83
Views
5
CrossRef citations to date
0
Altmetric
Original Research

Magnetic Fe3O4 nanoparticles and chemotherapy agents interact synergistically to induce apoptosis in lymphoma cells

, , , , &
Pages 999-1004 | Published online: 19 Nov 2010

Abstract

The purpose of this study was to investigate the potential effects of combination therapy using magnetic nanoparticles of Fe3O4 (MNP-Fe3O4) and chemotherapeutic drugs on lymphoma cells. Proliferation, inhibition, and viability of Raji cells were detected by MTT and trypan blue exclusion. The percentage of cells undergoing apoptosis was detected by flow cytometry using fluorescein isothiocyanate-annexin V and propidium iodide staining. p53 and nuclear factor-κB (NF-κB) protein levels were measured by Western blot. The results showed that proliferation of Raji cells was inhibited by adriamycin or daunorubicin in a dose-and time-dependent manner. Cell sensitivity was improved and the 50% inhibitory concentrations of adriamycin and daunorubicin decreased when combined with a MNP-Fe3O4 carrier. Interestingly, increased apoptosis in Raji lymphoma cells was accompanied by upregulation of p53 protein and downregulation of NF-κB protein. Furthermore, the combination of MNP-Fe3O4 with adriamycin or daunorubicin increased p53 protein levels and decreased NF-κB protein levels more than adriamycin or daunorubicin alone, indicating that MNP-Fe3O4 could enhance the effect of chemotherapeutic drugs on p53 and NF-κB. Similar results for cell apoptosis and protein expression were not observed for the groups treated with dexamethasone ± MNP-Fe 3O4 (P > 0.05). These findings suggest a potential clinical application for MNP-Fe3O4 in combination with daunorubicin or adriamycin in the treatment of lymphoma.

Introduction

Treatment of aggressive lymphoma is difficult, and the standard of care involves multiagent chemotherapeutic protocols incorporating adriamycin, an anthracycline antibiotic. Adriamycin and a similar compound, daunorubicin, originally isolated from streptomyces, are widely used and highly effective chemotherapeutic agents.Citation1 It has recently been reported that glucocorticoids such as dexamethasone induce apoptosis in lymphoid cells and therefore are commonly used in chemotherapy protocols for lymphoid malignancies.Citation2,Citation3 However, this effect is of questionable clinical relevance because the drug doses required for significant intercalation to occur are associated with unacceptable side effects.Citation4 To minimize dose-related side effects during chemotherapy, a promising approach is to combine conventional chemotherapy with new strategies to maximize the efficacy of chemotherapy by enhancing drug delivery and increasing the drug concentration reaching tumor cells.Citation5

In recent years, nanomaterials, defined as particles with diameters of less than 100 nm, have been widely applied in the fields of technology and medicine due to their unique physiochemical properties and tunable characteristics.Citation6,Citation7 Especially, it has been incorporated into clinical application in therapy and diagnosis owing to it both improves the traditional drug delivery route and resists the initiative excretion of drugs in tumor cells.Citation7,Citation8 Nowadays, the most promising biocompatible materials are magnetic nanoparticles of iron oxide (MNP-Fe3O4), which are feasible to produce, easy to functionalize, and show satisfactory water solubilization and degradation in vivo.Citation8 It has also been reported that MNP-Fe 3O4 can improve the sensitivity of anticancer drugs and reverse multidrug resistance, so could be used for targeted drug carriers with target orientation.Citation8,Citation9

Apoptosis is the main mechanism of cell death, and is mediated by a cell-intrinsic suicide program, with the relative balance of pro- and antiapoptotic signaling pathways determining the fate of the cell.Citation10 Nuclear factor-κB (NF-κB) transcription factors play a central role as mediators of immune and inflammatory responses, as well as in the control of cell proliferation and apoptosis.Citation11 In addition to the well known function of p53 as a cell cycle regulator, the regulation of cell proliferation and apoptosis are also closely associated with regulation of normal development.Citation5,Citation12,Citation13

The combination of MNP-Fe3O4 with different chemotherapeutics may provide new clinical options in the treatment of lymphoma. However, few in vitro data are available for the combination of MNP-Fe3O4 with other compounds, eg, adriamycin, daunorubicin, and dexamethasone, all of which are currently used in the treatment of lymphoma. The present study was designed to identify the cytotoxicity of this treatment strategy in a human Burkitt’s lymphoma cell line, also known as Raji cells, and to investigate possible underlying mechanisms by specifying a group of genes that can be altered significantly by the above mentioned agents.

Materials and methods

Materials

The main materials used in this study were daunorubicin (Pfizer, New York, NY), adriamycin (Hisun Phamaceutical Co, Zhejiang, China), dexamethasone (Xianju Pharmaceutical Co, Zhejiang, China), MTT (Sigma-Aldrich Co, St. Louis, MO), and the Annexin-V-FITC Apoptosis Detection Kit (Peking University Center for Human Disease Genomics, Beijing, China). MNP-Fe3O4 obtained from the State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China, were well distributed in RPMI 1640 medium (Invitrogen, Carlsbad, CA) containing 10% (v/v) heat-inactivated fetal bovine serum (HyClone, Logan, UT) by using ultrasound treatment to obtain an MNP-Fe3O4 colloidal suspension. Different concentrations of adriamycin, daunorubicin, and dexamethasone conjugated with 50 μM MNP-Fe3O4 were prepared by mechanical absorption polymerization at 4°C for 48 hours, as previously reported.Citation14

Cell lines and culture

The Raji lymphoma cell line was obtained from the Center for Human Disease Genomics of Peking University. The cells were cultured in RPMI 1640 medium containing 10% (v/v) heat-inactivated fetal bovine serum, 100 U/mL penicillin, and 100 μg/mL streptomycin at 37°C in a humidified 5% CO2 incubator, and passaged once every 2–3 days.

MTT assays

To measure the cell inhibition rate, 2 × 105/mL Raji cells were incubated in 96-well flat-bottomed plates (CoStar, Charlotte, NC). Different concentrations of adriamycin, daunorubicin, dexamethasone, MNP-Fe3O4, adriamycin + MNP-Fe3O4, daunorubicin + MNP-Fe3O4, dexamethasone + MNP-Fe3O4, and RPMI 1640 alone were added to these cells and cultured at 37°C for 6, 12, 24, 48, and 72 hours, respectively. After cells were added to each well with 5 μL MTT (5 mg/mL) and incubated at 37°C for four hours, dimethyl sulfoxide 100 μL was added to each well and the plates were gently shaken for 10 minutes. Reduction of MTT was quantified by optical densities (OD) at a measurement wavelength of 570 nm and a reference wavelength of 630 nm using a Thermo Scientific Varioskan Flash (Thermo Fisher Scientific, Rockford, IL). The cell inhibition ratio was calculated as (1-OD treated group/OD control group) × 100%. IC50 was defined as the concentration required for 50% inhibition of cell growth.

Trypan blue staining

Cell viability was tested using trypan blue staining. Briefly, adriamycin 0.1 μg/mL, daunorubicin 0.1 μg/mL, and dexamethasone 100 μg/mL alone and in combination with MNP-Fe3O4 50 μmol/L were incubated with Raji cells in 24-well plates for different lengths of time. Cells were obtained and stained with trypan blue solution 0.4%. Finally, at least 200 cells were counted under the microscope.

Apoptosis assay by flow cytometric assay

Apoptosis rates were measured by flow cytometric assay.Citation15 After incubation in MTT assays, cells were collected at 48 hours, washed twice with phosphate-buffered saline, suspended in 200 μL of binding buffer and 10 μL of Annexin V-FITC for 20 minutes in the dark, and thereafter 300 μL of binding buffer and 5 μL of propidium iodide were added to each sample. Analyses were done using FACSCalibur® flow cytometry (Becton Dickinson, San Antonio, TX) with Cell Quest software.

Western blot analysis

After the experimental treatment, total protein was isolated and subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis, and then transferred to a nitrocellulose membrane. The blots were stained with either p53 or NF-κB antibodies (1:10000) overnight at 4°C, and then with horseradish peroxidase-conjugated goat antirabbit or mouse secondary antibody (1:5000) for one hour at room temperature. The signal was detected using the Odyssey® Infrared Imaging System (LI-COR Biosciences, Lincoln, NE), the signal received was analyzed by the Odyssey software, and β-actin was used as the internal control.Citation16

Statistical analysis

All experiments were repeated three times. Data were expressed as mean ± standard deviation and analyzed using the Statistical Package for Social Science (SPSS Release 17.0; SPSS Inc, Chicago, IL). P < 0.05 was considered to indicate a statistically significant difference.

Results

Cell growth and inhibition

Inhibition of proliferation rate in Raji cells was enhanced by an increase in concentration and incubation time. After being coincubated with MNP-Fe3O4 for 48 hours, Raji cell proliferation was markedly inhibited. The IC50 of adriamycin decreased from 12.273 μg/mL to 8.529 μg/mL and that of daunorubicin from 2.695 μg/mL to 1.105 μg/mL. There were significant differences between the adriamycin + MNP-Fe3O4- treated group and the adriamycin-treated group (P < 0.05), and between the (daunorubicin + MNP-Fe3O4)-treated group and daunorubicin-treated group (), but there was no obvious enhancement in the dexamethasone + MNP-Fe3O4- treated group (data not shown). Similar results were found on trypan blue staining (see ).

Figure 1 Proliferation inhibitory ratio of Raji cells incubated with different drugs with or without MNP-Fe3O4. A) ADM incubated with Raji cells for 48 hours. B) DNR incubated with Raji cells for 48 hours. C) Either 0.1 μg/mL ADM or 0.1 μg/mL DNR ± 50 μmol/L MNP-Fe3O4 incubated with Raji cells for different lengths of time.

Abbreviations: MNP-Fe3O4, magnetic nanoparticles of Fe3O4; ADM, adriamycin; DNR, daunorubicin.

Figure 1 Proliferation inhibitory ratio of Raji cells incubated with different drugs with or without MNP-Fe3O4. A) ADM incubated with Raji cells for 48 hours. B) DNR incubated with Raji cells for 48 hours. C) Either 0.1 μg/mL ADM or 0.1 μg/mL DNR ± 50 μmol/L MNP-Fe3O4 incubated with Raji cells for different lengths of time.Abbreviations: MNP-Fe3O4, magnetic nanoparticles of Fe3O4; ADM, adriamycin; DNR, daunorubicin.

Table 1 Cell viability of different drugs with or without MNP-Fe3O4 for different times (%, mean ± SD)

Enhancement of apoptosis by adriamycin and daunorubicin

After being incubated with MNP-Fe3O4 for 48 hours, the proportion of apoptosis in Raji cells induced by the combination of MNP-Fe3O4 with adriamycin and with daunorubicin was 69.04 ± 0.38% and 55.36 ± 0.71%, respectively, and was increased significantly (P < 0.05) when compared with either adriamycin (38.37 ± 0.46%) or daunorubicin (39.49 ± 0.61%) used alone. However, there was no significant difference in the relative proportion of apoptotic cells between cells treated with dexamethasone + MNP-Fe3O4 and cells treated with dexamethasone alone (34.24 ± 0.88% versus 27.41 ± 0.23%, P > 0.05, ).

Figure 2 Apoptosis of Raji cells incubated with different drugs for 48 hours. A) control, B) MNP-Fe3O4, C) ADM, D) ADM + MNP-Fe3O4, E) DNR, F) DNR + MNP-Fe3O4, G) DEX, H) DEX + MNP-Fe3O4.

Abbreviations: MNP-Fe3O4, magnetic nanoparticles of Fe3O4; ADM, adriamycin; DNR, daunorubicin; DEX, dexamethasone.

Figure 2 Apoptosis of Raji cells incubated with different drugs for 48 hours. A) control, B) MNP-Fe3O4, C) ADM, D) ADM + MNP-Fe3O4, E) DNR, F) DNR + MNP-Fe3O4, G) DEX, H) DEX + MNP-Fe3O4.Abbreviations: MNP-Fe3O4, magnetic nanoparticles of Fe3O4; ADM, adriamycin; DNR, daunorubicin; DEX, dexamethasone.

Expression of p53 and NF-κB

Western blot assay revealed the p53 protein levels for the different treatment groups (). Combination of MNP-Fe3O4 with either adriamycin or daunorubicin increased the expression of p53 protein more effectively than adriamycin or daunorubicin alone. However, there was no significant difference in expression of p53 protein between cells treated with dexamethasone + MNP-Fe3O4 and cells treated with dexamethasone alone (P > 0.05). NF-κB protein levels were also influenced by adriamycin + MNP-Fe3O4 and daunorubicin + MNP-Fe3O4. Compared with adriamycin treatment alone, MNP-Fe3O4 + adriamycin decreased NF-κB protein levels more effectively (P < 0.05), as did MNP-Fe3O4 + daunorubicin when compared with daunorubicin alone, suggesting that MNP-Fe3O4 can enhance the effects of both adriamycin and daunorubicin. However, p53 protein expression in Raji cells was not significantly different (P > 0.05) between cells treated with dexamethasone ± MNP-Fe3O4.

Figure 3 Expressions of P53 and NF-κB protein in Raji cells. Lane 1) control; Lane 2) 0.1 μg/mL ADM; Lane 3) 0.1 μg/mL DNR; Lane 4) 100 μg/mL DEX; Lane 5) 50 μmol/L MNP-Fe3O4; Lane 6) 0.1 μg/mL ADM+ 50 μmol/L MNP-Fe3O4; Lane 7) 0.1μg/mL DNR+ 50 μmol/L MNP-Fe3O4; Lane 8) 100 μg/mL DEX+ 50 μmol/L MNP-Fe3O4.

Abbreviations: MNPs(Fe3O4), magnetic nanoparticles of Fe3O4; ADM, adriamycin; DNR, daunorubicin; DEX, dexamethasone.

Figure 3 Expressions of P53 and NF-κB protein in Raji cells. Lane 1) control; Lane 2) 0.1 μg/mL ADM; Lane 3) 0.1 μg/mL DNR; Lane 4) 100 μg/mL DEX; Lane 5) 50 μmol/L MNP-Fe3O4; Lane 6) 0.1 μg/mL ADM+ 50 μmol/L MNP-Fe3O4; Lane 7) 0.1μg/mL DNR+ 50 μmol/L MNP-Fe3O4; Lane 8) 100 μg/mL DEX+ 50 μmol/L MNP-Fe3O4.Abbreviations: MNPs(Fe3O4), magnetic nanoparticles of Fe3O4; ADM, adriamycin; DNR, daunorubicin; DEX, dexamethasone.

Discussion

Resistance of lymphoma cells to chemotherapy-induced apoptosis remains the most significant contributor to the development of multidrug resistance and treatment failure. Moreover, the toxicity of high-dose anticancer drugs limits the efficacy of many chemotherapeutic agents.Citation17 Strategies to reverse the resistance mechanism have been a hot topic in tumor therapy. The potential medical applications of MNP-Fe3O4 as a targeted-drug carrier with target orientation and sustained-release properties have been widely studied. It has been previously reported that MNP-Fe3O4 in combination with anticancer drugs could improve the efficacy of chemotherapeutic agents which has a synergistic effect in multidrug resistance.Citation18Citation21

To determine if the effects of MNP-Fe3O4 combined with low-dose chemotherapy are different from those of high-dose chemotherapy, we investigated the effects of a combination of chemotherapeutic agents (adriamycin, daunorubicin, dexamethasone) with MNP-Fe3O4 on proliferation and viability of Raji cells in vitro. Our results show that both adriamycin and daunorubicin can inhibit Raji cell proliferation effectively in a dose-and time-dependent manner. Furthermore, the effect on cell sensitivity may be further improved, and the IC50 values for adriamycin and daunorubicin were decreased when these agents were combined with (MNP-Fe3O4) as a carrier, so it may be inferred that MNP-Fe3O4 has synergism with adriamycin and daunorubicin.

Apoptosis is the consequence of a series of precisely regulated cellular events and is an important metabolic step in regulating cell numbers and growth. If apoptosis is blocked, cell metabolism becomes disordered, predisposing to development and growth of tumors.Citation22 We have investigated the cytotoxicity of MNP-Fe3O4 and chemotherapeutic agents to Raji cells in vitro by flow cytometry. The combination of adriamycin or daunorubicin with MNP-Fe3O4 exerted a potent cytotoxic effect on Raji cells and induced apoptosis, suggesting that MNP-Fe3O4 synergistically enhances the apoptosis induced by adriamycin or daunorubicin. However, similar results were not observed for dexamethasone.

It is well known that p53 is an important tumor suppressor leading to apoptosis in response to most DNA-damaging agents involving primarily the intrinsic pathway,Citation23 and that NF-κB also plays an important role. The NF-κB family of transcription factors, which regulate the expression of several genes, plays a critical role in numerous cellular processes and in tumor progression via activation of the NF-κB signal way.Citation24 p53 defects and NF-κB overexpression are major contributing factors to drug resistance, and have recently been linked to reduced chemosensitivity.Citation25 In most patients with lymphoma who have loss of p53 function and overexpression of NF-κB might help circumvent resistance to conventional therapies. Our results showed that either adriamycin or daunorubicin increased apoptosis of Raji cells and that this was accompanied by upregulation of p53 protein and downregulation of NF-κB protein. Interestingly, in the present study, MNP-Fe3O4 combined with adriamycin or daunorubicin increased upregulation of p53 protein and downregulation of NF-κB protein, but similar results were not found with dexamethasone. Our findings for NF-κB were similar to those reported by Ottonello et al, inhibition of NF-kB activation decreased the production of XIAP, an antiapoptotic molecule, leading to uncontrolled activity of regulators of apoptosis (eg, caspase 3).Citation26 Dexamethasone-induced apoptosis in human follicular lymphoma cells with Bcl-XL and dominant negative caspase 9 overexpression has been reported,Citation3 but this was not found for the Raji cells used in our study, and was not improved by combination with (MNP-Fe3O4), which needs to be explored further in the future. Overall, our results indicate that increased p53 expression and reduction of NF-κB induced by different activators contribute to inhibition of cell proliferation and induction of apoptosis in this system.

Conclusion

Our study demonstrates that concomitant treatment with adriamycin or daunorubicin and MNP-Fe3O4 could enhance proliferation and inhibition, as well as increase adriamycin- and daunorubicin-induced cell apoptosis in vitro, as a result of regulation of various proliferative and antiapoptotic gene products, including p53 and NF-κB. This would promote a potential clinical application for MNP-Fe3O4 in combination with daunorubicin or adriamycin against lymphoma. However, the effect and safety of such combinations need to be investigated further in the clinical setting.

Disclosure

The authors report no conflicts of interest in this work.

References

  • DoroshowJHChabnerBALongoDLAnthracyclines and anthracenedionesCancer Chemotherapy and Biotherapy3rd edPhiladelphia, PALippincott Williams and Wilkins2001
  • HerrIGasslerNFriessHBuchlerMWRegulation of differential pro- and anti-apoptotic signaling by glucocorticoidsApoptosis20071227129117191112
  • NuutinenURopponenASuorantaSDexamethasone-induced apoptosis and up-regulation of Bim is dependent on glycogen synthase kinase-3Leuk Res2009331714171719559478
  • SuiLLMooreMJTannockIFHillRPBristowRGHarringtonLPharmacology of anticancer drugsThe Basic Science of Oncology4th edNew York, NYMcGraw-Hill2005
  • TokimotoCPzadurRCancer management: A multidisciplinary approach: Medical, surgical, and radiation oncologyPrinciples of Chemotherapy8th edNew York, NYPPR2004
  • DuguetEVasseurSMornetSDevoisselleJMMagnetic nanoparticles and their applications in medicineNanomedicine (Lond)2006115716817716105
  • DaveSRGaoXHMonodisperse magnetic nanoparticles for biodetection, imaging, and drug delivery: A versatile and evolving technologyWiley Interdiscip Rev Nanomed Nanobiotechnol2009158360920049819
  • SchroederUSommerfeldPUlrichSSabelBANanoparticle technology for delivery of drugs across the blood-brain barrierJ Pharm Sci199887130513079811481
  • LinBLShenXDCuiSApplication of nanosized Fe3O4 in anticancer drug carriers with target-orientation and sustained-release propertiesBiomed Mater2007213213418458446
  • JarpeMBWidmannCKnallCAnti-apoptotic versus pro-apoptotic signal transduction: Checkpoints and stop signs along the road to deathOncogene199817147514829779994
  • KarinMGretenFRNF-kappa B: Linking inflammation and immunity to cancer development and progressionNat Rev Immunol2005574975916175180
  • LevineAJOrenMThe first 30 years of p53: Growing ever more complexNat Rev Cancer2009974975819776744
  • ReedJCRegulation of apoptosis by bcl-2 family proteins and its role in cancer and chemoresistanceCurr Opin Oncol199575415468547403
  • SunQChenBAWangXMPreparation of Fe3O4-magnetic nanoparticles loaded with adriamycin and its reversal of multidrug resistance in vitroZhongguo Shi Yan Xue Ye Xue Za Zhi20071574875117708796
  • YanFJiangYLiYMZhenXCenJFangWRReversal of P-glycoprotein and multidrug resistance-associated protein 1 mediated multidrug resistance in cancer cells by HZ08 isomers, tetrataisohydro-quinolin derivativesBiol Pharm Bull20083112631264
  • ZhaoYPChenGFengBMicroarray analysis of gene expression profile of multidrug resistance in pancreatic cancerChin Med J (Engl)20071201743175218028764
  • DevitaVTDose response in alive and wellJ Clin Oncol19864115711593525764
  • SomaCEDubernetCBentolilaDBenitaSCouvreurPReversion of multidrug resistance by co-encapsulation of doxorubicin and cyclosporine A in polyalkylcyanoacrylate nanoparticlesBiomaterials2000211710619673
  • ZhangRYWangXMWuCHSynergistic enhancement effect of magnetic nanoparticles on anticancer drug accumulation in cancer cellsNanotechnology2006173622362619661614
  • ChenBAChengJShenMFMagnetic nanoparticle of Fe3O4 and 5-bromotetrandrin interact synergistically to induce apoptosis by daunorubicin in leukemia cellsInt J Nanomedicine20094657119421371
  • WangJQChenBAChengJXuWLSunXCComparison of reversal effects of 5-bromotetrandrine and tetrandrine on P-glycoprotein-dependent resistance to adriamycin in human leukemia cell line K562/A02Ai Zheng20082749149518479598
  • KerrJFWinterfordCMHarmonBVApoptosis: Its significance in cancer and cancer therapyCancer199473201320268156506
  • KojimaKKonoplevaMSamudioIJMDM2 antagonists induce p53-dependent apoptosis in AML: Implications for leukemia therapyBlood20051063150315916014563
  • GhoshSMayMJKoppEBNF-kappa B and Rel proteins: Evolutionarily conserved mediators of immune responsesAnnu Rev Immunol1998162252609597130
  • TurcoMCRomanoMFPetrellaABisogniRTassonePVenutaSNF-kappa B/Rel-mediated regulation of apoptosis in hematologic malignancies and normal hematopoietic progenitorsLeukemia200418111714574329
  • OttonelloLBertolottoMMontecuccoFBianchiGDallegriFDelayed apoptosis of human monocytes exposed to immune complexes is reversed by oxaprozin: Role of the Akt/IkappaB kinase/nuclear factor kappaB pathwayBr J Pharmacol200915729430619338579