scholarly journals Protective Effects of Pretreatment with Quercetin Against Lipopolysaccharide-Induced Apoptosis and the Inhibition of Osteoblast Differentiation via the MAPK and Wnt/β-Catenin Pathways in MC3T3-E1 Cells

2017 ◽  
Vol 43 (4) ◽  
pp. 1547-1561 ◽  
Author(s):  
Chun Guo ◽  
Rui-Juan Yang ◽  
Ke Jang ◽  
Xiao-ling Zhou ◽  
Yu-zhen Liu

Background/Aims: Quercetin, a flavonoid found in onions and other vegetables, has potential inhibitory effects on bone resorption in vivo and in vitro. In our previous study, we found that quercetin treatment reversed lipopolysaccharide (LPS)-induced inhibition of osteoblast differentiation through the mitogen-activated protein kinase (MAPK) pathway in MC3T3-E1 cells. In this study, we investigated the underlying mechanisms of pretreatment with quercetin on apoptosis and the inhibition of osteoblast differentiation in MC3T3-E1 cells induced by LPS. Methods: MC3T3-E1 osteoblasts were treated with quercetin for 2 h; cells were then incubated with LPS in the presence of quercetin for the indicated times. Cell viability was measured using the Cell Counting Kit-8 (CCK-8) assay, and cell apoptosis was evaluated using Hoechst 33258 staining. The mRNA expression levels of osteoblast-specific genes, Bax and caspase-3 were determined by real-time quantitative polymerase chain reaction (qPCR). Protein levels of osteoblast-specific genes, caspase-3, Bax, cytochrome c, Bcl-2, Bcl-XL, phosphorylated MAPKs and Wnt/β-catenin were measured using Western blot assays. The MAPK and Wnt/β-catenin signalling pathways were blocked prior to pretreatment with quercetin. Results: Pretreatment with quercetin significantly restored LPS-suppressed bone mineralization and the mRNA and protein expression levels of osteoblast-specific genes such as Osterix (OSX), runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP) and osteocalcin (OCN) in a dose-dependent manner. Pretreatment with quercetin also inhibited osteoblast apoptosis, significantly restored the down-regulated expression of Bcl-2 and Bcl-XL and decreased the upregulated expression of caspase-3, Bax, and cytochrome c in MC3T3-E1 cells induced by LPS. Furthermore, pretreatment with quercetin not only decreased the abundance of phosphorylated p38 MAPK and increased the abundance of phosphorylated extracellular signal regulated kinase (ERK), but also triggered the Wnt/β-catenin pathway through enhancing expression of Wnt3 and β-catenin. Pretreatment with MAPK inhibitors or the Wnt/β-catenin inhibitor XAV939 blocked the protective effects of quercetin against LPS-induced apoptosis and the inhibition of osteoblast differentiation. Conclusions: Our findings suggest that pretreatment with quercetin may be a potential drug for preventing abnormal human bone loss induced by LPS in bacteria-induced bone diseases.

2007 ◽  
Vol 292 (6) ◽  
pp. F1710-F1717 ◽  
Author(s):  
Cheng Yang ◽  
Varsha Kaushal ◽  
Sudhir V. Shah ◽  
Gur P. Kaushal

Mcl-1 is an antiapoptotic member of the Bcl-2 family that plays an important role in cell survival. We demonstrate that proteasome-dependent regulation of Mcl-1 plays a critical role in renal tubular epithelial cell injury from cisplatin. Protein levels of Mcl-1 rapidly declined in a time-dependent manner following cisplatin treatment of LLC-PK1cells. However, mRNA levels of Mcl-1 were not altered following cisplatin treatment. Expression of other antiapoptotic members of the Bcl-2 family such as Bcl-2 and BclxL was not affected by cisplatin treatment. Cisplatin-induced loss of Mcl-1 occurs at the same time as the mitochondrial release of cytochrome c, activation of caspase-3, and initiation of apoptosis. Treatment of cells with cycloheximide, a protein synthesis inhibitor, revealed rapid turnover of Mcl-1. In addition, treatment with cycloheximide in the presence or absence of cisplatin demonstrated that cisplatin-induced loss of Mcl-1 results from posttranslational degradation rather than transcriptional inhibition. Overexpression of Mcl-1 protected cells from cisplatin-induced caspase-3 activation and apoptosis. Preincubating cells with the proteasome inhibitor MG-132 or lactacystin not only restored cisplatin-induced loss of Mcl-1 but also resulted in an accumulation of Mcl-1 that exceeded basal levels; however, Bcl-2 and BclxL levels did not change in response to MG-132 or lactacystin. The proteasome inhibitors effectively blocked cisplatin-induced mitochondrial release of cytochrome c, caspase-3 activation, and apoptosis. These studies suggest that proteasome regulation of Mcl-1 is crucial in the cisplatin-induced apoptosis via the mitochondrial apoptotic pathway and that Mcl-1 is an important therapeutic target in cisplatin injury to renal tubular epithelial cells.


2015 ◽  
Vol 2015 ◽  
pp. 1-19 ◽  
Author(s):  
Guy-Armel Bounda ◽  
Wang Zhou ◽  
Dan-dan Wang ◽  
Feng Yu

Objective. To study rhein-induced apoptosis signaling pathway and to investigate its molecular mechanisms in primary human hepatic cells.Results. Cell viability of HL-7702 cells treated with rhein showed significant decrease in dose-dependent manner. Following rhein treatment (25 μM, 50 μM, and 100 μM) for 12 h, the detection of apoptotic cells was significantly analyzed by flow cytometry and nuclear morphological changes by Hoechst 33258, respectively. Fatty degeneration studies showed upregulation level of the relevant hepatic markers (P< 0.01). Caspase activities expressed significant upregulation of caspase-3, caspase-9, and caspase-8. Moreover, apoptotic cells by rhein were significantly inhibited by Z-LEHD-FMK and Z-DEVD-FMK, caspase-9 inhibitor, and caspase-3 inhibitor, respectively. Overproduction of reactive oxygen species, lipid peroxidation, and loss of mitochondrial membrane potential were detected by fluorometry. Additionally, NAC, a ROS scavenger, significantly attenuated rhein-induced oxidative damage in HL-7702 cells. Furthermore, real-time qPCR results showed significant upregulation of p53, PUMA, Apaf-1, and Casp-9 and Casp-3 mRNA, with no significant changes of Fas and Cytochrome-c. Immunoblotting revealed significant Cytochrome-c release from mitochondria into cytosol and no change in Fas expression.Conclusion. Taken together, these observations suggested that rhein could induce apoptosis in HL-7702 cells via mitochondria-mediated signal pathway with involvement of oxidative stress mechanism.


2006 ◽  
Vol 34 (06) ◽  
pp. 1095-1103 ◽  
Author(s):  
Xiao-Shan Liu ◽  
Jikai Jiang

Matrine, a low toxic alkaloid purified from the Chinese herb Kushen, has been reported to induce apoptosis in leukemia K562 cells. In this study, the mechanism underling this apoptotic event was investigated. Treatment of K562 cells with matrine resulted in inhibition of cell survival more significantly than treatment of non-cancer fibroblast NIH3T3 cells. When K562 cells were incubated with matrine in higher than 0.2 mg/ml doses for 48 hours, the apoptotic cells were increased and both poly (ADP-ribose) polymerase (PARP) and caspase-3 were cleaved in a dose dependent manner. General caspase inhibitor (z-VAD-fmk) or caspase-3 inhibitor (z-DEVD-fmk) almost completely suppressed matrine-induced apoptosis. In addition, matrine increased proapoptotic protein bax and caused the release of cytochrome C. Taken together, the results suggest that matrine induces a cytochrome C-mediated, caspase-dependent apoptosis.


2021 ◽  
Author(s):  
qianqian chen ◽  
Yaqin Hou ◽  
Bingjie Hao ◽  
Zhou Ding ◽  
Qing Xia ◽  
...  

Abstract BackgroundNon-small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancers. Berberine (BBR), as an isoquinoline alkaloid, is commonly utilized in traditional Chinese medicine. Previous studies have proven that BBR possesses potential anti-tumor effect. However, the mechanism of on mitochondrial function in anti-NSCLC are still unknown.MethodsCell Counting Kit-8 (CCK-8), flow cytometry and western blotting were utilized to characterize the roles and relationships among BBR, ROS, ASK1, JNK, coxIV,caspase-3, cytochrome c ,bcl-2 and bax in NSCLC. Immunohistochemical (IHC) analysis was built to examine their expression in vivo.ResultsIn this study, we found that BBR potently suppressed NSCLC cells (A549 and PC9) growth by inducing apoptosis in a dose- and time-dependent manner. BBR induced apoptosis in NSCLC as evidenced by caspase-3 cleavage, cytochrome c release, and mitochondrial membrane depolarization. Furthermore, BBR induced ROS generation and ASK1 and JNK activation. To explore whether such apoptosis was linked to ROS production and ASK1 and JNK activation, we treated cells with a JNK inhibitor (SP600125), which significantly suppressed BBR-induced apoptosis. We further found that treating these cells with the anti-oxidant N-acetyl cysteine (NAC) was sufficient to both suppress ASK1 and JNK activation and to disrupt apoptotic induction.ConclusionsTogether, these data suggest that BBR induces NSCLC cells apoptosis via ROS-mediated ASK1/JNK and mitochondrial pathway activation.


Molecules ◽  
2020 ◽  
Vol 25 (3) ◽  
pp. 557 ◽  
Author(s):  
Yongjie Xiong ◽  
Qirun Yin ◽  
Erhui Jin ◽  
Huatao Chen ◽  
Shaojun He

Heat stress induces apoptosis in various cells. Selenium, an essential micronutrient, has beneficial effects in maintaining the cellular physiological functions. However, its potential protective action against chronic heat stress (CHS)-induced apoptosis in granulosa cells and the related molecular mechanisms are not fully elucidated. In this study, we investigated the roles of selenium in CHS-induced apoptosis in mouse granulosa cells and explored its underlying mechanism. The heat treatment for 6–48 h induced apoptosis, potentiated caspase 3 activity, increased the expression levels of apoptosis-related gene BAX and ER stress markers, glucose-regulated protein 78 (GRP78), and CCAAT/enhancer binding protein homologous protein (CHOP) in mouse granulosa cells. The treatment with ER stress inhibitor 4-PBA significantly attenuated the adverse effects caused by CHS. Selenium treatment significantly attenuated the CHS- or thapsigargin (Tg, an ER stress activator)-induced apoptosis, potentiation of caspase 3 activity, and the increased protein expression levels of BAX, GRP78, and CHOP. Additionally, treatment of the cells with 5 ng/mL selenium significantly ameliorated the levels of estradiol, which were decreased in response to heat exposure. Consistently, administering selenium supplement alleviated the hyperthermia-caused reduction in the serum estradiol levels in vivo. Together, our findings indicate that selenium has protective effects on CHS-induced apoptosis via inhibition of the ER stress pathway. The current study provides new insights in understanding the role of selenium during the process of heat-induced cell apoptosis.


2018 ◽  
Vol 47 (4) ◽  
pp. 1377-1388 ◽  
Author(s):  
Pan Lu ◽  
Shan Lei ◽  
Weisong Li ◽  
Yang Lu ◽  
Juan Zheng ◽  
...  

Background/Aims: Ketamine inhibits the proliferation of neural stem cells (NSCs) and disturbs normal neurogenesis. Dexmedetomidine provides neuroprotection against volatile anesthetic-induced neuroapoptosis and cognitive impairment in the developing brain. Whether it may protect NSCs from ketamine-induced injury remains unknown. In this study, we investigated the protective effects of dexmedetomidine on ketamine-exposed NSCs and explored the mechanisms potentially involved. Methods: Primary NSC cultures were characterized using immunofluorescence. Cell viability was determined using a Cell Counting Kit 8 assay. Proliferation and apoptosis were assessed with BrdU incorporation and TUNEL assays, respectively. Protein levels of cleaved caspase-3, phosphorylated protein kinase B (p-Akt), and glycogen synthase kinase-3β (p-GSK-3β) were quantified using western blotting. Results: Ket-amine significantly decreased NSC viability and proliferation and increased their apoptosis. Dexmedetomidine increased NSC proliferation and decreased their apoptosis in a dose-dependent manner. Furthermore, dexmedetomidine pretreatment notably augmented the viability and proliferation of ketamine-exposed NSCs and reduced their apoptosis. Moreover, dexmedetomidine lessened caspase-3 activation and increased p-Akt and p-GSK-3β levels in NSCs exposed to ketamine. The protective effects of dexmedetomidine on ketamine-exposed NSCs could be partly reversed by the PI3K inhibitor LY294002. Conclusions: Collectively, these findings indicate that dexmedetomidine may protect NSCs from ketamine-induced injury via the PI3K/Akt/GSK-3β signaling pathway.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4615-4615 ◽  
Author(s):  
Azhar R. Hussain ◽  
Naif A. Al-Jomah ◽  
Abdul K. Siraj ◽  
Manugaran S. Pulicat ◽  
Khaled A. Al-Hussein ◽  
...  

Abstract Primary effusion lymphoma (PEL) is an aggressive and fatal type of cancer. PEL cells produce a variety of autocrine cytokines and growth factors, which provides cyto-protection against conventional chemotherapeutic agents. In efforts to identify novel approaches to block the proliferation of PEL cells, we found that Sanguinarine, a natural compound isolated from the root plant Sanguinaria canadendid, that is being used as an anti-microbial agent, inhibited cell proliferation and induced apoptosis in a dose dependent manner in several PEL cell lines through a bax-dependent signaling pathway. Five PEL cell lines used in this study were treated with various doses of Sanguinarine ranging between 0.5–4μM inhibited cell proliferation in all the cell lines in a dose dependent manner (BC1 40–97%, BC3 46–93%, BCBL1 11–94%, BCP1 20–97% and HBL6 7–95%). Treatment with varying doses of Sanguinarine also induced apoptosis in all cell lines as determined by cell cycle analysis, annexinV/PI dual staining, TUNEL assay and DNA laddering. Sanguinarine treatment resulted in up-regulation of death receptor 5 (DR5) expression, activation of caspase-8 and Bid leading to Bax conformational changes and translocation to the mitochondrial causing loss of mitochondrial membrane potential as measured by JC1 staining and release of cytochrome c to the cytosole. Sanguinarine induced release of cytochrome c resulted in activation of caspase-3, followed by polyadenosin-5′-diphosphate-ribose polymerase (PARP) cleavage leading to inhibition of proliferation and induction of caspase-dependent apoptosis. Furthermore, pre-treatment of PEL cells with z-VAD-fmk, a universal inhibitor of caspases, abrogated caspase-3 and PARP activation and prevented cell death induced by Sanguinarine. Inhibitor of apoptosis proteins (IAPs), play an important role in protecting cells against apoptosis through their direct action on caspases-9 and -3. Treatment of PEL cells with Sanguinarine down-regulated the expression of IAPs; XIAP, cIAP1 and cIAP2. Taken altogether, our findings suggest that Sanguinarine induces apoptosis via up-regulation of DR5, activation of Bax in a caspase-dependent pathway and down-regulation of IAPs. These results provide the molecular basis and preliminary data for new treatment strategies that may incorporate Sanguinarine in regimens for primary effusion lymphoma treatment.


1999 ◽  
Vol 189 (1) ◽  
pp. 131-144 ◽  
Author(s):  
Glen MacDonald ◽  
Lianfa Shi ◽  
Christine Vande Velde ◽  
Judy Lieberman ◽  
Arnold H. Greenberg

Granzyme B (GraB) is required for the efficient activation of apoptosis by cytotoxic T lymphocytes and natural killer cells. We find that GraB and perforin induce severe mitochondrial perturbation as evidenced by the release of cytochrome c into the cytosol and suppression of transmembrane potential (Δψ). The earliest mitochondrial event was the release of cytochrome c, which occurred at the same time as caspase 3 processing and consistently before the activation of apoptosis. Granzyme K/perforin or perforin treatment, both of which kill target cells efficiently but are poor activators of apoptosis in short-term assays, did not induce rapid cytochrome c release. However, they suppressed Δψ and increased reactive oxygen species generation, indicating that mitochondrial dysfunction is also associated with this nonapoptotic cell death. Pretreatment with peptide caspase inhibitors zVAD-FMK or YVAD-CHO prevented GraB apoptosis and cytochrome c release, whereas DEVD-CHO blocked apoptosis but did not prevent cytochrome c release, indicating that caspases act both up- and downstream of mitochondria. Of additional interest, Δψ suppression mediated by GraK or GraB and perforin was not affected by zVAD-FMK and thus was caspase independent. Overexpression of Bcl-2 and Bcl-XL suppressed caspase activation, mitochondrial cytochrome c release, Δψ suppression, and apoptosis and cell death induced by GraB, GraK, or perforin. In an in vitro cell free system, GraB activates nuclear apoptosis in S-100 cytosol at high doses, however the addition of mitochondria amplified GraB activity over 15-fold. GraB- induced caspase 3 processing to p17 in S-100 cytosol was increased only threefold in the presence of mitochondria, suggesting that another caspase(s) participates in the mitochondrial amplification of GraB apoptosis. We conclude that GraB-induced apoptosis is highly amplified by mitochondria in a caspase-dependent manner but that GraB can also initiate caspase 3 processing and apoptosis in the absence of mitochondria.


2019 ◽  
Vol 19 (9) ◽  
pp. 673-682 ◽  
Author(s):  
Panpan Chang ◽  
Yuzi Tian ◽  
Aaron M. Williams ◽  
Umar F. Bhatti ◽  
Baoling Liu ◽  
...  

Background: Histone deacetylase (HDAC) 6 inhibitors have demonstrated significant protective effects in traumatic injuries. However, their roles in neuroprotection and underlying mechanisms are poorly understood. This study sought to investigate the neuroprotective effects of Tubastatin A (Tub-A), an HDAC6 inhibitor, during oxygenglucose deprivation (OGD) in HT22 hippocampal cells. Methods: HT22 hippocampal cells were exposed to OGD. Cell viability and cytotoxicity were assessed by cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) release assay. Cellular apoptosis was assessed by Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Mitochondria membrane potential was detected using JC-1 dye. Expressions of acetylated α-tubulin, α-tubulin, cytochrome c, VDAC, Bax, Bcl- 2, cleaved caspase 3, phosphorylated Akt, Akt, phosphorylated GSK3β and GSK3β were analyzed by Western blot analysis. Results: Tub-A induced acetylation of α-tubulin, demonstrating appropriate efficacy. Tub-A significantly increased cell viability and attenuated LDH release after exposure to OGD. Furthermore, Tub-A treatment blunted the increase in TUNEL-positive cells following OGD and preserved the mitochondrial membrane potential. Tub-A also attenuated the release of cytochrome c from the mitochondria into the cytoplasm and suppressed the ratio of Bax/Bcl-2 and cleaved caspase 3. This was mediated, in part, by the increased phosphorylation of Akt and GSK3β signaling pathways. Conclusion: HDAC 6 inhibition, using Tub-A, protects against OGD-induced injury in HT22 cells by modulating Akt/GSK3β signaling and inhibiting mitochondria-mediated apoptosis.


2020 ◽  
Vol 2020 ◽  
pp. 1-14
Author(s):  
Tian-Qi Wang ◽  
Xiao-Xiao Lai ◽  
Lu-Ting Xu ◽  
Yan Shen ◽  
Jian-Wei Lin ◽  
...  

Qingxin kaiqiao fang (QKF), a traditional Chinese medicine compound, has been applied to treat Alzheimer’s disease (AD) for many years and has exhibited remarkable effects. However, the underlying mechanism is still not explicit. The current study aims to investigate whether QKF exerts an antiapoptotic role through the p38 MAPK pathway in the course of AD. Network pharmacology analysis was applied to study the effective components, possible therapeutic targets, and AD-related pathway of QKF. Further, the AD cell model was established using amyloid-beta (Aβ)25-35 peptide and primary hippocampal neuronal cells extracted from newborn Sprague-Dawley rats. Microtubule-associated protein-2 (MAP-2) imaging was used to detect the morphology of hippocampal neurons. Western blot (WB) analysis was applied to detect the protein expression levels of p38 MAPK, p-p38 MAPK, Bcl-2, Bax, caspase-3, and cleaved caspase-3. Cell viability and apoptosis were determined using cell counting kit-8 (CCK-8) and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) assays, respectively. SB203580 and U46619 were used to detect changes in cell morphology, cell viability, and apoptosis upon inhibiting or activating p38 MAPK. Our present work showed that QKF protects hippocampal neuronal morphology, enhances cell viability, and reduces the number of TUNEL-positive cells. In addition, our results showed that QKF increased the expression levels of antiapoptotic proteins and decreased the expression of proapoptotic proteins. QKF at 25 mg·mL−1 best inhibited neuronal apoptosis among the three doses of QKF by suppressing p38 MAPK activity. Collectively, QKF plays an antiapoptotic role via the p38 MAPK pathway.


Sign in / Sign up

Export Citation Format

Share Document