Skip to main content
BMC is moving to Springer Nature Link. Visit this journal in its new home.

Nano delivery of MiR-146a and its effect study on genes involved in apoptosis and autophagy pathways in lung cancer and tuberculosis

BMC Biotechnology volume 25, Article number: 81 (2025) Cite this article

Abstract

Background

Tuberculosis (TB) and lung cancer (LC) are among the leading causes of death worldwide and present serious challenges in diagnosis and treatment. Therefore, developing new strategies for their treatment is crucial. MicroRNAs (miRNAs) are biological molecules that play a critical role in regulating essential processes, such as apoptosis and autophagy, in TB and LC by targeting specific genes. Recently, carbon nanotubes functionalized with Polyethyleneimine (CNT-PEI) to deliver miRNAs to target cells have been investigated to enhance therapeutic effects.

Methods

In this study, miR-146a was transfected into LC (A549), macrophages infected with TB (THP1), and healthy lung cells (MRC5) using CNT-PEI. Then, the expression of miR-146a and its target gene, TNF receptor-associated factor-6 (TRAF6), and other genes involved in apoptosis and autophagy pathways including BCL-2, IL-6, tumor necrosis factor-alpha (TNFα), were measured using Real-Time PCR. Finally, the effect of overexpression of miR-146a on these genes was investigated in all three cell lines.

Result

The results showed successful transfection of miR-146a using the CNT-PEI nano delivery system in LC and TB cell models. Then, increased expression of miR-146 increased apoptosis and autophagy by targeting the TRAF6 gene and affecting other genes such as BCL-2, IL-6, and TNFα through the NF-kB signaling pathway.

Conclusion

The findings suggest an important role for miR-146a in TB and LC, which regulates inflammatory responses and treats these diseases. However, further studies are needed on using CNT-PEI in vivo, as well as the balance between local anti-inflammatory and non-inflammatory factors.

Peer Review reports

Introduction

Lung cancer (LC), basically includes non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) [1]. LC is the second most common cancer in both men and women, with a total of 235,760 new cases and 131,880 deaths reported in 2021 [1]. Several genetic and environmental factors, such as smoking, asbestos, and radon, may play a role in causing LC [1]. Important genes that act as an oncogene in LC are EGFR, KRAS, BRAF, PIK3CA, RET, and ROS1 [2].

Apoptosis is an active and energy-dependent process, and its inhibition is linked to the development of cancers such as LC [3]. The BCL-2 gene can play roles in the induction and inhibition of apoptosis through the NF-κB signaling pathway [3]. The balance between apoptosis and anti-apoptotic processes in cancer cells is maintained by tumor necrosis factors (TNF) receptor-associated factor-6 (TRAF6) [4]. TRAF6 is associated with TNFα-induced cancer cell migration and invasion and is involved in IL-1 signaling, which leads to the activation of nuclear factor kappa B (NF-κB) [4, 5].

Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), remains a significant public health issue worldwide. In 2021, there were 10.6 million new TB cases and 1.6 million TB-related deaths [6]. An active TB infection causes fibrosis, irreversible scarring, and impaired immune function in the lung parenchyma [7]. In addition, TB susceptibility is influenced by genetic polymorphisms in innate immunity and inflammation genes, including Toll-like receptors and TNF, which are also linked to LC risk [7].

Autophagy is a normal physiological process that supports survival mechanisms in normal respiratory cells. It is also involved in the lysosomal degradation of microorganisms (e.g., TB), damaged organelles, and dysfunctional proteins [8]. Important genes in this pathway include IL-6, TNFα, and TRAF6, which are widely implicated in autophagy and autophagosome maturation in diseases such as TB [5, 9].

MicroRNAs (miRNAs) are small biological molecules found in plasma, serum, urine, and saliva, and have played critical roles in the diagnosis and treatment of diseases [10]. MiRNAs are small non-coding RNAs that range from 18 to 25 nucleotides in length. They bind to complementary sequences in the 3’ UTR of target transcribed mRNA, leading to translational repression, gene degradation, or silencing [11]. MiRNAs are involved in several biological processes, including regulation of cytokines, immune responses, gene expression, cell growth, migration, invasion, differentiation, autophagy, and apoptosis [11,12,13,14].

The miR-146 family consists of miR-146a and miR-146b, which are located in the chromosomal region 5q33.3 [15]. Evidence suggests that miR-146a is significantly altered in both TB and NSCLC and functions through common pathways such as regulating immune and cellular responses [16]. Studies have shown that in NSCLC cell lines and human tissue samples, miR-146a slows tumor growth by inhibiting the epithelial-mesenchymal transition process and enhancing the effect of anticancer drugs [17]. Previous studies have demonstrated that miR-146a targets several genes in LC cells, including CCND-1, EGFR, NFKB-1, TRAF6, IL-6, IL-8, and TNFβ. This targeting helps suppress their expression by disrupting NF-kB activity [9, 18,19,20,21,22]. In general, miR-146a affects important signaling pathways such as TNFα, NF-κB, MEK-1/2, and JNK-1/2, which are involved in the regulation of inflammation and cell growth [17]. MiR-146a does this by regulating the genes IRAK1 and TRAF6, which are downstream mediators of the IL-1α and TNFα signaling pathways and creates a negative feedback loop that prevents excessive immune stimulation and inflammation. This mechanism helps to maintain a balance of the immune response and prevents damage caused by chronic inflammation [17, 23]. Moreover, miR-146a-5p targets the 3’UTR of TRAF6 within the NF-κB pathway, leading to apoptosis by regulating the BCL-2 gene [3, 22]. Studies have shown that miR-146a expression in human monocytes is rapidly and dependently increased by NF-κB activation after exposure to mycobacteria and inflammatory stimuli [23]. MiR-146a also plays a role in the induction of endotoxin tolerance in monocytes and regulates the production of TNFα and other inflammatory cytokines [23]. So, in inflammatory states, overexpression of miR-146a increases apoptosis and autophagy, inhibits cell proliferation, and reduces cell migration [14, 17]. In addition to regulating inflammation, miR-146a plays a role in reducing fibrosis in lung and other inflammatory diseases [24]. By inhibiting the activation of NF-κB and TGF-β-related pathways, miR-146a prevents the conversion of fibroblasts into myofibroblasts and the production of extracellular matrix [24].

Carbon nanotubes (CNTs) are known as biocompatible polymers that possess a high capacity for drug and gene (plasmid DNA (pDNA), small interfering RNA, and miRNA) loading [25]. CNT functionalized with polyethyleneimine (PEI) (CNT-PEI) has a high carrying capacity and few side effects on healthy cells. Many studies have explored the various biomedical effects of CNT-PEI in several diseases and cell signaling pathways, including apoptosis and autophagy [26]. Recently, these nanoparticles have been used in the treatment and diagnosis of various diseases, such as breast and pancreatic cancer [27, 28].

In this study, miR-146a was upregulated using the novel CNT-PEI nano delivery system specifically applied to lung cancer and TB cell models. The investigation focused on how the overexpression of miR-146a affects genes such as BCL-2, IL-6, and TNFα via the TRAF6 gene within the NF-κB signaling pathways in LC and TB cell models.

Methods

The experimental protocols of this research were approved by the research committee of the Pasteur Institute of Iran with the ethical code of IR.PII.REC.1400.016.

Lentiviral vector preparation and cultivation

High copy number transformed lentiviral vectors pBON-Lenti-III-miR-eGFP with a length of 885 bp were purchased from Bon Yakhte Company (Fig. 1). They were presented into the DH5α strain of E. coli bacteria, exerting resistance to the Kanamycin antibiotic and its selective marker was Promycin (stored as lyophilized at -70 °C).

To culture, 5 ml (50 μg/ml) of Kanamycin antibiotic and 100 ml of the bacteria were added to the liquid LB culture medium. The bacterial culture was placed in a shaker incubator with a temperature of 37 °C and a speed of 100 rpm for 24 h. Then, bacteria were cultured linearly on LB agar medium, and a colony of the grown bacteria was cultured in 20 ml of liquid LB medium and incubated for 24 h at 37 °C and 100 rpm (Supplementary S1).

Fig. 1

The structure of pBON-Lenti-III-miR-eGFP vector [29]

DNA extraction

After 24 h of incubating bacteria, sediment was prepared from 20 ml of culture medium at 3800 rpm for 25 min. Then, DNA was extracted according to the Favorgen kit (No. FAPDE050) protocol and 50 μl Elution buffer, one-minute incubation at room temperature, and two minutes centrifugation at 18,000 g (Supplementary S2).

Transfection of cell lines

A549 and MRC5 cells were prepared from the national cell bank of the Pasteur Institute of Iran, and washed with PBS and a medium containing (DMEM + FBS 2%). 100 μl PBS and 5 μg of desired plasmid DNA were poured into six 1.5 microtubes and vortexed for one minute. Then, 600 μl PBS was poured into a sterile 1.5 microtube, and 36 μl multi-walled carbon nanotubes- polyethyleneimine (CNT-PEI, 2 mg/ml) was added to it; vortexed for one minute, and incubated for 10 min at room temperature. After that, 100 μl of the solution containing CNT-PEI and PBS was added to the solution containing pDNA (CNT-PEI-pDNA) and vortexed three times for three seconds each time. Subsequently, they were incubated for 25 min at room temperature, the CNT-PEI-pDNA composite was added dropwise to each well and the plate was vortexed well. The plate was placed in an incubator at 37 °C and 4 h after transfection, the medium of cells was removed and a complete medium (DMEM + Pen/Strep + FBS 10%) was added to each well. After 24 and 48 h of incubation, the cells were observed under a fluorescent microscope (Fig. 2). The MOC-2, miR-146a, and control wells (only containing CNT-PEI) were removed for flow cytometry apoptosis assay, and the broth and cell pellet were transferred to -70 °C. Re-transfection was performed following the latest protocol, and apoptosis levels in the cells were assessed after 24 h. To determine the percentage of apoptotic and necrotic cells, the Annexin V-FITC and propidium iodide (PI) double staining method was used [30]. First, the cells were washed with calcium-containing buffer and then incubated with Annexin V-FITC and PI solution for 15 to 30 min in the dark and at room temperature. After staining, the samples were analyzed using flow cytometry analysis software (FlowJo), and four cell groups were separated, including viable cells (Annexin V-/PI-), early apoptosis (Annexin V+/PI-), secondary or late apoptosis (Annexin V+/PI+), and necrotic cells (Annexin V-/PI+).

Cell infection with bacteria (MDR, XDR)

Pulmonary monocyte cells (THP1) were prepared from the national cell bank of the Pasteur Institute of Iran. They were used as a TB-infected model, so THP1 cells were differentiated into macrophages and infested with bacteria (MOI = 10). According to calculations, 70 μL of McFarland’s suspension was added to each well of the cell plate and incubated for 4 h in a 37 °C incubator [31].

RNA extraction, cDNA synthesis, and real-time PCR

RNA was extracted from the samples transfected with miR-146a, MOC P-Lenti, and treated with CNT-PEI (as a pDNA carrier) using a Trizol RNA extraction kit of Sinaclon (No. EX6101).

After RNA extraction, cDNA synthesis was performed to examine gene expression using the Real-Time PCR technique. For this purpose, the Parstous cDNA synthesis protocol (No. A101161) was used. The Master Mix kit (Green qPCR MasterMix 2X) of Yekta Tajiz Azma Super SYBR (No. YT2551) and specific primers (as shown in Table 1) were used to perform the Real-Time PCR process.

All Real-Time PCR reactions were performed in triplicate to ensure reproducibility and accuracy of the results. In addition, GAPDH and U6 were used as housekeeping genes and reference miRNA. Finally, the Melting Curve Analysis and the PCR product were electrophoresed (1% agarose gel) for final confirmation.

Table 1 Primer sequences used for Real-Time -PCR

Statistical analysis

One-way ANOVA method was used for statistical analysis of dose-response tests in each group and cell line using Graph Pad Prism software (version 9.0), and a p-value < 0.05 was considered significant.

Result

Fluorescent microscope observation

As shown in Fig. 2, single pDNA as a control was observed overnight after transfection by CNT-PEI in an acceptable number of cells transfected with miR-146a GFP. However, GFP was observed in a small number of cells transfected with MOC.

Fig. 2

Transfection of cells with CNT-PEI: A and B) miR-146a and C) MOC-Plenti. Acceptable results were observed in cells transfected with miR-146a, while only a small number of cells transfected with MOC showed GFP expression, after 24 h

Electrophoresis results

In the CNT-PEI-pDNA analysis with gel electrophoresis from a dilution of 1/8 to 1, no band was observed, which was considered the binding of pDNA to CNT-PEI (Fig. 3).

Fig. 3

The electrophoresis gel result of: A) miR-146a and MOC pDNA, B) CNT-PEI-pDNA, and single pDNA (as a control). No bands were observed in the CNT-PEI-pDNA analysis through gel electrophoresis for dilutions ranging from 1/8 to 1

Flow cytometry results

The flow cytometry results showed in A549 cell lines; which cells transfected with a miR-146a exhibited 23.06% apoptosis and 1.76% necrosis after 24 h (Fig. 4). The apoptosis rate in these cells increased to 39.7%, after 48 h which was 18.68% higher than the control condition (Fig. 5).

A few MOC-transfected cells were observed in the MRC5 (as control) cell lines after 24 h. However, flow cytometry results showed 8.07% and 27.2% apoptosis and almost 10% necrosis in cells transfected with miR-146a and MOC, respectively (Fig. 6).

Fig. 4

The results of flow cytometry in A549 cell lines showed 23.06% apoptosis and 1.76% necrosis in cells transfected with a plasmid containing miR-146a after 24 h. (Q1: Necrosis, Q2: Late Apoptosis, Q3: Early Apoptosis, Q4: Live Cells)

Fig. 5

The results of apoptosis by flow cytometry in A549 cell lines showed 39.7% apoptosis and 9.3% necrosis in cells transfected with miR-146a, while 21.02% apoptosis and 2.76% necrosis were observed in the control state after 48 h. (Q1: Necrosis, Q2: Late Apoptosis, Q3: Early Apoptosis, Q4: Live Cells)

Fig. 6

The flow cytometry results in MRC5 cell lines showed 8.07% apoptosis and 10.9% necrosis in cells transfected with miR-146a, compared to 27.2% apoptosis and 10.3% necrosis in cells transfected with MOC, after 48 h. (Q1: Necrosis, Q2: Late Apoptosis, Q3: Early Apoptosis, Q4: Live Cells)

Results of examining BCL-2 and TRAF6 genes and miR-146a in A549 cell line

Based on Fig. 7, the results regarding BCL-2 gene expression in A549 cells showed that the expression of the sample transfected with miR-146a, CNT-PEI, and P-Lenti, had increased compared to the control sample (non-transfected cell), respectively. Regarding TRAF6 gene expression, the results show that gene expression increased in the P-Lenti sample, while decreased in samples transfected with miR-146a and CNT-PEI compared to the control sample. A statistically significant difference was observed between the control and transfected groups (p-value < 0.001).

Fig. 7

In A549 cells, BCL-2 expression was increased in samples transfected with miR-146a, P-Lenti, and CNT-PEI compared to the control group. Whereas, TRAF6 expression was higher in P-Lenti samples but decreased in samples transfected with miR-146a and CNT-PEI compared to the control group (BCL2 and TRAF6 p-value = 0.0009 and 0.0001). From left to right: control sample (non-transfected cells), sample transfected with miR-146a, P-Lenti, and CNT-PEI

Results of examining BCL-2 and TRAF6 genes and miR-146a in the MRC5 cell line

After evaluating the results of the expression of BCL-2 and TRAF6 genes in the A549 cell line, the expression of these genes in the MRC5 cell line was also examined. In the MRC5 cell line, the expression levels of BCL-2 and TRAF6 genes were higher in samples transfected with CNT-PEI, P-Lenti, and miR-146a compared to the control sample. Moreover, the expression of each gene was higher in the cells transfected with CNT-PEI with a significant difference (p-value < 0.001) (Fig. 8).

Fig. 8

In MRC5 cells, BCL-2 and TRAF6 expression was higher in the CNT-PEI, P-Lenti, and miR-146a transfected groups compared to the control group (**** means p-value < 0.001). From left to right: control sample (non-transfected cells), sample transfected with miR-146a, P-Lenti, and CNT-PEI

Results of examining IL-6 and TNFα genes and miR-146a in THP1 cell line

According to Fig. 9, the fold change expression of the IL-6 and TNFα genes in THP1 cells decreased in those transfected with miR-146, P-Lenti, and CNT-PEI compared to non-transfected control cells, which exhibited the highest expression. A significant difference was found between the two groups (p-value < 0.001).

Fig. 9

The expression of the IL-6 and TNFα genes in the THP1 cell line decreased in cells transfected with miR-146, P-Lenti, and CNT-PEI compared to non-transfected cells (**** indicates p-value < 0.001). From left to right: control sample (non-transfected cells), sample transfected with miR-146a, P-Lenti, and CNT-PEI

Discussion

In this study, miR-146a transfection was well performed in different lung cell lines (A549, MRC5, and THP1). The results indicated that miR-146a overexpression causes apoptosis in LC and promotes autophagy in TB by targeting the TRAF6 gene, which influences genes such as BCL-2, IL-6, and TNFα through the NF-κB signaling pathway. Furthermore, the results of this study have demonstrated the complex and multifaceted role of TRAF6 and BCL-2 in regulating apoptosis and cell survival processes in different cell lines. In cancer cells, increased expression of TRAF6 can activate survival pathways and inhibit apoptosis, while its inhibition leads to increased apoptosis and decreased cell growth [32]. These findings are consistent with these results that TRAF6 expression in A549 cells was associated with increased apoptosis. The different results in TRAF6 and BCL-2 expression between A549 and MRC5 cells may be due to specific and biological differences of each cell, which requires further investigation at the signaling levels and pathways [32, 33].

In previous studies, the CNT nano delivery system has been used as a non-viral carrier in various cells, especially cancer, which has increased apoptosis, necrosis, and ultimately treatment [34, 35]. The CNT-PEI transfection system achieves DNA transfection and gene expression enhancement through the proton sponge effect [36]. This system has had many clinical applications for gene delivery, monoclonal antibodies, oligonucleotide, small interfering RNA, miRNA, etc. into the cell cytoplasm [37,38,39]. For example, Masotti, A. et al. [40] 2016 showed that CNT-PEI can deliver miRNA-503 into mouse endothelial cells and improve gene expression regulation and cell function. In another study, single-walled carbon nanotubes were non-covalently coated with PEI-SA (CNT-PEI-SA), and local delivery of siRNA to the skin of a melanoma mouse model was performed. This resulted in reduced tumor growth, low toxicity during delivery, and protection of siRNA from degradation in vivo models [41]. Zhang et al. [42] injected fluorescently labeled CNT-DNA conjugates into tumor-bearing mice and found that almost all tumor cells took up the conjugates.

A review of numerous studies showed that overexpression of miR-146 can activate the immune system. For example, studies on osteoarthritis (OA) showed that the expression of miR-146a-5p in the cartilage tissue of patients with OA inhibiting the expression of TRAF6 and suppressing the activation of the NF-κB signaling pathway can increase cell apoptosis [43]. In addition, overexpression of miR-146a in hepatocellular carcinoma cells inhibits proliferation and invasion and increases apoptosis by targeting TRAF6 [44]. Others had reported that miR-146a-5p was upregulated in pancreatic islets treated with proinflammatory cytokines and was associated with β-cell apoptosis and impaired insulin secretion [14]. Overexpression of miR-146a in NK/T cell lymphoma (SNK6 and YT) resulted in inhibition of NF-κB and TRAF6 activity, decreased cell proliferation, induced apoptosis, and increased chemosensitivity [45]. In a study conducted by Hu, Q. et al. [19], it was observed that the overexpression of miR-146a increases the survival of cervical cancer cells through the reduction of IRAK1 and TRAF6. Another study exhibited that miR-146a and BCL-2 increase hypoxia-induced autophagy [21].

MiR-146 by targeting genes such as IRAK1, TRAF6, TNFα, BCL-2, PTEN, KRAS, IL-6, and MAPK1 can control autophagy and apoptosis in TB and LC through NF-κB, PI3K/AKT, and MAPK pathways [3, 16]. Previous studies showed that miR-146a acts as an anti-cancer agent in LC by targeting EGFR, TGF-β, and NF-κB signaling, as well as IRAK-1, ATG-12, TRAF6, BCL-2, and JNK-2 genes [46,47,48]. In addition, more expression of miR-146a in NSCLC suppressed cell growth, inhibited cell migration, and induced cell apoptosis [22, 49, 50]. Chen et al. [46] demonstrated that upregulation of miR-146a significantly inhibited EGFR downstream signaling in NSCLC cell lines. The overexpression of miR-146a can prevent the expression of MIF through gene targeting; thereby inhibiting the proliferation of A549 cells and inducing apoptosis of cancer cells [51].

MiRNAs are associated with inflammatory responses by regulating the replication of TB and inducing pathogenesis by targeting the TRAF-6 signaling pathway [52]. Overexpression of miR-146a can modulate the inflammatory response by targeting TNF6 and IRAK1 [53]. Alijani E et al. [54] found that miR-146a and miR-155 were increased in people with TB compared to healthy people, and regulated the inflammatory response to reduce tissue damage infected with MTB. Liu Z. et al. [23] showed that overexpression of miR-146a enhances the killing ability of THP1 cells against intracellular M. bovis BCG and reduces the expression of the TNFα gene. Therefore, upregulation of miR-146 using CNT-PEI may have great potential for the treatment of TB and LC.

Conclusion

This study demonstrates the successful transfection of miR-146a into A549, TB-infected macrophages (THP1), and MRC5 using the CNT-PEI nano delivery system. Overexpression of miR-146a modulated key genes involved in apoptosis, autophagy, and inflammatory responses, specifically targeting TRAF6 and affecting BCL-2, IL-6, and TNF-α through the NF-κB signaling pathway. These findings highlight the critical regulatory role of miR-146a in the pathogenesis of TB and LC. These therapies can potentially enhance the efficacy and safety of various treatments. However, further research and development is needed to optimize this approach and identify the most effective miRNA targets. In addition, further studies are needed to investigate any potential off-target effects of these therapies and to assess their long-term safety and efficacy in clinical settings. Also, it is crucial to examine the balance between stimulatory and inhibitory factors in determining cell survival or death. Ultimately, with the advancements in nanotechnology and its application in miRNA-based therapies, this approach could revolutionize the treatment of various diseases and lead to better outcomes for patients.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Sakhi H, Arabi M, Ghaemi A, Movafagh A, Sheikhpour M. Oncolytic viruses in lung cancer treatment: a review Article. Immunotherapy. 2024;16(2):75–97.

    Article PubMed CAS Google Scholar

  2. Abolfathi H, Sheikhpour M, Mohammad Soltani B, Fahimi H. The comparison and evaluation of the miR-16, miR-155 and miR-146a expression pattern in the blood of TB and NSCLC patients: a research paper. Gene Rep. 2021;22:100967.

    Article CAS Google Scholar

  3. Abolfathi H, Arabi M, Sheikhpour M. A literature review of MicroRNA and gene signaling pathways involved in the apoptosis pathway of lung cancer. Respir Res. 2023;24(1):55.

    Article PubMed PubMed Central CAS Google Scholar

  4. Zhang XL, Dang YW, Li P, Rong MH, Hou XX, Luo DZ, et al. Expression of tumor necrosis factor receptor-associated factor 6 in lung cancer tissues. Asian Pac J Cancer Prevention: APJCP. 2014;15(24):10591–6.

    Article Google Scholar

  5. Starczynowski DT, Lockwood WW, Deléhouzée S, Chari R, Wegrzyn J, Fuller M, et al. TRAF6 is an amplified oncogene bridging the RAS and NF-κB pathways in human lung cancer. J Clin Investig. 2011;121(10):4095–105.

    Article PubMed PubMed Central CAS Google Scholar

  6. Yang L, Zhuang L, Ye Z, Li L, Guan J, Gong W. Immunotherapy and biomarkers in patients with lung cancer with tuberculosis: recent advances and future directions. iScience. 2023;26(10):107881.

    Article PubMed PubMed Central CAS Google Scholar

  7. Bhowmik S, Mohanto NC, Sarker D, Sorove AA. Incidence and risk of lung cancer in tuberculosis patients, and vice versa: A literature review of the last decade. Biomed Res Int. 2022;2022:1702819.

    Article PubMed PubMed Central Google Scholar

  8. Racanelli AC, Kikkers SA, Choi AMK, Cloonan SM. Autophagy and inflammation in chronic respiratory disease. Autophagy. 2018;14(2):221–32.

    Article PubMed PubMed Central Google Scholar

  9. Shao J, Ding Z, Peng J, Zhou R, Li L, Qian Q, et al. MiR-146a-5p promotes IL-1β-induced chondrocyte apoptosis through the TRAF6-mediated NF-kB pathway. Inflamm Res. 2020;69(6):619–30.

    Article PubMed CAS Google Scholar

  10. Ying H, FengYing S, YanHong W, YouMing H, FaYou Z, HongXiang Z, et al. MicroRNA-155 from sputum as noninvasive biomarker for diagnosis of active pulmonary tuberculosis. Iran J Basic Med Sci. 2020;23(11):1419–25.

    PubMed PubMed Central Google Scholar

  11. Ruan K, Fang X, Ouyang G, MicroRNAs. Novel regulators in the hallmarks of human cancer. Cancer Lett. 2009;285(2):116–26.

    Article PubMed CAS Google Scholar

  12. M K, S S, S M. Expression levels of candidate circulating MicroRNAs in pediatric tuberculosis. Pathogens Global Health. 2020;114(5):262–70.

  13. Bonafé GA, Boschiero MN, Sodré AR, Ziegler JV, Rocha T, Ortega MM. Natural plant compounds: does caffeine, dipotassium glycyrrhizinate, curcumin, and euphol play roles as antitumoral compounds in glioblastoma cell lines?? Front Neurol. 2021;12:784330.

    Article PubMed Google Scholar

  14. Krishnan P, Branco RCS, Weaver SA, Chang G, Lee CC, Syed F, et al. miR-146a-5p mediates inflammation-induced β cell mitochondrial dysfunction and apoptosis. bioRxiv: the preprint server for biology. 2024.

  15. Mao S, Wu J, Yan J, Zhang W, Zhu F. Dysregulation of miR-146a: a causative factor in epilepsy pathogenesis, diagnosis, and prognosis. Front Neurol. 2023;14:1094709.

    Article PubMed PubMed Central Google Scholar

  16. Sheikhpour M, Abolfathi H, Karimipoor M, Movafagh A, Shahsavani M. The common MiRNAs between tuberculosis and Non-Small cell lung cancer: A critical review. Tanaffos. 2021;20(3):197–208.

    PubMed PubMed Central Google Scholar

  17. Wani JA, Majid S, Khan A, Arafah A, Ahmad A, Jan BL, et al. Clinico-Pathological importance of miR-146a in lung cancer. Diagnostics (Basel). 2021;11(2).

  18. Li YL, Wang J, Zhang CY, Shen YQ, Wang HM, Ding L, et al. MiR-146a-5p inhibits cell proliferation and cell cycle progression in NSCLC cell lines by targeting CCND1 and CCND2. Oncotarget. 2016;7(37):59287–98.

    Article PubMed PubMed Central Google Scholar

  19. Hu Q, Song J, Ding B, Cui Y, Liang J, Han S. miR-146a promotes cervical cancer cell viability via targeting IRAK1 and TRAF6. Oncol Rep. 2018;39(6):3015–24.

    PubMed CAS Google Scholar

  20. https://mpd.bioinf.uni-sb.de/mirnas.html

  21. Zhang F, Wang J, Chu J, Yang C, Xiao H, Zhao C, et al. MicroRNA-146a induced by hypoxia promotes chondrocyte autophagy through Bcl-2. Cell Physiol Biochem. 2015;37(4):1442–53.

    Article PubMed CAS Google Scholar

  22. Liu X, Liu B, Li R, Wang F, Wang N, Zhang M, et al. miR-146a-5p plays an oncogenic role in NSCLC via suppression of TRAF6. Front Cell Dev Biology. 2020;8:847.

    Article Google Scholar

  23. Liu Z, Zhou G, Deng X, Yu Q, Hu Y, Sun H, et al. Analysis of MiRNA expression profiling in human macrophages responding to Mycobacterium infection: induction of the immune regulator miR-146a. J Infect. 2014;68(6):553–61.

    Article PubMed Google Scholar

  24. McMillan DH, Woeller CF, Thatcher TH, Spinelli SL, Maggirwar SB, Sime PJ et al. Attenuation of inflammatory mediator production by the NF-κB member RelB is mediated by microRNA-146a in lung fibroblasts. 2013;304(11):L774–81.

  25. Sheikhpour M, Golbabaie A, Kasaeian A. Carbon nanotubes: A review of novel strategies for cancer diagnosis and treatment. Mater Sci Engineering: C. 2017;76:1289–304.

    Article CAS Google Scholar

  26. Kamazani FM, Sotoodehnejad nematalahi F, Siadat SD, Pornour M, Sheikhpour M. A success targeted nano delivery to lung cancer cells with multi-walled carbon nanotubes conjugated to Bromocriptine. Sci Rep. 2021;11(1):24419.

    Article PubMed PubMed Central CAS Google Scholar

  27. Zomorodbakhsh S, Abbasian Y, Naghinejad M, Sheikhpour M. The effects study of Isoniazid conjugated multi-wall carbon nanotubes nanofluid on Mycobacterium tuberculosis. Int J Nanomed. 2020;15:5901–9.

    Article CAS Google Scholar

  28. Taghavi S, HashemNia A, Mosaffa F, Askarian S, Abnous K, Ramezani M. Preparation and evaluation of polyethylenimine-functionalized carbon nanotubes tagged with 5TR1 aptamer for targeted delivery of Bcl-xL ShRNA into breast cancer cells. Colloids Surf B. 2016;140:28–39.

    Article CAS Google Scholar

  29. https: //bonbiotech.ir/ /pbon-lenti-iii-mir-egfp/

  30. Lakshmanan I, Batra SK. Protocol for apoptosis assay by flow cytometry using Annexin V staining method. Bio-protocol. 2013;3(6).

  31. Sheikhpour M, Shokrgozar MA, Biglari A, Pornour M, Abdolrahimi F, Poorazar Dizaji S, et al. Gene expression and in vitro Pharmacogenetic studies of dopamine and serotonin gene receptors in tuberculosis. Tanaffos. 2021;20(2):126–33.

    PubMed PubMed Central Google Scholar

  32. Guo Y, Zhang X, Li J, Zhou Z, Zhu S, Liu W, et al. TRAF6 regulates autophagy and apoptosis of melanoma cells through c-Jun/ATG16L2 signaling pathway. MedComm. 2023;4(4):e309.

    Article PubMed PubMed Central CAS Google Scholar

  33. Xu H, Li L, Dong B, Lu J, Zhou K, Yin X et al. TRAF6 promotes chemoresistance to Paclitaxel of triple negative breast cancer via regulating PKM2-mediated Glycolysis. 2022.

  34. Fahira AI, Amalia R, Barliana MI, Gatera VA, Abdulah R. Polyethyleneimine (PEI) as a Polymer-Based Co-Delivery system for breast cancer therapy. Breast Cancer (Dove Med Press). 2022;14:71–83.

    PubMed CAS Google Scholar

  35. Moradian H, Fasehee H, Keshvari H, Faghihi S. Poly(ethyleneimine) functionalized carbon nanotubes as efficient nano-vector for transfecting mesenchymal stem cells. Colloids Surf B. 2014;122:115–25.

    Article CAS Google Scholar

  36. Nunes A, Amsharov N, Guo C, Van den Bossche J, Santhosh P, Karachalios TK, et al. Hybrid polymer-grafted multiwalled carbon nanotubes for in vitro gene delivery. Small. 2010;6(20):2281–91.

    Article PubMed CAS Google Scholar

  37. Wu H, Shi H, Zhang H, Wang X, Yang Y, Yu C, et al. Prostate stem cell antigen antibody-conjugated multiwalled carbon nanotubes for targeted ultrasound imaging and drug delivery. Biomaterials. 2014;35(20):5369–80.

    Article PubMed CAS Google Scholar

  38. Boussif O, Lezoualc’h F, Zanta MA, Mergny MD, Scherman D, Demeneix B, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci USA. 1995;92(16):7297–301.

    Article PubMed PubMed Central CAS Google Scholar

  39. Zhang QY, Ho PY, Tu MJ, Jilek JL, Chen QX, Zeng S, et al. Lipidation of polyethylenimine-based polyplex increases serum stability of bioengineered RNAi agents and offers more consistent tumoral gene knockdown in vivo. Int J Pharm. 2018;547(1–2):537–44.

    Article PubMed PubMed Central CAS Google Scholar

  40. Masotti A, Miller MR, Celluzzi A, Rose L, Micciulla F, Hadoke PW, et al. Regulation of angiogenesis through the efficient delivery of MicroRNAs into endothelial cells using polyamine-coated carbon nanotubes. Nanomedicine: Nanatechnol Biology Med. 2016;12(6):1511–22.

    Article CAS Google Scholar

  41. Siu KS, Chen D, Zheng X, Zhang X, Johnston N, Liu Y, et al. Non-covalently functionalized single-walled carbon nanotube for topical SiRNA delivery into melanoma. Biomaterials. 2014;35(10):3435–42.

    Article PubMed CAS Google Scholar

  42. Zhang Z, Yang X, Zhang Y, Zeng B, Wang S, Zhu T, et al. Delivery of telomerase reverse transcriptase small interfering RNA in complex with positively charged Single-Walled carbon nanotubes suppresses tumor growth. Clin Cancer Res. 2006;12(16):4933–9.

    Article PubMed CAS Google Scholar

  43. Zhong J-H, Li J, Liu C-F, Liu N, Bian R-X, Zhao S-M, et al. Effects of microRNA-146a on the proliferation and apoptosis of human osteoarthritis chondrocytes by targeting TRAF6 through the NF-κB signalling pathway. Biosci Rep. 2017;37(2).

  44. Zu Y, Yang Y, Zhu J, Bo X, Hou S, Zhang B, et al. MiR-146a suppresses hepatocellular carcinoma by downregulating TRAF6. Am J Cancer Res. 2016;6(11):2502–13.

    PubMed PubMed Central CAS Google Scholar

  45. Paik JH, Jang JY, Jeon YK, Kim WY, Kim TM, Heo DS, et al. MicroRNA-146a downregulates NFκB activity via targeting TRAF6 and functions as a tumor suppressor having strong prognostic implications in NK/T cell lymphoma. Clin Cancer Research: Official J Am Association Cancer Res. 2011;17(14):4761–71.

    Article CAS Google Scholar

  46. Yuan F, Zhang S, Xie W, Yang S, Lin T, Chen X. Effect and mechanism of miR-146a on malignant biological behaviors of lung adenocarcinoma cell line. Oncol Lett. 2020;19(6):3643–52.

    PubMed PubMed Central CAS Google Scholar

  47. Pang L, Lu J, Huang J, Xu C, Li H, Yuan G, et al. Upregulation of miR-146a increases cisplatin sensitivity of the non-small cell lung cancer A549 cell line by targeting JNK-2. Oncol Lett. 2017;14(6):7745–52.

    PubMed PubMed Central Google Scholar

  48. Wu Z, Lu H, Sheng J, Li L. Inductive microRNA-21 impairs anti-mycobacterial responses by targeting IL-12 and Bcl-2. FEBS Lett. 2012;586(16):2459–67.

    Article PubMed CAS Google Scholar

  49. Iacona JR, Lutz CS. miR-146a-5p: expression, regulation, and functions in cancer. Wiley Interdiscip Rev RNA. 2019;10(4):e1533.

    Article PubMed Google Scholar

  50. Chen G, Umelo IA, Lv S, Teugels E, Fostier K, Kronenberger P, et al. miR-146a inhibits cell growth, cell migration and induces apoptosis in non-small cell lung cancer cells. PLoS ONE. 2013;8(3):e60317.

    Article PubMed PubMed Central CAS Google Scholar

  51. Wang W-M, Liu J-C. Effect and molecular mechanism of mir-146a on proliferation of lung cancer cells by targeting and regulating MIF gene. Asian Pac J Trop Med. 2016;9(8):806–11.

    Article PubMed CAS Google Scholar

  52. Chauhan D, Davuluri KS. MicroRNAs associated with the pathogenesis and their role in regulating various signaling pathways during Mycobacterium tuberculosis infection. Front Cell Infect Microbiol. 2022;12.

  53. Li S, Yue Y, Xu W, Xiong S. MicroRNA-146a represses mycobacteria-induced inflammatory response and facilitates bacterial replication via targeting IRAK-1 and TRAF-6. PLoS ONE. 2013;8(12):e81438.

    Article PubMed PubMed Central Google Scholar

  54. Alijani E, Rad FR, Katebi A, Ajdary S. Differential expression of miR-146 and miR-155 in active and latent tuberculosis infection. Iran J Public Health. 2023;52(8):1749–57.

    PubMed PubMed Central Google Scholar

Download references

Funding

No financial support is relevant to this study.

Author information

Authors and Affiliations

  1. Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran (IPI), No. 69, Pasteur Ave, Tehran, 1316943551, Iran

    Mojgan Sheikhpour, Hanie Sakhi & Seyed Ali Nojoumi

  2. Departments of Biosciences, University of Milano, Via Celoria 26, Milan, I-30133, Italy

    Mobina Maleki

  3. Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran

    Abolfazl Movafagh

  4. National Cell Bank, Pasteur Institute of Iran, Tehran, Iran

    Leila Ghazizadeh

Authors
  1. Mojgan Sheikhpour
  2. Mobina Maleki
  3. Hanie Sakhi
  4. Abolfazl Movafagh
  5. Seyed Ali Nojoumi
  6. Leila Ghazizadeh

Contributions

Mojgan Sheikhpour: Conceptualization, Methodology, Validation, Resources, Data curation, Formal analysis, Writing – original draft, Writing – review & editing, Supervision, Project administration. Mobina Maleki: Conceptualization, Methodology, Validation, Data curation. Hanie Sakhi: Writing – original draft. Seyed Ali Nojoumi: Review & editing. Leila Ghazizadeh: Methodology.

Corresponding author

Correspondence to Mojgan Sheikhpour.

Ethics declarations

Ethical approval

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

About this article

Cite this article

Sheikhpour, M., Maleki, M., Sakhi, H. et al. Nano delivery of MiR-146a and its effect study on genes involved in apoptosis and autophagy pathways in lung cancer and tuberculosis. BMC Biotechnol 25, 81 (2025). https://doi.org/10.1186/s12896-025-01019-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Version of record:

  • DOI: https://doi.org/10.1186/s12896-025-01019-8

Keywords

BMC Biotechnology

ISSN: 1472-6750

Contact us

AltStyle によって変換されたページ (->オリジナル) /