References
Alexander SPH, Kelly E, Mathie A, Peters JA, Veale EL, Armstrong JF, et al. (2019). THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: Transporters. Br J Pharmacol 176 Suppl 1: S397-S493.
Alexander SPH, Roberts RE, Broughton BRS, Sobey CG, George CH, Stanford SC, et al. (2018). Goals and practicalities of immunoblotting and immunohistochemistry: A guide for submission to the British Journal of Pharmacology. Br J Pharmacol 175: 407-411.
Blanco-Garcia N, Asensio-Juan E, de la Cruz X, & Martinez-Balbas MA (2009). Autoacetylation regulates P/CAF nuclear localization. J Biol Chem 284: 1343-1352.
Canettieri G, Di Marcotullio L, Coni S, Greco A, & Gulino A (2010). Turning off the switch in medulloblastoma: the inhibitory acetylation of an oncogene. Cell Cycle 9: 2047-2048.
Canettieri G, Di Marcotullio L, Greco A, Coni S, Antonucci L, Infante P, et al. (2010). Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation. Nat Cell Biol 12: 132-142.
Chen J, Hu Y, Fang J, Chen L, Mu Y, Liu W, et al. (2020). GLI1, but not smoothened-dependent, signaling in hepatic progenitor cells promotes a ductular reaction, which aggravates liver fibrosis. Journal of Hepatology 73.
Chung SI, Moon H, Ju HL, Cho KJ, Kim DY, Han KH, et al. (2016). Hepatic expression of Sonic Hedgehog induces liver fibrosis and promotes hepatocarcinogenesis in a transgenic mouse model. J Hepatol 64:618-627.
Coni S, Antonucci L, D’Amico D, Di Magno L, Infante P, De Smaele E, et al. (2013). Gli2 Acetylation at Lysine 757 Regulates Hedgehog-Dependent Transcriptional Output by Preventing Its Promoter Occupancy. Plos One 8.
Coni S, Mancuso AB, Di Magno L, Sdruscia G, Manni S, Serrao SM, et al. (2017). Selective targeting of HDAC1/2 elicits anticancer effects through Gli1 acetylation in preclinical models of SHH Medulloblastoma. Sci Rep 7: 44079.
Curtis MJ, Alexander S, Cirino G, Docherty JR, George CH, Giembycz MA, et al. (2018). Experimental design and analysis and their reporting II: updated and simplified guidance for authors and peer reviewers. Brit J Pharmacol 175: 987-993.
Dennler S, Andre J, Alexaki I, Li A, Magnaldo T, ten Dijke P, et al. (2007). Induction of sonic hedgehog mediators by transforming growth factor-beta: Smad3-dependent activation of Gli2 and Gli1 expression in vitro and in vivo. Cancer Res 67: 6981-6986.
Ding N, Hah N, Yu RT, Sherman MH, Benner C, Leblanc M, et al.(2015). BRD4 is a novel therapeutic target for liver fibrosis. Proc Natl Acad Sci U S A 112: 15713-15718.
Falkenberg KJ, Newbold A, Gould CM, Luu J, Trapani JA, Matthews GM, et al. (2016). A genome scale RNAi screen identifies GLI1 as a novel gene regulating vorinostat sensitivity. Cell Death Differ 23: 1209-1218.
Friedman SL (2008). Mechanisms of hepatic fibrogenesis. Gastroenterology 134: 1655-1669.
Geng Y, Liu J, Xie Y, Jiang H, Zuo K, Li T, et al. (2018). Trichostatin A promotes GLI1 degradation and P21 expression in multiple myeloma cells. Cancer Manag Res 10: 2905-2914.
Gotic I, & Foisner R (2010). Multiple novel functions of lamina associated polypeptide 2 alpha in striated muscle. Nucleus-Phila 1: 397-401.
Guerrero-Juarez CF, & Plikus MV (2017). Gli-fully Halting the Progression of Fibrosis. Cell Stem Cell 20: 735-736.
Gulino A, Di Marcotullio L, Canettieri G, De Smaele E, & Screpanti I (2012). Hedgehog/Gli control by ubiquitination/acetylation interplay. Vitam Horm 88: 211-227.
Gurung B, Feng Z, & Hua X (2013). Menin directly represses Gli1 expression independent of canonical Hedgehog signaling. Mol Cancer Res 11: 1215-1222.
Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ, Ireland S, et al. (2018). The IUPHAR/BPS Guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucleic Acids Res 46: D1091-D1106.
Hernandez-Gea V, & Friedman SL (2011). Pathogenesis of Liver Fibrosis. Annu Rev Pathol-Mech 6: 425-456.
Hsu CC, Wu YC, Farh L, Du YC, Tseng WK, Wu CC, et al. (2012). Physalin B from Physalis angulata triggers the NOXA-related apoptosis pathway of human melanoma A375 cells. Food Chem Toxicol 50:619-624.
Katoh Y, & Katoh M (2009). Integrative genomic analyses on GLI1: positive regulation of GLI1 by Hedgehog-GLI, TGFbeta-Smads, and RTK-PI3K-AKT signals, and negative regulation of GLI1 by Notch-CSL-HES/HEY, and GPCR-Gs-PKA signals. Int J Oncol 35:187-192.
Kim JY, Shen S, Dietz K, He Y, Howell O, Reynolds R, et al.(2010). HDAC1 nuclear export induced by pathological conditions is essential for the onset of axonal damage. Nat Neurosci 13:180-U163.
Kim K, Kim KH, Cho HK, Kim HY, Kim HH, & Cheong J (2010). SHP (small heterodimer partner) suppresses the transcriptional activity and nuclear localization of Hedgehog signalling protein Gli1. Biochem J 427: 413-422.
Kramann R, Schneider RK, DiRocco DP, Machado F, Fleig S, Bondzie PA, et al. (2015). Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell 16: 51-66.
Lee YA, Wallace MC, & Friedman SL (2015). Pathobiology of liver fibrosis: a translational success story. Gut 64: 830-841.
Lilley E, Stanford SC, Kendall DE, Alexander SPH, Cirino G, Docherty JR, et al. (2020). ARRIVE 2.0 and the British Journal of Pharmacology: Updated guidance for 2020. Br J Pharmacol 177:3611-3616.
Liu YQ, Wang Z, Wang JN, Lam W, Kwong S, Li FR, et al. (2013). A histone deacetylase inhibitor, largazole, decreases liver fibrosis and angiogenesis by inhibiting transforming growth factor-beta and vascular endothelial growth factor signalling. Liver International 33:504-515.
Loh Z, Fitzsimmons RL, Reid RC, Ramnath D, Clouston A, Gupta PK, et al. (2019). Inhibitors of class I histone deacetylases attenuate thioacetamide-induced liver fibrosis in mice by suppressing hepatic type 2 inflammation. Br J Pharmacol 176: 3775-3790.
Ma YM, Han W, Li J, Hu LH, & Zhou YB (2015). Physalin B not only inhibits the ubiquitin-proteasome pathway but also induces incomplete autophagic response in human colon cancer cells in vitro. Acta Pharmacol Sin 36: 517-527.
Machado MV, & Diehl AM (2018). Hedgehog signalling in liver pathophysiology. J Hepatol 68: 550-562.
Magalhaes HI, Veras ML, Torres MR, Alves AP, Pessoa OD, Silveira ER, et al. (2006). In-vitro and in-vivo antitumour activity of physalins B and D from Physalis angulata. J Pharm Pharmacol 58:235-241.
Mazza D, Infante P, Colicchia V, Greco A, Alfonsi R, Siler M, et al. (2013). PCAF ubiquitin ligase activity inhibits Hedgehog/Gli1 signaling in p53-dependent response to genotoxic stress. Cell Death Differ 20: 1688-1697.
McMillin M, Galindo C, Pae HY, Frampton G, Di Patre PL, Quinn M, et al. (2014). Gli1 activation and protection against hepatic encephalopathy is suppressed by circulating transforming growth factor beta1 in mice. J Hepatol 61: 1260-1266.
Mederacke I, Dapito DH, Affo S, Uchinami H, & Schwabe RF (2015). High-yield and high-purity isolation of hepatic stellate cells from normal and fibrotic mouse livers. Nat Protoc 10: 305-315.
Miele E, Po A, Begalli F, Antonucci L, Mastronuzzi A, Marras CE, et al. (2017). beta-arrestin1-mediated acetylation of Gli1 regulates Hedgehog/Gli signaling and modulates self-renewal of SHH medulloblastoma cancer stem cells. BMC Cancer 17: 488.
Mirza AN, McKellar SA, Urman NM, Brown AS, Hollmig T, Aasi SZ, et al. (2019). LAP2 Proteins Chaperone GLI1 Movement between the Lamina and Chromatin to Regulate Transcription. Cell 176: 198-212 e115.
Nye MD, Almada LL, Fernandez-Barrena MG, Marks DL, Elsawa SF, Vrabel A, et al. (2014). The transcription factor GLI1 interacts with SMAD proteins to modulate transforming growth factor beta-induced gene expression in a p300/CREB-binding protein-associated factor (PCAF)-dependent manner. J Biol Chem 289: 15495-15506.
Park KC, Park JH, Jeon JY, Kim SY, Kim JM, Lim CY, et al. (2014). A new histone deacetylase inhibitor improves liver fibrosis in BDL rats through suppression of hepatic stellate cells. Br J Pharmacol 171: 4820-4830.
Seki E (2016). HEDGEHOG Signal in hepatocytes mediates macrophage recruitment: A new mechanism and potential therapeutic target for fatty liver disease. Hepatology 63: 1071-1073.
Soares MB, Brustolim D, Santos LA, Bellintani MC, Paiva FP, Ribeiro YM, et al. (2006). Physalins B, F and G, seco-steroids purified from Physalis angulata L., inhibit lymphocyte function and allogeneic transplant rejection. Int Immunopharmacol 6: 408-414.
Tan S, Liu H, Ke B, Jiang J, & Wu B (2020). The peripheral CB1 receptor antagonist JD5037 attenuates liver fibrosis via a CB1 receptor/beta-arrestin1/Akt pathway. Br J Pharmacol 177:2830-2847.
Vandenberghe I, Creancier L, Vispe S, Annereau JP, Barret JM, Pouny I, et al. (2008). Physalin B, a novel inhibitor of the ubiquitin-proteasome pathway, triggers NOXA-associated apoptosis. Biochem Pharmacol 76: 453-462.
Vidak S, Kubben N, Dechat T, & Foisner R (2015). Proliferation of progeria cells is enhanced by lamina-associated polypeptide 2 alpha (LAP2 alpha) through expression of extracellular matrix proteins. Gene Dev 29: 2022-2036.
Wu X, Wu X, Ma Y, Shao F, Tan Y, Tan T, et al. (2016). CUG-binding protein 1 regulates HSC activation and liver fibrogenesis. Nat Commun 7: 13498.
Yang Y, Yi L, Wang Q, Xie B, Sha C, & Dong Y (2018). Physalin B Suppresses Inflammatory Response to Lipopolysaccharide in RAW264.7 Cells by Inhibiting NF-κB Signaling. Journal of Chemistry 2018: 1-6.
Zhang F, Hao M, Jin H, Yao Z, Lian N, Wu L, et al. (2017). Canonical hedgehog signalling regulates hepatic stellate cell-mediated angiogenesis in liver fibrosis. Br J Pharmacol 174: 409-423.
Zhang F, Wang F, He J, Lian N, Wang Z, Shao J, et al. (2020). Reregulation of hepatic stellate cell contraction and cirrhotic portal hypertension by Wnt/beta-catenin signaling via interaction with Gli1. Br J Pharmacol.
Zhao SS, Wang JX, Wang YC, Shao RG, & He HW (2015). [Establishment and application of a high-throughput drug screening model based on COL1A1 promoter for anti-liver fibrosis]. Yao Xue Xue Bao 50:169-173.
Zheng Y, Chen J, Liu L, Liang X, & Hong D (2016). In vivo pharmacokinetics of and tissue distribution study of physalin B after intravenous administration in rats by liquid chromatography with tandem mass spectrometry. Biomed Chromatogr 30: 1278-1284.
Zheng Y, Luan L, Chen Y, Ren Y, & Wu Y (2012). Characterization of physalins and fingerprint analysis for the quality evaluation of Physalis alkekengi L. var. franchetii by ultra-performance liquid chromatography combined with diode array detection and electrospray ionization tandem mass spectrometry. J Pharm Biomed Anal 71:54-62.
Zupkovitz G, Tischler J, Posch M, Sadzak I, Ramsauer K, Egger G, et al. (2006). Negative and positive regulation of gene expression by mouse histone deacetylase 1. Mol Cell Biol 26: 7913-7928.
Figure legends
FIGURE 1 PB inhibits the proliferation and activation of HSCs. (A) The chemical structure of PB. (B-F) LX‐2 cells were treated with 2 ng·ml−1 TGFβ1 with or without different concentrations of PB for 24 hr after no FBS starvation. (B) COL1A1 luciferase activity was determined in LX-2 cells transfected with the pGL4.17-COL1A1 luciferase construct. (C) Cell proliferation was determined by CCK8 assay. (D)PB repressed theCOL1A1 , αSMA , TGFβ1 and TIMP1 in a dose‐dependent manner in LX‐2 cells. GADPH served as loading control. (E) PB dose‐dependently inhibited the protein levels of COL1A1 and α‐SMA in LX‐2 cells. The protein levels were normalized against β-actin. (F) Immunofluorescence staining for α-SMA (red) in LX-2 cells. (G-I) Mouse pHSCs were treated with different concentrations of PB for 24 hr after 7 days of isolation. (G) PB inhibited the col1a1 , and αsma in mouse pHSCs.β-actin served as loading control. (H) PB inhibited the protein levels of α‐SMA in mouse pHSCs. The protein levels were normalized against β-actin. (I) Immunofluorescence staining for α-SMA (red) in mouse pHSCs. The values are expressed as the mean ± SEM of five independent assays, *P < 0.05, **P < 0.01, ***P < 0.001, ****P< 0.0001, significantly different from the control group and #P < 0.05, ##P< 0.01, ###P< 0.001 significantly different from the TGFβ1 treatment group in LX‐2 cells and mouse pHSCs. ns, not significant.
FIGURE 2 PB markedly ameliorated the liver fibrosis in CCl4‐treated mice. (A) Liver histopathology was observed by H&E staining (magnification 200×). (B) Serum ALT, AST assays and (C) Liver hydroxyproline concentration assays demonstrated that PB reduced liver fibrosis in CCl4 mice. (D) The obtained liver sections were subjected to sirius red staining (magnification 200×; up panel), and Masson trichrome stain (magnification 200×; down panel). (E) mRNA expressions of αsma ,Col1a1 , Tgfb1 , and Timp1 in mice liver samples. (F) Western blot analysis and semi‐ quantitation of α‐SMA expressions. β-actin served as the loading control. The values are expressed as the mean ± SEM (n = 8 of each group), *P < 0.05, **P < 0.01, ***P < 0.001, significantly different from control group and ###P < 0.001, ####P < 0.0001, significantly different from CCl4‐corn oil group. ns, not significant.
FIGURE 3 PB markedly ameliorated the liver fibrosis in bile duct‐ligated (BDL) mice. (A) Liver pathological changes were detected by H&E staining (magnification 200×). (B) Serum ALT, AST assays and (C) Liver hydroxyproline concentration assays demonstrated that PB reduced liver fibrosis in BDL mice. (D) The degree of liver collagen accumulation was determined by sirius red straining (magnification 200×; up panel), and Masson trichrome stain (magnification 200×; down panel). (E) mRNA expressions ofαsma , Col1a1 , Tgfb1 , and Timp1 in mice liver sample. (F) Western blot analysis and semi‐quantitation of α‐SMA expressions. β-actin served as the loading control. The values are expressed as the mean ± SEM (n = 8 of each group), *P < 0.05, **P < 0.01, ***P < 0.001, significantly different from control group and ###P < 0.001, ####P < 0.0001, significantly different from BDL group. ns, not significant.
FIGURE 4 PB suppressed the GLI1 expression in vitro andin vivo . (A) PB repressed GLI-luciferase activities. (B-C) PB administration down‐regulated GLI1(B) mRNA and (C) protein expressions in LX‐2 cells. GADPH /β-actin served as loading control. (D-E) PB administration down‐regulated GLI1 mRNA(D) and (E) protein expressions in mouse pHSCs. β-actin /β-actin served as loading control. (F) PB administration down‐regulated GLI1 protein expressions in BDL livers. β-actin served as loading control. The in vitro values are expressed as the mean ± SEM of five independent assays, *P <0.05, **P < 0.01, ***P < 0.001, significantly different from the control group, #P < 0.05, ###P < 0.001significantly different from the TGFβ1 treatment group in LX‐2 cells. The in vivo values are expressed as the mean ± SEM (n = 8 of each group), # P<0.05; significantly different from sham group, *P<0.05; significantly different from BDL-NS group. ns, not significant.
FIGURE 5 Altering GLI1 expression influenced the antifibrotic activity of PB. (A-B) LX‐2 cells were transfected with 100 nM GLI1 siRNA1/2 or Ctrl siRNA (A) the mRNA expressions of fibrogenic gene were analyzed by RT-qPCR. GADPH served as loading control. (B) Western blot analysis and semi‐quantitation of CLI1, α‐SMA and Collagen α1(I) expressions. β-actin served as the loading control. (C) LX‐2 cells were transfected with 100 nM GLI1 siRNA1/2 or Ctrl siRNA and subsequently treated with 2 ng·ml−1 TGFβ1 with or without different concentrations of PB for 24 hr after no FBS starvation. The same protein expressions were detected by western blot with the indicated antibodies. β-actin was used as a loading control for all western blot assays. LX‐2 cells were transfected with (D) 2.5‐μg HA‐GLI1 or vector for 6 hr and subsequently treated with 2 ng·ml−1 TGFβ1 with or without different concentrations of PB for 24 hr after no FBS starvation. The protein expressions of HA and liver fibrosis markers were determined by immunoblotting with the indicated antibodies. The data are expressed as the mean ± SEM of five independent assays, *P <0.05, **P < 0.01, ***P < 0.001, significantly different from the Ctrl siRNA group, #P <0.05, ##P < 0.01, ###P < 0.001, significantly different from the pcDNA /Ctrl siRNA group +TGFβ1 group, ▲P < 0.05, significantly different from the pcDNA + TGFβ1 + PB 1-μM group.
FIGURE 6 PB blocked the nuclear localization of GLI1 in HSCs. (A) The nuclear and cytoplasmic extractions of LX‐2 cells were separated after PB treatment and analysed by western blot. (B)LX‐2 cells and (C) mouse pHSCs in dishes were fixed in 4% paraformaldehyde, stained with DAPI (blue) and GLI1 (red), and imaged by confocal microscopy, Scale bar: 20 μm. (D-E) The mRNA expressions of the GLI1 downstream genesHHIP , CYCLIN D , CYCLIN E and C-MYC (D) in LX-2 cells and (E) in BDL group. The values are expressed as the mean ± SEM of five independent assays, *P <0.05, **P < 0.01, ***P < 0.001, significantly different from the control group, #P < 0.05, ###P < 0.001, significantly different from the TGFβ1 treatment group. ns, not significant.
FIGURE 7 PB interrupted the LAP2α/HDAC1 complex. (A) HA-GLI1 was transfected into HEK293T cells. Whole cell extracts were immunoprecipitated with an anti-HA antibody. (B) Western blot analysis of GLI1, LAP2α and HDAC1 expression in LX-2 cells. β-actin served as a loading control. (C) Myc-LAP2α and HA-GLI1 were transfected into HEK293T cells. The interaction between Myc-LAP2α and HA-GLI1 was investigated using immunoprecipitation assays. (D) Myc-LAP2α and FLAG-HDAC1 were transfected into HEK293T cells. The interaction between Myc-LAP2α and FLAG-HDAC1 was investigated using immunoprecipitation assays. The interaction between LAP2α and HDAC1 was investigated using immunoprecipitation assays in LX‐2 cells (E) and (F) CCl4-mice (G) LX‐2 cells in dishes were fixed in 4% paraformaldehyde, stained with DAPI (blue), LAP2α (green) and HDAC1(red) and imaged by confocal microscopy, Scale bar: 20 μm. The values are expressed as the mean ± SEM of five independent assay.
FIGURE 8 LAP2α and HDAC1 were of significance to the antifibrotic effects of PB. (A-C) LX‐2 cells treated with PB (1 μM) and/or Vorinostat (10 μM, 6 hr) then (A) the protein expressions of GLI1 and fibrogenic markers were detected by immunoblotting with the indicated antibodies and (B) the protein expressions of GLI1 were detected by immunofluorescence staining. (C) HEK293T cells was transfected with HA-GLI1 and/or treated with PB (1 μM) or Vorinostat (10 μM, 6 hr), Whole cell extracts were immunoprecipitated with an anti-HA antibody. (D-G) 100‐nM LAP2α‐1/2 siRNA or Ctrl siRNA 48 hr. (D)The fibrotic mRNA and (E)protein expressions were detected by western blot and RT-qPCR. (F) The protein expressions of GLI1 were detected by immunofluorescence staining. (G) HEK293T cells was transfected with HA-GLI1 and/or treated with PB (1 μM) or si‐LAP2α, Whole cell extracts were immunoprecipitated with an anti-HA antibody. The values are expressed as the mean ± SEM of five independent assays, *P <0.05, **P < 0.01, ***P < 0.001, significantly different from the control group.
FIGURE 9 Physalin B (PB) disrupted the interaction between LAP2α and HDAC1, which enhanced the effect of GLI1 acetylation. The increase of GLI1 acetylation further inhibited GLI1 exerting effect on liver fibrosis.