Cistanche tubulosa phenylethanoid glycosides induce apoptosis in H22 hepatocellular carcinoma cells through both extrinsic and intrinsic signaling pathways (2024)

  • Research article
  • Open access
  • Published:

BMC Complementary and Alternative Medicine volume18, Articlenumber:275 (2018) Cite this article

  • 2244 Accesses

  • 15 Citations

  • 1 Altmetric

  • Metrics details

Abstract

Background

Cistanche tubulosa (Schenk) R. Wight is a traditional Chinese medicine that parasitizes the roots of the Tamarix plant and has been used to treat male impotence, sterility, body weakness, and as a tonic. However, its antitumor effect on hepatocellular carcinoma is still elusive. Here, we investigated the antitumor effect of C. tubulosa phenylethanoid glycosides (CTPG) on H22 hepatocellular carcinoma cells both in vitro and in vivo and its mechanisms.

Methods

The morphology, viability, apoptosis, cell cycle and mitochondrial membrane potential (Δψm) of H22 cells were analyzed by inverted microscopy, MTT assay and flow cytometry, respectively. The expression and activation of proteins in apoptosis pathway were detected by Western blot. The in vivo antitumor effect was evaluated in tumor mouse model established using male Kunming mice.

Results

CTPG treatment significantly suppressed H22 cell growth in a dose- and time-dependent manner, which was correlated with the increased apoptosis and cell cycle arrest at G0/G1 and G2/M phases. Moreover, the chromosomal condensation was observed in CTPG-treated H22 cells. CTPG treatment significantly increased Bax/Bcl-2 ratio, reduced Δψm and enhanced the release of cytochrome c. The levels of cleaved caspase-8 and caspase-9 in both extrinsic and intrinsic signaling pathways were significantly increased that sequentially activated caspase-7 and -3 to cleave PARP. Finally, CTPG inhibited the growth of H22 cells in mice and improved the survival rate of tumor mice.

Conclusions

These results suggested that CTPG suppressed H22 cell growth through both extrinsic and intrinsic apoptosis pathways.

Peer Review reports

Background

Liver cancer ranked sixth for cancer incidence and fourth for cancer deaths worldwide. Moreover, it ranked fourth for cancer incidence and first for cancer mortality in countries with low sociodemographic index [1]. In China, liver cancer is the third leading cause of cancer-related death in 2015 [2]. More than 90% of primary liver cancers are hepatocellular carcinoma (HCC) in the world [3]. Currently, hepatic resection is the main option for the therapy of HCC. However, less than 30% of patients with HCC met the criteria of curative hepatic resection and the over-all 5-year survival rate is still as low as 35-50% due to the high recurrence rate [4, 5]. The availability of treatment options for the patients with intermediate to advanced HCC is very limited. Sorafenib, a molecular targeted drug, has been approved by FDA as the first-line treatment for advanced HCC. However, sorafenib only prolongs about 3months of survival and the response rate is less than 4% [6, 7]. It is urgent to develop new drugs or strategies against HCC.

Traditional Chinese medicine (TCM) alone or combined with other strategies has been used to treat HCC and shown the clinical benefits including prolonged survival time, improved life quality, reduced adverse reactions, and so on [8, 9]. Cistanche, a kind of TCM, has various biological functions, such as anti-oxidation, anti-inflammation, anti-aging and neuroprotection [10, 11]. Phenylethanoid glycosides have been considered the major active components of Cistanche, which have diverse activities including anti-oxidation, anti-inflammation, hepatoprotection and neuroprotection [12,13,14,15]. Our group has reported that Cistanche tubulosa phenylethanoid glycosides (CTPG) could induce apoptosis in melanoma B16-F10 cells and inhibited the growth of tumor in mice [16]. In this study, we measured the antitumor effect of CTPG on HCC H22 cells both in vitro and in vivo and investigated its mechanisms. We found that CTPG induced apoptosis in H22 cells through both extrinsic and intrinsic signaling pathways and suppressed the growth of H22 tumor in mice.

Methods

Cell line

The mouse H22 hepatocellular carcinoma cells were obtained from the Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University (Urumqi, Xinjiang, China) and cultured in RPMI 1640 medium (Gibco) supplemented with 100U/ml penicillin and 100μg/ml streptomycin, and 10% heat-inactivated fetal bovine serum (Gibco) at 37°C in a humidified atmosphere of 5% CO2.

MTT assay

CTPG was purchased from Hetian Dichen Biotech Co., Ltd. (Hetian, Xinjiang, China) and the major compounds of CTPG were qualified and quantified by high performance liquid chromatography [16]. Cell viability was evaluated by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) (Sigma, St. Louis, MO, USA) assay. H22 cells were inoculated into 96-well plates at a density of 2 × 104 cells in 100μl medium per well and cultured at 37°C. After 24h, cells were treated with different concentrations of CTPG (0, 100, 200, 300 and 400μg/ml) or 0.3% DMSO (equal to that in 400μg/ml CTPG) for 24, 48 and 72h, respectively. After centrifugation at 1000rpm for 7min, supernatant was discarded and 100μl of MTT solution (5mg/ml in PBS) was added to each well. The plates were incubated at 37°C for 4h and 100μl DMSO was added to dissolve the formed formazan crystals. The OD490 values were detected by a 96-well microplate reader (Bio-Rad Laboratories, CA, USA). The cell viability was calculated according to the formula: Cell viability (%) = (ODtreated/ODuntreated) × 100%.

Detection of apoptosis

H22 cells were treated with different concentrations of CTPG (0, 100, 200, 300 and 400μg/ml) or 0.3% DMSO for 24h, and then stained with Annexin V-FITC/Propidium iodide (PI) Apoptosis Detection Kit (YEASEN, China) according to the manufacturer’s instructions. Samples were analyzed by flow cytometry (BD FACSCalibur, USA).

Detection of mitochondrial membrane potential

H22 cells were treated with different concentrations of CTPG (0, 200 and 400μg/ml) for 24h, and then stained with the membrane-permeable JC-1 dye (Beyotime,China) for 20min at 37°C. After washing twice with JC-1 buffer, samples were resuspended with 300μl of JC-1 buffer and analyzed by flow cytometry (BD FACSCalibur, USA).

Analysis of cell cycle

H22 cells were inoculated in 60mm culture dishes and treated with different concentrations of CTPG (0, 100, 200, 300 and 400μg/ml) or 0.3% DMSO for 24h. All cells were collected and washed twice with PBS. Cells were fixed in 70% ice-cold ethanol at − 20°C for 2h and washed twice with PBS, then re-suspended in 300μl Propidium iodide/RNase staining buffer (BD Biosciences). After 10min at room temperature, samples were collected by flow cytometry (BD FACSCalibur, USA) and cell cycle distribution was analyzed with the ModFit LT 3.0 software.

Hoechst 33,258 staining

The morphological changes of H22 cell nuclei were analyzed by membrane-permeable DNA-binding dye Hoechst 33,258 staining. H22 cells were seeded in 6-well plate at the concentration of 1 × 105 cells/well in 2ml medium. After 60%~ 70% confluence, the cells were treated with CTPG (0, 100, 200, 300 and 400μg/ml) for 24h. The cells were collected and fixed with 4% ice-cold Paraformaldehyde at 4°C for 10min. After washing with PBS, cells were stained with Hoechst 33,258 (Beyotime, China) at 4°C for 10min. Samples were observed by inverted fluorescence microscope (Nikon Eclipse Ti-E, Japan).

Western blot

Anti-caspase-3, anti-cleaved caspase-3, Anti-Bcl-2 and anti-Bax were purchased from Beyotime Biotech Co., Ltd. (Shanghai, China). Anti-caspase-7, anti-cleaved-caspase-7, anti-caspase-8, anti-cleaved-caspase-8, anti-caspase-9, anti-cleaved-caspase-9, anti-PARP, anti-cleaved PARP, anti-mouse IgG-HRP and anti-rabbit IgG-HRP were purchased from Cell Signaling Technology. Anti-β-actin was purchased from Beijing ComWin Biotech Co., Ltd. (Beijing, China).

H22 cells were treated with different concentrations of CTPG (0, 100, 200, 300 and 400μg/ml) or 0.3% DMSO for 24h. Cells were collected and lysed with the Cell Lysis Solution RIPA (Beijing ComWin Biotech Co., Ltd) for 30min on ice. Samples were spun down (12,000g for 15min at 4°C) to collect the supernatants and protein concentrations were measured by BCA Kit (Thermo Fisher Scientific, USA). Equal amount of protein in each sample was isolated by 12% SDS-PAGE and transferred to PVDF membranes (Biosharp, China). After blocking with TBST buffer contained 5% nonfat milk, membranes were incubated with corresponding primary antibodies and secondary antibodies conjugated to horseradish peroxidase (HRP), respectively. After washing with TBST, the target proteins were detected by ECL assay kit (Beyotime, China).

Animals and ethics statement

6-8weeks old male Kunming mice were purchased from Animal Laboratory Center, Xinjiang Medical University (Urumqi, Xinjiang, China). Mice were kept in a standard temperature-controlled, light-cycled animal facility of Xinjiang University. All animal studies were carried out according to the guidelines of the Animal Care and Use Committee of Xinjiang University. The protocol was approved by the Committee on the Ethics of Animal Experiments of Xinjiang Key Laboratory of Biological Resources and Genetic Engineering (BRGE-AE001), Xinjiang University.

Tumor mouse study

For induction of tumor mouse model, male Kunming mice were subcutaneously injected with 1 × 106 H22 cells in 100μl PBS into the right flank. After 3days, mice were randomly divided into 3 groups (7 mice/group). Control group was injected with 0.1ml DMSO subcutaneously around tumor. CTPG-200 and CTPG-400 groups were subcutaneously injected with 200 or 400mg/kg CTPG in 0.1ml DMSO around tumor. Mice were treated every 2days for up to 21days. Tumor sizes were measured using calipers up to 25days and tumor volume was calculated according to the formula: tumor volume (mm3) = (length×width2)/2. After 25days, survival of tumor mice was monitored every day until the end of this study.

Statistical analysis

Statistical significance was calculated by one-way analysis of variance among the treatment and control groups. All data were expressed as the mean ± standard deviation (S.D.). p < 0.05 was considered statistically significant.

Results

CTPG reduced the viability of H22 cells in vitro

In order to explore antitumor effect of CTPG on HCC, H22 cells were treated with different concentrations of CTPG (0, 100, 200, 300 and 400μg/ml) in vitro. After 24h, the morphology of H22 cells was observed using inverted microscope. We found that the morphology of H22 cells was dramatically changed by CTPG treatment. With increasing CTPG concentration, cells became small and round and cell number was also greatly reduced (Fig.1a). MTT assay was used to analyze the viability of H22 cells after CTPG treatment for 24, 48 and 72h, respectively. CTPG significantly reduced H22 cell viability in a dose-dependent and time-dependent manner (Fig.1b). CTPG at 300μg/ml arrived at the best inhibitory rate (Fig.1c). The values of IC50 of CTPG for H22 cells are 236μg/ml at 24h and 169.8μg/ml at 48h.

The effect of CTPG on the growth of H22 cells. H22 cells were treated with different concentrations of CTPG. (a) After 24h, the morphology of H22 cells was observed by inverted microscope. Inserted panels: 20 folds of magnification. (b) After 24, 48 and 72h, cell viability was detected by MTT assay. (c) The inhibitory rate of CTPG. * p < 0.05; *** p < 0.001 compared to control

Full size image

CTPG induced apoptosis in H22 cells

To investigate whether the decreased viability of H22 cells is mediated by the induction of apoptosis, H22 cells were treated with different concentrations of CTPG (0, 100, 200, 300 and 400μg/ml) for 24h and stained with PI and Annexin V. The flow cytometry results showed that CTPG significantly induced apoptosis of H22 cells (including early and late apoptosis) in a dose-dependent manner (Fig.2a). Although the high dose of CTPG also significantly increased necrosis of H22 cells, necrosis plays a minor role in the inhibition of H22 cell growth due to its lower proportion (8.3%) compared to that of apoptosis (52.6%). Further, total proteins of H22 cells were isolated after CTPG treatment and the expressions of anti-apoptotic B cell lymphoma 2 (Bcl-2) and pro-apoptotic BCL-2-associated X protein (Bax) were detected by Western blot. Grayscale scanning data showed that the expression levels of Bax and Bcl-2 were increased and decreased, respectively. The Bax/Bcl-2 ratio was significantly increased (Fig.2b). These results suggest that CTPG induces apoptosis in H22 cells.

CTPG induced apoptosis in H22 cells. Cells were treated with different concentrations of CTPG for 24h. (a) The apoptotic and necrotic H22 cells were detected by flow cytometry. The upper panel showed the individual dot plots and the lower panel showed the summary data. (b) Total protein was isolated to analyze the expressions of Bax and Bcl-2 by Western blot. ** p < 0.01; *** p < 0.001 compared to control

Full size image

CTPG induces chromosomal condensation and cell cycle arrest in H22 cells

It has reported that DNA damage and cell cycle arrest induced by drugs can inhibit tumor cell growth and cause apoptosis in tumor cells [17, 18]. To detect the morphology of nuclei in H22 cells after CTPG treatment for 24h, H22 cells were stained by Hoechst 33,342 and observed using inverted fluorescent microscopy. CTPG-treated cells showed a dose-dependent increase of brightly condensed chromatin of nuclei, while the untreated cells showed the hom*ogeneously stained nuclei (Fig.3a). Cell cycle distribution in H22 cells was further analyzed by PI staining after CTPG treatment for 24h. As shown in Fig.3b, CTPG treatment significantly increased the proportion of G0/G1- and G2/M-phase cells and significantly decreased the proportion of S-phase cells, suggesting that CTPG induced G0/G1 and G2/M-phase arrest in H22 cells. The high dose of CTPG also significantly increased the proportion of sub G1 cells.

CTPG induced chromosomal condensation and cell cycle arrest in H22 cells. H22 cells were treated with different concentrations of CTPG for 24h. (a) Cells were stained with Hoechst 33,342 and observed by inverted fluorescent microscopy. The arrows indicated the chromosomal condensation or fragmentation. (b) Cell cycle distribution in H22 cells was analyzed by flow cytometry. ** p < 0.01; *** p < 0.001 compared to control

Full size image

CTPG decreased mitochondrial membrane potential and increased the release of cytochrome c

Mitochondrial-dependent pathway plays an important role in the induction of apoptosis [19, 20]. The changes of mitochondrial membrane potential (Δψm) can be monitored by JC-1 staining due to JC-1 aggregate (red fluorescence) can disintegrate into monomer (green fluorescence) with the reduction of Δψm [21]. After CTPG treatment for 24h, H22 cells were stained by JC-1 dye. The flow cytometry data showed that the red fluorescence in FL-2 channel and green fluorescence in FL-1 channel were significantly decreased and increased upon CTPG treatment. The proportion of PEFITC+ cells were significantly increased (Fig.4a), suggesting that CTPG reduced the Δψm in H22 cells. This is consistent with the increased Bax/Bcl-2 ratio. Consequently, we observed the release of cytochrome c was significantly increased upon CTPG treatment (Fig.4b). These results indicated that CTPG might partially induce apoptosis in H22 cells via mitochondrial-dependent (intrinsic) pathway.

The reduction of Δψm and release of cytochrome c. H22 cells were treated with different concentrations of CTPG for 24h. (a) Cells were stained with JC-1 dye and analyzed by flow cytometry. The individual dot plots show the changes of JC-1 fluorescence. The summary data are shown in the lower panel. (b) Total protein was isolated to detect the release of cytochrome c by Western blot. * p < 0.05; *** p < 0.001 compared to control

CTPG activated caspase pathway and prevented DNA repair

Next, the activation of caspase induced by CTPG via both extrinsic and intrinsic signaling pathways was analyzed. After CTPG treatment for 24h, total proteins were isolated from H22 cells and the levels of pro- and cleaved-caspases were detected by Western blot. Compared with the untreated or the DMSO control, CTPG treatment significantly up-regulated not only the level of cleaved caspase-8 (extrinsic pathway) but also the level of cleaved caspase-9 (intrinsic pathway) (Fig.5). Sequentially, activated caspase-8 and -9 cleaved the downstream pro-caspase-3 and -7 that was observed in Fig.5. Activated caspase-3 cleaved the DNA repair enzyme of poly (ADP-ribose) polymerase (PARP) to prevent DNA repair and accumulate DNA damage as observed in Fig.3a. These results indicated that CTPG induced apoptosis in H22 cells through both extrinsic and intrinsic signaling pathways.

The levels of cleaved-caspases and cleaved-PARP. H22 cells were treated with different concentrations of CTPG for 24h. Total protein was isolated to detect the levels of cleaved-caspases and cleaved-PARP by Western blot

Full size image

CTPG suppresses the growth of H22 HCC in vivo and improves the survival rate of tumor mice

Finally, the antitumor effect of CTPG on HCC was evaluated in tumor mouse model, which was established by subcutaneous injection of H22 cells. After 3days of H22 cell injection, tumor mice were treated with CTPG for 8 times. The body weight of mice and tumor sizes were monitored at indicated time points. As shown in Fig.6a, the body weight of mice in each group has no significant difference, suggesting that the selected doses of CTPG have no obvious side effect. Interestingly, the tumor growth in mice treated with both 200mg/kg and 400mg/kg of CTPG was significantly inhibited (Fig.6b). Moreover, the two doses of CTPG treatment greatly improved the survival of tumor mice (3/7, 3/7) compared with control group (0/7) at the end of the experiment (Fig.6b). We also found that CTPG significantly enhanced the proliferation of splenocytes isolated from male Kunming mice in a dose-dependent manner (Fig.6c), suggesting that CTPG has immunostimulatory effect.

CTPG suppressed tumor growth in vivo. Tumor mouse model was established by injection of H22 cells. After 3days, tumor mice (7 mice/group) were treated with or without CTPG. Body weight of mice (a), tumor sizes and survival rate (b) were monitored at the indicated time points. (c) Splenocytes were isolated from Kunming mice and treated with different concentrations of CTPG for 24 and 48h. The proliferation of splenocytes was analyzed by MTT. * p < 0.05; *** p < 0.001 compared to control

Full size image

Discussion

TCM has been used to treat various diseases including cancers for a long history. It has been reported that TCM can induce apoptosis in different types of tumor cells through both extrinsic (death receptor-mediated) and intrinsic (mitochondria-dependent) signaling pathways to exert antitumor effects [22,23,24,25]. The two pathways can activate caspase-8 and -9, respectively [24, 26]. Here, we found that CTPG significantly suppressed H22 cell growth through induction of apoptosis and cell cycle arrest. The levels of cleaved caspase-8 and -9 were significantly up-regulated by CTPG treatment, suggesting that both extrinsic and intrinsic signaling pathways were involved in the induction of apoptosis. Our previous study showed that CTPG induced apoptosis in melanoma B16-F10 cells by mitochondria-dependent pathway that increased the level of cleaved caspase-9 but not caspase-8 [16]. CTPG might activate distinct signaling pathways in different types of tumor cells.

Mitochondrial membrane integrity is tightly regulated by the members of the BCL-2 protein family including Bax and Bcl-2 [27, 28]. The ratio of Bax to Bcl-2 plays a critical role in mitochondria-dependent apoptosis pathway [29]. In H22 cells treated by CTPG, Bax/Bcl-2 ratio was significantly up-regulated, which might cause the reduction of Δψm and the release of cytochrome c observed in this study. Consequently, pro-caspase-9 was cleaved and activated. Finally, the initiators of active caspase-8 and -9 activated the executioner of caspase-3 to cleave PARP to prevent DNA repair. Taken together, these results suggested that CTPG induced apoptosis in H22 cells through both extrinsic and intrinsic signaling pathways.

In tumor mouse model, CTPG significantly suppressed the growth of H22 HCC and greatly improved the survival of tumor mice. Interestingly, CTPG dose-dependently promoted the proliferation of splenocytes from Kunming mice, which is consistent with our previous study [16]. These results suggested that CTPG might suppress the growth of H22 HCC in mice through both direct antitumor effect and indirect immune enhancement.

Conclusions

CTPG suppressed the growth of H22 cells both in vitro and in vivo and induced apoptosis in H22 cells through both extrinsic and intrinsic signaling pathways. These data indicated that CTPG might be a potential candidate for the treatment of HCC.

Abbreviations

Bax:

BCL-2-associated X protein

Bcl-2:

B cell lymphoma 2

CTPG:

Cistanche tubulosa phenylethanoid glycosides

HCC:

hepatocellular carcinoma

HRP:

horseradish peroxidase

MTT:

3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide

PARP:

DNA repair enzyme of poly (ADP-ribose) polymerase

TCM:

traditional Chinese medicine

Δψm:

mitochondrial membrane potential

References

  1. Global Burden of Disease Cancer Collaboration, Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, Brenner H, Dicker DJ, Chimed-Orchir O, Dandona R, et al. Global, regional, and National Cancer Incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 Cancer groups, 1990 to 2015: a systematic analysis for the global burden of disease study. JAMA Oncol. 2017;3:524–48.

    Article Google Scholar

  2. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66:115–32.

    Article Google Scholar

  3. European Association For The Study Of The Liver, European Organisation For Research And Treatment Of Cancer. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56:908–43.

    Article Google Scholar

  4. Roayaie S, Obeidat K, Sposito C, Mariani L, Bhoori S, Pellegrinelli A, Labow D, Llovet JM, Schwartz M, Mazzaferro V. Resection of hepatocellular cancer ≤2 cm: results from two western centers. Hepatology. 2013;57:1426–35.

    Article Google Scholar

  5. Ting CT, Cheng YY, Tsai TH. Herb-drug interaction between the traditional Hepatoprotective formulation and Sorafenib on hepatotoxicity, Histopathology and Pharmaco*kinetics in Rats. Molecules. 2017;22:E1034.

    Article Google Scholar

  6. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–90.

    Article CAS Google Scholar

  7. Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, Luo R, Feng J, Ye S, Yang TS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:25–34.

    Article CAS Google Scholar

  8. Shi Z, Song T, Wan Y, Xie J, Yan Y, Shi K, Du Y, Shang L. A systematic review and meta-analysis of traditional insect Chinese medicines combined chemotherapy for non-surgical hepatocellular carcinoma therapy. Sci Rep. 2017;7:4355.

    Article Google Scholar

  9. Yang Z, Liao X, Lu Y, Xu Q, Tang B, Chen X, Yu Y. Add-on therapy with traditional Chinese medicine improves outcomes and reduces adverse events in hepatocellular carcinoma: a meta-analysis of randomized controlled trials. Evid Based Complement Alternat Med. 2017;2017:3428253.

    PubMed PubMed Central Google Scholar

  10. Lin LW, Hsieh MT, Tsai FH, Wang WH, Wu CR. Anti-nociceptive and anti-inflammatory activity caused by Cistanche deserticola in rodents. J Ethnopharmacol. 2002;83:177–82.

    Article Google Scholar

  11. Wu CR, Lin HC, Su MH. Reversal by aqueous extracts of Cistanche tubulosa from behavioral deficits in Alzheimer's disease-like rat model: relevance for amyloid deposition and central neurotransmitter function. BMC Complement Altern Med. 2014;14:202.

    Article Google Scholar

  12. Jiang Y, Tu PF. Analysis of chemical constituents in Cistanche species. J Chromatogr A. 2009;1216:1970–9.

    Article CAS Google Scholar

  13. Morikawa T, Pan Y, Ninomiya K, Imura K, Matsuda H, Yoshikawa M, Yuan D, Muraoka O. Acylated phenylethanoid oligoglycosides with hepatoprotective activity from the desert plant Cistanche tubulosa. Bioorg Med Chem. 2010;18:1882–90.

    Article CAS Google Scholar

  14. Nan ZD, Zeng KW, Shi SP, Zhao MB, Jiang Y, Tu PF. Phenylethanoid glycosides with anti-inflammatory activities from the stems of Cistanche deserticola cultured in Tarim desert. Fitoterapia. 2013;89:167–74.

    Article CAS Google Scholar

  15. Deng M, Zhao J, Tu P, Jiang Y, Li Z, Wang Y. Echinacoside resues the SHSY5Y neuronal cells from TNF α-induced apoptosis. Eur J Pharmacol. 2004;505:11–8.

    Article CAS Google Scholar

  16. Li J, Li J, Aipire A, Gao L, Huo S, Luo J, Zhang F. Phenylethanoid glycosides from Cistanche tubulosa inhibits the growth of B16-F10 cells both in vitro and in vivo by induction of apoptosis via mitochondria-dependent pathway. J Cancer. 2016;7:1877–87.

    Article CAS Google Scholar

  17. Shang HS, Chang CH, Chou YR, Yeh MY, Au MK, Lu HF, Chu YL, Chou HM, Chou HC, Shih YL, et al. Curcumin causes DNA damage and affects associated protein expression in HeLa human cervical cancer cells. Oncol Rep. 2016;36:2207–15.

    Article CAS Google Scholar

  18. Wang R, Zhang Q, Peng X, Zhou C, Zhong Y, Chen X, Qiu Y, Jin M, Gong M, Kong D. Stellettin B induces G1 arrest, apoptosis and autophagy in human non-small cell lung Cancer A549 cells via blocking PI3K/Akt/mTOR pathway. Sci Rep. 2016;6:27071.

    Article CAS Google Scholar

  19. Sinha K, Das J, Pal PB, Sil PC. Oxidative stress: the mitochondria-dependent and mitochondria-independent pathways of apoptosis. Arch Toxicol. 2013;87:1157–80.

    Article CAS Google Scholar

  20. Zhang YS, Shen Q, Li J. Traditional Chinese medicine targeting apoptotic mechanisms for esophageal cancer therapy. Acta Pharmacol Sin. 2016;37:295–302.

    Article Google Scholar

  21. Chong ZZ, Lin SH, Li F, Maiese K. The sirtuin inhibitor nicotinamide enhances neuronal cell survival during acute anoxic injury through AKT, BAD, PARP, and mitochondrial associated "anti-apoptotic" pathways. Curr Neurovasc Res. 2005;2:271–85.

    Article CAS Google Scholar

  22. Hu B, Wang SS, Du Q. Traditional Chinese medicine for prevention and treatment of hepatocarcinoma: from bench to bedside. World J Hepatol. 2015;7:1209–32.

    Article Google Scholar

  23. Hu B, An HM, Wang SS, Chen JJ, Xu L. Preventive and therapeutic effects of Chinese herbal compounds against hepatocellular carcinoma. Molecules. 2016;21:142.

    Article Google Scholar

  24. Xu H, Zhao X, Liu X, Xu P, Zhang K, Lin X. Antitumor effects of traditional Chinese medicine targeting the cellular apoptotic pathway. Drug Des Devel Ther. 2015;9:2735–44.

    CAS PubMed PubMed Central Google Scholar

  25. Li-Weber M. Targeting apoptosis pathways in cancer by Chinese medicine. Cancer Lett. 2013;332:304–12.

    Article CAS Google Scholar

  26. Xu G, Shi Y. Apoptosis signaling pathways and lymphocyte homeostasis. Cell Res. 2007;17:759–71.

    Article CAS Google Scholar

  27. Tait SW, Green DR. Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 2010;11:621–32.

    Article CAS Google Scholar

  28. Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol. 2012;13:780–8.

    Article CAS Google Scholar

  29. Martinou JC, Youle RJ. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell. 2011;21:92–101.

    Article CAS Google Scholar

Download references

Funding

This work was supported by the High Level Talent Introduction Project of Xinjiang Uygur Autonomous Region to J.L., the Chinese National Natural Science Foundation Grant (31460241) to J.L. and the Doctoral Start-up Fund of Xinjiang University (BS160261 to X.W. and BS150236 to Y.L.).

Availability of data and materials

The raw data for this study are available upon reasonable request to the corresponding author.

Author information

Authors and Affiliations

  1. Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, 666 Shengli Road, Urumqi, Xinjiang, 830046, China

    Pengfei Yuan,Adila Aipire,Yi Yang,Lijie Xia,Xinhui Wang,Yijie Li&Jinyao Li

  2. College of Life Science, Xinjiang Normal University, 102 Xinyi Road, Urumqi, 830054, Xinjiang, China

    Jinyu Li

  3. Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, China

    Jinyao Li

Authors

  1. Pengfei Yuan

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  2. Jinyu Li

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  3. Adila Aipire

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  4. Yi Yang

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  5. Lijie Xia

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  6. Xinhui Wang

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  7. Yijie Li

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  8. Jinyao Li

    View author publications

    You can also search for this author in PubMedGoogle Scholar

Contributions

JL and JL designed the experiments. PY, JL, AA and YY performed the experiments. LX, XW and YL analyzed the data. PY, JL and JL wrote the manuscript. All authors contributed and approved the final manuscript.

Corresponding authors

Correspondence to Jinyu Li or Jinyao Li.

Ethics declarations

Ethics approval

The animal study was approved by the Committee on the Ethics of Animal Experiments of Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, Xinjiang University.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Cistanche tubulosa phenylethanoid glycosides induce apoptosis in H22 hepatocellular carcinoma cells through both extrinsic and intrinsic signaling pathways (7)

Cite this article

Yuan, P., Li, J., Aipire, A. et al. Cistanche tubulosa phenylethanoid glycosides induce apoptosis in H22 hepatocellular carcinoma cells through both extrinsic and intrinsic signaling pathways. BMC Complement Altern Med 18, 275 (2018). https://doi.org/10.1186/s12906-018-2201-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-018-2201-1

Keywords

  • Cistanche tubulosa
  • Phenylethanoid glycosides
  • Apoptosis
  • Signaling pathway
  • Tumor mouse model
Cistanche tubulosa phenylethanoid glycosides induce apoptosis in H22 hepatocellular carcinoma cells through both extrinsic and intrinsic signaling pathways (2024)
Top Articles
‘ajji’: Film Review | Busan 2017
The Rise of Malayalam Movies Download Isaimini - The Digital Weekly
Matgyn
Washu Parking
O'reilly's Auto Parts Closest To My Location
Erskine Plus Portal
Midway Antique Mall Consignor Access
True Statement About A Crown Dependency Crossword
Matthew Rotuno Johnson
Mawal Gameroom Download
Inside California's brutal underground market for puppies: Neglected dogs, deceived owners, big profits
De Leerling Watch Online
Where does insurance expense go in accounting?
Fredericksburg Free Lance Star Obituaries
Gmail Psu
Samsung Galaxy S24 Ultra Negru dual-sim, 256 GB, 12 GB RAM - Telefon mobil la pret avantajos - Abonament - In rate | Digi Romania S.A.
Locate At&T Store Near Me
Harem In Another World F95
Craigslist In Flagstaff
Craigslist Southern Oregon Coast
Southwest Flight 238
What Individuals Need to Know When Raising Money for a Charitable Cause
Craigslist Wilkes Barre Pa Pets
Kohls Lufkin Tx
Sound Of Freedom Showtimes Near Movie Tavern Brookfield Square
Bleacher Report Philadelphia Flyers
Yale College Confidential 2027
Criglist Miami
HP PARTSURFER - spare part search portal
Little Einsteins Transcript
Darktide Terrifying Barrage
Darknet Opsec Bible 2022
Kacey King Ranch
Jambus - Definition, Beispiele, Merkmale, Wirkung
Goodwill Houston Select Stores Photos
Final Exam Schedule Liberty University
Otter Bustr
RALEY MEDICAL | Oklahoma Department of Rehabilitation Services
Discover Wisconsin Season 16
Kutty Movie Net
Shoecarnival Com Careers
QVC hosts Carolyn Gracie, Dan Hughes among 400 laid off by network's parent company
Gotrax Scooter Error Code E2
Dying Light Mother's Day Roof
Mega Millions Lottery - Winning Numbers & Results
Enjoy Piggie Pie Crossword Clue
Treatise On Jewelcrafting
Nfhs Network On Direct Tv
OSF OnCall Urgent Care treats minor illnesses and injuries
Fetllife Com
Www.card-Data.com/Comerica Prepaid Balance
Dinargurus
Latest Posts
Article information

Author: Melvina Ondricka

Last Updated:

Views: 5466

Rating: 4.8 / 5 (48 voted)

Reviews: 95% of readers found this page helpful

Author information

Name: Melvina Ondricka

Birthday: 2000-12-23

Address: Suite 382 139 Shaniqua Locks, Paulaborough, UT 90498

Phone: +636383657021

Job: Dynamic Government Specialist

Hobby: Kite flying, Watching movies, Knitting, Model building, Reading, Wood carving, Paintball

Introduction: My name is Melvina Ondricka, I am a helpful, fancy, friendly, innocent, outstanding, courageous, thoughtful person who loves writing and wants to share my knowledge and understanding with you.