Metformin induced protection against oxidative stress is associated with AKT/mTOR restoration in PC12 cells Behzad Khallaghi, Fatemeh Safarian, Sanaz Nasoohi, Abolhassan Ahmadiani, Leila Dargahi PII: DOI: Reference:

S0024-3205(16)30073-X doi: 10.1016/j.lfs.2016.02.024 LFS 14709

To appear in:

Life Sciences

Received date: Revised date: Accepted date:

31 May 2015 26 January 2016 7 February 2016

Please cite this article as: Khallaghi Behzad, Safarian Fatemeh, Nasoohi Sanaz, Ahmadiani Abolhassan, Dargahi Leila, Metformin induced protection against oxidative stress is associated with AKT/mTOR restoration in PC12 cells, Life Sciences (2016), doi: 10.1016/j.lfs.2016.02.024

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Metformin induced protection against oxidative stress is associated with AKT/mTOR restoration in PC12 cells

T

Behzad Khallaghi1, Fatemeh Safarian1, Sanaz Nasoohi2, Abolhassan Ahmadiani1, 3, Leila

RI P

Dargahi4* 1

Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; 3Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia; 4NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.

NU

SC

2

ED

MA

Behzad Khallaghi and Fatemeh Safarian, as co-first author, contributed equally to this work.

AC

CE

PT

*Corresponding author Leila Dargahi NeuroBiology Research Center, Shahid Beheshti University of Medical sciences, Tehran, Iran. Email Address: [email protected] P. O. BOX: 19615-1178 Telephone: +98-21-22429768-9 Fax: +98-21-22432047

1

ACCEPTED MANUSCRIPT Abstract

Aims:

T

Reactive oxygen species have been recognized to impair cell function through suppressing Akt

RI P

the well-known pro-survival molecule. Pile of concrete evidence imply metformin as an Insuline sensitizer may enhance Akt/mTOR activity however the significance of Akt/mTOR recruitment

SC

has not yet been revealed in metformin induced neuroprotection against oxidative stress. Main methods:

NU

In the current study using H2O2 induced injury in PC12 cells; we first examined metformin impact on cell death by MTT assay and visual assessment. Metformin pretreated cells were then

MA

subjected to immunoblotting as well as real time PCR to find PI3K, Akt, mTOR and S6K concurrent transcriptional and post-transcriptional changes. The proportions of phosphorylated to non-phosphorylated constituents of PI3K/Akt/mTOR/S6K were determined to address their

ED

activation upon metformin treatment. Key findings:

PT

According to cells morphology and MTT data metformin led to significant protection against H2O2 induced injury in 0.1 and 0.5 mM concentrations. Metformin induced protection concurred

CE

with elevated PI3K/Akt/mTOR/S6K activity as well as enhanced GSH levels. These changes paralleled with a profound decline in the corresponding transcripts as determined by rea time

Significance:

AC

PCR.

Taken together our experimentation support the hypothesis that Akt/mTOR/S6K cascade may contribute to metformin alleviating effect. The present work while highlighting metformin antioxidant characteristics, concludes that Akt/mTOR signaling might be central to the drug’s alleviating effects. Keywords: Metformin; Oxidative stress; Akt; mTOR

2

ACCEPTED MANUSCRIPT

1. Introduction

T

Metformin has been introduced as a neuroprotective agent by in vitro examinations [1] as

RI P

well as in vivo experiments describing pro-survival and anti-inflammatory impacts of metformin in Huntington's disease and multiple sclerosis experimental models [2, 3]. The pro-survival

SC

effects of metformin has been ascribed to the decreased intracellular production of reactive oxygen species (ROS) [4] and to the improved balance between pro-oxidants and antioxidants

NU

[5].

Empirically metformin has been also shown to stimulate mammalian target of rapamycin

MA

(mTOR) especially in low doses [6] in which it exerts significant cytoprotection in non-cancerous cells [7, 8]. mTOR regulates a variety of cellular responses to extracellular signals particularly nutrients availability and stress [9]. This is predominantly mediated through its best-studied

ED

down-stream effectors S6K1 (p70 ribosomal protein S6 kinase 1) and 4EBP1 (eIF4E binding protein). The serine/threonine protein kinase Akt, a downstream effector of PI3K, is a critical

PT

mediator of mTOR activity [10]. The prominence of Akt/mTOR integrity have been documented by several empirical evidences to promote cell survival [11, 12] and proliferation which is mainly

CE

mediated through S6K activation. In line with this, there are pile of concrete evidence indicating that Akt/mTOR inhibitors like Rapamycine may be regarded as therapeutic agents to impede

AC

cancerous proliferation of different cell types [13, 14]. Notably, ROS such as superoxide (O2−) and hydrogen peroxide (H2O2) could also regulate tyrosine phosphorylation [15, 16] and may suppress mTOR activity through Akt inhibition [17]. Nevertheless, there are also recent evidences implying ROS-induced repression of mTOR may be downstream to tuberous sclerosis complex (TSC) signaling node on the peroxisome which functions as a cellular sensor for ROS to regulate mTOR and then autophagy [18]. Taken together, Akt/mTOR signaling seems to get remarkably affected by both oxidative stress and metformin. Therefore, theoretically it could be assumed that Akt/Mtor may mediate anti-oxidant effects of the drug. Accordingly, in the present work we aimed to determine the correlation between Akt/mTOR activity and metformin induced protection against oxidative stress. Since the H2O2 induced insult was associated with dampened Akt/mTOR, the main purpose was to find whether metformin protective doses enhance Akt/mTOR activity 3

ACCEPTED MANUSCRIPT simultaneously. To this end PC12 cells as commonly used neural like cells were exposed to H2O2 in IC50 concentrations and effect of metformin pretreatment was determined at transcriptional and post-translational levels of PI3K/Akt/mTOR/S6K signaling components. The

T

concurrent changes in antioxidants’ status; catalase (CAT) and superoxide dismutase (SOD)

RI P

activity and glutathione (GSH) levels, were also evaluated in stressed cells exposed to protective concentrations of metformin that has been identified as an anti-oxidant agent [19].

SC

2. Experimental Procedures 2.1. Cell culture and treatment

NU

PC12 cells (National cell bank, Pasteur institute of Iran) were maintained in high glucose (4500 mg/L) DMEM containing heat inactivated fetal bovine serum (FBS) (10% v/v), Penicillin

MA

(100 units/ml) and Streptomycin (100 µg/ml) (all from Invitrogen) at 37 °C in 95% air and 5% carbon dioxide. PC12 cells, at passage number 8-10, were seeded in 96-well plates (JET BioFil)

ED

at a density of 1×104 cells per well in serum free DMEM medium (Invitrogen) and were allowed to attach and expand for 24 h. H2O2 at the concentration of 150 µM was used to induce oxidative

PT

stress in PC12 cells. Pre-treatment with varying concentrations of freshly prepared Metformin (0.1, 0.5, 1, 3, 5, 7 mM) solution was performed for 60 min prior to stress induction by H2O2.

CE

Cells were then subjected to experimental analyses following H2O2 exposure for 24 h. A negative control cell group containing culture medium alone was also included as reference cells.

AC

2.3. Cell survival assay

Cell viability was grossly assessed by the stress related changes as determined in three separated fiedls in each photomicrographs. The mitochondrial integrity was also evaluated by 3(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) reduction to purple formazan [20]. Briefly, cells were incubated with MTT solution (final concentration 0.1-7 mg/mL) for 3 h at 37 °C. The medium was carefully removed by aspiration and formazan crystals were dissolved in 100 µl of dimethyl sulfoxide (DMSO). The extent of the reduction of MTT was quantified by measuring the absorbance at 570 nm and was reported as the percentage of cell viability compared to control.

4

ACCEPTED MANUSCRIPT 2.4. Superoxide dismutase activity assay Superoxide dismutase (SOD) activity was analyzed by the methods of Kakkar et al [21]. Briefly the assay mixture containing appropriate amounts of nicotinamide adenine dinucleotide,

T

phenazine methosulphate and nitroblue tetrazolium (750, 186 and 300 µM respectively) were

RI P

subjected to react with SOD present in cellular lysate. Following adequate incubation in 30 οC (90 sec), the mixture was stirred after n-butanol addition. The measured absorbance at 560 nm

SC

was considered as the color intensity of amino blue tetrazolium formazan (ABTF), the chromogen in butanol. Samples which resulted in 50% inhibition of total nitroblue tetrazolium

NU

reduction to ABTF per each mg of protein, was considered to possess one unit of enzymatic activity.

MA

2.5. Catalase activity assay

Catalase (CAT) activity was determined by the method of Aebi [22]. Simply cell lysate was

ED

mixed with phosphate buffer (50mM, pH 7.0) and subjected to react with the freshly prepared H2O2 (30mM). The rate of decomposition of H2O2 measured by the mixture’s absorbance at

2.6. GSH level analysis

PT

240 nm was considered as the CAT activity.

CE

Total GSH levels were measured in whole cell lysates according to the rate of colorimetric change of 5,5′-dithiobis-2-nitrobenzoic acid (Ellman’s reagent) as determined by the

µmol/mg.

AC

spectrophotometric method and absorbance at 405 nm [23]. GSH concentration was expressed as

2.7. Immunoblot Analysis The cells were washed with PBS scraped in ice-cold RIPA buffer (25 mM Tris–HCl, pH 7.6, 150 mM NaCl, 0.1% SDS, 2 mM EDTA, 1% sodium deoxycholate, 1% NP-40, 0.5 mM PMSF), and incubated on ice for 30 minutes. The cellular debris was removed by centrifugation (8000g for 15 min) at 4 °C and the cell lysate was carefully transferred to the microcentrifuge tubes. Proteins in samples standardized by Bradford assay [24], were separated by SDS electrophoresis and transferred to PVDF membranes. To estimate the amount of target proteins, membranes were then incubated with specific primary antibodies against Akt1/2/3/3 (H-136) and p-Akt1/2/3 (Ser 473), PI3 Kinase Class III, phospho-PI3 Kinase p85 (Tyr458)/p55 (Tyr199), mTOR, phospho5

ACCEPTED MANUSCRIPT mTOR (Ser248), p70S6 Kinase and phospho-p70 S6 Kinase (Thr389). Antibodies were obtained from Cell Signaling Technology and prepared in 1:1000 v/v dilutions except for Akt1 and p-Akt1 which were purchased from Santa Cruz and used in 1/600 v/v dilution. To visualize protein

T

bands, the blots were incubated with horseradish peroxidase-linked secondary antibody (#7074)

RI P

(1:10000 v/v; Cell Signaling Technology) which is directly detectable by chemiluminescence kit reagent (Amersham). The attained scans were then analyzed semi-quantitatively using Image J

SC

software in proportion to β-actin band intensity as internal control. 2.8. Quantitative Real-Time PCR

NU

Total RNA was extracted from cultured cells using NucleoSpin II kit (Macherey-Nagel GmbH & Co KG) and cDNA was synthesized using Revert Aid first strand cDNA synthesis kit

MA

(Thermo Fisher Scientific Inc.), all according to the manufacturers’ instructions. Quantitative RTPCR was performed on an ABI 7500 real time PCR system (PE Applied Biosystems, Foster City, CA, USA) using Maxima SYBR Green/ROX qPCR Master Mix according to the manufacturer’s

ED

recommendations with primer pairs listed in Table 1. β-actin was used as the reference housekeeping gene. The relative expression of mRNA transcripts, standard error means and p-

PT

values for differences in fold-expression were determined by the relative expression ratio method

CE

via the Relative Expression Software Tool-Multiple Condition Solver (REST-MCS) [25]. 2.9. Statistical Analysis

AC

All results represent the mean value from at least three separate experiments each with at least two replicates. All the data with the exception of those obtained at real-time PCR, were statistically analyzed using SPSS software. Normality of data was verified using the ShapiroWilk test. Analysis was performed with one-way ANOVA followed by Tukey post-hoc test and a P-value < 0.05 was considered as significant. Data are presented as mean ±SEM with the exception of relative fold changes in gene expression data which are presented as mean ±SD.

3. Results

3.1. Metformin does not exert dose-dependent protection against H2O2-induced injury in PC-12 cell

6

ACCEPTED MANUSCRIPT According to the presumptive diverse roles of metformin as a pro-survival [1] or proapoptotic [26] agent and for the differences in the experimental setup (eg. different cell lines) a dose response assay was primarily performed to determine the protective metformin

T

concentrations. Photomicrographs show that the presence of metformin together with H2O2

RI P

attenuates morphological alterations caused by H2O2 as defined by the ratio of stressed round shaped cells (Fig. 1a,b). Nevertheless, such impact was not detectable in metformin

SC

concentrations higher than 0.5 mM. Similarly, metformin (at 5 discreet concentrations ranging from 0.1 to 5 mM) did not show consistent dose-dependent responses in MTT assay. That is

NU

while incubation of the PC12 cells with metformin at 1, 3, and 5 mM concentrations did not result in restoring cell viability, metformin exposure at 0.1 and 0.5 mM concentrations rendered

MA

PC12 cells nearly resistant to H2O2 (Fig. 1c). Given metformin alone (at 5 discrete concentrations ranging from 0.1 to 5 mM) did not change MTT assay results (data not shown), such alterations are not seemingly due to metformin impact on intact cells. H2O2 concentration

ED

used to cause oxidative stress (150 µM) was selected according to the previously reported H2O2

PT

LD50% of 150 µM in a 24 h period of exposure as identified by our previous data [27].

3.2. The H2O2 induced decline in CAT and GSH was reversed by metformin

CE

To evaluate cells’ antioxidant strength, metformin effects was assessed on SOD, CAT and GSH as major detoxifying agents which degrade superoxide and H2O2 [28, 29]. According to the

AC

obtained data the antioxidant activity of SOD and CAT and the content of GSH decreased by H2O2 (150 µM) treatment in PC12 cells with higher confidence interval for CAT. Metformin treatment was not associated with any change in H2O2 induced escalation in SOD activity. Instead, as obtained by CAT and GSH assessment, PC12 cells exhibited improved CAT and GSH while pretreated with metformin low concentrations. Noteworthy, the improving effect of metformin on CAT and GSH vanished in doses higher than 1 mM and 0.5 mM respectively (data not shown). This may be corroborated with our MTT data in which metformin could not preserve cells’ viability while applied in high concentrations. Thus in order to evaluate the molecular alterations concurrent with metformin alleviating effects we proceeded to next experiments with metformin at 0.1, 0.5 and 1mM dosages (Fig 2).

7

ACCEPTED MANUSCRIPT 3.3. Protective effects of metformin concurred with enhanced PI3K/Akt/mTOR/S6K activity Impact of oxidative stress on PI3K/Akt/mTOR/S6K may be extensively context sensitive

T

[17]. Several concrete evidences suggest ROS may suppress PI3K/S6K while others imply in

RI P

pro-survival concentrations, ROS might activate PI3K/S6K ROS [30, 31]. According to our data, in our set of experiments H2O2 effectively inhibited PI3K/S6K activation as determined by the

SC

proportion of phosphorylated to un-phosphorylated PI3K/S6K constituents and the corresponding down streams mTOR and S6K. The relatively coordinated changes in PI3K, Akt, mTOR and

NU

S6K phosphorylation was suggestive of profound impression by H202. Notably, metformin augmented PI3K/S6K activity in the low dosages (0.1 and 0.5 mM) while in 1 mM it left the

MA

H2O2 induced PI3K/S6K repression almost entirely unaffected or even slightly intensified. As presented in figure 3, nearly all corresponding changes due to 1 mM metformin share the property of showing significant decline compared to control while it is not the case for the lower

ED

concentrations (0.1 and 0.5 mM). Taken together with the MTT data, this may imply the remarkable pro-survival effect of metformin was almost coincident with the restoring PI3K/S6K

PT

pathway.

proteins activation

CE

3.4. Changes in PI3K/Akt/mTOR/S6K mRNA transcripts inversely correlate with that of

AC

Quantitative real time PCR results of three independent experiments showed there is a significant shift towards enhanced transcription of PI3K/Akt/mTOR/S6K cascade proteins with increasing dosages of metformin (0.1 to 1 mM). As shown in figure 4, seemingly the upward change of transcripts in 1 mM concentration of metformin is the only case of over-expression that was strong enough to markedly surpass the corresponding one in control that is determined by 1 in y axis. The relevant refinements were also performed in the legend and also the pertinent result section.The maximal mRNA transcript was detected in 1 mM which surpassed that of control

cells. Interestingly as a common future, metformin in all included doses (0.1, 0.5 and 1 mM) seemingly reversed the proportion of Akt and mTOR transcripts to the up-stream effector PI3K. Nevertheless, here we could not suggest any correlation with protective effects of metformin since the mRNA levels did not consistently exceed that of control group in both protective concentrations of metformin (i.e.; 0.1 and 0.5 Mm) (Fig4).

8

ACCEPTED MANUSCRIPT 4. Discussion

Metformin antioxidant properties have been shown to reserve endothelial function [32] and

T

protect against cardiovascular [33] and central [34] complications in diabetic subjects, however;

RI P

no evidence has been reported considering PI3K/S6K implication. Results of the present work demonstrate that PI3K/S6K activity is restored during metformin induced protection. In this

SC

connection, our data suggest metformin may enhance cell survival in the face of oxidative stress at least partly through strengthening antioxidant systems particularly GSH and CAT.

NU

Basically, mitochondrion is the primary target of metformin within the cell in which metformin transiently inhibits complex I in electron transport chain. In the view of diabetes

MA

therapy, this results in a drop in energy charge [35] and consequently in AMP/ATP proportion which in turn activates AMP-activated protein kinase (AMPK) which negatively regulates mTOR/ S6K activation [36, 37]. On the other side, the same mechanism may also account for the

ED

hampered ROS production in reverse-electron flux at respiratory-chain complex 1 by metformin [38]. Besides reducing ROS levels, metformin administration may also end with strengthened

PT

antioxidant systems [39, 40] which might contribute to the documented nephro-protective effects of metformin in diabetes mellitus [41].

CE

According to our findings, low dose metformin was efficient enough to retrieve CAT and GSH despite lack of SOD restoration. This is somehow verified by previous works describing

AC

metformin may enhance CAT activity and GSH levels [42, 43, 44], while there is not enough evidence supporting SOD augmentation by metformin [45]. Given that oxidative stress regulates the activity of the cell survival factor Akt [35] we sought to correlate Akt/mTOR cascade to H2O2 toxicity. Having in mind H2O2 impact on Akt activity may strictly depend on cell type as well as exposure conditions [17], our results described a marked decrease in phosphorylation of Akt/mTOR inactive constituents in H2O2 treated PC12 cells. This is in agreement with previous studies indicating H2O2 induces a drop in Akt kinase activity in PC12 cells [46] which may hypothetically take part in the induced injury [47, 48]. As an effector downstream to Insulin receptor, PI3K may get activated upon Insulin binding. Metformin is empirically considered as an Insulin sensitizer in spite that it is devoid of metabolites or conjugates that could activate Insulin receptors [46]. This is noteworthy that Insulin sensitizing property of metformin has been supported by in vitro [50] and in vivo [51] 9

ACCEPTED MANUSCRIPT evidences implying metformin enhances insulin function by improving Insulin receptors-linked tyrosine phosphorylation. In line with this, low dose metformin has been shown to affect neural injury induced by glucose-oxygen deprivation through modulating phosphorylation of β-subunit

T

tyrosine in Insulin receptor [52].

RI P

Our data also described a transient PI3K/S6K signaling activation in the low dose (0.1 or 0.5 mM) while it was coincident with significant protection. This primary PI3K/S6K augmentation

Akt/mTOR activation by low dose metformin [52].

SC

may be the result of neutralizing H2O2 effects [46] or more probably, the direct consequent of

NU

Unexpectedly, our data shows the slightly higher dose of metformin (1 mM) did not keep on reversing PI3K/S6K activity against H2O2 toxicity. Actually, in some cases PI3K/S6K

MA

suppression even enhanced in this concentration. Such intensified PI3K/S6K suppression might be ascribed to switching between downstream effectors. That is based on empirical evidences, metformin may reasonably suppress PI3K/S6K activity by AMPK stimulation following

ED

interfering with the mitochondrial electron transport as described before. This way, activated AMPK switches cells from an anabolic to a catabolic state, thus shutting down the ATP-

PT

consuming proliferative pathways like mTOR/S6K signaling [53]. The observed S6K suppression by metformin increasing doses may indicate that such interference with Akt/S6K

CE

signaling may commence even in very low concentrations of the drug. Noteworthy, AMPK also has been shown to exert pro-survival effects in various cell types

AC

[54, 55], but here we can’t add anything about AMPK involvement as we did not examine AMPK activity. However, this presumptive AMPK stimulation by metformin might explain the opposite changes observed in PI3K/S6K by metformin at relatively higher concentration of 1 Mm. Intriguingly, in contrary to post translational effects of metformin, our results on mRNA assay identified that the transcriptional levels of PI3K/S6K signaling molecules were enhanced by any enhancement in metformin exposure. There are concrete evidences implying downstream components like mTOR may negatively control the activity of Akt/mTOR signaling [56, 57]. In this view, the initially inversed PI3K/S6K transcription at metformin low concentrations may be accountable for the transient augmentation of Akt/mTOR signaling by the low concentration of metformin. Nonetheless the reverse correlation between transcriptional and translational levels of

10

ACCEPTED MANUSCRIPT PI3K/S6K constituents might be suggestive of a negative feedback loop, we did not find any

T

evidence implying relevant gene transcripts are regulated by activated Akt or PI3K.

RI P

5. Conclusion

Collectively, the present work shows metformin induced protection in PC12 cells is associated with Akt/mTOR restoration however using kinase inhibitors would substantially

SC

contribute to verify Akt/mTOR implication. While the absence of pro-survival effect of metformin was coincident with dismissing GSH elevation, it needs to be unraveled whether these

NU

anti-oxidants properties is a direct consequent of PI3K/S6K activity. Lastly regarding probable differences between the relevant intracellular cascades, examining alternative pathways and in

neuroprotective effects of the drug.

ED

Acknowledgements

MA

particular considering virtual neural cultures, may shed may shed light on literally

This study was supported financially by Neuroscience Research Center, Shahid Beheshti

PT

University of Medical Sciences. The authors thank Dr. Khodagholi F for valuable guidance on

References [1]

[2]

[3]

[4]

AC

presentation.

CE

PC12 experiments and Dr. Talebi for effective discussion on the real time PCR results

El-Mir M-Y, Detaille D, Gloria R, Delgado-Esteban M, Guigas B, Attia S, Fontaine E, Almeida A, Leverve X (2008) Neuroprotective role of antidiabetic drug metformin against apoptotic cell death in primary cortical neurons. Journal of Molecular Neuroscience 34:77-87 Nath N, Khan M, Paintlia MK, Hoda MN, Giri S (2009) Metformin attenuated the autoimmune disease of the central nervous system in animal models of multiple sclerosis. The Journal of immunology 182:8005-8014 Ma TC, Buescher JL, Oatis B, Funk JA, Nash AJ, Carrier RL, Hoyt KR (2007) Metformin therapy in a transgenic mouse model of Huntington's disease. Neuroscience letters 411:98103 Ouslimani N, Peynet J, Bonnefont-Rousselot D, Thérond P, Legrand A, Beaudeux J-L (2005) Metformin decreases intracellular production of reactive oxygen species in aortic endothelial cells. Metabolism 54:829-834

11

ACCEPTED MANUSCRIPT

[10] [11]

[12]

[13] [14] [15]

[16]

[17]

[18]

T

RI P

SC

NU

MA

[9]

ED

[8]

PT

[7]

CE

[6]

Srividhya S, Ravichandran M, Anuradha C (2002) Metformin attenuates blood lipid peroxidation and potentiates antioxidant defense in high fructose-fed rats. Journal of biochemistry, molecular biology, and biophysics: JBMBB: the official journal of the Federation of Asian and Oceanian Biochemists and Molecular Biologists (FAOBMB) 6:379385 Bhamra GS, Hausenloy DJ, Davidson SM, Carr RD, Paiva M, Wynne AM, Mocanu MM, Yellon DM (2008) Metformin protects the ischemic heart by the Akt-mediated inhibition of mitochondrial permeability transition pore opening. Basic research in cardiology 103:274-284 Lesan V, Ghaffari SH, Salaramoli J, Heidari M, Rostami M, Alimoghaddam K, Ghavamzadeh A (2014) Evaluation of Antagonistic Effects of Metformin with Cisplatin in Gastric Cancer Cells. International Journal of Hematology-Oncology and Stem Cell Research 8:12-19 Janjetovic K, Vucicevic L, Misirkic M, Vilimanovich U, Tovilovic G, Zogovic N, Nikolic Z, Jovanovic S, Bumbasirevic V, Trajkovic V (2011) Metformin reduces cisplatin-mediated apoptotic death of cancer cells through AMPK-independent activation of Akt. European journal of pharmacology 651:41-50 Swiech L, Perycz M, Malik A, Jaworski J (2008) Role of mTOR in physiology and pathology of the nervous system. Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics 1784:116-132 Hay N, Sonenberg N (2004) Upstream and downstream of mTOR. Genes & development 18:1926-1945 Hu X, Pandolfi PP, Li Y, Koutcher JA, Rosenblum M, Holland EC (2005) mTOR promotes survival and astrocytic characteristics induced by Pten/AKT signaling in glioblastoma. Neoplasia 7:356-368 Meikle L, Pollizzi K, Egnor A, Kramvis I, Lane H, Sahin M, Kwiatkowski DJ (2008) Response of a neuronal model of tuberous sclerosis to mammalian target of rapamycin (mTOR) inhibitors: effects on mTORC1 and Akt signaling lead to improved survival and function. The Journal of Neuroscience 28:5422-5432 Morgensztern D, McLeod HL (2005) PI3K/Akt/mTOR pathway as a target for cancer therapy. Anti-cancer drugs 16:797-803 Mei H, Wang Y, Lin Z, Tong Q (2013) The mTOR Signaling Pathway in Pediatric Neuroblastoma. Pediatric Hematology-Oncology 30:605-615 RAO R, Basuroy S, Rao V, Karnaky Jr K, Gupta A (2002) Tyrosine phosphorylation and dissociation of occludin–ZO-1 and E-cadherin–β-catenin complexes from the cytoskeleton by oxidative stress. Biochem J 368:471-481 Carballo M, Conde M, El Bekay R, Martı́n-Nieto J, Camacho MaJ, Monteseirı́n J, Conde J, Bedoya FJ, Sobrino F (1999) Oxidative stress triggers STAT3 tyrosine phosphorylation and nuclear translocation in human lymphocytes. Journal of Biological Chemistry 274:1758017586 Chen W-C, Hsieh S-R, Chiu C-H, Hsu B-D, Liou Y-M (2014) Molecular identification for epigallocatechin-3-gallate-mediated antioxidant intervention on the H2O2-induced oxidative stress in H9c2 rat cardiomyoblasts. Journal of biomedical science 21:56 Zhang J, Kim J, Alexander A, Cai S, Tripathi DN, Dere R, Tee AR, Tait-Mulder J, Di Nardo A, Han JM (2013) A tuberous sclerosis complex signalling node at the peroxisome regulates mTORC1 and autophagy in response to ROS. Nature cell biology 15:1186-1196

AC

[5]

12

ACCEPTED MANUSCRIPT

[25]

[26]

[27]

[28] [29]

[30] [31]

[32]

[33]

[34]

T

RI P

SC

NU

[24]

MA

[23]

ED

[22]

PT

[21]

CE

[20]

Hou X, Song J, Li X-N, Zhang L, Wang X, Chen L, Shen YH (2010) Metformin reduces intracellular reactive oxygen species levels by upregulating expression of the antioxidant thioredoxin via the AMPK-FOXO3 pathway. Biochemical and biophysical research communications 396:199-205 Mosmann T (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods:55-63 Kakkar P DB, Viswanathan PN (1984) A modified spectrophotometric assay of superoxide dismutase. Indian J Biochem Biophys:130-132 Aebi H (1974) Catalase. In: Methods of enzymatic analysis. Academic Press, New York, pp 674-684 Sedlak J, Lindsay R H (1968). Estimation of total, protein-bound, and nonprotein sulfhydryl groups in tissue with Ellman's reagent. Anal Biochem 25:192-205 Bradford M (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem:248-254 Pfaffl MW HG, Dempfle L (2002) Relative expression software tool (REST) for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res Ucbek A, Özünal ZG, Uzun Ö, GepdĐremen A (2014) Effect of metformin on the human T98G glioblastoma multiforme cell line. Experimental and Therapeutic Medicine 7:12851290 Safarian F, Khallaghi B, Ahmadiani A, Dargahi L (2014) Activation of S1P1 Receptor Regulates PI3K/Akt/FoxO3a Pathway in Response to Oxidative Stress in PC12 Cells. Journal of Molecular Neuroscience:1-11 Sampayo JN GM, Lithgow GJ. (2003) Oxidative stress and aging--the use of superoxide dismutase/catalase mimetics to extend lifespan. Biochem Soc Trans:1305-1307 Zhang L, Wei S, Tang J-M, Guo L-Y, Zheng F, Yang J-Y, Kong X, Huang Y-Z, Chen S-Y, Wang J-N (2013) PEP-1-CAT protects hypoxia/reoxygenation-induced cardiomyocyte apoptosis through multiple sigaling pathways. Journal of translational medicine 11:113 Sadidi M, Lentz SI, Feldman EL (2009) Hydrogen peroxide-induced Akt phosphorylation regulates Bax activation. Biochimie 91:577-585 Salsman S, Felts N, Pye QN, Floyd RA, Hensley K (2001) Induction of Akt Phosphorylation in Rat Primary Astrocytes by H< sub> 2 O< sub> 2 Occurs Upstream of Phosphatidylinositol 3-Kinase: No Evidence for Oxidative Inhibition of PTEN. Archives of biochemistry and biophysics 386:275-280 Alexánderson-Rosas E, de Jesús MA, Ochoa-López J, Calleja-Torres R, Sierra-Fernández C, Iñarra-Talboy F, Meave-González A, Alexánderson-Rosas G, González-Canudas J (2008) [Effects of the combined treatment with Metformin/Glimepiride on endothelial function of patients with type 2 diabetes mellitus. A positron emission tomography (PET) evaluation study]. Archivos de cardiologia de Mexico 79:249-256 Kapinya K, Nijjar P, Stanek M, Amanullah A (2008) Insulin‐sensitizing antihyperglycaemic medications are associated with better outcome in patients with diabetes undergoing cardiac stress testing. Internal medicine journal 38:259-264 Bhutada P, Mundhada Y, Bansod K, Tawari S, Patil S, Dixit P, Umathe S, Mundhada D (2011) Protection of cholinergic and antioxidant system contributes to the effect of berberine ameliorating memory dysfunction in rat model of streptozotocin-induced diabetes. Behavioural brain research 220:30-41

AC

[19]

13

ACCEPTED MANUSCRIPT

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48]

[49]

T

RI P

SC

NU

[40]

MA

[39]

ED

[38]

PT

[37]

CE

[36]

Owen M, DORAN E, Halestrap A (2000) Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J 348:607-614 Mihaylova MM, Shaw RJ (2011) The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nature cell biology 13:1016-1023 Pernicva I, Korbonits M (2014) Metformin [mdash] mode of action and clinical implications for diabetes and cancer. Nature Reviews Endocrinology 10:143-156 Batandier C, Guigas B, Detaille D, El-Mir M, Fontaine E, Rigoulet M, Leverve XM (2006) The ROS production induced by a reverse-electron flux at respiratory-chain complex 1 is hampered by metformin. Journal of bioenergetics and biomembranes 38:33-42 Pavlović D, Kocić R, Kocić G, Jevtović T, Radenković S, Mikić D, Stojanović M, Djordjević P (2000) Effect of four‐week metformin treatment on plasma and erythrocyte antioxidative defense enzymes in newly diagnosed obese patients with type 2 diabetes. Diabetes, Obesity and Metabolism 2:251-256 Ewis SA, Abdel‐Rahman MS (1995) Effect of metformin on glutathione and magnesium in normal and streptozotocin‐induced diabetic rats. Journal of applied toxicology 15:387-390 Alhaider AA, Korashy HM, Sayed-Ahmed MM, Mobark M, Kfoury H, Mansour MA (2011) Metformin attenuates streptozotocin-induced diabetic nephropathy in rats through modulation of oxidative stress genes expression. Chemico-biological interactions 192:233-242 Gallo A, Ceolotto G, Pinton P, Iori E, Murphy E, Rutter GA, Rizzuto R, Semplicini A, Avogaro A (2005) Metformin prevents glucose-induced protein kinase C-β2 activation in human umbilical vein endothelial cells through an antioxidant mechanism. Diabetes 54:11231131 Dai J, Liu M, Ai Q, Lin L, Wu K, Deng X, Jing Y, Jia M, Wan J, Zhang L (2014) Involvement of catalase in the protective benefits of metformin in mice with oxidative liver injury. Chemico-biological interactions 216:34-42 Uranga RM, Katz S, Salvador GA (2013) Enhanced phosphatidylinositol 3-kinase (PI3K)/Akt signaling has pleiotropic targets in hippocampal neurons exposed to iron-induced oxidative stress. Systems 13:14. Akinola O, Gabriel M, Suleiman A-A, Olorunsogbon F (2012) Treatment of alloxan-induced diabetic rats with metformin or glitazones is associated with amelioration of hyperglycaemia and neuroprotection. The Open Diabetes Journal 5:8 Rao YK, Shih H-N, Lee Y-C, Cheng W-T, Hung H-C, Wang H-C, Chen CJ, Tzeng Y-M, Lee M-J (2014) Purification of kavalactones from< i> Alpinia zerumbet and their protective actions against hydrogen peroxide-induced cytotoxicity in PC12 cells. Journal of bioscience and bioengineering Brunet A, Datta SR, Greenberg ME (2001) Transcription-dependent and-independent control of neuronal survival by the PI3K–Akt signaling pathway. Current opinion in neurobiology 11:297-305 L Hixon M, Paccagnella L, Millham R, Perez-Olle R, Gualberto A (2010) Development of inhibitors of the IGF-IR/PI3K/Akt/mTOR pathway. Reviews on recent clinical trials 5:189208 Davidson MB, Peters AL (1997) An overview of metformin in the treatment of type 2 diabetes mellitus. The American journal of medicine 102:99-110

AC

[35]

14

ACCEPTED MANUSCRIPT

[54]

[55]

[56]

T

AC

CE

PT

ED

[57]

RI P

[53]

SC

[52]

NU

[51]

PRYOR P, Liu S, CLARK A, Yang J, HOLMAN G, Tosh D (2000) Chronic insulin effects on insulin signalling and GLUT4 endocytosis are reversed by metformin. Biochem J 348:8391 Stith BJ, Goalstone ML, Espinoza R, Mossel C, Roberts D, Wiernsperger N (1996) The antidiabetic drug metformin elevates receptor tyrosine kinase activity and inositol 1, 4, 5trisphosphate mass in Xenopus oocytes. Endocrinology 137:2990-2999 Mielke J, Taghibiglou C, Wang Y (2006) Endogenous insulin signaling protects cultured neurons from oxygen–glucose deprivation-induced cell death. Neuroscience 143:165-173 Hardie DG (2011) Sensing of energy and nutrients by AMP-activated protein kinase. The American journal of clinical nutrition 93:891S-896S Pantovic A, Krstic A, Janjetovic K, Kocic J, Harhaji-Trajkovic L, Bugarski D, Trajkovic V (2013) Coordinated time-dependent modulation of AMPK/Akt/mTOR signaling and autophagy controls osteogenic differentiation of human mesenchymal stem cells. Bone 52:524-531 Leclerc GM, Leclerc GJ, Fu G, Barredo JC (2010) AMPK-induced activation of Akt by AICAR is mediated by IGF-1R dependent and independent mechanisms in acute lymphoblastic leukemia. J Mol Signal 5:15 Manning BD (2004) Balancing Akt with S6K implications for both metabolic diseases and tumorigenesis. The Journal of cell biology 167:399-403 Tamburini J, Chapuis N, Bardet V, Park S, Sujobert P, Willems L, Ifrah N, Dreyfus F, Mayeux P, Lacombe C (2008) Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways. Blood 111:379-382

MA

[50]

Figure legends Fig. 1. Effect of metformin on H2O2 induced cell injury as determined by MTT assay and morphological alterations. A 24 h period of H2o2 (150 µM) exposure caused a significant decline in PC12 cells viability. The representative photomicrographs of PC12 cells (a) demonstrate the corresponding substantial decline in round forms (b) due to protection induced by metformin at 0.1 and 0.5 mM concentrations. In the same concentrations MTT assay data (b) agrees the antioxidant activity of metformin. Values are expressed as mean ± SEM from three independent experiments, each of which including four replicates. *p < 0.05, **p < 0.01, ***p < 0.001 versus control; #p

mTOR restoration in PC12 cells.

Reactive oxygen species have been recognized to impair cell function through suppressing Akt the well-known pro-survival molecule. Pile of concrete ev...
924KB Sizes 0 Downloads 14 Views