Skip to main content
Erschienen in: BMC Complementary Medicine and Therapies 1/2018

Open Access 01.12.2018 | Research article

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

verfasst von: Pengfei Yuan, Jinyu Li, Adila Aipire, Yi Yang, Lijie Xia, Xinhui Wang, Yijie Li, Jinyao Li

Erschienen in: BMC Complementary Medicine and Therapies | Ausgabe 1/2018

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.
Abkürzungen
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

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 3 months 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 [1215]. 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 100 U/ml penicillin and 100 μg/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 100 μl medium per well and cultured at 37 °C. After 24 h, cells were treated with different concentrations of CTPG (0, 100, 200, 300 and 400 μg/ml) or 0.3% DMSO (equal to that in 400 μg/ml CTPG) for 24, 48 and 72 h, respectively. After centrifugation at 1000 rpm for 7 min, supernatant was discarded and 100 μl of MTT solution (5 mg/ml in PBS) was added to each well. The plates were incubated at 37 °C for 4 h and 100 μl 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 400 μg/ml) or 0.3% DMSO for 24 h, 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 400 μg/ml) for 24 h, and then stained with the membrane-permeable JC-1 dye (Beyotime,China) for 20 min at 37 °C. After washing twice with JC-1 buffer, samples were resuspended with 300 μl of JC-1 buffer and analyzed by flow cytometry (BD FACSCalibur, USA).

Analysis of cell cycle

H22 cells were inoculated in 60 mm culture dishes and treated with different concentrations of CTPG (0, 100, 200, 300 and 400 μg/ml) or 0.3% DMSO for 24 h. All cells were collected and washed twice with PBS. Cells were fixed in 70% ice-cold ethanol at − 20 °C for 2 h and washed twice with PBS, then re-suspended in 300 μl Propidium iodide/RNase staining buffer (BD Biosciences). After 10 min 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 2 ml medium. After 60%~ 70% confluence, the cells were treated with CTPG (0, 100, 200, 300 and 400 μg/ml) for 24 h. The cells were collected and fixed with 4% ice-cold Paraformaldehyde at 4 °C for 10 min. After washing with PBS, cells were stained with Hoechst 33,258 (Beyotime, China) at 4 °C for 10 min. 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 400 μg/ml) or 0.3% DMSO for 24 h. Cells were collected and lysed with the Cell Lysis Solution RIPA (Beijing ComWin Biotech Co., Ltd) for 30 min on ice. Samples were spun down (12,000 g for 15 min 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

6-8 weeks 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 100 μl PBS into the right flank. After 3 days, mice were randomly divided into 3 groups (7 mice/group). Control group was injected with 0.1 ml DMSO subcutaneously around tumor. CTPG-200 and CTPG-400 groups were subcutaneously injected with 200 or 400 mg/kg CTPG in 0.1 ml DMSO around tumor. Mice were treated every 2 days for up to 21 days. Tumor sizes were measured using calipers up to 25 days and tumor volume was calculated according to the formula: tumor volume (mm3) = (length×width2)/2. After 25 days, 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 400 μg/ml) in vitro. After 24 h, 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 72 h, respectively. CTPG significantly reduced H22 cell viability in a dose-dependent and time-dependent manner (Fig. 1b). CTPG at 300 μg/ml arrived at the best inhibitory rate (Fig. 1c). The values of IC50 of CTPG for H22 cells are 236 μg/ml at 24 h and 169.8 μg/ml at 48 h.

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 400 μg/ml) for 24 h 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 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 24 h, 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 homogeneously stained nuclei (Fig. 3a). Cell cycle distribution in H22 cells was further analyzed by PI staining after CTPG treatment for 24 h. 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 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 24 h, 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.

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 24 h, 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.

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 3 days 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 200 mg/kg and 400 mg/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.

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 [2225]. 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.

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.

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.
Open AccessThis 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.
Literatur
1.
Zurück zum Zitat 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.CrossRef 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.CrossRef
2.
Zurück zum Zitat 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.CrossRef 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.CrossRef
3.
Zurück zum Zitat 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.CrossRef 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.CrossRef
4.
Zurück zum Zitat 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.CrossRef 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.CrossRef
5.
Zurück zum Zitat Ting CT, Cheng YY, Tsai TH. Herb-drug interaction between the traditional Hepatoprotective formulation and Sorafenib on hepatotoxicity, Histopathology and Pharmacokinetics in Rats. Molecules. 2017;22:E1034.CrossRef Ting CT, Cheng YY, Tsai TH. Herb-drug interaction between the traditional Hepatoprotective formulation and Sorafenib on hepatotoxicity, Histopathology and Pharmacokinetics in Rats. Molecules. 2017;22:E1034.CrossRef
6.
Zurück zum Zitat 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.CrossRef 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.CrossRef
7.
Zurück zum Zitat 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.CrossRef 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.CrossRef
8.
Zurück zum Zitat 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.CrossRef 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.CrossRef
9.
Zurück zum Zitat 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.PubMedPubMedCentral 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.PubMedPubMedCentral
10.
Zurück zum Zitat 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.CrossRef 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.CrossRef
11.
Zurück zum Zitat 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.CrossRef 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.CrossRef
12.
Zurück zum Zitat Jiang Y, Tu PF. Analysis of chemical constituents in Cistanche species. J Chromatogr A. 2009;1216:1970–9.CrossRef Jiang Y, Tu PF. Analysis of chemical constituents in Cistanche species. J Chromatogr A. 2009;1216:1970–9.CrossRef
13.
Zurück zum Zitat 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.CrossRef 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.CrossRef
14.
Zurück zum Zitat 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.CrossRef 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.CrossRef
15.
Zurück zum Zitat 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.CrossRef 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.CrossRef
16.
Zurück zum Zitat 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.CrossRef 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.CrossRef
17.
Zurück zum Zitat 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.CrossRef 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.CrossRef
18.
Zurück zum Zitat 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.CrossRef 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.CrossRef
19.
Zurück zum Zitat 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.CrossRef 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.CrossRef
20.
Zurück zum Zitat Zhang YS, Shen Q, Li J. Traditional Chinese medicine targeting apoptotic mechanisms for esophageal cancer therapy. Acta Pharmacol Sin. 2016;37:295–302.CrossRef Zhang YS, Shen Q, Li J. Traditional Chinese medicine targeting apoptotic mechanisms for esophageal cancer therapy. Acta Pharmacol Sin. 2016;37:295–302.CrossRef
21.
Zurück zum Zitat 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.CrossRef 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.CrossRef
22.
Zurück zum Zitat 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.CrossRef 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.CrossRef
23.
Zurück zum Zitat 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.CrossRef 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.CrossRef
24.
Zurück zum Zitat 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.PubMedPubMedCentral 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.PubMedPubMedCentral
25.
Zurück zum Zitat Li-Weber M. Targeting apoptosis pathways in cancer by Chinese medicine. Cancer Lett. 2013;332:304–12.CrossRef Li-Weber M. Targeting apoptosis pathways in cancer by Chinese medicine. Cancer Lett. 2013;332:304–12.CrossRef
26.
Zurück zum Zitat Xu G, Shi Y. Apoptosis signaling pathways and lymphocyte homeostasis. Cell Res. 2007;17:759–71.CrossRef Xu G, Shi Y. Apoptosis signaling pathways and lymphocyte homeostasis. Cell Res. 2007;17:759–71.CrossRef
27.
Zurück zum Zitat Tait SW, Green DR. Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 2010;11:621–32.CrossRef Tait SW, Green DR. Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 2010;11:621–32.CrossRef
28.
Zurück zum Zitat Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol. 2012;13:780–8.CrossRef Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol. 2012;13:780–8.CrossRef
29.
Zurück zum Zitat Martinou JC, Youle RJ. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell. 2011;21:92–101.CrossRef Martinou JC, Youle RJ. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell. 2011;21:92–101.CrossRef
Metadaten
Titel
Cistanche tubulosa phenylethanoid glycosides induce apoptosis in H22 hepatocellular carcinoma cells through both extrinsic and intrinsic signaling pathways
verfasst von
Pengfei Yuan
Jinyu Li
Adila Aipire
Yi Yang
Lijie Xia
Xinhui Wang
Yijie Li
Jinyao Li
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Complementary Medicine and Therapies / Ausgabe 1/2018
Elektronische ISSN: 2662-7671
DOI
https://doi.org/10.1186/s12906-018-2201-1

Weitere Artikel der Ausgabe 1/2018

BMC Complementary Medicine and Therapies 1/2018 Zur Ausgabe