Open Access

A novel manganese complex selectively induces malignant glioma cell death by targeting mitochondria

  • Authors:
    • Ji Geng
    • Jing Li
    • Tao Huang
    • Kaidi Zhao
    • Qiuyun Chen
    • Wenjie Guo
    • Jing Gao
  • View Affiliations

  • Published online on: July 12, 2016     https://doi.org/10.3892/mmr.2016.5509
  • Pages: 1970-1978
  • Copyright: © Geng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Despite advances in treatment, malignant glioma commonly exhibits recurrence, subsequently leading to a poor prognosis. As manganese (Mn) compounds can be transported by the transferrin‑transferrin receptor system, the present study synthesized and examined the potential use of Adpa‑Mn as a novel antitumor agent. Adpa‑Mn time and dose‑dependently inhibited U251 and C6 cell proliferation; however, it had little effect on normal astrocytes. Apoptosis was significantly elevated following treatment with Adpa‑Mn, as detected by chromatin condensation, Annexin V/propidium iodide staining, cytochrome c release from mitochondria to the cytoplasm, and the activation of caspases‑9, ‑7 and ‑3 and poly (ADP‑ribose) polymerase. In addition, Adpa‑Mn enhanced fluorescence intensity of monodansylcadaverine and elevated the expression levels of the autophagy‑related protein microtubule‑associated protein 1 light chain 3. Pretreatment with the autophagy inhibitors 3‑methyladenine and chloroquine enhanced Adpa‑Mn‑induced cell inhibition, thus indicating that autophagy has an essential role in this process. Furthermore, evidence of mitochondrial dysfunction was detected in the Adpa‑Mn‑treated group, including disrupted membrane potential, elevated levels of reactive oxygen species (ROS) and depleted adenosine triphosphate. Conversely, treatment with the mitochondrial permeability transition inhibitor cyclosporin A reversed Adpa‑Mn‑induced ROS production, mitochondrial damage and cell apoptosis, thus suggesting that Adpa‑Mn may target the mitochondria. Taken together, these data suggested that Adpa‑Mn may be considered for use as a novel anti‑glioma therapeutic option.

Introduction

Malignant glioma, which is the most common type of primary brain tumor, is highly aggressive and metastatic. Survival ranges between 12 and 40 months from initial diagnosis, and between 6 and 18 months following recurrence, despite treatment with standard therapies, including surgery, chemotherapy and radiotherapy (13). Therefore, it is necessary to develop novel strategies for the treatment of glioma. Mitochondria are unique cellular organelles, the primary role of which is to generate adenosine triphosphate (ATP) through oxidative phosphorylation, in order to ensure survival (4). Previous studies have reported that mitochondria have a central role in regulating proliferation, apoptosis and autophagy in cancer cells (57). Therefore, disturbing mitochondrial metabolism or interfering with mitochondrial membrane permeabilization may be considered a promising therapeutic approach to cancer.

Autophagy is a lysosomal degradation pathway by which cells consume intracellular materials, including damaged organelles, proteins or hazardous substances, in order to maintain cellular homeostasis (8). Autophagy is upregulated in response to cellular stress, including starvation, mitochondrial stress, endoplasmic reticulum stress and pathogenic infection (9). The emerging role of autophagy in cancer is considered to be a double-edged sword. Autophagy has been reported to render cancer cells able to tolerate therapy-induced stress; however, autophagy can also digest organelles and limit tumorigenesis (10). Therefore, modulating the autophagic pathway may provide novel approaches to cancer therapy and prevention.

Manganese (Mn) is an essential mineral that can influence intracellular and extracellular metabolism associated with the mitochondria (11). Apoptosis is activated by Mn compounds, which induce the rupture of DNA and the release of cytochrome c from the mitochondrial intermembrane space to the cytosol (12). In addition, several enzymes have been identified, which naturally contain Mn, including pyruvate carboxylase, arginase and Mn superoxide dismutase (13,14). The main purpose of Mn is as a coactivator of superoxide dismutase in mitochondria (15). It has been reported that Mn (II) ions are predominantly transported by divalent metal transporter 1 (DMT-1) and the transferrin-transferrin receptor (Tf-TfR) system, which is highly expressed in various tumors (16). It has previously been demonstrated that Mn (II) ions can transport across the rat blood-brain barrier, through saturable and Tf-dependent transport mechanisms, to the brain (1719). Based on the aforementioned findings, the present study hypothesized that Mn-containing compounds may cross the blood-brain barrier and selectively kill glioma cells via DMT-1 or TfR.

The present study indicated that the complex [(Adpa) Mn(Cl)(H2O)], designated as Adpa-Mn, which is an inorganic compound comprised of Adpa [bis(2-pyridylmethyl) amino-2-propionic acid] and Mn, is a promising novel anti-glioma agent with potent selective activity in vitro. Furthermore, it was verified that Adpa-Mn induces apoptotic cell death and cytoprotective autophagy by triggering mitochondrial dysfunction.

Materials and methods

Materials

The compound Adpa-Mn was synthesized by Professor Chen Qiuyun (Fig. 1A) as previously described (20). Cisplatin was purchased from Jiangsu Hengrui Medicine Co., Ltd. (Lianyungang, China). 3-[4,5-dimehyl-2-thiazolyl]-2,5-diphenyl-2H-tetrazolium bromide (MTT) was purchased from Amresco, LLC (Solon, OH, USA). JC-1 for mitochondrial membrane potential, Dulbecco's modified Eagle's medium (DMEM) for U251 and C6, RPMI-1640 for rat astrocyte cells, trypsin-EDTA solution for cell detachment and the Annexin V/propidium iodide (PI) kit for apoptotic detection were purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Ferric citrate, 4′,6-diamidino-2-phenylindole (DAPI), desferrioxamine (DFO) and cyclosporin A (CsA) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Fetal bovine serum (FBS) was obtained from Sijiqing Biological Engineering Materials Co., Ltd. (Hangzhou, China). The 2′,7′-dichlorofluorescein-diacetate (DCFH-DA) kit was purchased from Beyotime Institute of Biotechnology (Nantong, China). Anti-microtubule-associated protein 1 light chain 3 (LC3; cat. no. 3868; 1:1,000), anti-poly (ADP-ribose) polymerase (PARP; cat. no. 9536; 1:1000), anti-caspase-9 (cat. no. 9508, 1:1,000), anti-caspase-7 (cat. no. 9494, 1:1,000), anti-caspase-3 (cat. no. 9665; 1:1,000) and anti-B-cell lymphoma 2 (Bcl-2;cat. no. 15071; 1:1,000), cytochrome c (cat. no. 4280, 1:1000), cytochrome c oxidase subunit IV (cat. no. 4850; 1:1,000) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA) and were diluted with 1% bovine serum albumin (BSA). Anti-β-actin antibody (cat. no. SC-8432; 1:200) was obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA), anti-DMT-1 antibody (cat. no. 20507-1-AP; 1:200) was purchased from Proteintech Group, Inc. (Rosemont, IL, USA), and anti-TfR antibody (cat. no. MA5-11441; 1:200) was purchased from Thermo Fisher Scientific, Inc. Monodansylcadaverin (MDC), 3-methyladenine (3-MA), N-acetylcysteine (NAC), ferric citrate, DFO, CsA and chloroquine (CQ) were purchased from Sigma-Aldrich. All other chemicals were of high purity, and were purchased from commercial sources. The study was approved by the ethics committee of Jiangsu University (Zhenjiang, China).

Cell culture

The U251 human glioma cell line, and the C6 rat glioma cell line were obtained from the Cancer Cell Repository (Shanghai Cell Bank; Shanghai, China). The cells were maintained in DMEM supplemented with 10% (v/v) heat-inactivated FBS and antibiotics (100 U/ml penicillin and 100 U/ml streptomycin; Beyotime Institute of Biotechnology) at 37°C in a humidified atmosphere containing 5% CO2. Primary astrocyte cells isolated from three newborn Sprague-Dawley rats were cultured in RPMI-1640 as described previously (21).

Cell viability assay

The cells were plated at a density of ~4×103 viable cells/well in 96-well plates in the presence of 5, 10, 20, 30, or 40 µM of the compound for 12, 24, or 36 h. MTT (1 mg/ml) was added for 3 h followed by dimethyl sulfoxide to dissolve the formazan product, and the U251 and C6 cell viabilities were measured using a 96-well plate reader (SpectraMax 190; Molecular Devices,Sunnyvale, CA, USA) at 490 nm.

Cell apoptosis assay

The cells were plated at a density of ~1×105 viable cells/well in 6-well plates. Cells were harvested following 5, 10, 20 µM Adpa-Mn treatments for 24 h. Cells were collected using trypsin-EDTA and incubated with Annexin V/PI at room temperature for 15 min in the dark, then analyzed using a FACSCalibur flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA). CellQuest (Becton Dickinson, Mountain View, CA, USA) was applied to analyze flow cytometric results. Annexin V+/PI and Annexin V+/PI+ cells were considered to be early and late phase apoptotic cells, respectively.

Visualization of MDC-labeled vacuoles

Autophagic vacuoles were labeled with MDC. Briefly, U251 cells grown on coverslips were incubated with 50 µM MDC in phosphate-buffered saline (PBS) at 37°C for 10 min. Alterations in cellular fluorescence were observed under a Nikon eclipse Ti fluorescence microscope (Nikon Corporation, Tokyo, Japan; excitation, 380–420 nm; emission, 450 nm).

Western blot analysis

Proteins were extracted from the cells using lysis buffer (30 mM Tris, pH 7.5; 150 mM sodium chloride; 1 mM phenylmethylsulfonyl fluoride; 1 mM sodium orthovanadate; 1% Nonidet P-40; 10% glycerol; and phosphatase and protease inhibitors; Roche Diagnostics, Basel, Switzerland). Then, 20 µg protein was separated by 15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis at a constant voltage setting of 110 V for 80 min and were electrophoretically transferred onto polyvinylidene fluoride membranes (Thermo Fisher Scientific Inc.) at a constant current setting of 300 mA for 90 min. Membrane blocking was performed with 1% BSA for 1 h. The membranes were probed with primary antibodies diluted with 1% BSA overnight at 4°C, followed by an incubation with horseradish peroxidase-conjugated goat anti-mouse (cat. no. sc-2969) and goat anti-rabbit (cat. no. sc-2768; Santa Cruz Biotechnology, Inc.) secondary antibodies for 2 h at room temperature. Detection was performed using a LumiGLO Chemiluminescent Substrate system (KPL, Gaithersburg, MD, USA).

Mitochondrial membrane potential assay

Alterations to mitochondrial membrane potential were measured using JC-1. U251 cells were washed with PBS and were incubated with 5 µg/ml JC-1 at 37°C for 30 min. Cells were then washed twice with PBS and were immediately assessed by fluorescence spectrometry (Spectra Max Gemini; Molecular Devices). A 488-nm filter was used to detect the excitation of JC-1. Emission filters of 535 and 595 nm were used to quantify the population of mitochondria exhibiting green (JC-1 monomers) and red (JC-1 aggregates) fluorescence. The ratio of red/green fluorescence was used to reflect the mitochondrial membrane potential.

Determination of intracellular ATP levels

ATP content was measured according to the luciferin-luciferase method, which is based on the requirement of ATP for luciferase to produce light. The cells were plated at a density of ~1×105 viable cells/well in 6-well plates. Cells were harvested following treatment with 20 µM Adpa-Mn for 6, 12 and 24 h and were assayed for ATP using a chemical luciferase ATP assay kit (Beyotime Institute of Biotechnology). The quantity of ATP in the experimental samples was calculated from a standard curve prepared with ATP, and was expressed as nmol/mg protein.

Statistical analysis

Differences between groups were analyzed using a two-tailed Student's t-test with GraphPad Prism version 5.00 for Windows (GraphPad Software, San Diego CA, USA) and differences among groups were analyzed using one-way analysis of variance. All values are expressed as the mean ± standard deviation and P<0.05 was considered to indicate a statistically significant difference. All experiments were repeated at least three times.

Results

Adpa-Mn exhibits significant and selective anti-glioma activity

To determine whether Adpa-Mn has an inhibitory effect on glioma cells, the U251 human glioma cell line and the C6 rat glioma cell line were subjected to an MTT assay. Treatment with Adpa-Mn inhibited U251 and C6 cell proliferation in a dose- and time-dependent manner (Fig. 1B and 1C).

Subsequently, it was determined whether Adpa-Mn exhibits cancer cell selectivity. Treatment with Adpa-Mn exhibited significant selectivity towards glioma cells (C6) over normal rat astrocytes (Fig. 1D and E), whereas cisplatin did not exert this selectivity (Fig. 1F). This preferential toxicity toward cancer cells over non-cancer cells suggests the potential use of this compound as an antitumor therapeutic option.

Due to the high expression of TfR on tumor cells, the cancer cell selectivity of Adpa-Mn may be due to its transport mechanism. As detected by western blotting and MTT assay, when the expression of TfR was inhibited by ferric citrate (100 µM) pretreatment for 24 h, the inhibitory effect of Adpa-Mn on U251 cells was significantly decreased. Conversely, when TfR expression was upregulated following DFO (10 µM) pretreatment for 24 h, Adpa-Mn induced U251 cell inhibition more efficiently (Fig. 1G and H).

Adpa-Mn induces apoptotic cell death

To investigate the potential anticancer mechanisms of Adpa-Mn, U251 malignant glioma cells were treated with various concentrations of Adpa-Mn for 24 h. Apoptotic cell death was detected using 4′,6-diamidino-2-phenylindole, a typical nuclear stain that can visualize nuclear condensation. The results demonstrated that nuclei became condensed and fragmented, and apoptotic bodies were detected following treatment with Adpa-Mn (Fig. 2A). Adpa-Mn-induced apoptosis was further detected using Annexin V/PI staining. As presented in Fig. 2B and C, Annexin V+ cells were abundantly increased from 10.1 to 48.7 and 52.5% following treatment with 10 and 20 µM Adpa-Mn, respectively. To determine the involvement of the caspase cascade in Adpa-Mn-induced apoptosis, the expression levels of caspases were detected by western blotting. The hallmarks of apoptosis, including PARP, caspase-9, caspase-7 and caspase-3, were activated/cleaved in U251 cells following treatment with Adpa-Mn in a dose-dependent manner (Fig. 2D). Consistent with the decreased expression of Bcl-2 (Fig. 2D), which is an integral membrane protein located mainly on the outer mitochondrial membrane that inhibits apoptosis, cytochrome c was transferred from the mitochondrial intermembrane space to the cytoplasm (Fig. 2E). These results suggest that Adpa-Mn may induce significant apoptosis of U251 cells.

Adpa-Mn induces protective autophagy

Cellular features of necrosis, apoptosis and autophagy frequently occur together in response to death signals and toxic stress (22). The present study determined whether Adpa-Mn was able to induce cell death via autophagy. Autophagy is characterized by the accumulation of vesicles and the formation of autophagosomes, which can be detected by the presence of membrane-bound LC3-phospholipid conjugates (23). MDC is a specific marker of autophagic vacuoles (Fig. 3A) and treatment with Adpa-Mn (10 and 20 µM) for 12 h triggered the accumulation of MDC-stained acidic vesicular organelles (AVO) in U251 cells. In addition, Adpa-Mn yielded a time- and dose-dependent increase in the expression levels of LC3 II (the processed form of LC3; Fig. 3B and C). In addition, the LC3 II/LC3 I ratio was increased in a dose- and time-dependent manner following treatment with Adpa-Mn. These data suggest that autophagy was enhanced following treatment with Adpa-Mn. It has previously been reported that apoptosis and autophagy are not mutually exclusive pathways, but have been shown to act in synergy and also to counter each other (24). Pretreatment with 3-MA, an inhibitor of autophagy, and CQ, an inhibitor of lysosomal degradation, decreased Adpa-Mn induced LC3 II accumulation and LC3 II/LC3 I ratio (Fig. 3C), and accelerated cell death (Fig. 3D and E). Autophagy inhibition decreased cell viability, which indicated that Adpa-Mn-induced autophagy exerts a protective role in U251 cells.

Adpa-Mn triggers mitochondrial dysfunction

Since mitochondria are critical in apoptosis and autophagy, the present study aimed to determine the effects of Adpa-Mn on mitochondrial function. JC-1 was used to evaluate mitochondrial membrane potential. Treatment with 5, 10 and 20 µM Adpa-Mn for 12 h led to a decrease in mitochondrial membrane potential, as detected by enhanced green intensity and reduced red intensity of JC-1 (Fig. 4A). Subsequently, the ratio of red and green fluorescence intensity was determined using a fluorescent microplate reader, which indicated a dose- and time-dependent decrease (Fig. 4B). DCFH-DA was used to determine the effects of Adpa-Mn on intracellular ROS generation in U251 cells. As presented in Fig. 4C and D, the fluorescence intensity of DCFH-DA per 1×104 cells was significantly elevated following treatment with Adpa-Mn in a dose- and time-dependent manner, thus suggesting that intracellular ROS production was increased. Furthermore, exposure of U251 cells to Adpa-Mn resulted in ATP depletion (Fig. 4E). These results suggest that Adpa-Mn triggers mitochondrial dysfunction in U251 cells.

Adpa-Mn induces cell death dependent on mitochondrial dysfunction

Due to the finding that Adpa-Mn triggers mitochondrial membrane potential breakdown and ROS generation, the present study aimed to elucidate the association between ROS, mitochondria and Adpa-Mn-induced cell death (Fig. 5). Pretreatment with CsA (2 µM), an inhibitor of the mitochondrial permeability transition pore (MPTP) and NAC, a ROS scavenger, reduced Adpa-Mn-induced ROS production (Fig. 5A), whereas only CsA pretreatment was able to reverse Adpa-Mn-induced membrane potential collapse (Fig. 5B). Furthermore, MTT assay and Annexin V/PI staining indicated that cell death or apoptosis triggered by Adpa-Mn was significantly reduced by CsA pretreatment, but was aggravated to some extent by NAC pretreatment (Fig. 5C and E). In addition, CsA and NAC were able to reduce LC3 II expression (Fig. 5D). These findings indicate that Adpa-Mn may disrupt the mitochondrial membrane to induce apoptosis and protective autophagy in U251 cells.

Discussion

The success of cisplatin in the treatment of patients with cancer has resulted in the hypothesis that other metal complexes may be considered as potential drugs in future chemotherapy regimens. The present study aimed to verify whether the designed Mn-compound, Adpa-Mn, may be used as an anti-glioma lead compound. In the present study, Adpa-Mn was demonstrated to be active against glioma cells, whereas it had little effect on normal astrocytes. In addition, Adpa-Mn time and dose-dependently induced mitochondrial dysfunction, as determined by mitochondrial collapse and ROS generation, finally resulting in apoptotic cell death.

Apoptosis is a type of cell death that is characterized by nuclear condensation and fragmentation, and apoptotic body emergence without plasma membrane breakdown. During apoptosis, numerous death signals converge on mitochondria, particularly the release of cytochrome c as a consequence of the increased permeability of the outer mitochondrial membrane, which subsequently activates downstream caspase signaling (25). In the present study, Adpa-Mn was revealed to induce apoptosis mediated by the mitochondrial pathway (Fig. 2).

Autophagy is a dynamic process, which is often termed autophagic flux, whereby autophagosomes are formed in response to stimuli, engulf the cellular content and damaged organelles, and fuse with lysosomes; the contents of the autophagosome are subsequently degraded (26). Autophagy has previously been referred to as a physiological process that has a protective role in cells that encounter environmental stress, including starvation and pathogenic infection (27). Excess autophagy can also act as a pro-death mechanism; therefore, it has been classified as type II programmed cell death or autophagic cell death (28). In U251 cells, the following characteristics of autophagy were detected following Adpa-Mn treatment: Formation of AVOs and elevated ratio of LC3-II to LC3-I. When combined with the autophagy inhibitors, CQ and 3-MA, cell viability was markedly inhibited (Fig. 3). These results suggested that autophagy may have a protective role during Adpa-Mn treatment, which is similar to what has been reported for other chemicals (29,30).

ROS are predominantly generated from mitochondria and have a central role in cell death processes, including apoptosis and autophagy (31,32). Under excessive oxidative stress, the accumulation of ROS reaches a threshold level that triggers opening of the MPTP or oxidation of the mitochondrial outer membrane, which in turn leads to the simultaneous collapse of mitochondrial membrane potential and a transient increase in ROS generation by the respiratory chain that transforms O2 into ATP (33,34). Adpa-Mn was able to induce mitochondrial dysfunction, including mitochondrial membrane potential collapse, ROS accumulation and ATP depletion. Conversely, pretreatment with NAC and CSA significantly inhibited Adpa-Mn-induced ROS accumulation. In addition, NAC and CSA hampered Adpa-Mn-induced autophagy, whereas CsA decreased, but NAC aggravated, apoptosis (Fig. 5). CsA was also able to prevent Adpa-Mn-induced mitochondrial membrane potential collapse, whereas NAC could not. These data suggested that Adpa-Mn treatment destroyed mitochondria, thus leading to apoptosis of U251 cells; however, ROS that originated from the damaged mitochondria triggered protective autophagy.

In our previous study, the Adpa-Mn complex exhibited high toxicity on cancer cell lines, but showed weak DNA binding and cleavage activity (20). In addition, Adpa-Mn was shown to induce apoptotic cell death of HeLa cells (35). Conversely, whereas Adpa-Mn-induced autophagy in HeLa cells acted as a form of cell death, it was protective in U251 cells. These findings suggested that there may be different mechanisms underlying autophagy regulation in HeLa and U251 cells, which require further investigation.

In conclusion, the present study demonstrated that Adpa-Mn exhibited selective inhibition on glioma cell proliferation coupled with mitochondria-mediated apoptosis and increased autophagy. In addition, Adpa-Mn-induced autophagy exerted protective effects on glioma cells. Therefore, Adpa-Mn alone, or combined with autophagy inhibitors, may be considered a novel option for the treatment of glioma.

Acknowledgments

The authors of the present would you like to thank the following: The National Natural Science Foundation of China (grant no. 81402938); the Natural Science Foundation of Jiangsu Province (grant nos. BK2012710 and 20140575); and the Grant of Jiangsu University (grant no. 13JDG064).

References

1 

Walker MD, Green SB, Byar DP, Alexander E Jr, Batzdorf U, Brooks WH, Hunt WE, MacCarty CS, Mahaley MS Jr, Mealey J Jr, et al: Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med. 303:1323–1329. 1980. View Article : Google Scholar : PubMed/NCBI

2 

Subach BR, Witham TF, Kondziolka D, Lunsford LD, Bozik M and Schiff D: Morbidity and survival after 1, 3-bis (2-chloroethyl)-1-nitrosourea wafer implantation for recurrent glioblastoma: A retrospective case-matched cohort series. Neurosurgery. 45:17–23. 1999. View Article : Google Scholar

3 

Chamberlain MC and Kormanik P: Salvage chemotherapy with taxol for recurrent anaplastic astrocytomas. J Neurooncol. 43:71–78. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Attardi G and Schatz G: Biogenesis of mitochondria. Annu Rev Cell Biol. 4:289–333. 1988. View Article : Google Scholar : PubMed/NCBI

5 

Bender A, Opel D, Naumann I, Kappler R, Friedman L, von Schweinitz D, Debatin KM and Fulda S: PI3K inhibitors prime neuroblastoma cells for chemotherapy by shifting the balance towards pro-apoptotic Bcl-2 proteins and enhanced mitochondrial apoptosis. Oncogene. 30:494–503. 2011. View Article : Google Scholar

6 

Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loeffler M, et al: Molecular characterization of mitochondrial apoptosis-inducing factor. Nature. 397:441–446. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, et al: Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 12:222–230. 2011. View Article : Google Scholar

8 

Mathew R and White E: Autophagy in tumorigenesis and energy metabolism: Friend by day, foe by night. Curr Opin Genet Dev. 21:113–119. 2011. View Article : Google Scholar : PubMed/NCBI

9 

He C and Klionsky DJ: Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 43:67–93. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Kimura T, Takabatake Y, Takahashi A and Isaka Y: Chloroquine in cancer therapy: A double-edged sword of autophagy. Cancer Rese. 73:3–7. 2013. View Article : Google Scholar

11 

Williams R: Free manganese (II) and iron (II) cations can act as intracellular cell controls. FEBS Lett. 140:3–10. 1982. View Article : Google Scholar : PubMed/NCBI

12 

Ansari KI, Kasiri S, Grant JD and Mandal SS: Apoptosis and anti-tumour activities of manganese(III)-salen and -salphen complexes. Dalton Trans. 8525–8531. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Kono Y and Fridovich I: Isolation and characterization of the pseudocatalase of Lactobacillus plantarum. J Biol Chem. 258:6015–6019. 1983.PubMed/NCBI

14 

Law NA, Caudle T and Pecoraro VL: Manganese redox enzymes and model systems: Properties, structures and reactivity. Adv Inorg Chem. 46:305–440. 1998. View Article : Google Scholar

15 

Luk E, Carroll M, Baker M and Culotta VC: Manganese activation of superoxide dismutase 2 in Saccharomyces cerevisiae requires MTM1, a member of the mitochondrial carrier family. Proc Natl Acad Sci USA. 100:10353–10357. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Calzolari A, Oliviero I, Deaglio S, Mariani G, Biffoni M, Sposi NM, Malavasi F, Peschle C and Testa U: Transferrin receptor 2 is frequently expressed in human cancer cell lines. Blood Cells Mol Dis. 39:82–91. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Aschner M and Aschner JL: Manganese transport across the blood-brain barrier: Relationship to iron homeostasis. Brain Res Bull. 24:857–860. 1990. View Article : Google Scholar : PubMed/NCBI

18 

Aschner M and Gannon M: Manganese (Mn) transport across the rat blood-brain barrier: Saturable and transferrin-dependent transport mechanisms. Brain Res Bull. 33:345–349. 1994. View Article : Google Scholar : PubMed/NCBI

19 

Li SJ, Jiang L, Fu X, Huang S, Huang YN, Li XR, Chen JW, Li Y, Luo HL, Wang F, et al: Pallidal index as biomarker of manganese brain accumulation and associated with manganese levels in blood: A meta-analysis. PloS One. 9:e939002014. View Article : Google Scholar : PubMed/NCBI

20 

Chen QY, Huang J, Li JF and Gao J: Synthesis, interaction with mitochondrial and cancer cells of a dinuclear manganese(II) complex: Mn2(Adpa)2Cl4. Chinese J Inorg Chem. 24:1789–1793. 2008.In Chinese.

21 

McCarthy KD and de Vellis J: Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol. 85:890–902. 1980. View Article : Google Scholar : PubMed/NCBI

22 

Lemasters JJ, Qian T, He L, Kim JS, Elmore SP, Cascio WE and Brenner DA: Role of mitochondrial inner membrane permeabilization in necrotic cell death, apoptosis, and autophagy. Antioxid Redox Signal. 4:769–781. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Tanida I, Minematsu-Ikeguchi N, Ueno T and Kominami E: Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy. 1:84–91. 2005. View Article : Google Scholar

24 

Eisenberg-Lerner A, Bialik S, Simon HU and Kimchi A: Life and death partners: Apoptosis, autophagy and the cross-talk between them. Cell Death Differ. 16:966–975. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Desagher S and Martinou JC: Mitochondria as the central control point of apoptosis. Trends Cell Biol. 10:369–377. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Thorburn A: Autophagy and its effects: Making sense of double-edged swords. PLoS Biol. 12:e10019672014. View Article : Google Scholar : PubMed/NCBI

27 

Klionsky DJ and Emr SD: Autophagy as a regulated pathway of cellular degradation. Science. 290:1717–1721. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L and Kroemer G: Cell death modalities: Classification and pathophysiological implications. Cell Death Differ. 14:1237–1243. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Han J, Hou W, Goldstein LA, Lu C, Stolz DB, Yin XM and Rabinowich H: Involvement of protective autophagy in TRAIL resistance of apoptosis-defective tumor cells. J Biol Chem. 283:19665–19677. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Li J, Hou N, Faried A, Tsutsumi S, Takeuchi T and Kuwano H: Inhibition of autophagy by 3-MA enhances the effect of 5-FU-induced apoptosis in colon cancer cells. Ann Surg Oncol. 16:761–771. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L and Elazar Z: Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J. 26:1749–1760. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Scherz-Shouval R and Elazar Z: ROS, mitochondria and the regulation of autophagy. Trends Cell Biol. 17:422–427. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Garlid KD and Beavis AD: Evidence for the existence of an inner membrane anion channel in mitochondria. Biochim Biophys Acta. 853:187–204. 1986. View Article : Google Scholar : PubMed/NCBI

34 

Zorov DB, Juhaszova M and Sollott SJ: Mitochondrial ROS-induced ROS release: An update and review. Biochim Biophys Acta. 1757:509–517. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Liu J, Guo W, Li J, Li X, Geng J, Chen Q and Gao J: Tumor-targeting novel manganese complex induces ROS-mediated apoptotic and autophagic cancer cell death. Int J Mol Med. 35:607–616. 2015.PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 14 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Geng J, Li J, Huang T, Zhao K, Chen Q, Guo W and Gao J: A novel manganese complex selectively induces malignant glioma cell death by targeting mitochondria. Mol Med Rep 14: 1970-1978, 2016
APA
Geng, J., Li, J., Huang, T., Zhao, K., Chen, Q., Guo, W., & Gao, J. (2016). A novel manganese complex selectively induces malignant glioma cell death by targeting mitochondria. Molecular Medicine Reports, 14, 1970-1978. https://doi.org/10.3892/mmr.2016.5509
MLA
Geng, J., Li, J., Huang, T., Zhao, K., Chen, Q., Guo, W., Gao, J."A novel manganese complex selectively induces malignant glioma cell death by targeting mitochondria". Molecular Medicine Reports 14.3 (2016): 1970-1978.
Chicago
Geng, J., Li, J., Huang, T., Zhao, K., Chen, Q., Guo, W., Gao, J."A novel manganese complex selectively induces malignant glioma cell death by targeting mitochondria". Molecular Medicine Reports 14, no. 3 (2016): 1970-1978. https://doi.org/10.3892/mmr.2016.5509