Skip to main content
Log in

Hyperoside attenuates pyrrolizidine alkaloids-induced liver injury by ameliorating TFEB-mediated mitochondrial dysfunction

  • Research Article
  • Published:
Archives of Pharmacal Research Aims and scope Submit manuscript

Abstract

Pyrrolizidine alkaloids (PAs) are potent hepatotoxins that can cause liver damage. Hyperoside (Hyp), a natural flavonoid, can be extracted from medicinal plants. Hyp displays hepatoprotective activity in various liver diseases. However, the potential effect and mechanism of action of Hyp in ameliorating PA-induced liver injury remain obscure. This study aimed to explore the protective effect of Hyp against PA-induced hepatotoxicity and its underlying mechanism. We established an in vitro model of PAs in mouse primary hepatocytes and developed a mouse model of acute PA toxicity to investigate the protective effect of Hyp. We found that Hyp notably attenuated PA-induced hepatotoxicity. RNA-sequencing showed that the beneficial effect of Hyp against PA-induced hepatotoxicity was associated with the transcription factor EB (TFEB)-peroxisome proliferator-activated receptor-γ coactivator-1-α (PGC1α) pathway. Our results confirmed that both the autophagy-lysosomal pathway and mitochondrial biogenesis were induced by Hyp through TFEB nuclear translocation in PA-induced liver injury. Furthermore, we demonstrated that activation of the mechanistic target of rapamycin complex 1 (mTORC1) by MHY 1485 decreased TFEB nuclear translocation and abrogated the protective effect of Hyp against PA-induced liver injury in mice. In contrast, inhibition of mTORC1 activity increased the level of TFEB and reduced hepatotoxicity induced by PAs in mouse livers. Likewise, Hyp-induced TFEB activation was validated in vitro. In conclusion, Hyp can activate the TFEB-mediated autophagy-lysosomal pathway and mitochondrial biogenesis through inhibition of mTORC1 activity, alleviating the liver injury induced by PAs, thus suggesting the potential value of Hyp in the treatment of PA-induced hepatotoxicity.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Data availability

The data that support the findings of this study are not openly available due to resasons of sensitivity and are available from the corresponding author upon reasonable request.

References

Download references

Acknowledgements

This work is financially supported by the National Natural Science Foundation of China (No. 81920108033; 82074011; 82130115) and the Program of Shanghai Municipal Health Commission (No. ZY (2021–2023)-0215).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Lili Ding or Li Yang.

Ethics declarations

Conflict of interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

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.

Supplementary file1 (DOCX 4736 KB)

12272_2023_1460_MOESM2_ESM.tif

Supplementary file2 Supplementary Fig. 1 Hyp attenuates PA-induced liver injury by enhancing autophagy in primary hepatocytes. A Primary hepatocytes were treated with TA for 12 h or 24 h. Hyp was added to the cultures 1 h before TA treatment. The expression of the proteins LC3-II and SQSTM1/p62 were measured by western blotting in primary hepatocytes. Bar graphs show summary data. B Primary hepatocytes were treated with TA and Hyp in the presence or absence of Baf A1. The expression of the protein LC3-II was measured by western blotting in primary hepatocytes. C Primary hepatocytes were transfected with Ad-mRFP-GFP-LC3 for 24 h. After transfection, cells were treated with TA for 12 h or 24 h, Hyp was added to the cultures 1 h before TA treatment. Representative images of Ad-mRFP-GFP-LC3 in primary hepatocytes. Scale bars, 200 μm. B-C Hyp, 50 μM. Data were shown as the means ± SD from at least three independent experiments and analyzed by one-way ANOVA. ###P < 0.001 vs. Vehicle; *P < 0.05, **P < 0.01, ***P < 0.001 vs. TA. (TIF 4416 KB)

12272_2023_1460_MOESM3_ESM.tif

Supplementary file3 Supplementary Fig. 2 Hyp attenuates PA-induced impairment of lysosomal function. A The expression of the protein LAMP1 was measured by western blotting in primary hepatocytes. Bar graphs show summary data. B The expression of the protein LAMP1 was measured by western blotting in mouse liver. Bar graphs show summary data (n = 3). B Hyp, 40 mg/kg. A Data are shown as the means ± SD from at least three independent experiments and analyzed by one-way ANOVA. B Data were shown as the means ± SEM and analyzed by one-way ANOVA. ###P < 0.001 vs. Vehicle; *P < 0.05, **P < 0.01, ***P < 0.001vs. TA. (TIF 1795 KB)

12272_2023_1460_MOESM4_ESM.tif

Supplementary file4 Supplementary Fig. 3 Hyp induces mitochondrial biogenesis through the TFEB-PGC1α pathway in PA-treated primary hepatocytes. A Total intracellular ATP level. B ΔΨm determined by JC-1 red/green fluorescence ratio. C Representative images of the mitochondria of primary hepatocytes stained with MitoTracker Green. D Primary hepatocytes were fractionated into mitochondria and cytosol, and the level of Cyt C was analyzed by western blotting. Fraction quality was verified by immunoblotting with markers for the mitochondria (COX IV) and cytoplasm (α-Tubulin). E Representative TEM micrographs showing Hyp ameliorates mitochondria damage in PAs-treated primary hepatocytes. Scale bars, 2 μm (wireframe indicates the magnified image). F The expression of the protein PGC1α was measured by western blotting in primary hepatocytes. Bar graphs show summary data. G Primary hepatocytes were transfected with scRNA or siTFEB for 24 h and treated with TA in the presence or absence of Hyp for 12 h or 24 h. The expression of the proteins TFEB and PGC1α were measured by western blotting. H The expression of the proteins CPT1α and PPARα were measured by western blotting in primary hepatocytes. Bar graphs show summary data. A-E, G, Hyp, 50 μM. Data were shown as the means ± SD from at least three independent experiments and analyzed by one-way ANOVA. ###P < 0.001 vs. Vehicle; *P < 0.05, **P < 0.01, ***P < 0.001 vs. TA. (TIF 2214 KB)

12272_2023_1460_MOESM5_ESM.tif

Supplementary file5 Supplementary Fig. 4 TFEB mediates the beneficial effects of Hyp in PA-induced liver injury. The mRNA levels of TFEB-associated genes (Map1lc3b, Uvrag, Wipi1, Lamp1, Pgc1α, Nrf1, Tfam) were evaluated by qRT-PCR. Data were shown as the means ± SEM and analyzed by one-way ANOVA or Student's t-test. ***P < 0.001 vs. TA; &&P < 0.01, &&&P < 0.001 vs. TA-Hyp (siRNA NC). (TIF 2275 KB)

12272_2023_1460_MOESM6_ESM.tif

Supplementary file6 Supplementary Fig. 5 Primary hepatocytes were transfected with a plasmid carrying TFEB or empty vector and then treated to 50 μM Hyp for 12 h or 24 h. Effects of Hyp on TFEB nuclear translocation in GFP-TFEB primary hepatocytes. Scale bars, 200 μm. Data were representative of at least three independent experiments. (TIF 1608 KB)

12272_2023_1460_MOESM7_ESM.tif

Supplementary file7 Supplementary Fig. 6 Inhibition of mTOR1 activity by Hyp attenuates PA-induced liver injury in primary hepatocytes. A The expression of the proteins p-mTORC and p-ERK1/2 were measured by western blotting in primary hepatocytes. Bar graphs show summary data. B The expression of the proteins p-S6 and p-eIF4E were measured by western blotting in primary hepatocytes. Bar graphs show summary data. C Primary hepatocytes treated with TA in the presence or absence of Torin1 for 12 h or 24 h. The expression of the nuclear and cytosolic TFEB proteins in primary hepatocytes were analyzed by western blotting. D Primary hepatocytes treated with TA and Hyp in the presence or absence of MHY 1485 for 12 h or 24 h. The expression of the nuclear and cytosolic TFEB proteins in primary hepatocytes were analyzed by western blotting. Data were shown as the means ± SD from at least three independent experiments and analyzed by one-way ANOVA. ##P < 0.01, ###P < 0.001 vs. Vehicle; *P < 0.05, **P < 0.01, ***P < 0.001 vs. TA. (TIF 4846 KB)

12272_2023_1460_MOESM8_ESM.tif

Supplementary file8 Supplementary Fig. 7 Torin1 attenuates PA-induced liver injury in mice. The mice were orally administered TA, and then were intraperitoneally injected with Torin1 once at 6 h after TA administration. A Serum ALT activity (n = 6). B Serum AST activity (n = 6). C Serum TBA amount (n = 6). D Representative images of H&E-stained liver sections (scale bars, 50 μm). E Nuclear fractions of TFEB in mouse liver was analyzed by western blotting (n = 3). F The mRNA levels of TFEB target genes (Map1lc3b, Uvrag, Wipi1, Lamp1, Pgc1α) were evaluated by qRT-PCR (n = 6). G Paraffin sections of mouse liver were stained with TFEB (red) and DAPI (blue). Scale bars, 50 μm (wireframe indicates the magnified image). Data were shown as the means ± SEM and analyzed by one-way ANOVA. A-C ***P < 0.001 vs. TA. F ###P < 0.001 vs. Vehicle; **P < 0.01, ***P < 0.001 vs. TA. (TIF 6776 KB)

12272_2023_1460_MOESM9_ESM.tif

Supplementary file9 Supplementary Fig. 8 Inhibition of mTORC1 activity by Hyp attenuates PA-induced liver injury in mice. Paraffin sections of mouse liver were stained with TFEB (red) and DAPI (blue). Scale bars, 50 μm (wireframe indicates the magnified image). (TIF 3939 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, J., Xiong, A., Wang, X. et al. Hyperoside attenuates pyrrolizidine alkaloids-induced liver injury by ameliorating TFEB-mediated mitochondrial dysfunction. Arch. Pharm. Res. 46, 694–712 (2023). https://doi.org/10.1007/s12272-023-01460-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12272-023-01460-3

Keywords

Navigation