 271:1976712019. View Article : Google Scholar

40 

Iori V, Iyer AM, Ravizza T, Beltrame L, Paracchini L, Marchini S, Cerovic M, Hill C, Ferrari M, Zucchetti M, et al: Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy. Neurobiol Dis. 99:12–23. 2017. View Article : Google Scholar

41 

Quinn EM, Wang JH, O'Callaghan G and Redmond HP: MicroRNA-146a is upregulated by and negatively regulates TLR2 signaling. PLoS One. 8:e622322013. View Article : Google Scholar : PubMed/NCBI

42 

Chen Y, Wu Z, Yuan B, Dong Y, Zhang L and Zeng Z: MicroRNA-146a-5p attenuates irradiation-induced and LPS-induced hepatic stellate cell activation and hepatocyte apoptosis through inhibition of TLR4 pathway. Cell Death Dis. 9:222018. View Article : Google Scholar : PubMed/NCBI

43 

Yang L, Boldin MP, Yu Y, Liu CS, Ea CK, Ramakrishnan P, Taganov KD, Zhao JL and Baltimore D: miR-146a controls the resolution of T cell responses in mice. J Exp Med. 209:1655–1670. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Lochhead RB, Ma Y, Zachary JF, Baltimore D, Zhao JL, Weis JH, O'Connell RM and Weis JJ: MicroRNA-146a provides feedback regulation of lyme arthritis but not carditis during infection with Borrelia burgdorferi. PLoS Pathog. 10:e10042122014. View Article : Google Scholar : PubMed/NCBI

45 

Bhatt K, Lanting LL, Jia Y, Yadav S, Reddy MA, Magilnick N, Boldin M and Natarajan R: Anti-inflammatory role of MicroRNA-146a in the pathogenesis of diabetic nephropathy. J Am Soc Nephrol. 27:2277–2288. 2016. View Article : Google Scholar :

46 

Ammari M, Presumey J, Ponsolles C, Roussignol G, Roubert C, Escriou V, Toupet K, Mausset-Bonnefont AL, Cren M, Robin M, et al: Delivery of miR-146a to Ly6Chigh monocytes inhibits pathogenic bone erosion in inflammatory arthritis. Theranostics. 8:5972–5985. 2018. View Article : Google Scholar

47 

Hsu YR, Chang SW, Lin YC and Yang CH: MicroRNA-146a alleviates experimental autoimmune anterior uveitis in the eyes of lewis rats. Mediators Inflamm. 2017:96013492017. View Article : Google Scholar

48 

Lv F, Huang Y, Lv W, Yang L, Li F, Fan J and Sun J: MicroRNA-146a ameliorates inflammation via TRAF6/NF-κB pathway in intervertebral disc cells. Med Sci Monit. 23:659–664. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Bi X, Zhou L, Liu Y, Gu J and Mi QS: MicroRNA-146a deficiency delays wound healing in normal and diabetic mice. Adv Wound Care (New Rochelle). 11:19–27. 2022. View Article : Google Scholar

50 

Tang Y, Luo X, Cui H, Ni X, Yuan M, Guo Y, Huang X, Zhou H, de Vries N, Tak PP, et al: MicroRNA-146A contributes to abnormal activation of the type I interferon pathway in human lupus by targeting the key signaling proteins. Arthritis Rheum. 60:1065–1075. 2009. View Article : Google Scholar : PubMed/NCBI

51 

He X, Tang R, Sun Y, Wang YG, Zhen KY, Zhang DM and Pan WQ: MicroR-146 blocks the activation of M1 macrophage by targeting signal transducer and activator of transcription 1 in hepatic schistosomiasis. EBioMedicine. 13:339–347. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Chen Y, Wang JJ, Li J, Hosoya KI, Ratan R, Townes T and Zhang SX: Activating transcription factor 4 mediates hyperglycaemia-induced endothelial inflammation and retinal vascular leakage through activation of STAT3 in a mouse model of type 1 diabetes. Diabetologia. 55:2533–2545. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Shirai T, Nazarewicz RR, Wallis BB, Yanes RE, Watanabe R, Hilhorst M, Tian L, Harrison DG, Giacomini JC, Assimes TL, et al: The glycolytic enzyme PKM2 bridges metabolic and inflammatory dysfunction in coronary artery disease. J Exp Med. 213:337–354. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Elsaeidi F, Bemben MA, Zhao XF and Goldman D: Jak/Stat signaling stimulates zebrafish optic nerve regeneration and overcomes the inhibitory actions of Socs3 and Sfpq. J Neurosci. 34:2632–2644. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Fasler-Kan E, Barteneva NS, Ketterer S, Wunderlich K, Reschner A, Nurzhanova A, Flammer J, Huwyler J and Meyer P: Human cytokines activate JAK-STAT signaling pathway in porcine ocular tissue. Xenotransplantation. 20:469–480. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Samardzija M, Wenzel A, Aufenberg S, Thiersch M, Remé C and Grimm C: Differential role of Jak-STAT signaling in retinal degenerations. FASEB J. 20:2411–2413. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Ye EA and Steinle JJ: miR-146a suppresses STAT3/VEGF pathways and reduces apoptosis through IL-6 signaling in primary human retinal microvascular endothelial cells in high glucose conditions. Vision Res. 139:15–22. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Guo H, Zhang Y, Liao Z, Zhan W, Wang Y, Peng Y, Yang M, Ma X, Yin G and Ye L: MiR-146a upregulates FOXP3 and suppresses inflammation by targeting HIPK3/STAT3 in allergic conjunctivitis. Ann Transl Med. 10:3442022. View Article : Google Scholar : PubMed/NCBI

59 

Li T, Li M, Xu C, Xu X, Ding J, Cheng L and Ou R: miR-146a regulates the function of Th17 cell differentiation to modulate cervical cancer cell growth and apoptosis through NF-κB signaling by targeting TRAF6. Oncol Rep. 41:2897–2908. 2019.PubMed/NCBI

60 

Ferrer-Marín F, Arroyo AB, Bellosillo B, Cuenca EJ, Zamora L, Hernández-Rivas JM, Hernández-Boluda JC, Fernandez-Rodriguez C, Luño E, García Hernandez C, et al: miR-146a rs2431697 identifies myeloproliferative neoplasm patients with higher secondary myelofibrosis progression risk. Leukemia. 34:2648–2659. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Sun W, Ma J, Zhao H, Xiao C, Zhong H, Ling H, Xie Z, Tian Q, Chen H, Zhang T, et al: Resolvin D1 suppresses pannus formation via decreasing connective tissue growth factor caused by upregulation of miRNA-146a-5p in rheumatoid arthritis. Arthritis Res Ther. 22:612020. View Article : Google Scholar : PubMed/NCBI

62 

Dai X, Mao C, Lan X, Chen H, Li M, Bai J, Deng J, Liang Q, Zhang J, Zhong X, et al: Acute Penicillium marneffei infection stimulates host M1/M2a macrophages polarization in BALB/C mice. BMC Microbiol. 17:1772017. View Article : Google Scholar : PubMed/NCBI

63 

Khan J, Sharma PK and Mukhopadhaya A: Vibrio cholerae porin OmpU mediates M1-polarization of macrophages/monocytes via TLR1/TLR2 activation. Immunobiology. 220:1199–1209. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Vinuesa E, Hotter G, Jung M, Herrero-Fresneda I, Torras J and Sola A: Macrophage involvement in the kidney repair phase after ischaemia/reperfusion injury. J Pathol. 214:104–113. 2008. View Article : Google Scholar

65 

Huen SC and Cantley LG: Macrophage-mediated injury and repair after ischemic kidney injury. Pediatr Nephrol. 30:199–209. 2015. View Article : Google Scholar

66 

Lee S, Huen S, Nishio H, Nishio S, Lee HK, Choi BS, Ruhrberg C and Cantley LG: Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol. 22:317–326. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Alikhan MA, Jones CV, Williams TM, Beckhouse AG, Fletcher AL, Kett MM, Sakkal S, Samuel CS, Ramsay RG, Deane JA, et al: Colony-stimulating factor-1 promotes kidney growth and repair via alteration of macrophage responses. Am J Pathol. 179:1243–1256. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Peng X, He F, Mao Y, Lin Y, Fang J, Chen Y, Sun Z, Zhuo Y and Jiang J: miR-146a promotes M2 macrophage polarization and accelerates diabetic wound healing by inhibiting the TLR4/NF-κB axis. J Mol Endocrinol. 69:315–327. 2022. View Article : Google Scholar : PubMed/NCBI

69 

Liu XS, Fan B, Szalad A, Jia L, Wang L, Wang X, Pan W, Zhang L, Zhang R, Hu J, et al: MicroRNA-146a mimics reduce the peripheral neuropathy in type 2 diabetic mice. Diabetes. 66:3111–3121. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Huang C, Liu XJ, QunZhou, Xie J, Ma TT, Meng XM and Li J: MiR-146a modulates macrophage polarization by inhibiting Notch1 pathway in RAW264.7 macrophages. Int Immunopharmacol. 32:46–54. 2016. View Article : Google Scholar : PubMed/NCBI

71 

Jiang M, Xiang Y, Wang D, Gao J, Liu D, Liu Y, Liu S and Zheng D: Dysregulated expression of miR-146a contributes to age-related dysfunction of macrophages. Aging Cell. 11:29–40. 2012. View Article : Google Scholar

72 

Li Z, Wang S, Zhao W, Sun Z, Yan H and Zhu J: Oxidized low-density lipoprotein upregulates microRNA-146a via JNK and NF-κB signaling. Mol Med Rep. 13:1709–1716. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Yang Y, Huang G, Xu Q, Zhao G, Jiang J, Li Y and Guo Z: miR-146a-5p attenuates allergic airway inflammation by inhibiting the NLRP3 inflammasome activation in macrophages. Int Arch Allergy Immunol. 183:919–930. 2022. View Article : Google Scholar : PubMed/NCBI

74 

Chen X, Su C, Wei Q, Sun H, Xie J and Nong G: Exosomes derived from human umbilical cord mesenchymal stem cells alleviate diffuse alveolar hemorrhage associated with systemic lupus erythematosus in mice by promoting M2 macrophage polarization via the microRNA-146a-5p/NOTCH1 axis. Immunol Invest. 51:1975–1993. 2022. View Article : Google Scholar : PubMed/NCBI

75 

Luo S, Ding X, Zhao S, Mou T, Li R and Cao X: Long non-coding RNA CHRF accelerates LPS-induced acute lung injury through microRNA-146a/Notch1 axis. Ann Transl Med. 9:12992021. View Article : Google Scholar : PubMed/NCBI

76 

Ren W, Xi G, Li X, Zhao L, Yang K, Fan X, Gao L, Xu H and Guo J: Long non-coding RNA HCG18 promotes M1 macrophage polarization through regulating the miR-146a/TRAF6 axis, facilitating the progression of diabetic peripheral neuropathy. Mol Cell Biochem. 476:471–482. 2021. View Article : Google Scholar

77 

Cobb BS, Hertweck A, Smith J, O'Connor E, Graf D, Cook T, Smale ST, Sakaguchi S, Livesey FJ, Fisher AG and Merkenschlager M: A role for Dicer in immune regulation. J Exp Med. 203:2519–2527. 2006. View Article : Google Scholar : PubMed/NCBI

78 

Smigielska-Czepiel K, van den Berg A, Jellema P, van der Lei RJ, Bijzet J, Kluiver J, Boots AM, Brouwer E and Kroesen BJ: Comprehensive analysis of miRNA expression in T-cell subsets of rheumatoid arthritis patients reveals defined signatures of naive and memory Tregs. Genes Immun. 15:115–125. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Zhang Y, Yang Y, Guo J, Cui L, Yang L, Li Y, Mou Y, Jia C, Zhang L and Song X: miR-146a enhances regulatory T-cell differentiation and function in allergic rhinitis by targeting STAT5b. Allergy. 77:550–558. 2022. View Article : Google Scholar

80 

Li B, Wang X, Choi IY, Wang YC, Liu S, Pham AT, Moon H, Smith DJ, Rao DS, Boldin MP and Yang L: miR-146a modulates autoreactive Th17 cell differentiation and regulates organ-specific autoimmunity. J Clin Invest. 127:3702–3716. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Wang J, Yang L, Wang L, Yang Y and Wang Y: Forkhead box p3 controls progression of oral lichen planus by regulating microRNA-146a. J Cell Biochem. 119:8862–8871. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Wang J, Zhai X, Guo J, Li Y, Yang Y, Wang L, Yang L and Liu F: Long non-coding RNA DQ786243 modulates the induction and function of CD4+ Treg cells through Foxp3-miR-146a-NF-κB axis: Implications for alleviating oral lichen planus. Int Immunopharmacol. 75:1057612019. View Article : Google Scholar

83 

Schmidt SV, Nino-Castro AC and Schultze JL: Regulatory dendritic cells: There is more than just immune activation. Front Immunol. 3:2742012. View Article : Google Scholar : PubMed/NCBI

84 

Tang H, Lai Y, Zheng J, Chen K, Jiang H and Xu G: MiR-146a promotes tolerogenic properties of dendritic cells and through targeting notch1 signaling. Immunol Invest. 49:555–570. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Du J, Wang J, Tan G, Cai Z, Zhang L, Tang B and Wang Z: Aberrant elevated microRNA-146a in dendritic cells (DC) induced by human pancreatic cancer cell line BxPC-3-conditioned medium inhibits DC maturation and activation. Med Oncol. 29:2814–2823. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Stickel N, Hanke K, Marschner D, Prinz G, Köhler M, Melchinger W, Pfeifer D, Schmitt-Graeff A, Brummer T, Heine A, et al: MicroRNA-146a reduces MHC-II expression via targeting JAK/STAT signaling in dendritic cells after stem cell transplantation. Leukemia. 31:2732–2741. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Jurkin J, Schichl YM, Koeffel R, Bauer T, Richter S, Konradi S, Gesslbauer B and Strobl H: miR-146a is differentially expressed by myeloid dendritic cell subsets and desensitizes cells to TLR2-dependent activation. J Immunol. 184:4955–4965. 2010. View Article : Google Scholar : PubMed/NCBI

88 

Karrich JJ, Jachimowski LC, Libouban M, Iyer A, Brandwijk K, Taanman-Kueter EW, Nagasawa M, de Jong EC, Uittenbogaart CH and Blom B: MicroRNA-146a regulates survival and maturation of human plasmacytoid dendritic cells. Blood. 122:3001–3009. 2013. View Article : Google Scholar : PubMed/NCBI

89 

Park H, Huang X, Lu C, Cairo MS and Zhou X: MicroRNA-146a and microRNA-146b regulate human dendritic cell apoptosis and cytokine production by targeting TRAF6 and IRAK1 proteins. J Biol Chem. 290:2831–2841. 2015. View Article : Google Scholar :

90 

Xu D, Han Q, Hou Z, Zhang C and Zhang J: miR-146a negatively regulates NK cell functions via STAT1 signaling. Cell Mol Immunol. 14:712–720. 2017. View Article : Google Scholar :

91 

Pesce S, Squillario M, Greppi M, Loiacono F, Moretta L, Moretta A, Sivori S, Castagnola P, Barla A, Candiani S and Marcenaro E: New miRNA signature heralds human NK cell subsets at different maturation steps: Involvement of miR-146a-5p in the regulation of KIR expression. Front Immunol. 9:23602018. View Article : Google Scholar : PubMed/NCBI

92 

Wang H, Zhang Y, Wu X, Wang Y, Cui H, Li X, Zhang J, Tun N, Peng Y and Yu J: Regulation of human natural killer cell IFN-γ production by MicroRNA-146a via targeting the NF-κB signaling pathway. Front Immunol. 9:2932018. View Article : Google Scholar

93 

Friedman SL, Sheppard D, Duffield JS and Violette S: Therapy for fibrotic diseases: Nearing the starting line. Sci Transl Med. 5:167sr12013. View Article : Google Scholar : PubMed/NCBI

94 

Rosenbloom J, Mendoza FA and Jimenez SA: Strategies for anti-fibrotic therapies. Biochim Biophys Acta. 1832:1088–1103. 2013. View Article : Google Scholar

95 

Kalluri R and Neilson EG: Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 112:1776–1784. 2003. View Article : Google Scholar : PubMed/NCBI

96 

McAnulty RJ: Fibroblasts and myofibroblasts: Their source, function and role in disease. Int J Biochem Cell Biol. 39:666–671. 2007. View Article : Google Scholar : PubMed/NCBI

97 

Beyer C, Schett G, Gay S, Distler O and Distler JHW: Hypoxia. Hypoxia in the pathogenesis of systemic sclerosis. Arthritis Res Ther. 11:2202009. View Article : Google Scholar : PubMed/NCBI

98 

Lokmic Z, Musyoka J, Hewitson TD and Darby IA: Hypoxia and hypoxia signaling in tissue repair and fibrosis. Int Rev Cell Mol Biol. 296:139–185. 2012. View Article : Google Scholar : PubMed/NCBI

99 

Santos A and Lagares D: Matrix stiffness: The conductor of organ fibrosis. Curr Rheumatol Rep. 20:22018. View Article : Google Scholar : PubMed/NCBI

100 

Parker MW, Rossi D, Peterson M, Smith K, Sikström K, White ES, Connett JE, Henke CA, Larsson O and Bitterman PB: Fibrotic extracellular matrix activates a profibrotic positive feedback loop. J Clin Invest. 124:1622–1635. 2014. View Article : Google Scholar : PubMed/NCBI

101 

Watson CJ, Collier P, Tea I, Neary R, Watson JA, Robinson C, Phelan D, Ledwidge MT, McDonald KM, McCann A, et al: Hypoxia-induced epigenetic modifications are associated with cardiac tissue fibrosis and the development of a myofibroblast-like phenotype. Hum Mol Genet. 23:2176–2188. 2014. View Article : Google Scholar

102 

Kanzler S, Lohse AW, Keil A, Henninger J, Dienes HP, Schirmacher P, Rose-John S, zum Büschenfelde KH and Blessing M: TGF-beta1 in liver fibrosis: An inducible transgenic mouse model to study liver fibrogenesis. Am J Physiol. 276:G1059–G1068. 1999.

103 

Zhu H, Li Y, Qu S, Luo H, Zhou Y, Wang Y, Zhao H, You Y, Xiao X and Zuo X: MicroRNA expression abnormalities in limited cutaneous scleroderma and diffuse cutaneous scleroderma. J Clin Immunol. 32:514–522. 2012. View Article : Google Scholar : PubMed/NCBI

104 

Jia C, Xiong M, Wang P, Cui J, Du X, Yang Q, Wang W, Chen Y and Zhang T: Notoginsenoside R1 attenuates atherosclerotic lesions in ApoE deficient mouse model. PLoS One. 9:e998492014. View Article : Google Scholar : PubMed/NCBI

105 

Morishita Y, Imai T, Yoshizawa H, Watanabe M, Ishibashi K, Muto S and Nagata D: Delivery of microRNA-146a with polyethylenimine nanoparticles inhibits renal fibrosis in vivo. Int J Nanomedicine. 10:3475–3488. 2015. View Article : Google Scholar : PubMed/NCBI

106 

Liu Z, Lu CL, Cui LP, Hu YL, Yu Q, Jiang Y, Ma T, Jiao DK, Wang D and Jia CY: MicroRNA-146a modulates TGF-β1-induced phenotypic differentiation in human dermal fibroblasts by targeting SMAD4. Arch Dermatol Res. 304:195–202. 2012. View Article : Google Scholar

107 

Zou Y, Cai Y, Lu D, Zhou Y, Yao Q and Zhang S: MicroRNA-146a-5p attenuates liver fibrosis by suppressing profibrogenic effects of TGFβ1 and lipopolysaccharide. Cell Signal. 39:1–8. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Skhirtladze C, Distler O, Dees C, Akhmetshina A, Busch N, Venalis P, Zwerina J, Spriewald B, Pileckyte M, Schett G and Distler JH: Src kinases in systemic sclerosis: Central roles in fibroblast activation and in skin fibrosis. Arthritis Rheum. 58:1475–1484. 2008. View Article : Google Scholar : PubMed/NCBI

109 

Hu M, Che P, Han X, Cai GQ, Liu G, Antony V, Luckhardt T, Siegal GP, Zhou Y, Liu RM, et al: Therapeutic targeting of SRC kinase in myofibroblast differentiation and pulmonary fibrosis. J Pharmacol Exp Ther. 351:87–95. 2014. View Article : Google Scholar : PubMed/NCBI

110 

Yuan BY, Chen YH, Wu ZF, Zhuang Y, Chen GW, Zhang L, Zhang HG, Cheng JC, Lin Q and Zeng ZC: MicroRNA-146a-5p attenuates fibrosis-related molecules in irradiated and TGF-beta1-treated human hepatic stellate cells by regulating PTPRA-SRC signaling. Radiat Res. 192:621–629. 2019. View Article : Google Scholar : PubMed/NCBI

111 

Sun Y, Li Y, Wang H, Li H, Liu S, Chen J and Ying H: miR-146a-5p acts as a negative regulator of TGF-β signaling in skeletal muscle after acute contusion. Acta Biochim Biophys Sin (Shanghai). 49:628–634. 2017. View Article : Google Scholar : PubMed/NCBI

112 

Liu W, Ma C, Li HY, Chen L, Yuan SS and Li KJ: MicroRNA-146a downregulates the production of hyaluronic acid and collagen I in Graves' ophthalmopathy orbital fibroblasts. Exp Ther Med. 20:382020. View Article : Google Scholar : PubMed/NCBI

113 

Amrouche L, Desbuissons G, Rabant M, Sauvaget V, Nguyen C, Benon A, Barre P, Rabaté C, Lebreton X, Gallazzini M, et al: MicroRNA-146a in human and experimental ischemic AKI: CXCL8-dependent mechanism of action. J Am Soc Nephrol. 28:479–493. 2017. View Article : Google Scholar :

114 

Xiao Y, Qiao W, Wang X, Sun L and Ren W: MiR-146a mediates TLR-4 signaling pathway to affect myocardial fibrosis in rat constrictive pericarditis model. J Thorac Dis. 13:935–945. 2021. View Article : Google Scholar : PubMed/NCBI

115 

Yoshimura A, Wakabayashi Y and Mori T: Cellular and molecular basis for the regulation of inflammation by TGF-beta. J Biochem. 147:781–792. 2010. View Article : Google Scholar : PubMed/NCBI

116 

Sisto M, Lorusso L, Ingravallo G, Tamma R, Ribatti D and Lisi S: The TGF-β1 signaling pathway as an attractive target in the fibrosis pathogenesis of Sjögren's syndrome. Mediators Inflamm. 2018:19659352018. View Article : Google Scholar

117 

Biernacka A, Dobaczewski M and Frangogiannis NG: TGF-β signaling in fibrosis. Growth Factors. 29:196–202. 2011. View Article : Google Scholar : PubMed/NCBI

118 

Meng XM, Nikolic-Paterson DJ and Lan HY: TGF-β: The master regulator of fibrosis. Nat Rev Nephrol. 12:325–338. 2016. View Article : Google Scholar : PubMed/NCBI

119 

He Y, Huang C, Sun X, Long XR, Lv XW and Li J: MicroRNA-146a modulates TGF-beta1-induced hepatic stellate cell proliferation by targeting SMAD4. Cell Signal. 24:1923–1930. 2012. View Article : Google Scholar : PubMed/NCBI

120 

Wrighton KH, Lin X and Feng XH: Phospho-control of TGF-beta superfamily signaling. Cell Res. 19:8–20. 2009. View Article : Google Scholar

121 

Hill CS: Transcriptional control by the SMADs. Cold Spring Harb Perspect Biol. 8:a0220792016. View Article : Google Scholar : PubMed/NCBI

122 

Kaufhold S and Bonavida B: Central role of Snail1 in the regulation of EMT and resistance in cancer: A target for therapeutic intervention. J Exp Clin Cancer Res. 33:622014. View Article : Google Scholar : PubMed/NCBI

123 

Zhang Q, Cai R, Tang G, Zhang W and Pang W: MiR-146a-5p targeting SMAD4 and TRAF6 inhibits adipogenensis through TGF-β and AKT/mTORC1 signal pathways in porcine intra-muscular preadipocytes. J Anim Sci Biotechnol. 12:122021. View Article : Google Scholar

124 

Milano G, Biemmi V, Lazzarini E, Balbi C, Ciullo A, Bolis S, Ameri P, Di Silvestre D, Mauri P, Barile L and Vassalli G: Intravenous administration of cardiac progenitor cell-derived exosomes protects against doxorubicin/trastuzumab-induced cardiac toxicity. Cardiovasc Res. 116:383–392. 2020.

125 

Sisto M, Ribatti D and Lisi S: Organ fibrosis and autoimmunity: The role of inflammation in TGFβ-dependent EMT. Biomolecules. 11:3102021. View Article : Google Scholar

126 

Onichtchouk D, Chen YG, Dosch R, Gawantka V, Delius H, Massagué J and Niehrs C: Silencing of TGF-beta signalling by the pseudoreceptor BAMBI. Nature. 401:480–485. 1999. View Article : Google Scholar : PubMed/NCBI

127 

Yan X, Lin Z, Chen F, Zhao X, Chen H, Ning Y and Chen YG: Human BAMBI cooperates with Smad7 to inhibit transforming growth factor-beta signaling. J Biol Chem. 284:30097–30104. 2009. View Article : Google Scholar : PubMed/NCBI

128 

Jiang Y, Xiang C, Zhong F, Zhang Y, Wang L, Zhao Y, Wang J, Ding C, Jin L, He F and Wang H: Histone H3K27 methyltransferase EZH2 and demethylase JMJD3 regulate hepatic stellate cells activation and liver fibrosis. Theranostics. 11:361–378. 2021. View Article : Google Scholar : PubMed/NCBI

129 

Liu C, Chen X, Yang L, Kisseleva T, Brenner DA and Seki E: Transcriptional repression of the transforming growth factor β (TGF-β) pseudoreceptor BMP and activin membrane-bound inhibitor (BAMBI) by nuclear factor κB (NF-κB) p50 enhances TGF-β signaling in hepatic stellate cells. J Biol Chem. 289:7082–7091. 2014. View Article : Google Scholar : PubMed/NCBI

130 

Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA and Schwabe RF: TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med. 13:1324–1332. 2007. View Article : Google Scholar : PubMed/NCBI

131 

Wiest R, Lawson M and Geuking M: Pathological bacterial translocation in liver cirrhosis. J Hepatol. 60:197–209. 2014. View Article : Google Scholar

132 

Pradere JP, Troeger JS, Dapito DH, Mencin AA and Schwabe RF: Toll-like receptor 4 and hepatic fibrogenesis. Semin Liver Dis. 30:232–244. 2010. View Article : Google Scholar : PubMed/NCBI

133 

Akira S and Takeda K: Toll-like receptor signalling. Nat Rev Immunol. 4:499–511. 2004. View Article : Google Scholar : PubMed/NCBI

134 

Maubach G, Lim MCC, Chen J, Yang H and Zhuo L: miRNA studies in in vitro and in vivo activated hepatic stellate cells. World J Gastroenterol. 17:2748–2773. 2011. View Article : Google Scholar : PubMed/NCBI

135 

Chen Y, Zeng Z, Shen X, Wu Z, Dong Y and Cheng JC: MicroRNA-146a-5p negatively regulates pro-inflammatory cytokine secretion and cell activation in lipopolysaccharide stimulated human hepatic stellate cells through inhibition of Toll-like receptor 4 signaling pathways. Int J Mol Sci. 17:10762016. View Article : Google Scholar : PubMed/NCBI

136 

Xiao L, Gu Y, Ren G, Chen L, Liu L, Wang X and Gao L: miRNA-146a mimic inhibits NOX4/P38 signalling to ameliorate mouse myocardial ischaemia reperfusion (I/R) injury. Oxid Med Cell Longev. 2021:63662542021. View Article : Google Scholar : PubMed/NCBI

137 

Li J, Jiang ZZ, Li YY, Tang WT, Yin J and Long XP: LncRNA CHRF promotes TGF-β1 induced EMT in alveolar epithelial cells by inhibiting miR-146a up-regulating L1CAM expression. Exp Lung Res. 47:198–209. 2021. View Article : Google Scholar : PubMed/NCBI

138 

Feng B, Chen S, Gordon AD and Chakrabarti S: miR-146a mediates inflammatory changes and fibrosis in the heart in diabetes. J Mol Cell Cardiol. 105:70–76. 2017. View Article : Google Scholar : PubMed/NCBI

139 

Chen Y, Yuan B, Chen G, Zhang L, Zhuang Y, Niu H and Zeng Z: Circular RNA RSF1 promotes inflammatory and fibrotic phenotypes of irradiated hepatic stellate cell by modulating miR-146a-5p. J Cell Physiol. 235:8270–8282. 2020. View Article : Google Scholar : PubMed/NCBI

140 

Du J, Niu X, Wang Y, Kong L, Wang R, Zhang Y, Zhao S and Nan Y: MiR-146a-5p suppresses activation and proliferation of hepatic stellate cells in nonalcoholic fibrosing steatohepatitis through directly targeting Wnt1 and Wnt5a. Sci Rep. 5:161632015. View Article : Google Scholar : PubMed/NCBI

141 

Zhang H, Wen H and Huang Y: MicroRNA-146a attenuates isoproterenol-induced cardiac fibrosis by inhibiting FGF2. Exp Ther Med. 24:5062022. View Article : Google Scholar : PubMed/NCBI

142 

Editorial Office: Erratum to MiR-146a mediates TLR-4 signaling pathway to affect myocardial fibrosis in rat constrictive pericarditis model. J Thorac Dis. 13:4623–4624. 2021. View Article : Google Scholar : PubMed/NCBI

143 

Ma C, Qi X, Wei YF, Li Z, Zhang HL, Li H, Yu FL, Pu YN, Huang YC and Ren YX: Amelioration of ligamentum flavum hypertrophy using umbilical cord mesenchymal stromal cell-derived extracellular vesicles. Bioact Mater. 19:139–154. 2022. View Article : Google Scholar : PubMed/NCBI

144 

Saferding V, Puchner A, Goncalves-Alves E, Hofmann M, Bonelli M, Brunner JS, Sahin E, Niederreiter B, Hayer S, Kiener HP, et al: MicroRNA-146a governs fibroblast activation and joint pathology in arthritis. J Autoimmun. 82:74–84. 2017. View Article : Google Scholar : PubMed/NCBI

145 

Jang SY, Park SJ, Chae MK, Lee JH, Lee EJ and Yoon JS: Role of microRNA-146a in regulation of fibrosis in orbital fibroblasts from patients with Graves' orbitopathy. Br J Ophthalmol. 102:407–414. 2018. View Article : Google Scholar

146 

Acharya PS, Majumdar S, Jacob M, Hayden J, Mrass P, Weninger W, Assoian RK and Puré E: Fibroblast migration is mediated by CD44-dependent TGF beta activation. J Cell Sci. 121:1393–1402. 2008. View Article : Google Scholar : PubMed/NCBI

147 

Clark RA, McCoy GA, Folkvord JM and McPherson JM: TGF-beta 1 stimulates cultured human fibroblasts to proliferate and produce tissue-like fibroplasia: A fibronectin matrix-dependent event. J Cell Physiol. 170:69–80. 1997. View Article : Google Scholar : PubMed/NCBI

148 

Saha B, Kodys K and Szabo G: Hepatitis C virus-induced monocyte differentiation into polarized M2 macrophages promotes stellate cell activation via TGF-β. Cell Mol Gastroenterol Hepatol. 2:302–316.e8. 2016. View Article : Google Scholar

149 

Tang PM, Zhou S, Li CJ, Liao J, Xiao J, Wang QM, Lian GY, Li J, Huang XR, To KF, et al: The proto-oncogene tyrosine protein kinase Src is essential for macrophage-myofibroblast transition during renal scarring. Kidney Int. 93:173–187. 2018. View Article : Google Scholar

150 

Long H, Wang X, Chen Y, Wang L, Zhao M and Lu Q: Dysregulation of microRNAs in autoimmune diseases: Pathogenesis, biomarkers and potential therapeutic targets. Cancer Lett. 428:90–103. 2018. View Article : Google Scholar : PubMed/NCBI

151 

Shumnalieva R, Kachakova D, Shoumnalieva-Ivanova V, Miteva P, Kaneva R and Monov S: Whole peripheral blood miR-146a and miR-155 expression levels in systemic lupus erythematosus patients. Acta Reumatol Port. 43:217–225. 2018.PubMed/NCBI

152 

Zhu Y, Xue Z and Di L: Regulation of MiR-146a and TRAF6 in the diagnose of lupus nephritis. Med Sci Monit. 23:2550–2557. 2017. View Article : Google Scholar : PubMed/NCBI

153 

Abou-Zeid A, Saad M and Soliman E: MicroRNA 146a expression in rheumatoid arthritis: Association with tumor necrosis factor-alpha and disease activity. Genet Test Mol Biomarkers. 15:807–812. 2011. View Article : Google Scholar : PubMed/NCBI

154 

Li N, Wang J, Yu W, Dong K, You F, Si B, Tang B, Zhang Y, Wang T and Qiao B: MicroRNA-146a inhibits the inflammatory responses induced by interleukin-17A during the infection of Helicobacter pylori. Mol Med Rep. 19:1388–1395. 2019.

155 

Li LJ, Gu YJ, Wang LQ, Wan W, Wang HW, Yang XN, Ma LL, Yang LH and Meng ZH: Serum exosomal microRNA-146a as a novel diagnostic biomarker for acute coronary syndrome. J Thorac Dis. 13:3105–3114. 2021. View Article : Google Scholar : PubMed/NCBI

156 

Yang K, He YS, Wang XQ, Lu L, Chen QJ, Liu J, Sun Z and Shen WF: MiR-146a inhibits oxidized low-density lipoprotein-induced lipid accumulation and inflammatory response via targeting toll-like receptor 4. FEBS Lett. 585:854–860. 2011. View Article : Google Scholar : PubMed/NCBI

157 

Wu W, Xuan Y, Ge Y, Mu S, Hu C and Fan R: Plasma miR-146a and miR-365 expression and inflammatory factors in patients with osteoarthritis. Malays J Pathol. 43:311–317. 2021.PubMed/NCBI

158 

Ghotloo S, Motedayyen H, Amani D, Saffari M and Sattari M: Assessment of microRNA-146a in generalized aggressive periodontitis and its association with disease severity. J Periodontal Res. 54:27–32. 2019. View Article : Google Scholar

159 

Sabbatinelli J, Giuliani A, Matacchione G, Latini S, Laprovitera N, Pomponio G, Ferrarini A, Svegliati Baroni S, Pavani M, Moretti M, et al: Decreased serum levels of the inflammaging marker miR-146a are associated with clinical non-response to tocilizumab in COVID-19 patients. Mech Ageing Dev. 193:1114132021. View Article : Google Scholar

160 

Cai P, Mu Y, Olveda RM, Ross AG, Olveda DU and McManus DP: Serum exosomal miRNAs for grading hepatic fibrosis due to schistosomiasis. Int J Mol Sci. 21:35602020. View Article : Google Scholar : PubMed/NCBI

161 

Cai P, Mu Y, Olveda RM, Ross AG, Olveda DU and McManus DP: Circulating miRNAs as footprints for liver fibrosis grading in schistosomiasis. EBioMedicine. 37:334–343. 2018. View Article : Google Scholar : PubMed/NCBI

162 

Chen J, Chen T, Zhou J, Zhao X, Sheng Q and Lv Z: MiR-146a-5p mimic inhibits NLRP3 inflammasome downstream inflammatory factors and CLIC4 in neonatal necrotizing enterocolitis. Front Cell Dev Biol. 8:5941432021. View Article : Google Scholar : PubMed/NCBI

163 

Wang Y, Zhang S and Benoit DSW: Degradable poly(ethylene glycol) (PEG)-based hydrogels for spatiotemporal control of siRNA/nanoparticle delivery. J Control Release. 287:58–66. 2018. View Article : Google Scholar : PubMed/NCBI

164 

Niemiec SM, Hilton SA, Wallbank A, Azeltine M, Louiselle AE, Elajaili H, Allawzi A, Xu J, Mattson C, Dewberry LC, et al: Cerium oxide nanoparticle delivery of microRNA-146a for local treatment of acute lung injury. Nanomedicine. 34:1023882021. View Article : Google Scholar : PubMed/NCBI

165 

Chen B, Yoo K, Xu W, Pan R, Han XX and Chen P: Characterization and evaluation of a peptide-based siRNA delivery system in vitro. Drug Deliv Transl Res. 7:507–515. 2017. View Article : Google Scholar : PubMed/NCBI

166 

Su Y, Sun B, Gao X, Liu S, Hao R and Han B: Chitosan hydrogel doped with PEG-PLA nanoparticles for the local delivery of miRNA-146a to treat allergic rhinitis. Pharmaceutics. 12:9072020. View Article : Google Scholar : PubMed/NCBI

167 

Chiabotto G, Ceccotti E, Tapparo M, Camussi G and Bruno S: Human liver stem cell-derived extracellular vesicles target hepatic stellate cells and attenuate their pro-fibrotic phenotype. Front Cell Dev Biol. 9:7774622021. View Article : Google Scholar : PubMed/NCBI

168 

Liang YC, Wu YP, Li XD, Chen SH, Ye XJ, Xue XY and Xu N: TNF-α-induced exosomal miR-146a mediates mesenchymal stem cell-dependent suppression of urethral stricture. J Cell Physiol. 234:23243–23255. 2019. View Article : Google Scholar : PubMed/NCBI

169 

Shafei S, Khanmohammadi M, Ghanbari H, Nooshabadi VT, Tafti SHA, Rabbani S, Kasaiyan M, Basiri M and Tavoosidana G: Effectiveness of exosome mediated miR-126 and miR-146a delivery on cardiac tissue regeneration. Cell Tissue Res. 390:71–92. 2022. View Article : Google Scholar : PubMed/NCBI

170 

Carreras-Badosa G, Maslovskaja J, Periyasamy K, Urgard E, Padari K, Vaher H, Tserel L, Gestin M, Kisand K, Arukuusk P, et al: NickFect type of cell-penetrating peptides present enhanced efficiency for microRNA-146a delivery into dendritic cells and during skin inflammation. Biomaterials. 262:1203162020. View Article : Google Scholar : PubMed/NCBI

171 

Wang WX, Prajapati P, Vekaria HJ, Spry M, Cloud AL, Sullivan PG and Springer JE: Temporal changes in inflammatory mitochondria-enriched microRNAs following traumatic brain injury and effects of miR-146a nanoparticle delivery. Neural Regen Res. 16:514–522. 2021. View Article : Google Scholar :

172 

Bobba CM, Fei Q, Shukla V, Lee H, Patel P, Putman RK, Spitzer C, Tsai M, Wewers MD, Lee RJ, et al: Nanoparticle delivery of microRNA-146a regulates mechanotransduction in lung macrophages and mitigates injury during mechanical ventilation. Nat Commun. 12:2892021. View Article : Google Scholar : PubMed/NCBI

173 

Fouad MR, Salama RM, Zaki HF and El-Sahar AE: Vildagliptin attenuates acetic acid-induced colitis in rats via targeting PI3K/Akt/NFκB, Nrf2 and CREB signaling pathways and the expression of lncRNA IFNG-AS1 and miR-146a. Int Immunopharmacol. 92:1073542021. View Article : Google Scholar

174 

Pan Y, Wang J, Xue Y, Zhao J, Li D, Zhang S, Li K, Hou Y and Fan H: GSKJ4 protects mice against early sepsis via reducing proinflammatory factors and up-regulating MiR-146a. Front Immunol. 9:22722018. View Article : Google Scholar : PubMed/NCBI

175 

Sun W, Ma M and Yu H and Yu H: Inhibition of lncRNA X inactivate-specific transcript ameliorates inflammatory pain by suppressing satellite glial cell activation and inflammation by acting as a sponge of miR-146a to inhibit Nav 1.7. J Cell Biochem. 119:9888–9898. 2018. View Article : Google Scholar : PubMed/NCBI

176 

Zhou Z, Zhu Y, Gao G and Zhang Y: Long noncoding RNA SNHG16 targets miR-146a-5p/CCL5 to regulate LPS-induced WI-38 cell apoptosis and inflammation in acute pneumonia. Life Sci. 228:189–197. 2019. View Article : Google Scholar : PubMed/NCBI

177 

Dai L, Zhang G, Cheng Z, Wang X, Jia L, Jing X, Wang H, Zhang R, Liu M, Jiang T, et al: Knockdown of LncRNA MALAT1 contributes to the suppression of inflammatory responses by up-regulating miR-146a in LPS-induced acute lung injury. Connect Tissue Res. 59:581–592. 2018. View Article : Google Scholar : PubMed/NCBI

178 

Zhu D, Hu B, Zhou Y, Sun X, Chen J, Chen L, Ji Z, Zhu J and Duan Y: microRNA-146a is involved in rSjP40-inhibited activation of LX-2 cells by targeting Smad4 expression. J Cell Biochem. 119:9249–9253. 2018. View Article : Google Scholar : PubMed/NCBI

179 

Madhavan D, Cuk K, Burwinkel B and Yang R: Cancer diagnosis and prognosis decoded by blood-based circulating microRNA signatures. Front Genet. 4:1162013. View Article : Google Scholar : PubMed/NCBI

180 

Li Y, Zhang S, Zhang C and Wang M: LncRNA MEG3 inhibits the inflammatory response of ankylosing spondylitis by targeting miR-146a. Mol Cell Biochem. 466:17–24. 2020. View Article : Google Scholar : PubMed/NCBI

181 

Stenvang J, Petri A, Lindow M, Obad S and Kauppinen S: Inhibition of microRNA function by antimiR oligonucleotides. Silence. 3:12012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2023
Volume 51 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liao Z, Zheng R and Shao G: Mechanisms and application strategies of miRNA‑146a regulating inflammation and fibrosis at molecular and cellular levels (Review). Int J Mol Med 51: 7, 2023
APA
Liao, Z., Zheng, R., & Shao, G. (2023). Mechanisms and application strategies of miRNA‑146a regulating inflammation and fibrosis at molecular and cellular levels (Review). International Journal of Molecular Medicine, 51, 7. https://doi.org/10.3892/ijmm.2022.5210
MLA
Liao, Z., Zheng, R., Shao, G."Mechanisms and application strategies of miRNA‑146a regulating inflammation and fibrosis at molecular and cellular levels (Review)". International Journal of Molecular Medicine 51.1 (2023): 7.
Chicago
Liao, Z., Zheng, R., Shao, G."Mechanisms and application strategies of miRNA‑146a regulating inflammation and fibrosis at molecular and cellular levels (Review)". International Journal of Molecular Medicine 51, no. 1 (2023): 7. https://doi.org/10.3892/ijmm.2022.5210