Loading

Journal of Pharmacology and Clinical Toxicology

The Effect of Pegylated Liposomal Doxorubicin HCl on Cellular Mitochondrial Function

Research Article | Open Access | Volume 6 | Issue 1

  • 1. Department of Geriatrics, University of Arkansas for Medical Sciences, USA
+ Show More - Show Less
Corresponding Authors
Jeanne Y. Wei, UAMS Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, 4301 W. Markham, # 748, Little Rock, AR 72205, USA, Tel: 501-603-1261 Fax: 501-686-5300
Abstract

Pegylated liposomal doxorubicin HCl (Lipo-doxo) is one of the leading approved nanoparticle chemotherapeutic drugs widely used to treat variety of cancers and has demonstrated the most significant reduction in risk for cardiotoxicity compared to non-liposomal, conventional doxorubicin. Despite the fact that Lipo-doxo may target mitochondria either directly or indirectly, few data are available regarding cellular bioenergetics on the regulation of mitochondrial fission and fusion genes. In this work, C2C12 myoblast cells have been used as an in-vitro model to study cellular bioenergetics, variations in gene expressions, and biochemical alterations induced by Lipo-doxo under high glucose (25 mM) and normal glucose (5.5 mM) conditions. In C2C12 myoblast cells, Lipo-doxo treatment significantly reduced both mitochondrial oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) under both glucose conditions in a dose dependent manner. Furthermore, Lipo-doxo treatment dysregulated the expression of mitochondrial fission and fusion genes. This dysregulation of mitochondrial fission and fusion genes may influence transformation of the network’s connectivity. Using confocal microscopy of C2C12 myoblast cells, we showed that treatment of C2C12 cells with 30 µg/mL Lipo-doxo under high glucose and normal glucose  induced cellular defects. This study provided a better characterization of the effect of Lipo-doxo induced cardiotoxicity.

Keywords

• Nanoparticle drug-delivery

• Lipo-doxo

• C2C12 myoblasts

• Mitochondrial damage

Citation

Ameer FS, Zhang X, Azhar G, Wei JY (2018) The Effect of Pegylated Liposomal Doxorubicin HCl on Cellular Mitochondrial Function. J Pharmacol Clin Toxicol 6(1):1099.

ABBREVIATIONS

 Lipo-doxo: Pegylated Liposomal Doxorubicin HCl; PGC-1α: Peroxisome Proliferator-Activated Receptor Gamma Coactivator1alpha; PGC-1β: Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1beta; DRP: Dynamin-Related Protein 1; FIS1: Fission1; MFN1: Mitofusin1; MFN2: Mitofusin2; OCR: Oxygen Consumption Rate; ECAR: Extracellular Acidification Rate; qRT-PCR: Quantitative Reverse Transcription PCR; DMEM: Dulbecco’s Modified Eagle’s Medium; FBS: Fetal Bovine Serum; PBS: Phosphate Buffer Saline; NA: Numerical Aperture; cDNA: Complementary DNA; 2-DG: 2-deoxy-D-Glucose; FCCP: Trifluoromethoxy C

INTRODUCTION

Epidemiological evidence suggests that diabetics are at significantly higher risk for different types of cancer [1,2]. The presence of high blood sugars or hyperglycemia during chemotherapy for hematologic and solid tumors has been shown to correlate with increased toxicity for cancer cells as well as normal tissues. Data suggest that better glycemic control during chemotherapy might maintain the toxicity for cancer cells while protecting normal, non-cancerous cells from chemotherapy.

induced damage and improving the outcome of cancer patients [3,4].

Doxorubicin is a very commonly used drug for breast cancer treatment and the use of non-liposomal doxorubicin has been associated with significant cardiotoxicity [2,3]. The current use of polyethylene glycol coated (pegylated) liposomal doxorubicin HCl (Lipo-doxo), has a better safety profile and it is one of the leading approved nanoparticle products used in cancer therapy [2,3]. Pegylated liposomes are less extensively taken up by cells of the reticuloendothelial system and have a tendency to “leak” drug while in circulation, resulting in localization of doxorubicin via leaky vasculature in the tumor tissues [5,6]. It is well known that the cardiac muscle has a high concentration of mitochondria and is very vulnerable to damage from oxidative stress [7-9]. In mitochondria, doxorubicin is reduced to a reactive semiquinone by complex I, reoxidizing to the original form by univalent reduction of oxygen [10].

Mitochondrial morphology is very dynamic in nature with mitochondrial fission and fusion processes [11, 12]. A number of proteins regulate these processes including, dynaminrelated protein 1 (DRP1) and fission1 (FIS1) for mitochondrial fission and nuclear-encoded mitochondrial proteins, MFN1 and MFN2 for fusion [13-15]. Impairment of the mitochondria by chemotherapeutic agents such as doxorubicin could severely affect cellular homeostasis and result in cardiac dysfunction with increased morbidity and mortality [16]. In the setting of diabetes or high blood sugars, the toxic effect of chemotherapy could be enhanced. Hence, the optimal nutrient conditions to protect noncancerous cells from detrimental effects chemotherapy requires further investigation.

The current study has several objectives: 1) to analyze mitochondrial respiration and glycolysis using a flux analyzer in C2C12 cells treated with different concentrations of Lipodoxo under glucose stress conditions; 2) to evaluate the gene expression in C2C12 myoblast cells treated with Lipo-doxo; 3) to assess alterations in cell morphology in C2C12 cells in response to Lipo-doxo treatment.

MATERIALS AND METHODS

Reagents

Dulbecco’s modified Eagle’s medium (DMEM), glutamine, penicillin, streptomycin, and fetal bovine serum (FBS) were purchased from Gibco-Invitrogen (Grand Island, NY, USA). Pegylated liposomal doxorubicin (Doxil) was purchased from FormuMax Scientific (Silicon Valley, CA, USA). Mito Tracker Red CMXRos was purchased from ThermoFisher Scientific (Eugene, OR, USA).

Cell culture

The DMEM media containing 10% (v/v) FBS with two different clinically relevant glucose concentrations were used in the present study: normal (100 mg/dL or 5.5 mM) and high (450 mg/dL or 25 mM). Mouse musmusculus myoblast (C2C12) cells were purchased from American Type Culture Collection (Manassas, VA). C2C12 cells were cultured in BD Falcon 75 cm2 flasks, containing DMEM with 2 mM L-glutamine, supplemented with 10% (v/v) FBS, and penicillin G and streptomycin. Cell cultures were incubated at 37 °C in 5% CO2 . Primary cultures received new medium 24 h after seeding and were sub-cultured upon reaching more than 80% confluence by the use of 0.25% trypsin-EDTA, inactivated by trypsin neutralizing solution (Invitrogen).

Mitochondrial function assay

Mitochondrial function was assayed using an XF96 Extracellular Flux Analyzer (Seahorse Bioscience, North Billerica, MA) following manufacturer’s instructions. Briefly, C2C12 myoblast cells were seeded in XF96 cell culture plates (Seahorse Bioscience) in 80 µL of high glucose medium (or normal glucose medium) and cultured to 80% confluence overnight at 37 °C in a humidified incubator with 5% CO2 . Prior to the Lipo-doxo treatment, the growth medium in the well of the XF cell plate was replaced with fresh growth medium. Cells not exposed to Lipodoxo served as controls in each experiment. When cells reached sub-confluence, they were pre-treated for 24 h with 0, 15, and 30 µg/mL Lipo-doxo. Following treatment, the culture supernatant was removed and cells were washed three times with phosphatebuffered saline (PBS). Prior to the assay, the growth medium in the well of the XF cell plate was replaced with the appropriate assay medium. The sensor cartridge was calibrated, and the cell plate was incubated at 37 °C for one hour. All experiments were performed at 37 °C.

Measurement of oxygen consumption rate (OCR)

Mitochondrial respiration was measured using an XF Cell Mito Stress Test Kit (Seahorse Biosciences Billerica, MA, USA) following the manufacturer’s instructions. The following compounds were injected: oligomycin (2 μM), an ATP synthetase inhibitor; FCCP (0.5 μM), an uncoupler reagent; rotenone (0.5 μM), a mitochondrial complex I inhibitor; and antimycin A (0.5 μM), a mitochondrial complex III inhibitor.

Measurement of extra cellular acidification rate (ECAR)

The glycolysis assay was performed using an XF Glycolysis Stress test Kit (Seahorse Biosciences Billerica, MA) following the manufacturer’s instructions. Glycolysis, glycolytic capacity, and non-glycolytic acidification were calculated as for ECAR. The following compounds were injected: glucose (10 mM); oligomycin (2 μM), an ATP synthetase inhibitor; 2-deoxy-D-glucose (50 mM), glycolysis inhibitor.

Quantitative reverse transcription PCR (qRT-PCR)

C2C12 cells were cultured in six-well plates to 80% confluence in normal glucose medium, after which the cells were either remained in normal glucose medium, or were subjected to high glucose treatment. After that, the cells were pre-treated for 24 h with 30 µg/mL Lipo-doxo. Following incubation, total cellular RNA was extracted from cells using miRNeasy Mini Kit and RNase-free DNase I digestion (Qiagen, Valencia, CA) in accordance with the manufacture’s recommendations for mammalian cells. RNA with a high RNA integrity number and an A260 and A280 absorbance ratio ranging from 1.8 to 2.1 was utilized for cDNA synthesis. To quantitate the expression of the mRNA, real-time qRT-PCR was performed. To select a proper internal loading control for real-time qRT-PCR, we examined the expression of 5S ribosomal RNA (5S RNA) and 18S in Lipo-doxo treated versus untreated C2C12 cells, we found that the expression of 5S RNA remained unchanged in Lipo-doxo treated versus untreated C2C12 cells, while 18S expression changed significantly in Lipo-doxo treated versus untreated C2C12 cells. Therefore, 5S RNA was used as an internal loading control. The PCR amplification was performed in a 7900HT Fast Sequence Detector System (Applied Biosystems). CT values were automatically obtained. Relative expression values were obtained by normalizing CT values of the mRNA genes in comparison with CT values of the 5S gene (which was documented to remain unchanged across all the groups), using the delta-delta CT method.

Confocal microscopy

Briefly, C2C12 cells were seeded onto six-well plates and grown in normal glucose DMEM medium supplemented with 10% FBS at 37 °C and 5% CO2 for 24 h. Upon attachment and 80% confluence in normal glucose medium, the cells either remained in normal glucose medium, or were subjected to high glucose treatment. Next, C2C12 cells were treated with 30 µg/mL Lipo-doxo for 24 h. Prior to the Lipo-doxo treatment, the growth medium in the six-well plate was replaced with fresh growth medium. After treatment, the cells were washed with PBS twice and incubated in 150 nM Mito Tracker Red CMXRos (Invitrogen/ Molecular Probes, Eugene, OR) in growth medium at 37°C for 1 h. The cells were then washed and fixed in 3.7% formaldehyde for 10 min at room temperature. The fixative was removed by washing with PBS and fresh PBS was replaced for fixed cell imaging. All images were acquired using a 60X/1.4 NA oil objective at room temperature using a Nikon confocal microscope C2/C2si (Nikon, Inc.). A 1 mW and 561 nm wavelength solid-state helium–neon laser was used as the excitation source for confocal microscopy.

Statistical analysis

Data are given as mean ± SD, with n denoting the number of experiments unless otherwise indicated. The differentially expressed mRNAs with at least 1.5-fold changed were identified using a t-test with a cut off p value of either p < 0.05 or p < 0.01.

RESULTS

Lipo-doxo repressed the mitochondrial oxygen consumption rate (OCR)

We investigated the mitochondrial oxidative phosphorylation in C2C12 cells treated with different concentrations of Lipodoxo under high glucose (Figure 1A)

 Effect of Lipo-doxo on oxygen consumption rate (OCR) under high glucose (25 mM) vs. normal glucose (5.5 mM) conditions. Lipo-doxo  induces metabolic alterations in C2C12 cells. (A) Kinetic OCR responses of Lipo-doxo treated C2C12 cells under high glucose condition. (B) Basal  respiration, maximal respiration, and spare respiration capacity were calculated for Lipo-doxo treated and untreated C2C12 cell line under high  glucose condition. (C) Kinetic OCR responses of Lipo-doxo treated C2C12 cells under normal glucose condition. (D) Basal respiration, maximal  respiration, and spare respiration capacity were calculated for Lipo-doxo treated and untreated C2C12 cell line under normal glucose condition.  Asterisks (**) indicate statistically significant differences (p< 0.01) between Lipo-doxo untreated and treated C2C12 cells under high and normal  glucose conditions. Note that there were clear differences between Lipo-doxo untreated and treated C2C12 cells under high and normal glucose  conditions for basal respiration, which was significantly decreased in Lipo-doxo treated C2C12 cell under high glucose conditions. After 24 h of Lipodoxo incubation, OCR was measured under basal conditions followed by the sequential addition of oligomycin (2 ?M), FCCP (0.5 ?M), and rotenone/ antimycin A (0.5 ?M), as indicated.

Figure 1 Effect of Lipo-doxo on oxygen consumption rate (OCR) under high glucose (25 mM) vs. normal glucose (5.5 mM) conditions. Lipo-doxo induces metabolic alterations in C2C12 cells. (A) Kinetic OCR responses of Lipo-doxo treated C2C12 cells under high glucose condition. (B) Basal respiration, maximal respiration, and spare respiration capacity were calculated for Lipo-doxo treated and untreated C2C12 cell line under high glucose condition. (C) Kinetic OCR responses of Lipo-doxo treated C2C12 cells under normal glucose condition. (D) Basal respiration, maximal respiration, and spare respiration capacity were calculated for Lipo-doxo treated and untreated C2C12 cell line under normal glucose condition. Asterisks (**) indicate statistically significant differences (p< 0.01) between Lipo-doxo untreated and treated C2C12 cells under high and normal glucose conditions. Note that there were clear differences between Lipo-doxo untreated and treated C2C12 cells under high and normal glucose conditions for basal respiration, which was significantly decreased in Lipo-doxo treated C2C12 cell under high glucose conditions. After 24 h of Lipodoxo incubation, OCR was measured under basal conditions followed by the sequential addition of oligomycin (2 μM), FCCP (0.5 μM), and rotenone/ antimycin A (0.5 μM), as indicated.

and normal glucose (Figure 1C) conditions using the Seahorse XF96 Extracellular Flux Analyzer that measures the oxygen consumption rate (OCR). The seeding density and concentrations of the injection compounds were optimized. The mitochondrial stress test used four drugs that interrogate mitochondrial function. Oligomycin inhibits Complex V of the electron transport chain, and injection of this compound showed how much of the OCR was due to ATP synthesis. Subsequent injection of the uncoupler FCCP, a photonophore, allowed protons to move into the mitochondria matrix independent of the ATP synthase. In order to maintain the membrane potential, protons move back into the intermembrane space by increasing the flow of electrons across the electron transport chain to the maximum speed. Thus, injection of FCCP reveals the maximal respiratory capacity of the cells. The ratio of maximal OCR as compared to basal OCR is known as spare respiratory capacity. Rotenone and antimycin A block Complex I and III, respectively, completely inhibit the electron transport chain, and as such any remaining oxygen consumption after these drugs are introduced, is non-mitochondrial.

Here we observed the expected responses in OCR (Figure 1A and Figure 1C) as the C2C12 cells were treated with each successive compound. Compared with vehicle–treated cells, the Lipo-doxo treated cells exhibited a reduction in maximal respiratory capacity, as evidenced by reduced oxygen consumption under high glucose (Figure 1A) and normal glucose (Figure 1C) conditions. Both 15 µg/mL and 30 µg/mL Lipo-doxo concentrations significantly repressed basal respiration of the C2C12 cells at the resting state under high glucose (Figure 1A) and normal glucose (Figure 1C) conditions. When ATPase activity was blocked by oligomycin, ATP production was decreased in both empty vector-treated and Lipo-doxo treated C2C12 cells, which was further decreased in 15 µg/mL and 30 µg/mL Lipodoxo treated cells. This can be used to distinguish the percentage of oxygen consumption devoted to ATP synthesis and the percentage of oxygen consumption required to overcome the natural proton leak across the inner mitochondrial membrane. Subsequently, FCCP was injected to stimulate maximal capacity of cells for mitochondrial respiration. As shown previously, FCCP acted as an uncoupling agent. Since the cells had to overcome the proton leak across the inner mitochondrial membrane, OCR increased significantly as more O2 was consumed to pump the excess protons back across the mitochondrial membrane. Finally, rotenone inhibited mitochondrial Complex I and Complex III, respectively, which caused the flow of electrons to cease in the electron transport chain, and thus the consumption of O2 was drastically reduced. As shown in Figure 1B and Figure 1D data analysis revealed that the different concentrations of Lipo-doxo used in this study repressed basal respiration and maximal respiration under high and normal glucose conditions. Furthermore, the spare respiration capacity was up-regulated for 15 µg/mL Lipo-doxo treated C2C12 cells under high glucose condition. However, there was no significant change in the spare respiration capacity for 30 µg/mL Lipo-doxo treated C2C12 cells under nutrient stress conditions. These results suggested that C2C12 cells under normal glucose condition with or without treatment of Lipo-doxo, possessed higher mitochondrial oxidative phosphorylation activities than C2C12 cells under high glucose condition.

Lipo-doxo altered the glycolytic rate

Previous studies have shown that glycolysis account for ~80% of total ECAR in a number of cancer cells as determined through either removing glucose from the assay medium or adding glycolytic pathway inhibitor such as hexokinase inhibitor 2-DG [17]. The remaining 20% of the ECAR can be attributed to other metabolic processes, such as the tricarboxylic acid cycle CO2 evolution. The OCR response to glucose, monitored concurrently with ECAR serves as an indicator of whether glucose is also catabolized through mitochondrial respiration (Data not shown).

To determine glycolysis, glycolytic capacity, and nonglycolytic acidification of the C2C12 cells treated with 15 µg/ mL and 30 µg/mL Lipo-doxo under high glucose (Figure 2A)

 Effect of Lipo-doxo on extracellular acidification rate (ECAR) under high glucose (25 mM) vs. normal glucose (5.5 mM) conditions. (A)  Kinetic ECAR responses in Lipo-doxo treated and untreated C2C12 cells under high glucose condition. (B) Glycolysis, glycolytic capacity, and nonglycolytic acidification were calculated for Lipo-doxo treated and untreated C2C12 cells under high glucose condition. (C) Kinetic ECAR responses  in Lipo-doxo treated C2C12 cells under normal glucose condition. (D) Glycolysis, glycolytic capacity, and non-glycolytic acidification were calculated  for Lipo-doxo treated and untreated C2C12 cells under normal glucose condition. After 24 h of Lipo-doxo incubation, ECAR was measured under  basal conditions followed by the sequential addition of 10 mM glucose, 2 ?Moligomycin, and 50 mM 2-Deoxy-D-glucose (2-DG). A series of ECAR  in C2C12 cells in the presence or absence of Lipo-doxo were examined by Seahorse XF96 analyzer. ECAR following the addition of glucose defines  glycolysis and ECAR following oligomycin represents maximum glycolytic capacity. Data are shown as mean ± standard deviation, n = 3.

Figure 2 Effect of Lipo-doxo on extracellular acidification rate (ECAR) under high glucose (25 mM) vs. normal glucose (5.5 mM) conditions. (A) Kinetic ECAR responses in Lipo-doxo treated and untreated C2C12 cells under high glucose condition. (B) Glycolysis, glycolytic capacity, and nonglycolytic acidification were calculated for Lipo-doxo treated and untreated C2C12 cells under high glucose condition. (C) Kinetic ECAR responses in Lipo-doxo treated C2C12 cells under normal glucose condition. (D) Glycolysis, glycolytic capacity, and non-glycolytic acidification were calculated for Lipo-doxo treated and untreated C2C12 cells under normal glucose condition. After 24 h of Lipo-doxo incubation, ECAR was measured under basal conditions followed by the sequential addition of 10 mM glucose, 2 μMoligomycin, and 50 mM 2-Deoxy-D-glucose (2-DG). A series of ECAR in C2C12 cells in the presence or absence of Lipo-doxo were examined by Seahorse XF96 analyzer. ECAR following the addition of glucose defines glycolysis and ECAR following oligomycin represents maximum glycolytic capacity. Data are shown as mean ± standard deviation, n = 3.

and normal glucose (Figure 2C) conditions, we measured ECAR while consecutively injection glucose, oligomycin, and 2-DG. As shown in Figure 2A, glucose addition to C2C12 cells caused an instant increase in kinetic ECAR responses and triggered a glycolytic flux of 35 ± 2.7 mpH/min for control cells, 25 ± 1.2 mpH/min for 15 µg/mL Lipo-doxo treated cells, and 14 ± 2.8 mpH/min for 30 µg/ mL Lipo-doxo treated cells under high glucose conditions. The subsequent addition of oligomycin caused a further increase in ECAR to 47 ± 4.1 mpH/min for control cells, 45 ± 3.1 mpH/min for 15 µg/mL Lipo-doxo treated cells, and 37 ± 3.5 mpH/min for 30 µg/mL Lipo-doxo treated cells under high glucose conditions, indicating an elevated glucose flux toward lactate and revealing the glycolysis capacity of C2C12 cells treated with different concentrations of Lipo-doxo. The final addition of glycolysis inhibitor 2-DG abolished the overall glycolysis. Similar results were obtained under normal glucose condition (Figure 2C). The calculated glycolysis, glycolytic capacity, and non-glycolytic acidification for C2C12 cells treated with different concentrations of Lipo-doxo under high and normal glucose conditions are shown in Figure 2B and Figure 2D. Among two different glucose concentration tested in the present study, Lipo-doxo treated and untreated C2C12 cells under high glucose conditions showed higher values for glycolysis, glycolytic capacity, and nonglycolytic acidification (Figure 2B), as compared with the normal glucose conditions (Figure 2D). The concentration of oligomycin and glucose used in this study were optimized to get maximal inhibition of respiration (data not shown).

Dysregulation of mitochondrial fission and fusion genes expression as a result of Lipo-doxo treatment

The effect of Lipo-doxo on the expression of PGC-1α gene and PGC-1β gene, which are upstream regulators of mitochondrial genes, were examined. As shown in Figure 3A1,

 Quantitative reverse transcription PCR (qRT-PCR) validation of changes in the expression of selected genes involved in mitochondrial  biogenesis and function. Effect of 30 µg/mL Lipo-doxo for 24 h on (A) PGC-1? and ?, (B) MFN1 and 2, and (C) FIS1 and DRP1 genes under high  glucose and normal glucose conditions. Cells were harvested for the analysis of gene expression by qRT-PCR, and the data were normalized to those  for 5S. * p < 0.05 and ** p < 0.01. Data are expressed as the mean ± standard deviation of three independent experiments.

Figure 3 Quantitative reverse transcription PCR (qRT-PCR) validation of changes in the expression of selected genes involved in mitochondrial biogenesis and function. Effect of 30 µg/mL Lipo-doxo for 24 h on (A) PGC-1α and β, (B) MFN1 and 2, and (C) FIS1 and DRP1 genes under high glucose and normal glucose conditions. Cells were harvested for the analysis of gene expression by qRT-PCR, and the data were normalized to those for 5S. * p < 0.05 and ** p < 0.01. Data are expressed as the mean ± standard deviation of three independent experiments.

under high glucose stress condition treatment with C2C12 cells with 30 μg/ mL Lipo-doxo for 24 h up-regulated the expression of PGC-1α mRNA level (p < 0.01, n = 3), but the expression of PGC-1β mRNA level (p < 0.01, n = 3) was down-regulated. Similar effect was observed under normal glucose condition (Figure 3A2).

The PGC-1α is upstream regulator of mitochondrial genes, including mitofusin genes. The MFN1 and MFN2 participate in the regulation of the mitochondrial network [18,19]. As shown in Figure 3B1, we detected the expression levels of MFN1 and MFN2 and observed that MFN1 (p<0.01, n= 3) was downregulated upon 30 µg/mL Lipo-doxo treatment for 24 h under high glucose condition. However, there is no significant change in the expression of MFN2 (NS, n = 3) upon 30 µg/mL Lipo-doxo treatment for 24 h under high glucose condition. As shown in Figure 3B2, under normal glucose condition, when C2C12 cells were treated with 30 µg/mL Lipo-doxo for 24 h MFN1 (p< 0.05, n= 3) was down-regulated and MFN2 was up regulated (n = 3p< 0.01).

As shown in Figure 3C1, treatment with 30 µg/mL Lipo-doxo for 24 h under high glucose condition repressed the expression of both FIS1 (p < 0.05, n = 3) and DRP1 (p <0.01, n = 3) mRNA level. Similar results were observed under normal glucose condition (Figure 3C2) for FIS1 (p < 0.01, n = 3) and DRP1 (p < 0.01, n = 3) mRNA level.

Lipo-doxo altered the mitochondrial morphology and mitochondrial membrane potential

C2C12 myoblast cells were star-shaped, with a prominent single central nucleus, with numerous nucleoli. By using confocal microscopy, alterations in mitochondrial morphology were evaluated using Mito tracker red (Figure 4).

 Change in mitochondrial morphology in Lipo-doxo exposed C2C12 cells under different glucose concentrations treatment. Exposure to 30  µg/mL Lipo-doxo under high glucose and normal glucose conditions induces fine structural changes of mitochondria in C2C12 myoblast cells. Cells  were stained with Mito tracker red. Confocal microscopy images of C2C12 cells treated with (A, C) 0 µg/mL and (B, D) 30 µg/mL of Lipo-doxo under  high glucose (top) and normal glucose (bottom) conditions for 24 h. Quantitation of the intensity of Mito tracker red staining under (E) high glucose  and (F) normal glucose conditions. *Denotes statistical difference between a Lipo-doxo treated and the untreated control (p < 0.05, n=3). Note that  Mito tracker red fluorescence appears to be diminished with Lipo-doxo treatment under high and normal glucose conditions for 24 h. However,  mitochondria still maintained some degree of membrane polarization as evidenced by their ability to stain with Mito tracker red. The scale bar of  each image is 50 µm. Images were acquired using a 60X oil immersion objective with a NA of 1.40. All images have the same magnification.

Figure 4 Change in mitochondrial morphology in Lipo-doxo exposed C2C12 cells under different glucose concentrations treatment. Exposure to 30 µg/mL Lipo-doxo under high glucose and normal glucose conditions induces fine structural changes of mitochondria in C2C12 myoblast cells. Cells were stained with Mito tracker red. Confocal microscopy images of C2C12 cells treated with (A, C) 0 µg/mL and (B, D) 30 µg/mL of Lipo-doxo under high glucose (top) and normal glucose (bottom) conditions for 24 h. Quantitation of the intensity of Mito tracker red staining under (E) high glucose and (F) normal glucose conditions. *Denotes statistical difference between a Lipo-doxo treated and the untreated control (p < 0.05, n=3). Note that Mito tracker red fluorescence appears to be diminished with Lipo-doxo treatment under high and normal glucose conditions for 24 h. However, mitochondria still maintained some degree of membrane polarization as evidenced by their ability to stain with Mito tracker red. The scale bar of each image is 50 µm. Images were acquired using a 60X oil immersion objective with a NA of 1.40. All images have the same magnification.

Figure 4A and 4B shows control C2C12 myoblasts with intact cell membranes and filamentous polarized mitochondria under high glucose and normal glucose conditions, respectively. Treatment with 30 µg/mL Lipo-doxo caused breakage of the mitochondrial network under both glucose concentrations (Figure 4C and 4D). Furthermore, loss of the mitochondrial Mito tracker red signal could be seen due to the mitochondrial depolarization induced by Lipo-doxo (Figure 4C and 4D). However, for 30 µg/mL Lipodoxo concentration tested, the mitochondrial network still accumulated Mito tracker red, demonstrated the retention of a transmembrane electric potential (Figure 4C and 4D).

Perturbation of mitochondrial membrane potential might be associated with various physiological and pathophysiologic processes inside cells, and its determination is widely used for the characterization of cellular and mitochondrial metabolism and viability, as well as apoptosis. Mito tracker red is a mitochondrion selective dye that accumulates in mitochondria in a membrane potential dependent manner. When used in appropriately designed experiments, Mito tracker red has been validated as a noncytotoxic and sensitive indicator of relative changes in mitochondrial membrane potential in several cultured cell types. The effect of 30 µg/mL Lipo-doxo for 24 h on mitochondrial membrane potential was evaluated in C2C12 cells under high glucose and normal glucose conditions. Cells were exposed to 30 µg/mL Lipo-doxo for 24 h and assayed for Mito tracker red uptake using confocal microscope. Mitochondrial staining was punctate in untreated cells (Figure 4A and 4B), indicative of selective uptake and concentration of Mito tracker red by healthy, actively respiring mitochondria. Once taken up by mitochondria, a mildly thiol-reactive chloromethyl moiety is responsible for keeping the dye associated with the organelle [20]. The brightness of the fluorescent intensity was reduced in cells exposed to Lipo-doxo (Figure 4C and 4D) indicates a significant reduction of mitochondrial membrane potential in C2C12 cells (Figure 4E and 4F). Furthermore, some of the cells but not all treated with 30 µg/mL Lipo-doxo for 24 h, exhibited more diffuse Mito tracker red staining throughout the cytosol, a pattern indicative of mitochondrial membrane depolarization (data not shown). This diffuse staining pattern was the result of a lack of healthy mitochondria and thereby reduced uptake of the probe, resulting in diffusion of Mito tracker red throughout the cytoplasm [21].

Treatment with 30 µg/mL Lipo-doxo under high glucose (Figure 5B)

 Lipo-doxo induces cytoplasm vacuolization under different glucose conditions. DIC images of C2C12 cells treated with (A, C) 0 µg/mL and  (B, D) 30 µg/mL of Lipo-doxo under high glucose (top) and normal glucose (bottom) conditions for 24 h. The scale bar of each image is 10 µm. All  images have the same magnification. Arrows indicate Lipo-doxo induced cytoplasmic vacuolization under different glucose conditions.

Figure 5 Lipo-doxo induces cytoplasm vacuolization under different glucose conditions. DIC images of C2C12 cells treated with (A, C) 0 µg/mL and (B, D) 30 µg/mL of Lipo-doxo under high glucose (top) and normal glucose (bottom) conditions for 24 h. The scale bar of each image is 10 µm. All images have the same magnification. Arrows indicate Lipo-doxo induced cytoplasmic vacuolization under different glucose conditions.

and normal glucose (Figure 5D) conditions for 24 h led to alterations in cellular morphology and the cytoplasm appeared to be vacuolated. Nuclear alterations induced by Lipodoxo exposure were also explored in the present work. 30 µg/ mL Lipo-doxo cause an increase in the area of cell nuclei under high glucose and normal glucose conditions. The increase in nuclear area was clearly observed by microscopy (Figure 5B and 5D) and was accompanied by what appeared to be nucleoli disorganization. An increase in nuclear size was observed regardless of the degree of vacuolization exhibited by C2C12 cells.

DISCUSSION

The present study has several major findings. In C2C12 myoblast cells, treatment with Lipo-doxo repressed mitochondrial oxygen consumption rate and extracellular acidification rate under high glucose and normal glucose conditions. The presence of Lipo-doxo induced mitochondrial morphological alterations in C2C12 cells and reduced mitochondrial membrane potential. Lipo-doxo treatment down-regulated the expression of MFN1, PGC-1β, FIS1 and DRP1 genes, but up-regulated the expression of MFN2 and PGC-1α genes (Table 1).

Table 1: Summary of differences between C2C12 myoblast cells treated with 30 µg/mL Lipo-doxo, under normal glucose and high glucose conditions.

  Normal Glucose (5 mM) High Glucose (25 mM)
Lipo-doxo treated Mean (±SD) Lipo-doxo treated Mean (±SD)
Oxygen consumption rate (pmol/min)
Basal respiration 82.6 ± 0.8 73.7 ± 4.0
Maximal respiration 284.1 ± 16.6 183.2 ± 13.8
Spare respiration capacity 201.5 ± 16.2 109.5 ± 10.5
Extra-cellular acidification rate (mpH/min)
Glycolysis 12.1 ± 3.0 13.8 ± 2.8
Glycolytic capacity 31.5 ± 4.9 37.6 ± 3.5
Non-glycolytic acidification 6.5 ± 0.8 12.2 ± 2.7
Real-time qRT-PCR (Fold change)
MFN1 0.74 ± 0.14 0.77 ± 0.04
MFN2 1.10 ± 0.03 1.13 ± 0.15
PGC-1α 2.88 ± 0.20 5.10 ±0.10
PGC-1β 0.26 ± 0.03 0.25 ± 0.01
DRP1 0.37 ± 0.01 0.35 ± 0.16
FIS1 0.92 ± 0.01 0.80 ± 0.09
Mitochondrial membrane potential (Normalized 
fluorescence intensity)
0.85 ± 0.11 0.60 ± 0.12
Abbreviations: Lipo-doxo: Pegylated Liposomal Doxorubicin HCl; PGC-1α: Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1alpha; 
PGC-1β: Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1beta; DRP1: Dynamin-Related Protein 1; FIS1: Fission1; MFN1: Mitofusin1; 
MFN2: Mitofusin2; qRT-PCR: Quantitative Reverse Transcription PCR; SD: Standard Deviation
 

In the recent years, targeting mitochondria emerged as an attractive strategy to control mitochondrial dysfunction related diseases. Despite the desire to direct therapeutics to the mitochondria, the actual task is more difficult due to the highly complex nature of the mitochondria. Only a handful of nanoparticles based on metal oxides, gold nanoparticles, dendrons, carbon nanotubes, and liposomes were recently engineered to target mitochondria. Most of these materials face tremendous challenges when administered in vivo due to their limited biocompatibility. Among the numerous types of nanocarriers, liposomes emerged as promising delivery systems [22-27].

Our results suggest that the reduced OCR after Lipo-doxo treatment may be due to Complex I damage, and could explain the decline in OCR of C2C12 cells treated with Lipo-doxo for 24 h. Previous reports have shown mitochondrial dysfunction [28], and inactivation of mitochondrial systems such as complexes I, III, IV, and V, and also phosphate carrier [29], and ATP-ADP translocase, partly due to binding of doxorubicin to cardiolipin of the inner mitochondrial membrane [30]. The spare respiratory capacity of mitochondria in response to oxidative stress and energetic insufficiency is important [31,32]. The maximal respiration mainly depends on the function of the substrate supplementation [33]. Souid and coworkers reported that oxidative phosphorylation is enhanced in Jurkat and HL-60 cells exposed to 1 µM non-liposomal conventional doxorubicin for 30 min [34]. They also demonstrated that incubation of cells with high concentrations of non-liposomal conventional doxorubicin (5-20 µM) for 1 h caused a significant inhibition of mitochondrial respiration [35]. Interestingly, C2C12 cells treated with Lipo-doxo under different glucose concentrations significantly changed the sensitivity of mitochondria to the mitochondrial uncoupler FCCP. The sensitivity to FCCP may depend on the conformational state of the mitochondrial inner membrane, such as changes in its fluidity and regularity. We also observed a significant decrease

in ATP levels in Lipo-doxo treated C2C12 cells (data not shown), which points to the major role of mitochondrial oxidative phosphorylation in maintaining energy status. We also assessed the differences in glycolysis-induced ECAR among the Lipo-doxo treated and untreated C2C12 cells under high glucose and normal glucose conditions (Figure 2). The results of the extracellular flux study showed a significant decrease in extracellular acidification rates in glycolysis, glycolytic capacity, and non-glycolytic acidification (Figure 2B and 2D) under normal glucose and high glucose conditions.

Interestingly, we noted that treatment of C2C12 myoblast cells with 30 µg/mL Lipo-doxo for 24 h up-regulated the expression of MFN2. In agreement with this finding, other studies tend to support the idea that increased MFN2 in cardiomyocytes predicts an adverse outcome, promoting cell death through apoptotic or autophagic mechanisms [9]. PGC-1α and PGC-1β are important positive regulators of mitochondrial activity and biogenesis in skeletal muscle [36]. Key mitochondrial processes, such as organelle biogenesis and uncoupling, are differentially regulated by these homologues. It has been shown that PGC-1α stimulates MFN2 gene promoter activity, and increases the expression of MFN2 mRNA and protein expression in both skeletal muscle and brown adipose tissue in conditions associated with enhanced energy expenditure [37]. It is likely that the up-regulation of PGC1α gene by Lipo-doxo treatment may have affected the expression of MFN2 gene expression. Cells lacking DRP1 contain highly interconnected mitochondrial nets that are formed by ongoing fusion in the absence of fission activity [11]. Previous work using animal models have demonstrated that MFN1 and MFN2 are essential to mitochondrial remodelling during postnatal cardiac development [38]. Mutations in the mitochondrial fission and fusion genes are associated with several human diseases [39].

The mitochondrial membrane potential is a useful tool to monitor changes in the cell’s capacity to generate ATP by oxidative phosphorylation [40]. According to the literature, repression of MFN2 caused changes in mitochondrial morphology, reduced the mitochondrial membrane potential, cellular respiration, glucose oxidation and proton leak as well as oxidation of pyruvate and fatty acids [41]. Wallace et al., reported that nonliposomal conventional doxorubicin altered the organization of H9C2 myoblast cardiac sarcomericmyocin [42]. A relation between non-liposomal conventional doxorubicin treatment and alterations in myocardial structural proteins and regulatory protein expression after in vivo treatment of rats has also been reported [43].

We also observed an increase in nuclear area after Lipo-doxo treatment under high glucose and normal glucose conditions. Similar nuclear swelling has been described after etoposide treatment of neuronal cells [44]. Changes in nuclear morphology, including chromatin de-condensation, were also observed by electron microscopy [42]. The vacuoles observed in the present study could have originated from sarcoplasmic reticular enlargement [45] or autophagic cell death [46]. Such vacuoles are known to contain products of lipid metabolism or peroxidation [47].

Epidemiological studies have revealed that diabetes is correlated with increased cancer risk [1,2], and that diabetics treated with metformin have reduced risk of developing various types of cancer [48,49]. Metformin is an extensively used and well-tolerated drug for treating individuals with type 2 diabetes. Metformin inhibits growth of breast cancer cell lines [50,51], blocks cellular transformation in an inducible model system [52,53], and has anti-tumor effects in mouse xenografts [52-55]. Furthermore, in mouse xenografts involving a human breast cancer cell line, co-injection of metformin and doxorubicin intra-peritoneally near the tumor increases the rate of tumor regression compared to treatment with doxorubicin alone, and this combinatorial therapy prevents relapse for at least several months [52]. The results from the present study support the notion that Lipo-doxo is more toxic under high glucose compared to normal glucose conditions (Table 1). This implies that chemotherapy drugs cause damage not only to cancer cells, but also normal cells especially at high doses under high glucose conditions. Our results are in agreement with previous studies which showed that short-term starvation can selectively sensitize cancer cells to chemotherapeutics, while simultaneously protecting normal cells from their side effects via the insulin-like growth factor 1 pathway and the regulation of the glucose levels [9].

CONCLUSION

The present data suggest that treatment of C2C12 myoblast cells with Lipo-doxo down-regulated the expression of MFN1, PGC-1β, FIS1, and DRP1 genes and up-regulated the expression of PGC-1α and MFN2 genes, and likely thereby altered the mitochondrial membrane potential and reduced mitochondrial function. Lipo-doxo treatment also significantly reduced both mitochondrial oxygen consumption rate and extra cellular acidification rate under high glucose and normal glucose conditions in a dose dependent manner. There were several morphological alterations after treatment of C2C12 myoblasts with 30 µg/mL Lipo-doxo for 24 h under high glucose stress conditions including nuclear swelling and the formation of vacuoles. This study may help in the development of new studies on therapeutic targets for preserving mitochondria and preventing myocardial damage associated with the use of Lipodoxo in treatment of a variety of cancers.

ACKNOWLEDGMENTS

This study was supported in part by the Claude D. Pepper Older American Independence Center grant (P30AG28718). We thank Laura Gocio for the assistance with the manuscript preparation.

REFERENCES

1. Larsson SC, Mantzoros CS, Wolk A. Diabetes mellitus and risk of breast cancer: A meta-analysis. Int J Cancer. 2007; 121: 856-862.

2. Hsu IR, Kim SP, Kabir M, Bergman RN. Metabolic syndrome, hyperinsulinemia, and cancer. Am J Clin Nutr. 2007; 86: 867-871.

3. Duan W, Shen X, Lei J, Xu Q, Yu Y, Li R, et al. Hyperglycemia, a Neglected Factor during Cancer Progression. BioMed Res Int. 2014; 2014: 10.

4. Brunello A, Kapoor R, Extermann M. Hyperglycemia During Chemotherapy for Hematologic and Solid Tumors Is Correlated With Increased Toxicity. Am J Clin Oncol. 2011; 34: 292-296.

5. Rose PG. Pegylated liposomal doxorubicin: optimizing the dosing schedule in ovarian cancer. Oncologist. 2005; 10: 205-214.

6. Rahman AM, Yusuf SW, Ewer MS. Anthracycline-induced cardiotoxicity and the cardiac-sparing effect of liposomal formulation. Int J Nanomed. 2007; 2: 567-583.

7. Zhang S, Liu X, Bawa-Khalfe T, Lu L-S, Lyu YL, Liu LF, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med. 2012; 18: 1639-1642.

8. Carvalho FS, Burgeiro A, Garcia R, Moreno AJ, Carvalho RA, Oliveira PJ. Doxorubicin-induced cardiotoxicity: from bioenergetic failure and cell death to cardiomyopathy. Medl Res Rev. 2014; 34: 106-135.

9. Todorova VK, Makhoul I, Siegel ER, Wei J, Stone A, Carter W, et al. Biomarkers for Presymptomatic Doxorubicin-Induced Cardiotoxicity in Breast Cancer Patients. PLoS ONE. 2016; 11: e0160224.

10. Xu MF, Tang PL, Qian ZM, Ashraf M. Effects by doxorubicin on the myocardium are mediated by oxygen free radicals. Life Sci. 2001; 68: 889-901.

11. Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol. 2010; 11: 872-884.

12. Frazier Ann E, Kiu C, Stojanovski D, Hoogenraad Nicholas J, Ryan Michael T. Mitochondrial morphology and distribution in mammalian cells. Biol Chem. 2006; 387: 1551-1558.

13. Song M, Mihara K, Chen Y, Scorrano L, Dorn Ii Gerald W. Mitochondrial fission and fusion factors reciprocally orchestrate mitophagic culling in mouse hearts and cultured fibroblasts. Cell Metab. 2015; 21: 273- 285.

14. Dietrich Marcelo O, Liu Z-W, Horvath Tamas L. Mitochondrial dynamics controlled by mitofusins regulate agrp neuronal activity and diet-induced obesity. Cell. 2013; 155: 188-199.

15. Ranieri M, Brajkovic S, Riboldi G, Ronchi D, Rizzo F, Bresolin N, et al. Mitochondrial fusion proteins and human diseases. Neurol Res Int. 2013; 2013: 11.

16. Nunnari J, Suomalainen A. Mitochondria: in sickness and in health. Cell. 2012; 148: 1145-1159.

17. Wu M, Neilson A, Swift AL, Moran R, Tamagnine J, Parslow D, et al. Multiparameter metabolic analysis reveals a close link between attenuated mitochondrial bioenergetic function and enhanced glycolysis dependency in human tumor cells. Am J Physiol Cell Physiol. 2007; 292: C125-C136.

18. Meeusen S, McCaffery JM, Nunnari J. Mitochondrial fusion intermediates revealed in vitro. Science. 2004; 305: 1747-1752.

19. Koshiba T, Detmer SA, Kaiser JT, Chen H, McCaffery JM, Chan DC. Structural basis of mitochondrial tethering by mitofusin complexes. Science. 2004; 305: 858-862.

20. Macho A, Decaudin D, Castedo M, Hirsch T, Susin SA, Zamzami N, et al. Chloromethyl-X-rosamine is an aldehyde-fixable potential-sensitive fluorochrome for the detection of early apoptosis. Cytometry. 1996; 25: 333-340.

21. Krysko DV, Roels F, Leybaert L, D’Herde K. Mitochondrial transmembrane potential changes support the concept of mitochondrial heterogeneity during apoptosis. J Histochem Cytochem. 2001; 49: 1277-1284.

22. Weissig V. From Serendipity to Mitochondria-Targeted Nanocarriers. Pharm Res. 2011; 28: 2657.

23. Vaidya B, Paliwal R, Rai S, Khatri K, Goyal AK, Mishra N, et al. Cellselective mitochondrial targeting: A new approach for cancer therapy. Cancer Ther. 2009; 7: 141-148.

24. Wang X, Shao N, Zhang Q, Cheng Y. Mitochondrial targeting dendrimer allows efficient and safe gene delivery. J Mater Chem B. 2014; 2: 2546-2553.

25. Battigelli A, Russier J, Venturelli E, Fabbro C, Petronilli V, Bernardi P, et al. Peptide-based carbon nanotubes for mitochondrial targeting. Nanoscale. 2013; 5: 9110-9117.

26. Chang HI, Yeh MK. Clinical development of liposome-based drugs: formulation, characterization, and therapeutic efficacy. Int J Nanomed. 2012; 7: 49-60.

27. Yamada Y, Akita H, Kogure K, Kamiya H, Harashima H. Mitochondrial drug delivery and mitochondrial disease therapy – An approach to liposome-based delivery targeted to mitochondria. Mitochondrion. 2007; 7: 63-71.

28. Green PS, Leeuwenburgh C. Mitochondrial dysfunction is an early indicator of doxorubicin-induced apoptosis. Biochim Biophys Acta (BBA). 2002; 1588: 94-101.

29. Papadopoulou LC, Theophilidis G, Thomopoulos GN, Tsiftsoglou AS. Structural and functional impairment of mitochondria in adriamycininduced cardiomyopathy in mice: suppression of cytochrome c oxidase II gene expression. Biochem Pharmacol. 1999; 57: 481-489.

30. Goormaghtigh E, Huart P, Praet M, Brasseur R, Ruysschaert JM. Structure of the adriamycin-cardiolipin complex. Biophys Chem. 1990; 35: 247-257.

31. Brand Martin D, Nicholls David G. Assessing mitochondrial dysfunction in cells. Biochem J. 2011; 435: 297-312.

32. Hill BG, Dranka BP, Zou L, Chatham JC, Darley-Usmar VM. Importance of the bioenergetic reserve capacity in response to cardiomyocyte stress induced by 4-hydroxynonenal. Biochem J. 2009; 424: 99-107.

33. Hill BG, Benavides GA, Lancaster JR, Ballinger S, Dell’Italia L, Zhang J, et al. Integration of cellular bioenergetics with mitochondrial quality control and autophagy. Biol Chem. 2012; 393: 1485-1512.

34. Souid AK, Penefsky HS, Sadowitz PD, Toms B. Enhanced cellular respiration in cells exposed to doxorubicin. Mol Pharm. 2006; 3: 307- 321.

35. Souid AK, Tacka KA, Galvan KA, Penefsky HS. Immediate effects of anticancer drugs on mitochondrial oxygen consumption. Biochem Pharmacol. 2003; 66: 977-987.

36. Liesa M, Borda-d’Água B, Medina-Gómez G, Lelliott CJ, Paz JC, Rojo M, et al. Mitochondrial fusion is increased by the nuclear coactivator PGC-1β. PLoS ONE. 2008; 3: e3613.

37. Martin OJ, Lai L, Soundarapandian MM, Leone TC, Zorzano A, Keller MP, et al. A role for PGC-1 coactivators in the control of mitochondrial dynamics during postnatal cardiac growth. Circ Res. 2014; 114: 626- 636.

38. Papanicolaou KN, Kikuchi R, Ngoh GA, Coughlan KA, Dominguez I, Stanley WC, et al. Mitofusins 1 and 2 are essential for postnatal metabolic remodeling in heart. Circ Res. 2012; 111: 1012-1026.

39. Wallace DC. A Mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet. 2005; 39: 359.

40. Perry SW, Norman JP, Barbieri J, Brown EB, Gelbard HA. Mitochondrial membrane potential probes and the proton gradient: a practical usage guide. BioTechniques. 2011; 50: 98-115.

41. Eisner V, Lenaers G, Hajnóczky G. Mitochondrial fusion is frequent in skeletal muscle and supports excitation–contraction coupling. J Cell Biol. 2014; 205: 179-195.

42. Sardão VA, Oliveira PJ, Holy J, Oliveira CR, Wallace KB. Morphological alterations induced by doxorubicin on H9c2 myoblasts: nuclear, mitochondrial, and cytoskeletal targets. Cell Biol Toxicol. 2009; 25: 227-243.

43. Dudnakova TV, Lakomkin VL, Tsyplenkova VG, Shekhonin BV, Shirinsky VP, Kapelko VI. Alterations in myocardial cytoskeletal and regulatory protein expression following a single Doxorubicin injection. J Cardiovascular pharmacol. 2003; 41: 788-794.

44. Kim JE, Han BS, Choi WS, Eom DS, Lee EH, Oh TH, et al. Temporospatial sequence of cellular events associated with etoposide-induced neuronal cell death: Role of antiapoptotic protein Bcl-XL. J Neurosci Res. 2001; 66: 1074-1082. 

45. Iwasaki T, Suzuki T. Ultrastructural alterations of the myocardium induced by doxorubicin. Virchows Archiv B. 1991; 60: 35-39.

46. Yan L, Vatner DE, Kim SJ, Ge H, Masurekar M, Massover WH, et al. Autophagy in chronically ischemic myocardium. PNAS USA. 2005; 102: 13807-13812.

47. Terman A, Brunk UT. Autophagy in cardiac myocyte homeostasis, aging, and pathology. Cardiovasc Res. 2005; 68: 355-365.

48. Evans JMM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005; 330: 1304-1305.

49. Jiralerspong S, Palla SL, Giordano SH, Meric-Bernstam F, Liedtke C, Barnett CM, et al. Metformin and Pathologic Complete Responses to Neoadjuvant Chemotherapy in Diabetic Patients With Breast Cancer. J Clin Oncol. 2009; 27: 3297-3302.

50. Alimova IN, Liu B, Fan Z, Edgerton SM, Dillon T, Lind SE, et al. Metformin inhibits breast cancer cell growth, colony formation and induces cell cycle arrest in vitro. Cell Cycle. 2009; 8: 909-915.

51. Liu B, Fan Z, Edgerton SM, Deng XS, Alimova IN, Lind SE, et al. Metformin induces unique biological and molecular responses in triple negative breast cancer cells. Cell Cycle. 2009; 8: 2031-2040.

52. Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K. Metformin Selectively Targets Cancer Stem Cells, and Acts Together with Chemotherapy to Block Tumor Growth and Prolong Remission. Cancer Res. 2009; 69: 7507-7511.

53. Hirsch HA, Iliopoulos D, Joshi A, Zhang Y, Jaeger SA, Bulyk M, et al. A Transcriptional Signature and Common Gene Networks Link Cancer with Lipid Metabolism and Diverse Human Diseases. Cancer Cell. 2010; 17: 348-361.

54. Anisimov VN, Egormin PA, Piskunova TS, Popovich IG, Tyndyk ML, Yurova MN, et al. Metformin extends life span of HER-2/neu transgenic mice and in combination with melatonin inhibits growth of transplantable tumors in vivo. Cell Cycle. 2010; 9: 188-197.55.

55. Dowling RJO, Zakikhani M, Fantus IG, Pollak M, Sonenberg N. Metformin Inhibits Mammalian Target of Rapamycin–Dependent Translation Initiation in Breast Cancer Cells. Cancer Res. 2007; 67: 10804-10812.

Ameer FS, Zhang X, Azhar G, Wei JY (2018) The Effect of Pegylated Liposomal Doxorubicin HCl on Cellular Mitochondrial Function. J Pharmacol Clin Toxicol 6(1):1099.

Received : 04 Jan 2018
Accepted : 31 Jan 2018
Published : 31 Jan 2018
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X