Open Access

Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene

  • Authors:
    • Roberto Gambari
    • Chiara Papi
    • Jessica Gasparello
    • Enzo Agostinelli
    • Alessia Finotti
  • View Affiliations

  • Published online on: February 25, 2025     https://doi.org/10.3892/etm.2025.12835
  • Article Number: 85
  • Copyright: © Gambari 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

The coronavirus disease‑19 (COVID‑19) pandemic has been a very significant health issue in the period between 2020 and 2023, forcing research to characterize severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) sequences and to develop novel therapeutic approaches. Interleukin‑6 (IL‑6) and IL‑8 are considered significant therapeutic targets for COVID‑19 and emerging evidence has suggested that microRNAs (miRNAs/miRs) serve a key role in regulating these genes. MiRNAs are short, 19‑25 nucleotides in length, non‑coding RNAs that regulate gene expression at the post‑transcriptional level through the sequence‑selective recognition of the 3'‑untranslated region (3'‑UTR) of the regulated mRNAs, eventually repressing translation, commonly, via mRNA degradation. For example, among several miRNAs involved in the regulation of the COVID‑19 ‘cytokine storm’, miR‑93‑5p can inhibit IL‑8 gene expression by directly targeting the 3'‑UTR of IL‑8 mRNA. In addition, miR‑93‑5p can regulate Toll‑like receptor‑4 (TLR4) and interleukin‑1 receptor‑associated kinase 4 (IRAK4) expression, thus affecting the nuclear factor‑κB (NF‑κB) pathway and the expression of NF‑κB‑regulated genes, such as IL‑6, IL‑1β and other hyper‑expressed genes during the COVID‑19 ‘cytokine storm’. In the present study, the results provided preliminary evidence suggesting that the miR‑93‑5p‑based miRNA therapeutics could be combined with the anti‑inflammatory aged garlic extract (AGE) to more effectively inhibit IL‑8 gene expression. The human bronchial epithelial IB3‑1 cell line was employed as experimental model system. IB3‑1 cells were stimulated with the BNT162b2 COVID‑19 vaccine and transfected with pre‑miR‑93‑5p in the absence or in the presence of AGE, to verify the inhibitory effects on the BNT162b2‑induced expression of the IL‑8 gene. The accumulation of IL‑8 mRNA was assessed by RT‑qPCR; the release of IL‑8 protein was determined by Bio‑Plex assay. In addition, the possible applications of TLR4/NF‑κB inhibitory agents (such as miR‑93‑5p and AGE) for treating human pathologies at a hyperinflammatory state, such as COVID‑19, cystic fibrosis and other respiratory diseases, were summarized.

Introduction

The coronavirus disease-19 (COVID-19) pandemic has been considered as very important health issue between 2020 and 2023. Therefore, several studies have focused on characterizing the sequences of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and developing novel therapeutic options (1-3). Despite the development, validation, approval and worldwide use of anti-SARS-CoV-2 vaccines (4,5), specific anti-viral agents targeting the life cycle of SARS-CoV-2 and improving COVID-19-related clinical manifestations are still needed (6-9). In this respect, the majority of patients with COVID-19 show moderate symptoms; however, a consistent number of patients develop severe COVID-19, which is generally characterized by a hyperinflammatory state (10-13). Additionally, inflammation is considered as a significant feature of ‘long COVID-19’ (14) and post-acute sequelae of SARS-CoV-2 infection (15).

It has been clearly shown that following lung infection, SARS-CoV-2 promotes immune responses, which are characterized by the high production of inflammatory cytokines. This event, also known as ‘cytokine storm’ (16,17), can occur due to the activation of several transcription factors, such as nuclear factor kappa-B (NF-κB) and signal transducer and activator of transcription 3 (STAT-3), which in turn regulate the expression of numerous inflammation-related genes, including vascular endothelial growth factor (VEGF), monocyte chemoattractant protein 1 (MCP-1), interleukin-8 (IL-8) and IL-6 (18-21). Among the proteins which have been proposed to play a key role in the ‘cytokine storm’ phenomenon in patients with COVID-19, IL-1α, IL-1β, IL-2, IL-6, IL-7, IL-8, IL-9, IL-10, basic fibroblast growth factor, granulocyte-macrophage colony-stimulating factor (GM-CSF), G-CSF, platelet-derived growth factor, VEGF, interferon gamma, interferon-inducible protein 10, MCP-1, macrophage inflammatory protein 1-alpha (MIP-1-α), MIP-1-β and tumor necrosis factors (TNFs) are the most studied ones (22).

Examples of clinical trials on IL-6- and IL-8-targeting drugs for the treatment of patients with COVID-19 are summarized in Table I. These trials supported the concept that this approach could be considered for the development of possible therapeutic strategies against COVID-19. The above speculation has been also sustained by several published reports, such as those by Comarmond et al (23), Ghosn et al (24), Pomponio et al (25) and Castelnovo et al (26). Therefore, Comarmond et al (23) reported that patients suffering from inflammatory rheumatic and musculoskeletal diseases and treated with anti-IL6 therapy based on tocilizumab or sarilumab, displayed a less severe form of COVID-19. Although the association between COVID-19 outcomes and the duration of pre-existing anti-IL6 therapy was not analyzed in depth, the results of the study suggested that the anti-IL6 therapy was associated with less frequent hospitalization and need for respiratory support (23). Additionally, the pilot study by Pomponio et al (25) indicated that the efficacy of tocilizumab, an anti-IL6 therapy, could improve the respiratory function of patients. However, this result warrants further investigation in randomized trials. Castelnovo et al (26) collected data from patients with COVID-19 treated with the anti-IL6 drugs, tocilizumab and sarilumab. The analysis revealed that the anti-IL6 drugs could be effective in treating the severe forms of COVID-19. Therefore, it was hypothesized that these drugs could reduce inflammation and the risk of multi-organ failure-related mortality (26). Furthermore, a meta-analysis by Tharmarajah et al (27) analyzed the outcomes of IL-6 inhibition in the treatment of COVID-19. In general, the aforementioned studies on anti-IL6 therapies suggested that despite anti-IL6 monotherapy could display several benefits, this approach could be of great interest if combined with other anti-inflammatory drugs. Previous large clinical trials demonstrated that when tocilizumab was used in combination with dexamethasone to treat severe COVID-19-associated Acute Respiratory Distress Syndrome, provided significant clinical improvement. In this respect, Segú-Vergés et al (28) employed artificial intelligence approaches to evaluate the potential of tocilizumab in combination with corticosteroid therapy in treating COVID-19. This in silico study suggested that the administration of tocilizumab and dexamethasone could induce a synergistic effect, thus strongly reducing inflammation and associated pathological processes, eventually supporting the beneficial effects of the combined therapy in patients with COVID-19 in critical clinical conditions.

Table I

Examples of clinical trials for COVID-19 based on IL-6 and IL-8 targeting.

Table I

Examples of clinical trials for COVID-19 based on IL-6 and IL-8 targeting.

Clinical trial numberTitleBioactive molecule
NCT04381052Study for the Use of the IL-6 Inhibitor Clazakizumab in Patients With Life-threatening COVID-19 InfectionClazakizumab
NCT04343989A Randomized Placebo-controlled Safety and Dose- finding Study for the Use of the IL-6 Inhibitor Clazakizumab in Patients With Life-threatening COVID-19 InfectionClazakizumab
NCT04322773Anti-IL6 Treatment of Serious COVID-19 Disease with Threatening Respiratory FailureTwo human monoclonal antibodies against IL-6 receptor, tocilizumab and sarilumab
NCT04486521Clinical Outcome of Anti-IL6 vs. Anti-IL6 Corticosteroid Combination in Patients With SARS-CoV-2 Cytokine Release SyndromeCortocosteroid combinations
NCT04347226Anti-Interleukin-8 for patients with COVID-19BMS-986253
NCT04878055Study on Efficacy and Safety of Reparixin in the Treatment of Hospitalized Patients With Severe COVID-19 PneumoniaReparixin

[i] COVID-19, Coronavirus disease 2019; IL, interleukin; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.

Currently, less studies have been published regarding the effect of anti-IL-8 therapies against COVID-19. Two IL-8 inhibitors are being currently evaluated for the treatment of COVID-19. HuMax-IL-8 (BMS986253), a human monoclonal antibody targeting IL-8, was studied for the first time on several types of cancer (29). Reparixin, an allosteric inhibitor of IL-8 activity, has been investigated for its safety and efficacy in patients with severe COVID-19 pneumonia (30). Promising results were obtained in a phase II trial (REPAVID-19). Reparixin was also found to effectively improve biochemical and biophysical parameters in a murine model of liposaccharide (LPS)-induced acute lung injury (31). A murine model of COVID-19 was also employed and the results demonstrated a clear clinical improvement following IL-8-like signaling inhibition with reparixin (32). Taken together, these studies supported that IL-8 could be considered as a promising therapeutic target in the treatment of severe COVID-19 and therefore more anti-IL-8 therapies should be developed in basic and applied research.

MicroRNAs (miRNAs/miRs) are short, non-coding RNAs, 19-25 nucleotides in length, which are involved in regulating gene expression at the post-transcriptional level. Commonly, miRNAs act via specifically binding to the 3'-untraslated region (3'-UTR) of their target mRNAs, thus promoting mRNA degradation (33-37). Importantly, a single miRNA can target several mRNAs, while the 3'-UTR of a single mRNA can encompass several sequences recognized by different miRNAs. It has been reported that >60% of mammalian mRNAs can be targeted by miRNAs (37). A previous study supported that the release of key proteins of the COVID-19-related ‘cytokine storm’ could be strongly inhibited via mimicking the biological activity of miRNAs (38). In the above study, it was hypothesized that among possible anti-inflammatory agents, ‘miRNA targeting’ should be carefully considered as a significant strategy (38). Therefore, the general working hypothesis was that targeting the miRNA network could serve a significant role for the development of therapeutic approaches against the SARS-CoV-2-induced pro-inflammatory responses. This hypothesis was based on several publications, showing that miRNA overexpression could inhibit the expression of ‘cytokine storm’-related proteins in COVID-19(38). The above study demonstrated that a molecule mimicking miR-93-5p could modulate the expression of IL-8 gene via targeting its RNA transcript. The first evidence of the effect of miR-93-5p on regulating IL-8 gene expression was reported by Fabbri et al (39). This study demonstrated that miR-93-5p could bind to the IL-8 3'-UTR. In addition, it was found that the binding sites of miR-93-5p on IL-8 3'-UTR were conserved within different species, thus supporting its molecular evolution (39). Accordingly, Gasparello et al (40) showed that the treatment of bronchial epithelial IB3-1 cells with the SARS-CoV-2 spike protein enhanced the secretion of IL-8. In addition, IL-8 synthesis and extracellular release could be strongly reduced by a molecule mimicking miR-93-5p. The effect of miR-93-5p on directly affecting IL-8 expression is presented in Fig. 1. This figure illustrates the possible interaction between miR-93-5p and the 3'-UTR of IL-8 RNA, and the expected (experimentally validated) effects of miR-93-5p on IB3-1 cells (39,40). This study also revealed that other genes coding for proteins involved in the COVID-19 ‘cytokine storm’ are regulated by miR-93-5p, including IL-6, G-CSF, GM-CSF and IL-1β (40). More particularly, the results of the above study demonstrated that the expression of these genes was upregulated and downregulated following cell transfection with anti-miR-93-5p and pre-miR-93-5p, respectively, thus highlighting the key role of miR-93-5p in regulating the expression of these genes (40).

Fabbri et al (39) showed that the inhibitory effect of ago-miRNAs on the expression of pro-inflammatory genes was mediated by the interaction of these ago-miRNAs with the 3'-UTR of the target pro-inflammatory mRNAs. This finding was also verified in the miR-93-5p/IL-8 axis. In addition, the ago-miRNA molecule could interact and down-regulate the expression of other target pro-inflammatory genes. Furthermore, all IL-1α, IL-6 and IL-8 genes are upregulated by NF-κB. Eventually, all ago-miRNAs mimicking the biological activity of miRNAs on downregulating NF-κB, such as that of miR-130a, miR-146a/b, miR-218 and miR-451, could also inhibit the expression levels of NF-κB and other NF-κB-regulated genes (41). The focus of applied research on miRNA therapeutics has been discussed in several review articles, describing the results of both pre-clinical and clinical studies (42-45). However, a general and clear consensus on the potential biomedical applications of this approach is still missing (44,45). In addition, clinical studies using both PROOFS-FIG2 anti-miRNA (46) and miRNA mimicking (47) molecules are ongoing.

The experimental system, which could be employed to study the effects of potential inhibitors on the expression of pro-inflammatory genes is described in Fig. 2. According to the above experimental system, the upregulation of pro-inflammatory genes could be achieved following treatment of target cell lines, and particularly bronchial epithelial IB3-1 cells, with the spike RNA-based BNT162b2 vaccine. The approach was the same with that described in other experimental model systems, which aimed to uncover the effects of the BNT162b2 vaccine on the gene expression and functions of eukaryotic cells (48,49). The most significant characteristics of this approach are the following: i) The cellular uptake of spike mRNA could upregulate and promote the release of S-protein; and ii) the resulted NF-κB upregulation, could be also associated with the increased expression of other NF-κB-regulated genes, including those coding proteins associated with ‘cytokine storm’ and those involved in the hyperinflammation state of other human diseases, such as cystic fibrosis. Regarding the roles of the BNT162b2 vaccine in the expression of NF-κB and NF-κB-dependent genes, it should be noted that according to the study by Gasparello et al (50) on the effects of purified SARS-CoV-2 S-protein on IB3-1 cells, these should be expected. In this respect, BNT162b2 vaccine is composed of a liposomal vaccine vector which is used to carry the spike mRNA. Since lipid-based nanoparticles are known to stimulate inflammatory responses through endosomal Toll-like receptor (TLR)-7 and TLR-8 pathways (51-53), it was suggested that both lipid formulation and spike mRNA are involved in the activity of BNT162b2 on regulating the expression of pro-inflammatory genes. In any case, in the experiments based on the use of the BNT162b2 vaccine on IB3-1 cells, the effects of the anti-inflammatory compounds could be studied. Interestingly, in the majority of cases, the inducing effects of BNT162b2 on IB3-1 cells were more potent compared with those of other control stimuli, such as SARS-CoV-2 spike (40,50), Pseudomonas aeruginosa infection (39), TNF-a (54,55) and LPS (56). Although the experimental condition may vary in these studies, the data available strongly suggest that the BNT162b2 treatment of IB3-1 cells is an efficient strategy to induce the expression of pro-inflammatory genes.

A previous study by Gao et al (57) indicated that miR-93-5p could suppress inflammation in an LPS-induced acute lung injury mouse model via targeting the TLR4/myeloid differentiation factor 88 (MyD88)/NF-κB signaling pathway. In the above study, a luciferase construct, composed of a partial sequence of the TLR4 3'-UTR, encompassing the binding sites of miR-93, and sub-cloned into a dual-luciferase reporter vector (pmirGLO), was used to verify that TLR4 was a direct target of miR-93-5p (57). The results demonstrated that RAW264.7 cell transfection with miR-93 mimics inhibited the luciferase activity directed by the TLR4-3'UTR reporter plasmid. By contrast, cell co-transfection with a miR-93 inhibitor enhanced luciferase activity. These findings were consistent with those reported by Fabbri et al (39). This study investigated the effects of miR-93-5p on luciferase activity using a vector carrying the 3'-UTR of human IL-8 mRNA. In both studies, the control experiments demonstrated that there were no changes in luciferase activity in cells transfected with mutant TLR4(57) and IL-8(39) 3'-UTRs, lacking the miR-93 binding sites, thus indicating that there was a direct interaction between miR-93 and the 3'-UTR of IL-8 and TLR4 (39,57).

The possible effects of miR-93-5p on TLR4 are of great significance in applied research, regarding its possible role as inhibitor of SARS-CoV-2 infection and SARS-CoV-2-associated pro-inflammatory gene expression. In fact, several previous studies showed that the SARS-CoV-2 spike protein could activate TLR4, which in turn promoted the synthesis of proinflammatory cytokines (58-60). In this context, the molecular effect of TLR4 in COVID-19 should be considered as a significant regulatory factor of SARS-CoV-2 infection (61). The interaction between SARS-CoV-2 spike protein and TLR4, and its activation, was also reported by Zhao et al (62) and Patra et al (63). The above studies suggested that the binding of SARS-CoV-2 spike protein to TLR4, resulting in its activation, upregulates angiotensin-converting enzyme 2, thus promoting virus entry and hyperinflammation. These studies supported that targeting human TLRs could be a strategy to combat COVID-19. Accordingly, Das et al (64) performed in silico experiments to identify novel anti-SARS-CoV-2 agents from bioactive phytocompounds targeting the viral spike glycoprotein and human TLR4. The above study analyzed a group of 30 phytocompounds, previously demonstrated to retain antiviral activity. These compounds were theoretically screened, using molecular docking, MD simulations and ADME analysis, for their binding efficacy to SARS-CoV-2 spike protein and TLR4. The in silico results showed that cajaninstilbene acid and papaverine could block human TLR4, thus suggesting that these phytocompounds could mitigate SARS-CoV-2-induced proinflammatory responses (64). The significance of this approach was also verified by Sahanic et al (65), reporting that SARS-CoV-2 could activate the TLR4/MyD88 pathway in human macrophages, thus indicating that this effect could be associated with strong pro-inflammatory responses in severe COVID-19. Accordingly, Nakazawa et al (66) found that SARS-CoV-2 could directly impair renal cells via promoting pro-inflammatory responses, while inhibition of TLR4 and IL-1R could prevent SARS-CoV-2 mediated kidney injury. The interplay between TLR4 and SARS-CoV-2, which is involved in the complex mechanisms of inflammation and severity in COVID-19 infections, has been recently reviewed by Asaba et al (67). This study provided novel insights into the unique role of TLR4 in SARS-CoV-2 infection, thus highlighting its potential in the development of novel treatment strategies for COVID-19.

Materials and methods

Materials

All chemicals and reagents were of analytical grade. Aged Garlic Extract (AGE) was obtained by Wakunaga Pharmaceutical Co., Ltd. (Hiroshima, Japan). The Pre-miR miRNA Precursor (PM10951, hsa-miR-93-5p), and the Pre-miR miRNA Precursor Negative Control #1 were both purchased from Ambion-ThermoFisher (cat no. AM17100 and cat no. AM17110, respectively).

Cell culture

Human bronchial epithelial IB3-1 cells (40,50) were cultured as previously described (39). Cells were transfected with 100 nM hsa-miR-93-5p precursor and pre-miR-93 negative control (NEG93) (40) using the Lipofectamine RNAiMAX Transfection Reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The AGE powder used in our experiments was prepared by lyophilization. The concentration of AGE employed in this study was 0.5 mg/ml, in agreement with the study by Gasparello et al (68). The AGE powder was freshly dissolved in complete cell culture medium prior to each experiment.

Treatment of IB3-1 cells with the BNT162b2 vaccine

The BNT162b2 vaccine (Comirnaty; Lot. no. FP8191) was obtained from the Hospital Pharmacy of the University of Padova. For treatment with the BNT162b2 vaccine, IB3-1 cells were seeded at a density of 200,000 cells/ml and were then treated with 0.5 µg/ml of the vaccine for 24 h (48,49). When needed, cells were transfected with pre-miR-93-5p and/or co-treated with AGE.

mRNA quantitative analyses

For the quantification of the relative mRNA expression levels, total RNA was reverse transcribed into cDNA, as described by Gasparello et al (40). Quantitative (q)PCR analysis for IL-6 (assay ID, Hs.PT.58.40226675), IL-8 (assay ID, Hs.PT.58.38869678.g) and IL-1β (assay ID, Hs.PT.58.1518186) was performed using the corresponding assay kits, as previously described (40). PCR amplification was carried out as previously described using the CFX96 Touch Real-Time PCR Detection System (Bio-Rad Laboratories, Inc.) (40,69). RT-qPCR analysis for BNT162b2 Spike mRNA was performed according to the study by Fertig et al (70) (as reported in Data S1). Relative IL-8 gene expression was calculated using the comparative cycle threshold method (ΔΔCq method) and the endogenous control human β-actin was used as normalizer, described previously (39).

Statistical analysis

The differences among different groups were compared using one-way ANOVA (analyses of variance between groups, http://vassarstats.net/anova1u.html). Prism (v. 9.02) by GraphPad software was also employed (followed by Bonferroni's post-hoc tests). Differences were considered statistically significant when P<0.05 and highly significant when P<0.01 (1,4,68,70).

Results

Effect of BNT162b2 vaccine on NF-κB expression

The BNT162b2-treated IB3-1 bronchial cell model is characterized by the BNT162b2-induced expression of pro-inflammatory genes with a higher efficiency compared with SARS-CoV-2 spike protein (40). As a control, in this model spike protein is produced following cell treatment with the BNT162b2 vaccine. The intracellular content of SARS-CoV-2 spike protein should be increased based on the concentrations of BNT162b2 used to treat IB3-1 cells. Herein, a significant increase (P<0.01) in the content of intracellular BNT162b2 Spike mRNA was observed following cell treatment with 1 µg/ml BNT162b2 vaccine for 24 h (Fig. S1). Quantitative RT-qPCR analysis for BNT162b2 Spike mRNA was performed according to the study by Fertig et al (70). In this study, primers were designed to be specific to the publicly available, codon-optimised vaccine mRNA sequence (70). Fully in agreement with the RT-qPCR data shown in Supplementary Fig. S1, full-length S-protein was detected by western blot analysis (data not shown). Since previous studies demonstrated that S-protein could promote pro-inflammatory gene expression via activating the NF-κB pathway in several cellular systems (69,71,72), in the present study NF-κB was upregulated in IB3-1 cells treated with the BNT162b2 vaccine (data not shown).

AGE as a possible anti-inflammatory agent for tackling ‘cytokine storm’ in COVID-19

The AGE powder used in our experiments was prepared by lyophilization. The concentration of AGE employed in this study was 0.5 mg/ml, in agreement with the study by Gasparello et al, who found that this concentration is optimal to obtain inhibition of pro-inflammatory genes in bronchial epithelial IB3-1 cells stimulated with either the SARS-CoV-2 S-protein or the BNT162b2 vaccine (68). The AGE powder was freshly dissolved in complete cell culture medium prior to each experiment. The analysis of the organosulfur compound content in the AGE powder was performed using a GC-MS chromatography and has been reported by Gasparello et al (68). The relative expression levels of the pro-inflammatory genes, IL-6, IL-8 and IL-1β, were detected after 3 days treatment by reverse transcription-qPCR. As shown in Fig. 3, the mRNA expression levels of the aforementioned cytokines were increased following IB3-1 cell treatment with 0.5 µg/ml BNT162b2 (white bars). However, cell transfection with 100 nM pre-miR-93-5p displayed the opposite effect (grey bars).

AGE as a potential anti-inflammatory agent for tackling ‘cytokine storm’ in COVID-19: A pilot study exploring the effects of a combined treatment with pre-miR-93-5p

The present study aimed to explore the possible effects of the combination of pre-miR-93 and AGE in mitigating ‘cytokine storm’ in COVID-19, since a previous study demonstrated that the AGE constituent S-allyl-cysteine (SAC) could interact with TLR4(71). For example, docking analysis performed by Gasparello et al (68), predicted that the SAC/TLR4 interaction was based on hydrogen bonds, involving His685, Tyr657, Ser659 and Arg722. This effect could affect, at least in principle, TLR4 activity, since TIR domain is essential for NF-κB activation (73-76). The results of the pilot experiments, where cells were co-treated with AGE and pre-miR-93-5p, are shown in Fig. 4. In preparing this experiment, we first verified the transfection efficiency of Lipofectamine-delivered pre-miR-93-5p, according with the procedure described by Gasparello et al (40); this validation was performed by RT-qPCR analysis of the miR-93-5p sequences accumulated in pre-miR-93-5p transfected IB3-1 cells. The list of the assays employed for miRNA detection is reported in Table SI. A representative experiment is shown in Supplementary Fig. S2. We always found high transfection efficiency similar to that shown in Fig. S2, with no exception. Another important control was the formal demonstration of the specificity of the effects of transfection of IB3-1 cells with the pre-miR-93-5p. This was demonstrated by comparing the effects of pre-miR93-5p to those of the pre-miR93 negative control, as reported in the representative experiments shown in Fig. S3 and in Table SII. The results concerning the effects of a combined treatment based on AGE and pre-miR-93-5p is shown in Fig. 4. The results obtained revealed a sharp increase in IL-8 gene expression levels following IB3-1 cell treatment with the BNT162b2 vaccine for three days. However, the BNT162b2-induced IL-8 gene expression was reduced after treatment of cells with pre-miR-93-5p, AGE or pre-miR-93-5p plus AGE. Interestingly, the inhibition of IL-8 gene expression was more potent in cells treated with the combination of pre-miR-93-5p and AGE.

Effects of miRNA therapeutics combined with approaches based on the use of natural products: Proposed mechanism of action of miR-93-5p and future perspectives

The extent of complementarity between miR-93-5p and the 3'-UTRs of TLR4 and IL-8 mRNAs are shown in Fig. 5A (39,57). The number of hydrogen bonds generated between miR-93-5p and the 3'-UTRs of TLR4 and IL-8 mRNAs were very similar, thus supporting that both TLR4 and IL-8 mRNAs could be direct targets of miR-93-5p. In addition, miR-93-5p could significantly affect NF-κB signaling via directly interacting with IL-1 receptor-associated kinase 4 (IRAK4), a kinase known to activate NF-κB in the TLR signaling pathways (77,78). Accordingly, the possible mechanism of action of miR-93-5p is depicted in Fig. 5B. In this theoretical model, the mRNA expression levels of IL-8 could be directly inhibited by miR-93-5p, as suggested by Fabbri et al (39). Additionally, IL-8 expression could be indirectly suppressed via the inhibition of the TLR4/NF-κB pathway. This finding could be due to the direct miR-93-5p/TLR-4-3'-UTR-mediated TLR4 downregulation (57) and the direct interaction (and consequent inhibition) with IRAK4(77). In this case, NF-κB inhibition resulted in the inhibitory effects of miR-93-5p on NF-κB-regulated genes, such as IL-1β and IL-6. Data sustaining the direct interaction between miR-93-5p and the 3'-UTRs of IL-8, TLR4 and IRAK4 have been also reported in the studies by Fabbri et al (39), Gao et al (57) and Xu et al (77).

Discussion

The present study reported preliminary evidence suggesting that the miR-93-5p based ‘miRNA therapeutics’ combined with AGE could be considered as an efficient approach for inhibiting IL-8 expression. This could be of significant interest, since the TLR4/NF-κB-related inhibitory agents, such as miR-93-5p, could be applied in the treatment of human pathologies associated with a hyperinflammatory state, such as COVID-19 (45-49) and cystic fibrosis (62,63), which are characterized by IL-8 upregulation.

Emerging evidence has suggested that IL-8, TLR4 and IRAK4 can be directly targeted by miR-93-5p (39,57,77). Accordingly, the proposed mechanism of action of miR-93-5p (Fig. 5B), indicated that the direct targeting of the IL-8 3'-UTR by miR-93-5p could inhibit IL-8 expression. At the same time, the indirect suppression of IL-8 expression could be achieved via blocking the TLR4/NF-κB pathway, possibly through the direct interaction between miR-93-5p and IRAK4 or TLR4. Although several studies have supported the direct interaction between miR-93-5p and the 3'-UTR of IL-8, TLR4 and IRAK4 (39,57,77), future studies are still needed to verify the possible interactions of miR-93-5p with TLR4 and IRAK4 in the BNT162b2 based experimental model system, particularly in cells also treated with pre-miR-93-5p and AGE.

The most significant limitation of the current study was that the expression levels of only a few pro-inflammatory genes (IL-1β, IL-6 and IL-8) were detected in BNT162b2-treated IB3-1 cells and cells co-treated with both miR-93-5p mimics and AGE (IL-8). Therefore, the present study should be considered as an exploratory pilot study based on a proof-of-principle, suggesting that a combined treatment with pre-miR-93-5p and AGE could be most effective in inhibiting the expression of pro-inflammatory genes. Future studies, focusing on more proteins involved in the ‘cytokine storm’ in COVID-19 and on the hyperinflammation state of other human pathologies, such as cystic fibrosis and chronic obstructive pulmonary disease, should be performed. In addition, the current preliminary study could promote the design of other experimental studies on the effects of pre-miR-93-5p in combination with other extracts from medicinal plants, including the bioactive constituents of AGE (SAC and S1PC), on regulating IL-8 expression. Although the BNT162b2 treated cells are not a stringent model system for viral infection, the results of this study are expected to stimulate experimental efforts to determine whether the combination of pre-miR-93-5p and AGE exerts the same effects in SARS-CoV-2-infected cells (68).

The preliminary data of the present study suggested that pre-miR-93-5p could be considered as a double-acting agent, since it could directly inhibit IL-8 mRNA translation and indirectly block NF-κB signaling via inhibiting TRL4 and IRAK4. Further studies on other pro-inflammatory genes could determine whether the inhibitory effects of the pre-miR-93/AGE combined treatment on IL-8 expression could be also generalized to other genes involved in the COVID-19-related hyper-inflammatory state. This experimental plan could also verify the association between the inhibitory effects on pro-inflammatory genes and corresponding alterations on the NF-κB pathway. Another significant limitation of the current study was that only one ago-miRNA molecule, namely pre-miR-93 ago-miRNA, was employed. However, it has been reported that other miRNAs can interact with TLRs, such as miR-145-5p, and NF-κB, such as miR-130a, miR-146a/b, miR-218 and miR-451 (79-82). Studies based on molecules mimicking the function(s) of these miRNAs should be also considered to fully understand the interplay between miRNAs, NF-κB, TLR4 and ‘cytokine storm’-related pro-inflammatory genes. In addition, these future studies could allow the identification of novel pre-miRNA molecules, which could be useful for verifying the hypothesis that pre-miRNA molecules in combination with natural products, such as AGE and other compounds isolated from AGE, could possibly enhance their inhibitory activity on inflammation.

Supplementary Material

Data S1
(A) Representative examples showing the RT-qPCR analysis of SARS-CoV-2 S-protein mRNA. (B) Content of the SARS-CoV-2 mRNA (fold increase with respect to trace amount of hybridizable material found in control cells, taken as 1). The content of SARS-CoV-2 S-protein mRNA has been evaluated by RT-qPCR using as described by Fertig et al (70) and further detailed in Data S1. Endogenous β-actin was used as internal control. Results represent the average ± standard deviation of three independent experiments. RT-qPCR, reverse transcription-quantitative PCR; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Representative examples showing the increase of the content of miR-93-5p (fold increase with respect to endogenous miR-93-5p) in IB3-1 cells transfected with the pre-miR-93-5p. The content of miR-93-5p has been evaluated by RT-qPCR using the assays described in Table SI. The relative expression of miR-93-5p was calculated as elsewhere described (39) using human Let-7c-5p (white) and U6 (grey) RNAs as normalizers. Results represent the average ± standard deviation of five independent experiments. RT-qPCR, reverse transcription-quantitative PCR; miR, microRNA; n.s., not significant.
Representative examples showing the increase of the expression of pro-inflammatory genes upon treatment of IB3-1 cells with 5 nm SARS-CoV2 S-protein and the effect of transfection with 200 nM pre-miR-93-5p (agomiR-93) and miR-93 negative control using (A) Bio-plex analysis and (B) RT-qPCR. The relative expression of IL-8 mRNA was calculated as elsewhere described (1,4) using human β-actin as normalizer. Results represent the average ± standard deviation of four independent experiments. Modified from the study reported by Gasparello et al (1). Copyright can be found at: https://pmc.ncbi.nlm.nih.gov/articles/PMC8492649/. IL, interleukin; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2. Further information is included in Table SII.
List of assays employed for miRNA detection.
Effects of pre-miR-93-5p on IB3-1 exposed to SARS-CoV-2 S-protein.

Acknowledgements

The authors wish to thank Dr. Takahiro Ogawa and Dr. Toshiaki Matsutomo (Wakunaga Pharmaceutical Co., Ltd.) for organizing the shipment of the AGE extract.

Funding

Funding: This work was funded by the Italian Ministery of University and Research (MUR)-FISR COVID-miRNA Peptide Nucleic Acid Project (grant no. FISR2020IP_04128), by FIRD 2024 funds from the University of Ferrara. (grant no. FIRD-Finotti 2024) and by the Interuniversity Consortium for Biotechnologies, Italy (grant nos. CIB-Unife-2020 and CIB-Unife-2021).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

AF, EA and RG designed the experimental plan. JG, CP, AF were involved in the methodology development. JG and CP performed the experiments. AF, CP, JG and RG curated the data and analysed the results. AF and RG were responsible for funding acquisition. AF and RG supervised the investigation. RG wrote the original draft. AF, RG and EA reviewed and edited the draft. All authors read and approved the final version of the manuscript. RG and AF confirm the authenticity of all the raw data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The company Wakunaga Pharmaceutical Co., Ltd. both provided the aged garlic extract used in this study and also paid for the publication charges.

References

1 

Killerby ME, Biggs HM, Midgley CM, Gerber SI and Watson JT: Middle east respiratory syndrome coronavirus transmission. Emerg Infect Dis. 26:191–198. 2020.PubMed/NCBI View Article : Google Scholar

2 

Murakami N, Hayden R, Hills T, Al-Samkari H, Casey J, Del Sorbo L, Lawler PR, Sise ME and Leaf DE: Therapeutic advances in COVID-19. Nat Rev Nephrol. 19:38–52. 2023.PubMed/NCBI View Article : Google Scholar

3 

Narayanan SA, Jamison DA Jr, Guarnieri JW, Zaksas V, Topper M, Koutnik AP, Park J, Clark KB, Enguita FJ, Leitão AL, et al: A comprehensive SARS-CoV-2 and COVID-19 review, Part 2: Host extracellular to systemic effects of SARS-CoV-2 infection. Eur J Hum Genet. 32:10–20. 2024.PubMed/NCBI View Article : Google Scholar

4 

Tulimilli SV, Dallavalasa S, Basavaraju CG, Kumar Rao V, Chikkahonnaiah P, Madhunapantula SV and Veeranna RP: Variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and vaccine effectiveness. Vaccines (Basel). 10(1751)2022.PubMed/NCBI View Article : Google Scholar

5 

Watson OJ, Barnsley G, Toor J, Hogan AB, Winskill P and Ghani AC: Global impact of the first year of COVID-19 vaccination: A mathematical modelling study. Lancet Infect Dis. 22:1293–1302. 2022.PubMed/NCBI View Article : Google Scholar

6 

Walker PGT, Whittaker C, Watson OJ, Baguelin M, Winskill P, Hamlet A, Djafaara BA, Cucunubá Z, Olivera Mesa D, Green W, et al: The impact of COVID-19 and strategies for mitigation and suppression in low- and middle-income countries. Science. 369:413–422. 2020.PubMed/NCBI View Article : Google Scholar

7 

Villamagna AH, Gore SJ, Lewis JS and Doggett JS: The need for antiviral drugs for pandemic coronaviruses from a global health perspective. Front Med (Lausanne). 7(596587)2020.PubMed/NCBI View Article : Google Scholar

8 

Mikulska M, Sepulcri C, Dentone C, Magne F, Balletto E, Baldi F, Labate L, Russo C, Mirabella M, Magnasco L, et al: Triple combination therapy with 2 antivirals and monoclonal antibodies for persistent or relapsed severe acute respiratory syndrome coronavirus 2 infection in immunocompromised patients. Clin Infect Dis. 77:280–286. 2023.PubMed/NCBI View Article : Google Scholar

9 

Meyerowitz EA and Li Y: Review: The landscape of antiviral therapy for COVID-19 in the era of widespread population immunity and omicron-lineage viruses. Clin Infect Dis. 78:908–917. 2024.PubMed/NCBI View Article : Google Scholar

10 

Selickman J, Vrettou CS, Mentzelopoulos SD and Marini JJ: COVID-19-related ARDS: Key mechanistic features and treatments. J Clin Med. 11(4896)2022.PubMed/NCBI View Article : Google Scholar

11 

Silva MJA, Ribeiro LR, Gouveia MIM, Marcelino BDR, Santos CSD, Lima KVB and Lima LNGC: Hyperinflammatory response in COVID-19: A systematic review. Viruses. 15(553)2023.PubMed/NCBI View Article : Google Scholar

12 

Conti P, Caraffa A, Gallenga CE, Ross R, Kritas SK, Frydas I, Younes A and Ronconi G: Coronavirus-19 (SARS-CoV-2) induces acute severe lung inflammation via IL-1 causing cytokine storm in COVID-19: A promising inhibitory strategy. J Biol Regul Homeost Agents. 34:1971–1975. 2020.PubMed/NCBI View Article : Google Scholar

13 

Zawawi A, Naser AY, Alwafi H and Minshawi F: Profile of circulatory cytokines and chemokines in human coronaviruses: A systematic review and meta-analysis. Front Immunol. 12(666223)2021.PubMed/NCBI View Article : Google Scholar

14 

Altmann DM, Whettlock EM, Liu S, Arachchillage DJ and Boyton RJ: The immunology of long COVID. Nat Rev Immunol. 23:618–634. 2023.PubMed/NCBI View Article : Google Scholar

15 

Ghorra N, Popotas A, Besse-Hammer T, Rogiers A, Corazza F and Nagant C: Cytokine profile in patients with postacute sequelae of COVID-19. Viral Immunol. 37:346–354. 2024.PubMed/NCBI View Article : Google Scholar

16 

Song P, Li W, Xie J, Hou Y and You C: Cytokine storm induced by SARS-CoV-2. Clin Chim Acta. 509:280–287. 2020.PubMed/NCBI View Article : Google Scholar

17 

Sefik E, Qu R, Junqueira C, Kaffe E, Mirza H, Zhao J, Brewer JR, Han A, Steach HR, Israelow B, et al: Inflammasome activation in infected macrophages drives COVID-19 pathology. Nature. 606:585–593. 2022.PubMed/NCBI View Article : Google Scholar

18 

Majidpoor J and Mortezaee K: Interleukin-6 in SARS-CoV-2 induced disease: Interactions and therapeutic applications. Biomed Pharmacother. 145(112419)2022.PubMed/NCBI View Article : Google Scholar

19 

Santa Cruz A, Mendes-Frias A, Oliveira AI, Dias L, Matos AR, Carvalho A, Capela C, Pedrosa J, Castro AG and Silvestre R: Interleukin-6 is a biomarker for the development of fatal severe acute respiratory syndrome coronavirus 2 pneumonia. Front Immunol. 12(613422)2021.PubMed/NCBI View Article : Google Scholar

20 

Zizzo G, Tamburello A, Castelnovo L, Laria A, Mumoli N, Faggioli PM, Stefani I and Mazzone A: Immunotherapy of COVID-19: Inside and beyond IL-6 signalling. Front Immunol. 13(795315)2022.PubMed/NCBI View Article : Google Scholar

21 

Li L, Li J, Gao M, Fan H, Wang Y, Xu X, Chen C, Liu J, Kim J, Aliyari R, et al: Interleukin-8 as a biomarker for disease prognosis of coronavirus disease-2019 patients. Front Immunol. 11(602395)2021.PubMed/NCBI View Article : Google Scholar

22 

Yu Q, Zhou X, Kapini R, Arsecularatne A, Song W, Li C, Liu Y, Ren J, Münch G, Liu J and Chang D: Cytokine storm in COVID-19: Insight into pathological mechanisms and therapeutic benefits of chinese herbal medicines. Medicines (Basel). 11(14)2024.PubMed/NCBI View Article : Google Scholar

23 

Comarmond C, Drumez E, Labreuche J, Hachulla E, Thomas T, Flipo RM, Seror R, Avouac J, Balandraud N, Desbarbieux R, et al: COVID-19 presentation and outcomes in patients with inflammatory rheumatic and musculoskeletal diseases receiving IL6-receptor antagonists prior to SARS-CoV-2 infection. J Transl Autoimmun. 6(100190)2023.PubMed/NCBI View Article : Google Scholar

24 

Ghosn L, Chaimani A, Evrenoglou T, Davidson M, Graña C, Schmucker C, Bollig C, Henschke N, Sguassero Y, Nejstgaard CH, et al: Interleukin-6 blocking agents for treating COVID-19: A living systematic review. Cochrane Database Syst Rev. 3(CD013881)2021.PubMed/NCBI View Article : Google Scholar

25 

Pomponio G, Ferrarini A, Bonifazi M, Moretti M, Salvi A, Giacometti A, Tavio M, Titolo G, Morbidoni L, Frausini G, et al: Tocilizumab in COVID-19 interstitial pneumonia. J Intern Med. 289:738–746. 2021.PubMed/NCBI View Article : Google Scholar

26 

Castelnovo L, Tamburello A, Lurati A, Zaccara E, Marrazza MG, Olivetti M, Mumoli N, Mastroiacovo D, Colombo D, Ricchiuti E, et al: Anti-IL6 treatment of serious COVID-19 disease: A monocentric retrospective experience. Medicine (Baltimore). 100(e23582)2021.PubMed/NCBI View Article : Google Scholar

27 

Tharmarajah E, Buazon A, Patel V, Hannah JR, Adas M, Allen VB, Bechman K, Clarke BD, Nagra D, Norton S, et al: IL-6 inhibition in the treatment of COVID-19: A meta-analysis and meta-regression. J Infect. 82:178–185. 2021.PubMed/NCBI View Article : Google Scholar

28 

Segú-Vergés C, Artigas L, Coma M and Peck RW: Artificial intelligence assessment of the potential of tocilizumab along with corticosteroids therapy for the management of COVID-19 evoked acute respiratory distress syndrome. PLoS One. 18(e0280677)2023.PubMed/NCBI View Article : Google Scholar

29 

Dominguez C, McCampbell KK, David JM and Palena C: Neutralization of IL-8 decreases tumor PMN-MDSCs and reduces mesenchymalization of claudin-low triple-negative breast cancer. JCI Insight. 2(e9496)2017.PubMed/NCBI View Article : Google Scholar

30 

Piemonti L, Landoni G, Voza A, Puoti M, Gentile I, Coppola N, Nava S, Mattei A, Marinangeli F, Marchetti G, et al: Efficacy and safety of reparixin in patients with severe COVID-19 pneumonia: A phase 3, randomized, double-blind placebo-controlled study. Infect Dis Ther. 12:2437–2456. 2023.PubMed/NCBI View Article : Google Scholar

31 

Zarbock A, Allegretti M and Ley K: Therapeutic inhibition of CXCR2 by Reparixin attenuates acute lung injury in mice. Br J Pharmacol. 155:357–364. 2008.PubMed/NCBI View Article : Google Scholar

32 

Kaiser R, Leunig A, Pekayvaz K, Popp O, Joppich M, Polewka V, Escaig R, Anjum A, Hoffknecht ML, Gold C, et al: Self-sustaining IL-8 loops drive a prothrombotic neutrophil phenotype in severe COVID-19. JCI Insight. 6(e150862)2021.PubMed/NCBI View Article : Google Scholar

33 

Shang R, Lee S, Senavirathne G and Lai EC: microRNAs in action: Biogenesis, function and regulation. Nat Rev Genet. 24:816–833. 2023.PubMed/NCBI View Article : Google Scholar

34 

Fazi   and Nervi C: MicroRNA: Basic mechanisms and transcriptional regulatory networks for cell fate determination. Cardiovasc Res. 79:553–561. 2008.PubMed/NCBI View Article : Google Scholar

35 

Ivey KN and Srivastava D: microRNAs as developmental regulators. Cold Spring Harb Perspect Biol. 7(a008144)2015.PubMed/NCBI View Article : Google Scholar

36 

Chaudhuri K and Chatterjee R: MicroRNA detection and target prediction: Integration of computational and experimental approaches. DNA Cell Biol. 26:321–337. 2007.PubMed/NCBI View Article : Google Scholar

37 

Doench JG and Sharp PA: Specificity of microRNA target selection in translational repression. Genes Dev. 18:504–511. 2004.PubMed/NCBI View Article : Google Scholar

38 

Gasparello J, Finotti A and Gambari R: Tackling the COVID-19 ‘cytokine storm’ with microRNA mimics directly targeting the 3'UTR of pro-inflammatory mRNAs. Med Hypotheses. 146(110415)2021.PubMed/NCBI View Article : Google Scholar

39 

Fabbri E, Borgatti M, Montagner G, Bianchi N, Finotti A, Lampronti I, Bezzerri V, Dechecchi MC, Cabrini G and Gambari R: Expression of microRNA-93 and interleukin-8 during pseudomonas aeruginosa-mediated induction of proinflammatory responses. Am J Respir Cell Mol Biol. 50:1144–1155. 2014.PubMed/NCBI View Article : Google Scholar

40 

Gasparello J, d'Aversa E, Breveglieri G, Borgatti M, Finotti A and Gambari R: In vitro induction of interleukin-8 by SARS-CoV-2 spike protein is inhibited in bronchial epithelial IB3-1 cells by a miR-93-5p agomiR. Int Immunopharmacol. 101(108201)2021.PubMed/NCBI View Article : Google Scholar

41 

Wu J, Ding J, Yang J, Guo X and Zheng Y: MicroRNA roles in the nuclear factor kappa B signaling pathway in cancer. Front Immunol. 9(546)2018.PubMed/NCBI View Article : Google Scholar

42 

Hanna J, Hossain G and Kocerha J: The potential for microRNA therapeutics and clinical research. Front Genet. 10(478)2019.PubMed/NCBI View Article : Google Scholar

43 

Momin MY, Gaddam RR, Kravitz M, Gupta A and Vikram A: The challenges and opportunities in the development of MicroRNA therapeutics: A multidisciplinary viewpoint. Cells. 10(3097)2021.PubMed/NCBI View Article : Google Scholar

44 

Diener C, Keller A and Meese E: Emerging concepts of miRNA therapeutics: From cells to clinic. Trends Genet. 38:613–626. 2022.PubMed/NCBI View Article : Google Scholar

45 

Rupaimoole R and Slack FJ: MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 16:203–222. 2017.PubMed/NCBI View Article : Google Scholar

46 

Lee EC, Valencia T, Allerson C, Schairer A, Flaten A, Yheskel M, Kersjes K, Li J, Gatto S, Takhar M, et al: Discovery and preclinical evaluation of anti-miR-17 oligonucleotide RGLS4326 for the treatment of polycystic kidney disease. Nat Commun. 10(4148)2019.PubMed/NCBI View Article : Google Scholar

47 

Reid G, Kao SC, Pavlakis N, Brahmbhatt H, MacDiarmid J, Clarke S, Boyer M and van Zandwijk N: Clinical development of TargomiRs, a miRNA mimic-based treatment for patients with recurrent thoracic cancer. Epigenomics. 8:1079–1085. 2016.PubMed/NCBI View Article : Google Scholar

48 

Zurlo M, Gasparello J, Verona M, Papi C, Cosenza LC, Finotti A, Marzaro G and Gambari R: The anti-SARS-CoV-2 BNT162b2 vaccine suppresses mithramycin-induced erythroid differentiation and expression of embryo-fetal globin genes in human erythroleukemia K562 cells. Exp Cell Res. 433(113853)2023.PubMed/NCBI View Article : Google Scholar

49 

Cosenza LC, Marzaro G, Zurlo M, Gasparello J, Zuccato C, Finotti A and Gambari R: Inhibitory effects of SARS-CoV-2 spike protein and BNT162b2 vaccine on erythropoietin-induced globin gene expression in erythroid precursor cells from patients with β-thalassemia. Exp Hematol. 129(104128)2024.PubMed/NCBI View Article : Google Scholar

50 

Gasparello J, D'Aversa E, Papi C, Gambari L, Grigolo B, Borgatti M, Finotti A and Gambari R: Sulforaphane inhibits the expression of interleukin-6 and interleukin-8 induced in bronchial epithelial IB3-1 cells by exposure to the SARS-CoV-2 spike protein. Phytomedicine. 87(153583)2021.PubMed/NCBI View Article : Google Scholar

51 

Verbeke R, Hogan MJ, Loré K and Pardi N: Innate immune mechanisms of mRNA vaccines. Immunity. 55:1993–2005. 2022.PubMed/NCBI View Article : Google Scholar

52 

Sartorius R, Trovato M, Manco R, D'Apice L and De Berardinis P: Exploiting viral sensing mediated by Toll-like receptors to design innovative vaccines. NPJ Vaccines. 6(127)2021.PubMed/NCBI View Article : Google Scholar

53 

Delehedde C, Even L, Midoux P, Pichon C and Perche F: Intracellular routing and recognition of lipid-based mRNA nanoparticles. Pharmaceutics. 13(945)2021.PubMed/NCBI View Article : Google Scholar

54 

Dechecchi MC, Nicolis E, Norez C, Bezzerri V, Borgatti M, Mancini I, Rizzotti P, Ribeiro CM, Gambari R, Becq F and Cabrini G: Anti-inflammatory effect of miglustat in bronchial epithelial cells. J Cyst Fibros. 7:555–565. 2008.PubMed/NCBI View Article : Google Scholar

55 

Gambari R, Borgatti M, Lampronti I, Fabbri E, Brognara E, Bianchi N, Piccagli L, Yuen MCW, Kan CW, Hau DKP, et al: Corilagin is a potent inhibitor of NF-kappaB activity and downregulates TNF-alpha induced expression of IL-8 gene in cystic fibrosis IB3-1 cells. Int Immunopharmacol. 13:308–315. 2012.PubMed/NCBI View Article : Google Scholar

56 

De Stefano D, Ungaro F, Giovino C, Polimeno A, Quaglia F and Carnuccio R: Sustained inhibition of IL-6 and IL-8 expression by decoy ODN to NF-κB delivered through respirable large porous particles in LPS-stimulated cystic fibrosis bronchial cells. J Gene Med. 13:200–208. 2011.PubMed/NCBI View Article : Google Scholar

57 

Gao H, Xiao D, Gao L and Li  : MicroRNA-93 contributes to the suppression of lung inflammatory responses in LPS-induced acute lung injury in mice via the TLR4/MyD88/NF-κB signaling pathway. Int J Mol Med. 46:561–570. 2020.PubMed/NCBI View Article : Google Scholar

58 

Silva-Aguiar RP, Teixeira DE, Peruchetti DB, Peres RAS, Alves SAS, Calil PT, Arruda LB, Costa LJ, Silva PL, Schmaier AH, et al: Toll like receptor 4 mediates the inhibitory effect of SARS-CoV-2 spike protein on proximal tubule albumin endocytosis. Biochim Biophys Acta Mol Basis Dis. 1870(167155)2024.PubMed/NCBI View Article : Google Scholar

59 

Chakraborty C, Mallick B, Bhattacharya M and Byrareddy S: SARS-CoV-2 omicron spike shows strong binding affinity and favourable interaction landscape with the TLR4/MD2 compared to other variants. J Genet Eng Biotechnol. 22(100347)2024.PubMed/NCBI View Article : Google Scholar

60 

Fontes-Dantas FL, Fernandes GG, Gutman EG, De Lima EV, Antonio LS, Hammerle MB, Mota-Araujo HP, Colodeti LC, Araújo SMB, Froz GM, et al: SARS-CoV-2 spike protein induces TLR4-mediated long-term cognitive dysfunction recapitulating post-COVID-19 syndrome in mice. Cell Rep. 42(112189)2023.PubMed/NCBI View Article : Google Scholar

61 

Choudhury A and Mukherjee S: In silico studies on the comparative characterization of the interactions of SARS-CoV-2 spike glycoprotein with ACE-2 receptor homologs and human TLRs. J Med Virol. 92:2105–2113. 2020.PubMed/NCBI View Article : Google Scholar

62 

Zhao Y, Kuang M, Li J, Zhu L, Jia Z, Guo X, Hu Y, Kong J, Yin H, Wang X and You F: SARS-CoV-2 spike protein interacts with and activates TLR41. Cell Res. 31:818–820. 2021.PubMed/NCBI View Article : Google Scholar

63 

Patra R, Chandra Das N and Mukherjee S: Targeting human TLRs to combat COVID-19: A solution? J Med Virol. 93:615–617. 2021.PubMed/NCBI View Article : Google Scholar

64 

Das NC, Labala R, Patra R, Chattoraj A and Mukherjee S: In silico identification of new anti-SARS-CoV-2 agents from bioactive phytocompounds targeting the viral spike glycoprotein and human TLR4. Lettr Drug Des Discov. 19:175–191. 2022.

65 

Sahanic S, Hilbe R, Dünser C, Tymoszuk P, Löffler-Ragg J, Rieder D, Trajanoski Z, Krogsdam A, Demetz E, Yurchenko M, et al: SARS-CoV-2 activates the TLR4/MyD88 pathway in human macrophages: A possible correlation with strong pro-inflammatory responses in severe COVID-19. Heliyon. 9(e21893)2023.PubMed/NCBI View Article : Google Scholar

66 

Nakazawa D, Takeda Y, Kanda M, Tomaru U, Ogawa H, Kudo T, Shiratori-Aso S, Watanabe-Kusunoki K, Ueda Y, Miyoshi A, et al: Inhibition of Toll-like receptor 4 and Interleukin-1 receptor prevent SARS-CoV-2 mediated kidney injury. Cell Death Discov. 9(293)2023.PubMed/NCBI View Article : Google Scholar

67 

Asaba CN, Ekabe CJ, Ayuk HS, Gwanyama BN, Bitazar R and Bukong TN: Interplay of TLR4 and SARS-CoV-2: Unveiling the complex mechanisms of inflammation and severity in COVID-19 infections. J Inflamm Res. 17:5077–5091. 2024.PubMed/NCBI View Article : Google Scholar

68 

Gasparello J, Papi C, Marzaro G, Macone A, Zurlo M, Finotti A, Agostinelli E and Gambari R: Aged garlic extract (AGE) and its constituents S-allyl-cysteine (SAC) inhibit the expression of pro-inflammatory genes induced in bronchial epithelial IB3-1 cells by exposure to the SARS-CoV-2 spike protein and the BNT162b2 vaccine. Molecules. 29(5938)2024.PubMed/NCBI View Article : Google Scholar

69 

Gasparello J, Marzaro G, Papi C, Gentili V, Rizzo R, Zurlo M, Scapoli C, Finotti A and Gambari R: Effects of Sulforaphane on SARS-CoV-2 infection and NF-κB dependent expression of genes involved in the COVID-19 ‘cytokine storm’. Int J Mol Med. 52(76)2023.PubMed/NCBI View Article : Google Scholar

70 

Fertig TE, Chitoiu L, Marta DS, Ionescu VS, Cismasiu VB, Radu E, Angheluta G, Dobre M, Serbanescu A, Hinescu ME and Gherghiceanu M: Vaccine mRNA can be detected in blood at 15 days post-vaccination. Biomedicines. 10(1538)2022.PubMed/NCBI View Article : Google Scholar

71 

Zhou Q, Zhang L, Dong Y, Wang Y, Zhang B, Zhou S, Huang Q, Wu T and Chen G: The role of SARS-CoV-2-mediated NF-κB activation in COVID-19 patients. Hypertens Res. 47:375–384. 2024.PubMed/NCBI View Article : Google Scholar

72 

Forsyth CB, Zhang L, Bhushan A, Swanson B, Zhang L, Mamede JI, Voigt RM, Shaikh M, Engen PA and Keshavarzian A: The SARS-CoV-2 S1 spike protein promotes MAPK and NF-κB activation in human lung cells and inflammatory cytokine production in human lung and intestinal epithelial cells. Microorganisms. 10(1996)2022.PubMed/NCBI View Article : Google Scholar

73 

Zhang G and Ghosh S: Toll-like receptor-mediated NF-kappaB activation: A phylogenetically conserved paradigm in innate immunity. J Clin Invest. 107:13–19. 2001.PubMed/NCBI View Article : Google Scholar

74 

Gargiulo S, Gamba P, Testa G, Rossin D, Biasi F, Poli G and Leonarduzzi G: Relation between TLR4/NF-κB signaling pathway activation by 27-hydroxycholesterol and 4-hydroxynonenal, and atherosclerotic plaque instability. Aging Cell. 14:569–581. 2015.PubMed/NCBI View Article : Google Scholar

75 

Muir A, Soong G, Sokol S, Reddy B, Gomez MI, Van Heeckeren A and Prince A: Toll-like receptors in normal and cystic fibrosis airway epithelial cells. Am J Respir Cell Mol Biol. 30:777–783. 2004.PubMed/NCBI View Article : Google Scholar

76 

Greene CM, Carroll TP, Smith SGJ, Taggart CC, Devaney J, Griffin S, O'neill SJ and McElvaney NG: TLR-induced inflammation in cystic fibrosis and non-cystic fibrosis airway epithelial cells. J Immunol. 174:1638–1646. 2005.PubMed/NCBI View Article : Google Scholar

77 

Xu Y, Jin H, Yang X, Wang L, Su L, Liu K, Gu Q and Xu X: MicroRNA-93 inhibits inflammatory cytokine production in LPS-stimulated murine macrophages by targeting IRAK4. FEBS Lett. 588:1692–1698. 2014.PubMed/NCBI View Article : Google Scholar

78 

Tian F, Yuan C, Hu L and Shan S: MicroRNA-93 inhibits inflammatory responses and cell apoptosis after cerebral ischemia reperfusion by targeting interleukin-1 receptor-associated kinase 4. Exp Ther Med. 14:2903–2910. 2017.PubMed/NCBI View Article : Google Scholar

79 

Wei L and Zhao D: M2 macrophage-derived exosomal miR-145-5p protects against the hypoxia/reoxygenation-induced pyroptosis of cardiomyocytes by inhibiting TLR4 expression. Ann Transl Med. 10(1376)2022.PubMed/NCBI View Article : Google Scholar

80 

Jin C, Wang A, Liu L, Wang G, Li G and Han Z: miR-145-5p inhibits tumor occurrence and metastasis through the NF-κB signaling pathway by targeting TLR4 in malignant melanoma. J Cell Biochem. 120:11115–11126. 2019.PubMed/NCBI View Article : Google Scholar

81 

Ma X, Becker Buscaglia LE, Barker JR and Li Y: MicroRNAs in NF-kappaB signaling. J Mol Cell Biol. 3:159–166. 2011.PubMed/NCBI View Article : Google Scholar

82 

Ghafouri-Fard S, Abak A, Fattahi F, Hussen BM, Bahroudi Z, Shoorei H and Taheri M: . The interaction between miRNAs/lncRNAs and nuclear factor-κB (NF-κB) in human disorders. Biomed Pharmacother. 138(111519)2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2025
Volume 29 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gambari R, Papi C, Gasparello J, Agostinelli E and Finotti A: Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene. Exp Ther Med 29: 85, 2025.
APA
Gambari, R., Papi, C., Gasparello, J., Agostinelli, E., & Finotti, A. (2025). Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene. Experimental and Therapeutic Medicine, 29, 85. https://doi.org/10.3892/etm.2025.12835
MLA
Gambari, R., Papi, C., Gasparello, J., Agostinelli, E., Finotti, A."Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene". Experimental and Therapeutic Medicine 29.4 (2025): 85.
Chicago
Gambari, R., Papi, C., Gasparello, J., Agostinelli, E., Finotti, A."Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene". Experimental and Therapeutic Medicine 29, no. 4 (2025): 85. https://doi.org/10.3892/etm.2025.12835