Everolimus targets mTORC1 inducing NSCLC cells to secrete miR-7-5p-loaded exosomes in Rab27A and Rab27B dependent manners
In multiple NSCLC cell lines, including A549, H358, H520 and SPC-A1, the level of miR-7-5p was significantly down-regulated after Everolimus exposing. And it lasted more than 48 hours without changing of autophagy in NSCLC cell lines (Fig. 1a and S1a). In order to find the reason for the reduction of miR-7-5p, we further analyzed the lung squamous cell carcinoma mRNA data of TCGA, and divided the data into two groups named mTORlow group and mTORhigh group according to the mean value of mTOR level in the samples. The GSEA on the mRNA profiles changes revealed negative associations between mTOR and gene sets involved in extracellular exosomes (Fig. 1b). Consequently, we focused on the exosomes derived from NSCLC cells in the culture medium. They were isolated and identified by transmission electron microscope (TEM) (Fig. 1c) and NanoSight Analysis (Fig. 1d). The size and morphology of exosomes were similar between two groups, presented as 50-100nm round like. It was also found the fluorescent labelled exosomal marker CD63 in the cytoplasm was significantly weakened after Everolimus treating (Fig. 1e). Meanwhile, expression of exosomal markers HSP70, TSG101, CD63 and CD9 significantly enriched in exosomes accompanied by down-regulation of intracellular exosomal markers in A549 cells by Western blotting (Fig. 1f). To further determine whether there were association between the declination of intracellular miR-7-5p and elevation of release of exosomes induced by Everolimus, we used exosomes inhibitor GW4869 to block the release of exosomes. When the secretion of exosomes was inhibited by GW4869, miR-7-5p distribution was reversed (Fig. 1g), which indicated that Everolimus induced the secretion of miR-7-5p-loaded exosomes into the surrounding extracellular environment, resulting in the loss of miR-7-5p in intracellular.
Furthermore, we performed siTSC1/2 to active 13 but Everolimus to inhibit the activity of mTORC1. After confirming the expression of mTOR, TSC1 and TSC2 following indicated treatments (Figure S1b), we discovered miR-7-5p was more enriched in exosomes than in intracellular when Everolimus was employed. On the contrary, miR-7-5p located in intracellular rather than in exosomes when siTSC1/2 was performed. When Everolimus was employed in siTSC1/2-A549 cells, miR-7-5p would be enriched in exosomes again (Fig. 1h). It confirmed that the activity of mTORC1 controlled the redistribution of miR-7-5p.
Rab27A and Rab27B are two closely related Rab small GTPases, which play key roles in the secretion of exosomes in many types of cells14. The knockdown of Rab27A and Rab27B by siRNA would reduce exosome secretion of HeLa cells. And Rapamycin targeting mTORC1 stimulated the release of exosome dependent on Rab27A 7. Here, we found that the release of miR-7-5p-loaded exosomes was significantly reduced to almost undetectable when Rab27A or Rab27B was knocked down (Figure S1c), accompanied by the enrichment of intracellular miR-7-5p. When Everolimus were performed after Rab27A or Rab27B knocking down, the exocytosis of miR-7-5p loaded exosomes was also suppressed (Fig. 1i). This revealed that the release of miR-7-5p-loaded exosomes induced by Everolimus depended on Rab27A and Rab27B.
Loss of intracellular miR-7-5p induces phosphorylation of MNK/eIF4E axis, but supplement of extra exosomal miR-7-5p can reverse it
With the gradual deepening of understanding, it had been found that there were feedbacks of multiple kinases during the employment of Everolimus, such as hyperphosphorylation of eIF4E at the Ser209 site, which led to the development of drug resistance15. The mTOR inhibitor Rapalog induced eIF4E phosphorylation was done by kinases termed MAP kinase-interacting kinases (Mnks) independently of the conventional Mnk-activating MAPK pathway16. Coincidentally, MNK1 was a potential targeted gene of miR-7-5p by bioinformatics analysis (Pictar, TargetScan, and Tarbase) (Fig. 2a). It could directly bind to the 3'UTR of mRNA of MNK1 by the luciferase reporter gene test (Fig. 2b). When miR-7-5p was elevated by mimics, the MNK1 would be down-regulated. On the contrary, inhibition of miR-7-5p by inhibitor could up-regulate the MNK1 as Figure S1d & 2c. The two NSCLC cell lines A549 and SPC-A1 were utilized to conduct the following experiments due to that miR-7-5p level was lower but MNK1 expression was higher compared with the human bronchial epithelial (HBE) cell and other NSCLC cell lines (Figure S1e). Therefore, a lentivirus-based system (LV-miR-7-5p and LV-NC) was used to conduct stable miR-7-5p overexpressed A549 and SPC-A1 cell lines. When Everolimus posed on stable miR-7-5p A549 and SPC-A1 cells, overexpressed miR-7-5p was able to inhibit Everolimus induced upregulation of MNK1 and p-eIF4ES209 without affection of downstream of mTOR, including p-S6S235/236 and p-4EBP1Thr37/46 (Fig. 2d). Simultaneously, Considering higher miR-7-5p can inhibit protein ubiquitinal degradation 17, and MNK1 protein can be degraded by ubiquitination in addition to be regulated by phosphorylation18, the stable miR-7-5p and control A549 cells were treated with protein synthesis inhibitor cycloheximide (CHX) to calculate the half-life of MNK1 protein. As show in Fig. 2e and 2f, the MNK1 protein decreased by ~ 50% within 7.785 hours in the control A549 cells, and 7.278 hours in the stable miR-7-5p A549 cells. The half-life of the MNK1 protein between the two groups was relatively close, which indicated that miR-7-5p had only few impacts on MNK1 protein stability. So, it could completely inhibit the activation of the MNK/eIF4E axis induced by Everolimus. Hence, we tried to use miR-7-5p-loaded exosomes as a candidate therapy. Here, we used stable miR-7-5p overexpressed A549 cells derived exosomes. They were able to be internalized by the A549 cells (Fig. 2g), and the rate of internalization is greater than the rate of secretion induced by Everolimus (Fig. 2h). The same as stable upregulation of miR-7-5p, they suppressed the phosphorylation of MNK/eIF4E axis without activation of downstream of mTOR (Fig. 2i), which suggested that miR-7-5p-loaded exosomes derived from tumor cells might be served as a new mechanism to alleviate the resistance of Everolimus.
The declined miR-7-5p and elevated MNK1 are associated with poor prognosis of NSCLC
To investigate the clinical significance of miR-7-5p and MNK1 in NSCLC specimens, we firstly used qPCR confirmed that the levels of miR-7-5p declined in cancer tissues compared to paired adjacent tissues (Fig. 3a), while the MNK1 levels significantly elevated in cancer tissues rather than paired adjacent tissues (Fig. 3b) in 34 pairs fresh tissues. There was a negative correlation between the mRNA levels of miR-7-5p and MNK1 in NSCLC (Fig. 3c, R=-0.3004, P = 0.0128). Then, we performed the in-situ hybridization and immunohistochemistry in 318 cases of paraffin-embedded NSCLC tissues (including 161 cases of lung adenocarcinoma and 157 cases of lung squamous cell carcinoma) and 90 cases of adjacent tissues respectively to further detect miR-7-5p and MNK1. Positive miR-7-5p and MNK1 were mainly located in the cytoplasm (Fig. 3d). Strongly positive miR-7-5p was observed in normal alveolar epithelial cells, accompanied by weak staining of MNK1. In adenocarcinoma and squamous cell carcinoma tissues, miR-7-5p was declined, while MNK1 was elevated (Fig. 3e).
Univariate analysis showed that lower miR-7-5p was more prone to occur lymph node metastasis (LNM) (P = 0.042, Table 1). And compared with the primary focus, the levels of miR-7-5p were lower in metastases (Fig. 3f). Besides, lower miR-7-5p (P = 0.048) or positive MNK1 (P = 0.007) had poor survival status (Table 1). Higher miR-7-5p combined with negative MNK1 would behave as a better survival status (P = 0.004) (Table 1). We also calculated OS for NSCLC patients with lower miR-7-5p, higher MNK1 and the combined index of miR-7-5p and MNK1 respectively (Fig. 3g). NSCLC patients tended to have poor prognosis if miR-7-5p was lower (Fig. 3g, upper), or MNK was positive (Fig. 3g, middle). Optimal outcomes were observed if combined higher miR-7-5p and negative MNK1 was positive. But lower miR-7-5p and positive MNK1 coexist with a herald of the worst prognosis. And the prognosis was intermediate in other expression patterns with no significant statistical difference (Fig. 3g, lower). Multivariate regression analysis showed that besides LNM status, clinical stages, gender and pathological grades, miR-7-5p and MNK1 are also independent prognostic factors for NSCLC (Table 2). Correlation analysis discovered that there was a significant negative correlation between the miR-7-5p and MNK1 (Table 3, r=-0.143, P = 0.011). The human NSCLC clinical data supported the concept that miR-7-5p appeared as a tumor suppressor, but MNK1 had an oncogenic function. The balance of the two biomarkers plays an important role in the pathogenesis of NSCLC. Therefore, restoring the levels of miR-7-5p not only has a tumor suppressor effect, but also reverses the phosphorylation of the MNK/eIF4E axis.
Table 1
Analysis of the association between expression of hsa-miR-7-5p and MNK and clinicopathological features of NSCLC (n = 318).
Clinicopathological
features (n)
|
miR-7-5p
|
MNK
|
miR-7-5p/MNK#
|
Low (%)
|
High (%)
|
P-value
|
Po (%)
|
Ne (%)
|
P-value
|
N− (%)
|
P+ (%)
|
P-value
|
Age (years)
|
|
|
|
|
|
|
|
|
|
≤ 55 (n = 149)
|
70 (49.3)
|
72 (50.7)
|
0.859
|
68 (47.9)
|
74 (53.1)
|
0.740
|
38 (26.8)
|
104 (73.2)
|
0.159
|
> 55 (n = 169)
|
85(48.3)
|
91 (51.7)
|
81 (46.0)
|
95 (54.0)
|
60 (34.1)
|
116 (65.9)
|
Gender
|
|
|
|
|
|
|
|
|
|
Male (n = 241)
|
116 (48.1)
|
125(51.9)
|
0.701
|
108 (44.8)
|
133 (55.2)
|
0.197
|
79 (32.8)
|
162 (67.2)
|
0.180
|
Female (n = 77)
|
39 (50.6)
|
38 (49.4)
|
41 (53.2)
|
36 (46.8)
|
19 (24.7)
|
58 (75.3)
|
LNM status
|
|
|
|
|
|
|
|
|
|
NO LNM (n = 131)
|
54 (41.9)
|
75 (58.1)
|
0.042*
|
55 (42.6)
|
74 (57.4)
|
0.213
|
47 (36.4)
|
82 (63.6)
|
0.073
|
LNM (n = 187)
|
101 (53.4)
|
88 (46.6)
|
94 (49.7)
|
95 (50.3)
|
51 (27.0)
|
138 (73.0)
|
Differentiation
|
|
|
|
|
|
|
|
|
|
Well/Moderate (n = 145)
|
71 (49.0)
|
74 (51.0)
|
0.942
|
74 (51.0)
|
71 (49.0)
|
0.172
|
42 (29.0)
|
103 (71.0)
|
0.513
|
Poor (n = 173)
|
84 (48.6)
|
89 (51.4)
|
75 (43.4)
|
98 (56.6)
|
56 (32.4)
|
117 (67.6)
|
Clinical stages
|
|
|
|
|
|
|
|
|
|
StageⅠ-Ⅱ (n = 166)
|
76 (45.8)
|
90(54.2)
|
0.270
|
77 (46.4)
|
89 (53.6)
|
0.861
|
56 (33.7)
|
110 (66.3)
|
0.239
|
Stage Ⅲ (n = 152)
|
79 (52.0)
|
73 (48.0)
|
72 (47.4)
|
80 (52.6)
|
42 (27.6)
|
110 (72.4)
|
Survival status
|
|
|
|
|
|
|
|
|
|
Alive (n = 210)
|
94(44.8)
|
116(55.2)
|
0.048*
|
87 (41.4)
|
123 (58.6)
|
0.007**
|
76 (36.2)
|
134 (63.8)
|
0.004**
|
Dead (n = 108)
|
61 (56.5)
|
47 (43.5)
|
62 (57.4)
|
46 (42.6)
|
22 (20.4)
|
86 (79.6)
|
*Chi-square test, statistically significant difference (*P < 0.05, **P < 0.01). |
Abbreviations: ADC, adenocarcinoma; H, High expression; L, Low expression; Po, Positive expression; Ne, Negative expression; LNM, lymph node metastasis; NSCLC: non-small cell lung cancer; SCC, squamous cell carcinoma; miR-7-5p/MNK#, N−, the higher miR-7-5p combined with negative expression of MNK, |
P+, other combination of expression of these two factors. |
Table 2
Summary of multivariate of Cox proportional regression for overall survival in 318 cases of NSCLC.
Parameter
|
B
|
SE
|
Wald
|
Sig.
|
Exp (B)
|
95.0% CI for Exp (B)
|
Lower
|
upper
|
Age
|
− .153
|
.199
|
.593
|
.441
|
.858
|
.580
|
1.268
|
Gender
|
− .521
|
.249
|
4.357
|
.037*
|
.594
|
.364
|
.969
|
Histological types
|
− .040
|
.215
|
.034
|
.853
|
.961
|
.630
|
1.465
|
LNM status
|
− .976
|
.252
|
15.020
|
.000***
|
.377
|
.230
|
.617
|
Clinical stages
|
− .634
|
.216
|
8.632
|
.003***
|
.530
|
.347
|
.810
|
Pathological grades
|
− .486
|
.202
|
5.801
|
.016*
|
.615
|
.414
|
.913
|
hsa-miR-7-5p
|
.394
|
.199
|
4.007
|
.045*
|
1.491
|
1.008
|
2.204
|
MNK
|
− .464
|
.206
|
5.095
|
.024*
|
.629
|
.420
|
.941
|
Abbreviations: CI, confidence interval; LNM, lymph node metastasis; NSCLC: non-small cell lung cancer. |
Table 3
The pairwise association between expression of miR-7-5p and MNK in 318 cases of NSCLC
|
|
MNK
|
|
|
Positive (%)
|
Negative (%)
|
P-value
|
miR-7-5p
|
|
|
|
High (%)
|
65(39.9)
|
98(60.1)
|
0.011*
|
Low (%)
|
84(54.2)
|
71(45.8)
|
(r = -0.143)
|
*Spearman’s rank correlation test, statistically significant difference (*P < 0.05) |
Exosomal miR-7-5p sensitizes the anti-cancer effect of Everolimus in vitro.
Exogenous miR-7-5p could supply the deficiency of it. Further contrast among the groups of Everolimus, exogenous miR-7-5p and combined of them, we discovered that Everolimus alone or stable miR-7-5p expression could inhibit tumor cell proliferation (Fig. 4a & S2a), Clone formation (Fig. 4b & S2b) and 2D&3D migration capabilities (Fig. 4c-d & S2c-d, respectively). When Everolimus was carried out to pose on stable miR-7-5p cells, the inhibitory effect was the most significant.
As the functions of exosomes mediated by their contents are more widely recognized, they have been proposed as a potential cell-based alternative therapy which have received increasing attention19. Due to the direct sorting and packaging of nucleic acids into exosomes may not provide the functionally active contents into recipient cells effectively20. We tried to use exosomes derived from tumor cells directly to improve miR-7-5p in Everolimus treated NSCLC cells. Like exogenous miR-7-5p, exosomal miR-7-5p could inhibit the cell proliferation (Fig. 4e & S2e), Clone formation (Fig. 4f & S2f) and 2D&3D migration capabilities (Fig. 4g-h & S2g-h, respectively). And further usage of miR-7-5p-loaded exosomes combined with Everolimus showed a more obvious tumor suppressive effect, which indicated that exo-miR-7-5p could be internalized by NSCLC cells and cooperated with Everolimus to enhance the anticancer effect of it in vitro.
Exosomal miR-7-5p sensitizes the anti-cancer effect of Everolimus in vivo.
We used LV-miR-7-5p and LV-NC A549 cells to construct nude mouse subcutaneous tumor models respectively. Once the tumor volume reached 50-100mm3, they would be divided into four groups and received indicated treatments. The growth rate of the LV-miR-7-5p group was significantly slower than the control group, as well as the tumor volume. It was worth noting that although Everolimus had a strong inhibitory effect on tumor growth in the early stage, its growth rate tended to accelerate in the later period, resulting in the relatively bigger volume than LV-miR-7-5p combined Everolimus group. The proliferation rate of tumor cells and tumor volume always kept at a lowest level when LV-miR-7-5p was performed with Everolimus (Fig. 5a-d). Further, in spite of that Everolimus could suppress the Ki-67 index of tumors than the control groups no matter combined with LV-NC or LV-miR-7-5p, the activation of MNK1/eIF4E axis was only observed in the LV-NC combined Everolimus group, (Fig. 5e), meant restoring the levels of miR-7-5p could be used as an effective method to reverse the activation of MNK/eIF4E axis induced by Everolimus.
Currently, the treatment of cancers highlights the need for more effective and innovative therapies. Because of safer biological behaviors, exosomes have the value of translational medicine for clinical treatment purposes20. Using tumor-derived exosomes will provide additional competitive advantages for the selective delivery of anticancer therapies not only to the primary tumor but also to the premetastatic niche and even to metastasis, owing to their intrinsic organotropic tumor-homing properties21. We further established the abdominal metastasis models accepting the indicated treatments to monitor tumors in vivo and the exosomal miR-7-5p in the blood. Tumor involvement was observed in all groups (Fig. 5f & S3). Notably, the exo-miR-7-5p combined with Everolimus group had the weakest fluorescence signal (Fig. 5g), and high miR-7-5p in the blood (Fig. 5h).
The miR-7-5p combined with Everolimus can induce apoptosis via dual abrogation of MNK/eIF4E and mTOR in NSCLC
The function of eIF4E is relay on the subcellular locations of it. The initiation of the translation of most mRNAs is controlled by intracytoplasmic eIF4E. And if it locates in the nucleus, the export of certain mRNAs to the cytoplasm will depend on it 22. In order to further explore the specific value and mechanism of the combined targeting of the MNK/eIF4E axis and the mTOR pathway in the treatment of NSCLC, the first synthetic small molecule MNK1 inhibitor CGP57380 was performed to block the MNK/eIF4E axis. We found p-MNK1Thr197/202 was mainly distributed in the nucleus and was inhibited. In spite of p-eIF4ES209 was located both in the cytoplasm and nucleus, CGP57380 mainly suppressed the phosphorylation of intracytoplasmic eIF4E (Fig. 6a, Figure S4a). Besides, it still has characteristics such as poor specificity and relatively weak affinity, which limits its value in clinical treatment 23. Fortunately, both endogenous and exosomal miR-7-5p could restore the intracellular tumor suppressor miR-7-5p and reverse the feedback activation of MNK/eIF4E axis more specific and safer. When Everolimus was performed in stable LV-miR-7-5p A549 and SPC-A1 cells (Fig. 6b, Figure S4b) or combined with the miR-7-5p-loaded exosomes derived from A549 cells (Fig. 6c, Figure S4c), the activation of the MNK/eIF4E axis was eliminated.
Further flow cytometry analysis indicated the apoptosis rate of miR-7-5p mimics or Everolimus treatment alone was less than the combined group (Fig. 6d, Figure S4d). And representative microscopic images of liver metastatic lesions among all groups in previous abdominal metastasis models observed obvious necrosis of metastases in exo-miR-7-5p combined with Everolimus group (Fig. 6e), which suggested that the miR-7-5p in combination with Everolimus could significantly strengthen the therapeutic effect of Everolimus through inducing cell death. Mechanismly, caspase-3, cleaved caspase-3, DR4, DR5, pro-apoptotic proteins Bak, Bad and Bax were significantly up-regulated but anti-apoptotic protein Bcl-xL was down-regulated (Fig. 6f-h, Figure S4f-h) when Everolimus posed on the stable LV-miR-7-5p NSCLC cells, indicating it could induce the co-activation of the inherent mitochondrial apoptosis pathway and the death receptor pathway.