Regulation of opioid tolerance by let-7 family microRNA targeting the mu opioid receptor - PubMed (original) (raw)

Regulation of opioid tolerance by let-7 family microRNA targeting the mu opioid receptor

Ying He et al. J Neurosci. 2010.

Abstract

MicroRNA has emerged as a critical regulator of neuronal functions. This study aimed to test whether let-7 microRNAs can regulate the mu opioid receptor (MOR) and opioid tolerance. Employing bioinformatics, we identified a let-7 binding site in the 3'-untranslated region (UTR) of MOR mRNA, which was experimentally confirmed as a direct target of let-7. The repressive regulation of MOR by let-7 was revealed using a LNA-let-7 inhibitor to knockdown let-7 in SH-SY5Y cells. Conversely, morphine significantly upregulated let-7 expression in SH-SY5Y cells and in a mouse model of opioid tolerance. The LNA-let-7 inhibitor decreased brain let-7 levels and partially attenuated opioid antinociceptive tolerance in mice. Although chronic morphine treatment did not change overall MOR transcript, polysome-associated mRNA declined in a let-7-dependent manner. let-7 was identified as a mediator translocating and sequestering MOR mRNA to P-bodies, leading to translation repression. These results suggest that let-7 plays an integral role in opioid tolerance.

PubMed Disclaimer

Figures

Figure 1.

Figure 1.

Interaction of let-7 with the 3′-UTR of the MOR. A, Sequence alignment of the let-7 family miRNAs and the predicted sequence pairing with a region of MOR mRNA 3′-UTR. The nucleotides in red are the positions that are identical among all members of the let-7 family. The predicted MOR mRNA 3′-UTR sequences that may interact with let-7 seed match are in green (bold). B, A schematic plot of the luciferase constructs. Solid bar indicates the location of the putative let-7 binding site within the 3′-UTR of MOR mRNA. The LNA-let-7 inhibitor and scrambled-LNA control are shown below the let-7a alignment with the position of LNA nucleotides underlined in bold. C, Luciferase reporter assays were performed in HEK293 cells transfected with the reporter plasmids psiCHECK/MOR1UTR or psiCHECK/MOR1UTRmut together with the LNA-let-7 inhibitor (0, 1, 5, 25 pmol) or scrambled-LNA control (25 pmol). The Renilla luciferase activity was normalized to the internal control firefly luciferase activity. *p < 0.05, ***p < 0.001 vs the 3′-UTR control (n = 3).

Figure 2.

Figure 2.

Constitutive repression of MOR expression by let-7 miRNAs in SH-SY5Y cells. SH-SY5Y cells were transfected with the LNA-let-7 inhibitor (0, 2, 10, 50, 250 pmol) or scrambled-LNA control (250 pmol). Cells were lysed for Western blot analysis 48 h later. The anti-MOR antibody did not detect 65 kDa MOR bands in the null deletion mice (supplemental Fig. S1, available at

www.jneurosci.org

as supplemental material). Histogram graph was constructed from the representative figure shown and two other experiments. ***p < 0.001 vs the control.

Figure 3.

Figure 3.

Increased let-7 expression and decreased MOR expression in SH-SY5Y cells during the development of opioid tolerance. SH-SY5Y cells with or without retinoic acid (RA) pretreatment (5 μ

m

, 6 d) were incubated with morphine (1 μ

m

, 24–48 h) to induce a tolerant state. The transcripts of let-7a (A), let-7c (B), and let-7g (C), but not miR-134 (D), were increased as determined by the real time RT-PCR. MOR expression was decreased as measured by Western blot analysis (E, F). *p < 0.05, **p < 0.01, ***p < 0.001 compared with 0 h without RA; ##p < 0.01, ###p < 0.001 compared with 0 h with RA (n = 3).

Figure 4.

Figure 4.

Increased let-7 expression in the mouse brain during the development of opioid tolerance. Mice were implanted with morphine pellets to induce opioid tolerance. On day 0, 1, 3 and 5, brain let-7a (A), let-7c (B), let-7g (C), and miR-134 (D) transcripts were analyzed by quantitative RT-PCR. The development of opioid tolerance was confirmed by following significantly reduced morphine-antinociception in a tail-flick assay (E). *p < 0.05, ***p < 0.001 vs day 0 group (n = 5).

Figure 5.

Figure 5.

Fluorescence in situ hybridization (FISH) of let-7 before and after the treatment with morphine (75 mg/pellet/mouse). The brain cortex was used for the analysis of let-7 mRNA by FISH. Samples were costained using anti-MOR antibody (1:5000) to determine the level of let-7 in MOR-expressing cells. The let-7 transcript was increased after morphine treatment. Moreover, the increase was observed in MOR-expressing cells (arrows indicate representative cells with costaining of MOR and let-7), but not in MOR-negative cells (only DAPI nuclear staining, but no MOR staining). Fifteen prefrontal cortex slices from 3 mice were studied for each condition. In the control group, 78 MOR-expressing cells and 60 MOR-negative cells were imaged. In the morphine group, let-7 upregulation occurred in 78/85 MOR-expressing cells; no cell showed let-7 response in MOR-negative cells (0/65).

Figure 6.

Figure 6.

Effect of the LNA-let-7 inhibitor on morphine antinociception, morphine antinociceptive tolerance, and let-7 expression in mice. A, The LNA-let-7 inhibitor potentiated morphine antinociception in naive mice, suggesting constitutive repression of MOR by let-7. B, The LNA-let-7 inhibitor dose-dependently attenuated morphine antinociceptive tolerance. C–E, Morphine-induced let-7 upregulation was dose-dependently reduced by the LNA-let-7 inhibitor (0.3–3.0 nmol, twice/day, for 3 d). *p < 0.05, **p < 0.01, ***p < 0.001 vs the control group; #p < 0.05, ##p < 0.01, ###p < 0.001 vs the morphine group (n = 4).

Figure 7.

Figure 7.

The total and polysome-associated μ opioid receptor transcript after the treatment with morphine in SH-SY5Y cells. SH-SY5Y cells were treated with morphine (1 μ

m

, 48 h). A, MOR transcript was not altered by morphine (n = 5 each group). B, Polysome-associated MOR mRNA was decreased by morphine. The change was significantly attenuated by the LNA-let-7 inhibitor. **p < 0.01 vs the control group; #p < 0.05, vs the morphine group (n = 5 each group). C, Colocalization of MOR mRNA with hDcp1a, a P-body marker, after, but not before, morphine treatment. Blue (DAPI) identifies nuclei.

Figure 8.

Figure 8.

A schematic plot for the proposed mechanism of MOR regulation by let-7. Pri- and pre-let-7 were produced and exported into the cytosol where the mature let-7 was incorporated into the RISC. The latter recruits and sequesters MOR mRNA to P-bodies that are deprived of translational machinery, effectively reducing polysome-bound MOR mRNA and leading to translation repression.

Similar articles

Cited by

References

    1. Ambros V. The functions of animal microRNAs. Nature. 2004;431:350–355. - PubMed
    1. Bernstein MA, Welch SP. mu-Opioid receptor down-regulation and cAMP-dependent protein kinase phosphorylation in a mouse model of chronic morphine tolerance. Brain Res Mol Brain Res. 1998;55:237–242. - PubMed
    1. Bhargava HN, Gulati A. Down-regulation of brain and spinal cord mu-opiate receptors in morphine tolerant-dependent rats. Eur J Pharmacol. 1990;190:305–311. - PubMed
    1. Bhattacharyya SN, Habermacher R, Martine U, Closs EI, Filipowicz W. Relief of microRNA-mediated translational repression in human cells subjected to stress. Cell. 2006;125:1111–1124. - PubMed
    1. Boyerinas B, Park SM, Shomron N, Hedegaard MM, Vinther J, Andersen JS, Feig C, Xu J, Burge CB, Peter ME. Identification of let-7-regulated oncofetal genes. Cancer Res. 2008;68:2587–2591. - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources