PGK1 depletion activates Nrf2 signaling to protect human osteoblasts from dexamethasone

Jinqian Liang, Xiang-Yang Zhang, Yun-Fang Zhen, Chong Chen, Haining Tan, Jianhua Hu, Ming-Sheng Tan, Jinqian Liang, Xiang-Yang Zhang, Yun-Fang Zhen, Chong Chen, Haining Tan, Jianhua Hu, Ming-Sheng Tan

Abstract

Activation of nuclear-factor-E2-related factor 2 (Nrf2) cascade can alleviate dexamethasone (DEX)-induced oxidative injury and death of human osteoblasts. A recent study has shown that phosphoglycerate kinase 1 (PGK1) inhibition/depletion will lead to Kelch-like ECH-associated protein 1 (Keap1) methylglyoxal modification, thereby activating Nrf2 signaling cascade. Here, in OB-6 osteoblastic cells and primary human osteoblasts, PGK1 silencing, by targeted shRNA, induced Nrf2 signaling cascade activation, causing Nrf2 protein stabilization and nuclear translocation, as well as increased expression of ARE-dependent genes (HO1, NQO1, and GCLC). Functional studies demonstrated that PGK1 shRNA largely attenuated DEX-induced oxidative injury and following death of OB-6 cells and primary osteoblasts. Furthermore, PGK1 knockout, by the CRISPR/Cas9 method, similarly induced Nrf2 signaling activation and protected osteoblasts from DEX. Importantly, PGK1 depletion-induced osteoblast cytoprotection against DEX was almost abolished by Nrf2 shRNA. In addition, Keap1 shRNA mimicked and nullified PGK1 shRNA-induced anti-DEX osteoblast cytoprotection. At last we show that PGK1 expression is downregulated in human necrotic femoral head tissues of DEX-taking patients, correlating with HO1 depletion. Collectively, these results show that PGK1 depletion protects human osteoblasts from DEX via activation of Keap1-Nrf2 signaling cascade.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1. PGK1 silencing activates Nrf2 signaling…
Fig. 1. PGK1 silencing activates Nrf2 signaling in human osteoblasts.
Expression of listed genes (mRNAs and proteins) in stable OB-6 osteoblastic cells, with applied PGK1 shRNA (“sh-PGK1-S1/S2”) or the non-sense control shRNA (“sh-C”), were tested by qPCR and western blotting assays (ac, f). The relative NQO1 activity was tested as well (g). OB-6 cells were treated with MG-132 (10 μM) or plus sh-PGK1-S1 infection (“+sh-PGK1-S1”), after 24 h Nrf2 and Erk1/2 (the loading control) protein expression in total cell lysates was shown (d). Stable OB-6 cells with sh-PGK1-S1 were treated with cycloheximide (CHX, 100 μg/mL) for 12 and 24 h, Nrf2 and Erk1/2 protein expression in total cell lysates was shown (e). The primary human osteoblasts were transduced with lentiviral PGK1 shRNA (“sh-PGK1-S1”) or the lentiviral non-sense control shRNA (“sh-C”), expression of listed genes (hj) and the relative NQO1 activity (k) were tested. Expression of listed proteins was quantified, normalized to the loading control (a, ce, h, i). Data were expressed as mean ± standard deviation (SD, n = 5). *p< 0.05 vs. “sh-C” cells. Experiments were repeated four times, with similar results obtained.
Fig. 2. PGK1 silencing protects human osteoblasts…
Fig. 2. PGK1 silencing protects human osteoblasts from DEX-induced death and apoptosis.
Stable OB-6 osteoblastic cells (af) or primary human osteoblasts (gi) with applied PGK1 shRNA (“sh-PGK1-S1/S2”) or the non-sense control shRNA (“sh-C”) were treated with or without DEX (1 μM) for applied time periods, cell viability (MTT assay; a and g) and cell death (LDH medium release; b and h) were tested; Cell apoptosis activation was tested by the listed assays (cf, i). Expression of listed proteins was quantified, normalized to the loading control (d). Data were expressed as mean ± standard deviation (SD, n = 5). “Ctrl” represents untreated control group (Same for all Figures). *p < 0.05 vs. “sh-C” cells with “Ctrl” treatment. #p < 0.05 vs. “sh-C” cells with DEX treatment. Experiments in this figure were repeated four times, and similar results were obtained.
Fig. 3. PGK1 silencing attenuates DEX-induced oxidative…
Fig. 3. PGK1 silencing attenuates DEX-induced oxidative stress and programmed necrosis in human osteoblasts.
Stable OB-6 human osteoblastic cells (af) or primary human osteoblasts (g, h) with applied PGK1 shRNA (“sh-PGK1-S1/S2”) or the non-sense control shRNA (“sh-C”), were treated with or without DEX (1 μM) for applied time periods, oxidative stress (a, b, g) and programmed necrosis (cf, h) were tested by the listed assays mentioned in the text. For mitochondrial depolarization assay, JC-1 green intensity was examined at 550 nm via a fluorescence spectrofluorometer (results normalized to the control level), and the representative JC-1 fluorescence images, merging both green and red fluorescence pictures, were presented (c, d). Expression of listed proteins was quantified, normalized to the loading control (e, f). Data were expressed as mean ± standard deviation (SD, n = 5). *p < 0.05 vs. “sh-C” cells with “Ctrl” treatment. #p < 0.05 vs. “sh-C” cells with DEX treatment. Experiments in this figure were repeated four times, and similar results were obtained. Bar = 100 μm (c).
Fig. 4. PGK1 knockout activates Nrf2 signaling…
Fig. 4. PGK1 knockout activates Nrf2 signaling and protects osteoblasts from DEX.
Stable OB-6 osteoblastic cells (ag) or primary human osteoblasts (hk) with the CRISPR/Cas9-PGK1 knockout construct (“ko-PGK1”) or the CRISPR/Cas9 sgRNA control construct (“sg-C”) were established, expression of listed genes (a, b, h) was shown. The relative NQO1 activity was also tested (c). Cells were further treated with or without DEX (1 μM) for applied time periods, cellular superoxide levels were shown (d); cell viability (e, i), death (f, j), and apoptosis (g, k) were examined by MTT, LDH release, and Histone-DNA ELISA assays, respectively. Expression of listed proteins was quantified, normalized to the loading control (a, h). Data were expressed as mean ± standard deviation (SD, n = 5). *p < 0.05 vs. “sg-C” cells with “Ctrl” treatment. #p < 0.05 vs. “sg-C” cells with DEX treatment. Experiments in this figure were repeated three times, and similar results were obtained.
Fig. 5. PGK1 depletion-induced osteoblast cytoprotection against…
Fig. 5. PGK1 depletion-induced osteoblast cytoprotection against DEX is through activation of Keap1-Nrf2 cascade.
OB-6 cells with the CRISPR/Cas9-PGK1-knockout construct (“ko-PGK1”) were further transduced with Nrf2 shRNA (“+sh-Nrf2”) or the non-sense control shRNA (“+sh-C”), stable cells were established via selection by puromycin, relative expression of Nrf2 pathway genes was shown (a, b). The NQO1 activity was also tested (c). Above cells and the control cells (with CRISPR/Cas9 sgRNA control construct/sg-C) were further treated with or without DEX (1 μM) for applied time periods, cell viability (d), death (e) and apoptosis (f) were examined by MTT, LDH release, and Histone-DNA ELISA assays, respectively. Expression of the listed genes in the stable OB-6 cells with the non-sense control shRNA (“sh-C”), the lentiviral Keap1 shRNA (“sh-Keap1”), or together with the PGK1 shRNA (“sh-PGK1-S1”, sh-Keap1 + sh-PGK1) was shown (g, h), the relative NQO1 activity was tested as well (i). The above cells were treated with or without DEX (1 μM) for applied time periods, cell viability (j), death (k), and apoptosis (l) were tested similarly. Expression of listed proteins was quantified, normalized to the loading control (b, g). Data were expressed as mean ± standard deviation (SD, n = 5). *p < 0.05 vs. “ko-PGK1” cells (a, c); *p < 0.05 (df); #p < 0.05 vs. “sh-C” cells (hl). *p < 0.05 vs. “Ctrl” treatment (jl). p > 0.05 stands for no statistical difference. Experiments in this figure were repeated three times, and similar results were obtained.
Fig. 6. PGK1 downregulation in human necrotic…
Fig. 6. PGK1 downregulation in human necrotic femoral head tissues correlates with HO1 depletion.
mRNA (a, b) and protein (ce) expression of PGK1 and HO1 in necrotic femoral head tissues (“N”) and surrounding normal femoral head tissues (“S”) of 12 different DEX-taking patients was tested by qPCR and western blotting assays. f The proposed signaling carton of this study. Data were expressed as mean ± standard deviation (SD, n = 12). *p < 0.05 vs. “S” tissues. Experiments in this figure were repeated three times, and similar results obtained.

References

    1. Schacke H, Docke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids. Pharm. Ther. 2002;96:23–43. doi: 10.1016/S0163-7258(02)00297-8.
    1. Ding H, et al. Dexamethasone-induced apoptosis of osteocytic and osteoblastic cells is mediated by TAK1 activation. Biochem. Biophys. Res. Commun. 2015;460:157–163. doi: 10.1016/j.bbrc.2015.02.161.
    1. Yun SI, Yoon HY, Jeong SY, Chung YS. Glucocorticoid induces apoptosis of osteoblast cells through the activation of glycogen synthase kinase 3beta. J. Bone Min. Metab. 2009;27:140–148. doi: 10.1007/s00774-008-0019-5.
    1. Fan JB, et al. miR-135b expression downregulates Ppm1e to activate AMPK signaling and protect osteoblastic cells from dexamethasone. Oncotarget. 2016;7:70613–70622.
    1. Guo S, Chen C, Ji F, Mao L, Xie Y. PP2A catalytic subunit silence by microRNA-429 activates AMPK and protects osteoblastic cells from dexamethasone. Biochem. Biophys. Res. Commun. 2017;487:660–665. doi: 10.1016/j.bbrc.2017.04.111.
    1. Zhao S, et al. MicroRNA-200a activates Nrf2 signaling to protect osteoblasts from dexamethasone. Oncotarget. 2017;8:104867–104876.
    1. Liu G, et al. microRNA-19a protects osteoblasts from dexamethasone via targeting TSC1. Oncotarget. 2018;9:2017–2027.
    1. Li ST, et al. SC79 rescues osteoblasts from dexamethasone though activating Akt-Nrf2 signaling. Biochem Biophys. Res. Commun. 2016;479:54–60. doi: 10.1016/j.bbrc.2016.09.027.
    1. Li W, Kong AN. Molecular mechanisms of Nrf2-mediated antioxidant response. Mol. Carcinog. 2009;48:91–104. doi: 10.1002/mc.20465.
    1. Nguyen T, Yang CS, Pickett CB. The pathways and molecular mechanisms regulating Nrf2 activation in response to chemical stress. Free Radic. Biol. Med. 2004;37:433–441. doi: 10.1016/j.freeradbiomed.2004.04.033.
    1. Ma Q, He X. Molecular basis of electrophilic and oxidative defense: promises and perils of Nrf2. Pharm. Rev. 2012;64:1055–1081. doi: 10.1124/pr.110.004333.
    1. Itoh K, Mimura J, Yamamoto M. Discovery of the negative regulator of Nrf2, Keap1: a historical overview. Antioxid. Redox Signal. 2010;13:1665–1678. doi: 10.1089/ars.2010.3222.
    1. Nguyen T, Nioi P, Pickett CB. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J. Biol. Chem. 2009;284:13291–13295. doi: 10.1074/jbc.R900010200.
    1. Liu W, et al. Icariside II activates EGFR-Akt-Nrf2 signaling and protects osteoblasts from dexamethasone. Oncotarget. 2017;8:2594–2603.
    1. Xu YY, et al. Activation of AMP-activated protein kinase by compound 991 protects osteoblasts from dexamethasone. Biochem Biophys. Res. Commun. 2018;495:1014–1021. doi: 10.1016/j.bbrc.2017.11.132.
    1. Xu D, et al. microRNA-455 targets cullin 3 to activate Nrf2 signaling and protect human osteoblasts from hydrogen peroxide. Oncotarget. 2017;8:59225–59234.
    1. Zhang Y, et al. Macrophage-associated PGK1 phosphorylation promotes aerobic glycolysis and tumorigenesis. Mol. Cell. 2018;71:201–215 e207. doi: 10.1016/j.molcel.2018.06.023.
    1. Li X, Zheng Y, Lu Z. PGK1 is a new member of the protein kinome. Cell Cycle. 2016;15:1803–1804. doi: 10.1080/15384101.2016.1179037.
    1. Bollong MJ, et al. A metabolite-derived protein modification integrates glycolysis with KEAP1-NRF2 signalling. Nature. 2018;562:600–604. doi: 10.1038/s41586-018-0622-0.
    1. Xu Y, et al. Hepcidin increases intracellular Ca2+ of osteoblast hFOB1.19 through L-type Ca2+ channels. Regul. Pept. 2011;172:58–61. doi: 10.1016/j.regpep.2011.08.009.
    1. Fan JB, et al. microRNA-7 inhibition protects human osteoblasts from dexamethasone via activation of epidermal growth factor receptor signaling. Mol. Cell Biochem. 2019;460:113–121. doi: 10.1007/s11010-019-03575-y.
    1. Wu F, et al. miR-1273g silences MAGEA3/6 to inhibit human colorectal cancer cell growth via activation of AMPK signaling. Cancer Lett. 2018;435:1–9. doi: 10.1016/j.canlet.2018.07.031.
    1. Fan JB, et al. EGFR trans-activation mediates pleiotrophin-induced activation of Akt and Erk in cultured osteoblasts. Biochem. Biophys. Res. Commun. 2014;447:425–430. doi: 10.1016/j.bbrc.2014.04.002.
    1. Zhen YF, et al. P53 dependent mitochondrial permeability transition pore opening is required for dexamethasone-induced death of osteoblasts. J. Cell Physiol. 2014;229:1475–1483. doi: 10.1002/jcp.24589.
    1. Ye XT, Huang H, Huang WP, Hu WL. LncRNA THOR promotes human renal cell carcinoma cell growth. Biochem. Biophys. Res. Commun. 2018;501:661–667. doi: 10.1016/j.bbrc.2018.05.040.
    1. Brooks MM, Neelam S, Fudala R, Gryczynski I, Cammarata PR. Lenticular mitoprotection. Part A: monitoring mitochondrial depolarization with JC-1 and artifactual fluorescence by the glycogen synthase kinase-3beta inhibitor, SB216763. Mol. Vis. 2013;19:1406–1412.
    1. Zitka O, et al. Redox status expressed as GSH:GSSG ratio as a marker for oxidative stress in paediatric tumour patients. Oncol. Lett. 2012;4:1247–1253. doi: 10.3892/ol.2012.931.
    1. Zhang X, Yang H, Zhao L, Li G, Duan Y. Circular RNA PRKCI promotes glioma cell progression by inhibiting microRNA-545. Cell Death Dis. 2019;10:616. doi: 10.1038/s41419-019-1863-z.
    1. Zhu LQ, et al. Salinomycin activates AMP-activated protein kinase-dependent autophagy in cultured osteoblastoma cells: a negative regulator against cell apoptosis. PLoS ONE. 2013;8:e84175. doi: 10.1371/journal.pone.0084175.
    1. Zhang XY, Shan HJ, Zhang P, She C, Zhou XZ. LncRNA EPIC1 protects human osteoblasts from dexamethasone-induced cell death. Biochem. Biophys. Res. Commun. 2018;503:2255–2262. doi: 10.1016/j.bbrc.2018.06.146.
    1. Ruan JW, Yao C, Bai JY, Zhou X. Z. microRNA-29a inhibition induces Gab1 upregulation to protect OB-6 human osteoblasts from hydrogen peroxide. Biochem. Biophys. Res. Commun. 2018;503:607–614. doi: 10.1016/j.bbrc.2018.06.048.
    1. Fan JB, et al. Long non-coding RNA MALAT1 protects human osteoblasts from dexamethasone-induced injury via activation of PPM1E-AMPK signaling. Cell Physiol. Biochem. 2018;51:31–45. doi: 10.1159/000495159.
    1. Fan JB, et al. EGFR-AKT-mTOR activation mediates epiregulin-induced pleiotropic functions in cultured osteoblasts. Mol. Cell Biochem. 2015;398:105–113. doi: 10.1007/s11010-014-2210-4.
    1. Zhu CY, Yao C, Zhu LQ, She C, Zhou XZ. Dexamethasone-induced cytotoxicity in human osteoblasts is associated with circular RNA HIPK3 downregulation. Biochem. Biophys. Res. Commun. 2019;516:645–652. doi: 10.1016/j.bbrc.2019.06.073.
    1. Zhao S, Chen C, Wang S, Ji F, Xie Y. MHY1485 activates mTOR and protects osteoblasts from dexamethasone. Biochem. Biophys. Res. Commun. 2016;481:212–218. doi: 10.1016/j.bbrc.2016.10.104.
    1. Liu H, et al. K6PC-5 activates SphK1-Nrf2 signaling to protect neuronal cells from oxygen glucose deprivation/re-oxygenation. Cell Physiol. Biochem. 2018;51:1908–1920. doi: 10.1159/000495716.
    1. Suzuki T, Yamamoto M. Molecular basis of the Keap1-Nrf2 system. Free Radic. Biol. Med. 2015;88:93–100. doi: 10.1016/j.freeradbiomed.2015.06.006.
    1. Miura G. Playing KEAP1-away. Nat. Chem. Biol. 2019;15:2. doi: 10.1038/s41589-018-0197-y.
    1. Gourlay M, Franceschini N, Sheyn Y. Prevention and treatment strategies for glucocorticoid-induced osteoporotic fractures. Clin. Rheumatol. 2007;26:144–153. doi: 10.1007/s10067-006-0315-1.
    1. Inkielewicz-Stepniak I, Radomski MW, Wozniak M. Fisetin prevents fluoride- and dexamethasone-induced oxidative damage in osteoblast and hippocampal cells. Food Chem. Toxicol. 2012;50:583–589. doi: 10.1016/j.fct.2011.12.015.
    1. Guo S, et al. Activating AMP-activated protein kinase by an alpha1 selective activator compound 13 attenuates dexamethasone-induced osteoblast cell death. Biochem. Biophys. Res. Commun. 2016;471:545–552. doi: 10.1016/j.bbrc.2016.02.036.
    1. Furfaro AL, et al. The Nrf2/HO-1 axis in cancer cell growth and chemoresistance. Oxid. Med. Cell Longev. 2016;2016:1958174. doi: 10.1155/2016/1958174.

Source: PubMed

3
Suscribir