580 Research report

FK506 attenuates intracerebroventricular streptozotocininduced neurotoxicity in rats Rimpi B. Arora, Kushal Kumar and Rahul R. Deshmukh Upregulation in calcineurin (CaN) signaling has been implicated in various neurodegenerative disorders. In the present study, we have investigated the effect of FK506 – a CaN inhibitor – on streptozotocin (STZ)induced experimental dementia of the Alzheimer’s type in rats. STZ was administered intracerebroventricularly to induce a cognitive deficit and oxidative stress. Nonimmunosuppressive doses (0.5 and 1 mg/kg postoperatively) of FK506 (tacrolimus) were administered for 21 day in STZ-treated rats. Cognitive functions were assessed using the Morris water maze and passive avoidance tasks. Malondialdehyde and nitrite glutathione levels, as well as acetylcholinesterase activity, were determined to evaluate oxidative stress and cholinergic functions. Lactate dehydrogenase levels were estimated and histological analysis of the dentate gyrus and the CA1 region of the hippocampus was carried out to identify degenerative changes. STZ produced significant deterioration of cognitive functions, oxidative stress, and degenerative changes in the cortical and hippocampal

brain regions. FK506 dose-dependently attenuated STZ-induced cognitive deficits, oxidative stress, and degenerative changes in the cortex and hippocampus. These results suggest a potential role of CaN signaling in degenerative processes, and that inhibition of CaN may be useful in the treatment of neurodegenerative disorders such as Alzheimer’s disease. Behavioural c 2013 Wolters Kluwer Health | Pharmacology 24:580–589  Lippincott Williams & Wilkins.

Introduction

2008), Huntington’s disease (Xifiro et al., 2008; Dudek et al., 2010), and AD (Dineley et al., 2007).

Alzheimer’s disease (AD) is an age-related neurodegenerative disorder (Reed et al., 2009). Accumulating data indicate that disturbances in several aspects of cellular metabolism appear pathologically important in AD. Among these, increased brain insulin resistance (Watson and Craft, 2006; Hoyer and Lannert, 2007; SalkovicPetrisic, 2008) and decreased glucose utilization (Balbo et al., 2002; Steen et al., 2005; De la Torre, 2008) and energy metabolism are observed in the early stages of the disease (De la Monte and Wands, 2005; Hoyer and Lannert, 2007; Salkovic-Petrisic and Hoyer, 2007). Decreased energy metabolism has also been implicated in the activation of numerous signaling pathways and Ca2 + toxicity (Muller et al., 1998). One intracellular signaling element that has recently attracted attention as a potential modulator of both learning and memory function, as well as cell degeneration, is the Ca2 + /calmodulin-dependent protein serine– threonine phosphatase 2B, also known as calcineurin (CaN), which is the most abundant phosphatase in the brain (Mansuy, 2003; Liu et al., 2007) and is activated following Ca2 + influx (Liu, 2009). CaN has been implicated in the pathogenesis of various diseases such as cardiac hypertrophy (Nakamura et al., 2008), congenital heart disease (Groetzner et al., 2005), epilepsy (VazquezLopez et al., 2006), Parkinson’s disease (Wright et al., c 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins 0955-8810 

Behavioural Pharmacology 2013, 24:580–589 Keywords: cognitive dysfunction, FK506, oxidative stress, rat, sporadic dementia, streptozotocin, tacrolimus Department of Pharmacology, Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India Correspondence to Rahul R. Deshmukh, MPharm, PhD, Department of Pharmacology, Neuropharmacology Division, ISF College of Pharmacy, Moga 142001, Punjab, India E-mail: [email protected] Received 22 January 2013 Accepted as revised 4 July 2013

CaN has been reported to negatively regulate learning and memory (Eto et al., 2008). Inhibition of CaN signaling has been reported to improve associative learning and memory in Tg2876 APP transgenic mice (Dineley et al., 2007) and spatial memory in 3-nitropropionic acidinduced neurotoxic insult (Kumar and Kumar, 2009). CaN is specifically inhibited by certain drugs such as cyclosporine-A and FK506 in the presence of cyclophilin and FK506-binding protein (FKBP-12 and FKBP-52), respectively, resulting in immunosuppressant activity (Van Rossum et al., 2009). In addition, nonimmunosuppressive doses of FK506 have also been demonstrated to provide neuroprotection without affecting inflammatory mechanisms in various models such as experimental stroke (Butcher et al., 1997), spinal-cord injury (Voda et al., 2005), Parkinson’s disease (Wright et al., 2008) and Huntington’s disease (Kumar and Kumar, 2009). Despite these findings, the role of CaN and beneficial effects of FK506 in experimental sporadic dementia of Alzheimer’s type remain elusive. Intracerebroventricular administration of streptozotocin (ICV-STZ) in rats is commonly used to study experimental sporadic dementia of the Alzheimer’s type. ICV-STZ has been reported to produce behavioral, neurochemical, DOI: 10.1097/FBP.0b013e32836546db

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

FK506 against STZ-induced neurotoxicity Arora et al. 581

biochemical, and histopathological changes similar to those in an aging brain and is considered to be a suitable animal model of sporadic dementia of the Alzheimer’s type (Prickaerts et al., 1995; Sharma and Gupta, 2001, 2002; Salkovic-Petrisic, 2008; Ishrat et al., 2009). ICV-STZ administration has also been reported to increase the level of cytoplasmic calcium ions (Muller et al., 1998), and the pharmacological blockade of L-type calcium channels (lercanidipine) has been reported to attenuate behavioral and biochemical alterations in ICV-STZ-injected rats (Sonkusare et al., 2005). Dysregulation in Ca2 + homeostasis has been observed in neurodegenerative disorders including AD. Moreover, dysregulation of calcium signaling has been reported to upregulate CaN signaling mechanisms (Foster et al., 2001), which may perhaps contribute to the degenerative changes and subsequent cognitive decline associated with sporadic AD. Thus, in the present study, we have investigated the potential effect of FK506, a CaN inhibitor, on STZ-induced neurotoxicity.

Methods Subjects

The experiments were carried out on male Wistar rats (220–250 g) procured from the Indian Institute of Integrative Medicine, Jammu, India. The animals were kept in polyacrylic cages and maintained under standard housing conditions (room temperature 22±21C and relative humidity of 60–65%), with lights on at 07.00 h and off at 19.00 h, at the ‘Central Animal House’ of ISF College of Pharmacy, Moga, Punjab (India). Food, in the form of dry pellets, and water were freely available. All behavioral experiments were carried out between 10.00 and 16.00 h. The protocol was reviewed and approved by the ‘Institutional Animal Ethics Committee’, and the animal experiments were carried out in accordance with the Indian National Science Academy Guidelines for use and care of animals. Intracerebroventricular infusion of streptozotocin

Rats were anesthetized with ketamine (100 mg/kg, intraperitoneally) and xylazine (5 mg/kg, intraperitoneally). The head was placed in position in the stereotaxic apparatus and a midline saggital incision was made in the scalp. Two holes were drilled through the skull for placement of infusion cannulae into the lateral cerebral ventricles using the following coordinates: 0.8 mm posterior to bregma; 1.5 mm lateral to the saggital suture; 3.6 mm ventral from the surface of the brain (Paxinos and Watson, 1986). After cannula placement, animals were injected with gentamicin (5 mg/kg) and were placed in individual cages. Animals were observed for 1 week, and special care was taken by administering sweetened milk daily during this resting phase for recovery (Fig. 1). After the resting period, STZ was dissolved in citrate buffer (pH 4.4) and slowly infused (1 ml/min), using a Hamilton microsyringe pump (Quintessential stereotaxic injector; Stoelting Co., Wood Dale, Illinois, USA), into each

cerebral ventricle (bilateral ICV) in a volume of 10 ml on days 1 and 3 (Deshmukh et al., 2009). Experimental groups

Animals were divided into five groups, each comprising 10 animals. The treatment schedule and the interval for estimation of various parameters are presented in Fig. 1. Group1, which served as a double-vehicle control, received citrate buffer (pH 4.4) ICV at a volume of 10 ml in each ventricle on days 1 and 3 and 0.5% carboxymethyl cellulose (0.5 ml intraperitoneally, as a vehicle for FK506) for 21 days. The animals in group 2 were treated with 1 mg/kg FK506 postoperatively. Rats in group 3 were infused with ICV-STZ at a dose of 3 mg/kg. Groups 4 and 5 received FK506 at doses of 0.5 and 1 mg/kg, respectively, postoperatively, 1 h after STZ administration; this was started on the first post operative day and continued once daily for a period of 21 days. The nonimmunosuppressive doses of FK506 were selected on the basis of earlier reports in which significant antioxidant and neuroprotective properties were demonstrated at doses ranging from 0.2 to 3 mg/kg (Sakai et al., 1991; Singh et al., 2003a, Gold et al., 2004; Wright et al., 2008). Behavioral assessment Passive avoidance task

On days 14 and 15 after ICV-STZ infusion, the rats were tested for memory retention using a passive avoidance apparatus. The apparatus (Type-7552; Ugo Basile, Comerio, Italy) consisted of a chamber illuminated with a 40-W bulb and a dark chamber, separated by a guillotine door. The floor of the dark chamber consisted of a metal grid with a shock scrambler. During the acquisition trial, the rat was placed in the illuminated chamber. After an initial habituation period of 60 s, the guillotine door was opened and the time taken by the rat to enter the dark chamber was noted. The latency to step into the dark compartment was recorded as the initial trial latency (ITL) or preshock latency. As soon as the rat entered the dark chamber, it was given a mild foot shock of 0.5 mA for 2 s through the grid floor. The rat was allowed to remain in the dark compartment for 5 s and then taken out. After a 24-h interval, a retention trial was performed and retention trial latency (RTL) or postshock latency to step into the dark compartment was noted. The latency was recorded to a maximum of 600 s (Deshmukh et al., 2009). Short latencies indicated poorer retention. Morris water maze

Spatial learning and memory were tested in a Morris water maze (MWM; Morris, 1984), which consisted of a circular water tank (180 cm diameter, 60 cm height) filled with water (25±11C) to a depth of 40 cm. Milk powder was used to render the water opaque. Four equally spaced locations around the edge of the pool (North, South, East, and West) were used as start points, which divided

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

582

Behavioural Pharmacology 2013, Vol 24 No 7

Fig. 1

CI

Rest

−1

−7

PAL

ICV-STZ Day1

Day 2

Day 3

Day 14

Day15

MWM

Probe test

Day 17–20

Day 21

Tacrolimus (0.5 and 1 mg/kg, per oral) LA

SAC

Experimental procedure and treatment schedule. CI, cannula implantation; ICV, intracerebroventricular; LA, locomotor activity; MWM, Morris water maze; PAL, passive avoidance latency; SAC, sacrificed; STZ, streptozotocin.

the pool into four quadrants. An escape platform (10 cm in diameter) was placed 2 cm below the water surface in the middle of the north-east quadrant of the pool and kept in the same position throughout the experiment. Before training started, the rats were allowed to swim freely in the pool for 120 s without a platform. Animals underwent a training session consisting of four trials per session (once from each starting point) for 4 days (days 17, 18, 19, and 20), each trial having a ceiling time of 120 s and an intertrial interval of B30 s. After climbing onto the hidden platform, the animals were allowed to remain there for 30 s before commencement of the next trial. If the rat failed to locate the hidden platform within the maximum time of 120 s, it was gently placed onto the platform and allowed to remain there for the same interval of time. Twenty-four hours after the acquisition phase, a probe test (day 21) was conducted by removing the platform. Rats were allowed to swim freely in the pool for 60 s and the percentage of time spent in the target quadrant, which had previously contained the hidden platform, was recorded. The time spent in the target quadrant indicated the degree of memory consolidation that had taken place after learning (Deshmukh et al., 2009). The time taken to locate the hidden platform and the time spent in the target quadrant during the probe trial was measured by an experimenter who was blind to treatment, using a stopwatch. Spontaneous locomotor activity

Each animal was tested for spontaneous locomotor activity on day 21 after the first ICV-STZ infusion. Each animal was observed over a period of 10 min in a square closed arena equipped with infrared light sensitive photocells, using a digital photoactometer (INCO, Ambala, Haryana, India) (Deshmukh et al., 2009). Biochemical parameters Brain homogenate preparation

All biochemical parameters were measured in brain homogenates. On day 22 after the first STZ infusion, animals were killed by decapitation and brains were removed and rinsed with ice-cold isotonic saline. Brain

tissue samples were then homogenized with ice-cold 0.1 mol/l phosphate buffer (pH 7.4) in a volume 10 times the weight of the tissue. The whole brain homogenate was centrifuged at 10 000g for 15 min and aliquots of the supernatant were separated and used for biochemical estimation. Protein estimation

Protein levels in all brain samples were estimated using the method developed by Lowry et al. (1951), with BSA (1 mg/ml) as a standard. Acetylcholinesterase assay

The quantitative measurement of acetylcholinesterase (AChE) activity in the brain was performed according to the method described by Ellman et al. (1961). The assay mixture contained 0.05 ml of supernatant, 3 ml of 0.01 mol/l sodium phosphate buffer (pH 8), 0.10 ml of acetylthiocholine iodide, and 0.10 ml of 5,50 -dithiobis(2nitrobenzoic acid) (Ellman reagent). The change in absorbance was measured immediately at 412 nm spectrophotometrically. AChE activity in the supernatant was expressed as nmol/mg protein. Estimation of malondialdehyde

The quantitative measurement of malondialdehyde (MDA), the end product of lipid peroxidation, was performed according to the method developed by Wills (1966). The amount of MDA was measured after its reaction with thiobarbituric acid at 532 nm, using a spectrophotometer (UV-1700; Shimadzu, Columbia, Maryland, USA). The concentration of MDA was determined from a standard curve and expressed as nmol/mg protein. Estimation of reduced glutathione

Reduced glutathione (GSH) levels in the brain were estimated according to the method described by Ellman (1959). One milliliter of supernatant was precipitated with 1 ml of 4% sulfosalicylic acid and cold digested at 41C for 1 h. The samples were centrifuged at 1200g for 15 min. To 1 ml of the supernatant, 2.7 ml of phosphate buffer (0.1 mol/l, pH 8) and 0.2 ml of DTNB were added. The yellow color that developed was measured immediately at 412 nm using a spectrophotometer.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

FK506 against STZ-induced neurotoxicity Arora et al. 583

The concentration of GSH in the supernatant was determined from a standard curve and expressed as mmol/mg protein.

prepared afresh by suspending in 0.5% sodium carboxymethyl cellulose. All other chemicals used in the study were of analytical grade. Solutions of the drug and chemicals were freshly prepared before use.

Estimation of nitrite

The accumulation of nitrite in the supernatant, an indicator of the production of nitric oxide (NO), was determined by a colorimetric assay using the Greiss reagent [0.1% N-(1-naphthyl) ethylenediamine dihydrochloride, 1% sulfanilamide, and 2.5% phosphoric acid], as described by Green et al. (1982). Equal volumes of the supernatant and Greiss reagent were mixed, the mixture was incubated for 10 min at room temperature in the dark, and the absorbance was determined at 540 nm spectrophotometrically. The concentration of nitrite in the supernatant was determined from the sodium nitrite standard curve and expressed as mmol/mg protein.

Statistical analysis

Assessment of histological changes

On day 14 after the first ICV-STZ injection, the mean initial latency in the acquisition trial remained unchanged among all the groups [F(4,49) = 2.21, NS]. However, the mean retention latency on day 15 was significantly decreased (P < 0.001) in the ICV-STZ control group compared with vehicle-treated animals (Fig. 2). The ICV-STZ-induced decrease in the mean retention latency was significantly increased on FK506 treatment [F(4,49) = 174.3, P < 0.001], indicating protection against the effect of STZ treatment. The maximum improvement in retention latency by FK506 treatment on day 15 was observed at a dose of 1 mg/kg (P < 0.001). Moreover, control rats treated with FK506 (1 mg/kg) alone did not show any significant difference in retention latency as compared with vehicle-treated rats (P > 0.05).

The brains were rapidly removed and fixed by immersion in 10% formalin. Subsequently they were embedded in paraffin wax, cut into 5-mm-thick sections, and stained with cresyl violet acetate (Nissl stain; Shoham et al., 2003). Hippocampal brain sections were examined under bright field illumination (AHBT-51, Olympus VanoxAHBT; Olympus America, Melville, New York, USA). Drugs

STZ, acetylthiocholine iodide, and DTNB were purchased from Sigma-Aldrich (St Louis, Missouri, USA). FK506 was provided as an ex-gratia sample by M/S Panacea Biotec Ltd. (Mohali, Punjab, India). STZ was diluted in citrate buffer (pH 4.4) and FK506 was always

The results are expressed as mean±SD. The behavioral (for individual time points) and biochemical values were analyzed by one-way analysis of variance, followed by Tukey’s post-hoc test for multiple comparisons, using GraphPad Prism statistical software (version 5.0; GraphPad Prism software Inc., La Jolla, California, USA). A P-value of less than 0.05 was set to be statistically significant.

Results Effect of FK506 on memory performance in the passive avoidance task in ICV-STZ-treated rats

Fig. 2

Transfer latency (s)

600

Day-14 aquisition trial

Day-15 retention trial

∗#

400 ∗

200 @

g)

g)

m (1 FK

(0

.5

m

ST Z FK

e Pe

rs

cl e FK

m

hi Ve

g)

g) (1 FK

(0

.5

m

ST Z FK

e Pe rs

FK

Ve hi

cl

e

0

Effect of FK506 on performance of ICV-STZ-treated rats in the passive avoidance task. Values are expressed as mean±SD; @P < 0.05 versus vehicle; *P < 0.05 versus STZ; #P < 0.05 versus FK506 (0.5 mg/kg). FK, FK506; ICV, intracerebroventricular; STZ, streptozotocin.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

584 Behavioural Pharmacology 2013, Vol 24 No 7

Effect of FK506 on memory performance in the MWM task in ICV-STZ-treated rats

Effect of FK506 on brain GSH levels in ICV-STZ-treated rats

On day 17 of the acquisition trial, experimental animals from all the groups took a similar amount of time to locate the hidden platform in the MWM task [F(4,49) = 1.109, P > 0.05], but on days 18, 19, and 20 of the acquisition phase, STZ-injected rats showed a significant impairment in learning relative to the vehicle control group [F(4,49) = 59.07, 34.12, and 71.83, respectively, P < 0.001] (Fig. 3a). Moreover, during the probe trial [F(4,49) = 39.64, P < 0.01], STZ-treated rats also failed to remember the precise location of the platform, spending significantly less time in the target quadrant as compared with vehicletreated animals (P < 0.001; Fig. 3b). Chronic administration of FK506 in STZ-treated rats significantly and dosedependently attenuated STZ-induced acquisition and retention deficits in the MWM task. In the probe trial, FK506-treated rats spent more time in the target quadrant, as compared with STZ-treated control rats, indicating improved consolidation of memory (P < 0.05).

STZ administration in rats produced a significant depletion in GSH levels in the brain [F(4,49) = 83.90, P < 0.001] (Table 1) as compared with vehicle treatment (P < 0.001). Chronic administration of FK506 significantly restored GSH levels in STZ-treated rats (P < 0.01) without affecting GSH levels in control rats.

Effect of FK506 on spontaneous locomotor activity in ICV-STZ-treated rats

Effect of FK506 on the neuronal morphology of the dentate gyrus and the CA1 region of the hippocampus in ICV-STZ-treated rats

Spontaneous locomotor activity on day 21 did not differ significantly among groups [F(4,49) = 0.41, NS] (Fig. 4). Effect of FK506 on brain AChE activity in ICV-STZtreated rats

Ventricular administration of STZ produced a significant increase in brain AChE activity relative to vehicle controls [F(4,34) = 63.28, P < 0.001] (Fig. 5). Chronic administration of FK506 (0.5 and 1 mg/kg) dose-dependently attenuated the STZ-induced increase in AChE activity (P < 0.01). However, FK506 did not affect AChE activity in control rats (P > 0.05). Effect of FK506 on MDA levels in ICV-STZ-treated rats

Administration of STZ in rats produced a pronounced elevation in MDA levels, as compared with vehicle treatment [F(4,34) = 92.38, P < 0.001] (Table 1). Chronic administration of FK506 dose-dependently attenuated the STZ-induced elevation in MDA levels (P < 0.001). However, FK506 treatment did not produce any significant effect in normal animals when compared with vehicle controls (P > 0.05). Effect of FK506 on brain nitrite level in ICV-STZ-treated rats

STZ-treated rats showed a significant increase in brain nitrite levels as compared with vehicle control-treated animals [F(4,34) = 98.44, P < 0.001] (Table 1). The STZinduced increase in nitrite levels was significantly and dose-dependently attenuated by FK506 (0.5 and 1 mg/kg, P < 0.01), whereas FK506 administration did not produce any significant effect (P > 0.05) in control animals as compared with vehicle-treated animals.

Effect of FK506 on brain lactate dehydrogenase levels in ICV-STZ-treated rats

STZ produced a significant elevation in brain lactate dehydrogenase (LDH) levels [F(4,34) = 59.67, P < 0.001] (Fig. 6) as compared with vehicle treatment, indicating cellular damage. FK506 (0.5 and 1 mg/kg) dose-dependently attenuated the STZ-induced elevation in LDH levels (P < 0.01). However, FK506 treatment did not alter LDH levels in the brains of normal rats as compared with vehicle treatment (P > 0.05).

Cresyl violet-stained hippocampal dentate gyrus and CA1 sections of vehicle-treated animals showed healthy neurons (Fig. 7a, b), which were robust in shape, had a pale and spherical or slightly oval nucleus, and had a single large nucleolus with a clear, visible cytoplasm. Brain sections from rats treated with FK506 (1 mg/kg, postoperatively) also showed healthy neurons in the dentate gyrus and CA1 region (Fig. 7c, d). Similar morphology and neuronal density in the dentate gyrus and the hippocampal CA1 region were observed in the vehicle-treated and FK506-treated groups (Fig. 7a–d). Neuronal degeneration may present a continuum of neurodegenerative morphologies. In this study, brain sections from ICV-STZ-treated rats showed pyknotic neurons that were darkly stained with no nucleus or visible nucleolus, and a few of the cells were shrunken and sickle shaped. Photomicrographs of the dentate gyrus and CA1 region of the hippocampus of ICV-STZ-treated groups showed a marked decrease in cell bodies (Fig. 7e, f). Other neurons showed a smaller nucleus with condensed chromatin and a swollen cytoplasm. Further, STZ administration markedly decreased neuronal density and increased the number of pyknotic neurons. Chronic treatment with FK506 (0.5 mg, Fig. 7g, h; 1 mg, Fig. 7i, j) attenuated STZ-induced cell loss and pyknosis; however, some degenerating cells with changes in morphology were observed in the FK506 treatment groups. Sections from rats treated with the higher dose of FK506 showed rounded and open nuclei with marked protection from STZ-induced neurodegenerative morphologies (Fig. 7i, j). There was marked improvement in neuronal density and reduction in pyknotic neurons following chronic FK506 treatment in STZ-treated rats, indicating a neuroprotective potential, in line with behavioral and biochemical effects of FK506.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

FK506 against STZ-induced neurotoxicity Arora et al. 585

Fig. 3

(a)

120

Vehicle

FK per se

STZ

FK (0.5 mg)

FK (1 mg)

Mean escape latency (s)

100 @

80



@

@

∗# 60

∗ ∗#



40

∗# 20 0 Day 17

Time spent in target quadrant (%)

(b)

Day 18

Day 19

Day 20

100

80 ∗# 60

40

∗ @

20

0 Vehicle

FK per se

STZ

FK (0.5 mg)

FK (1 mg)

Effect of FK506 on performance of ICV-STZ-treated rats in the Morris water maze. (a) Escape latency and (b) time spent in the target quadrant. Values are expressed as mean±SD; @P < 0.05 versus vehicle, *P < 0.05 versus STZ, #P < 0.05 versus FK506 (0.5 mg/kg). FK, FK506; ICV, intracerebroventricular; STZ, streptozotocin. Effect of tacrolimus on time spent in target quadrant in ICV-STZ-treated rats is expressed as mean±SD. @ P < 0.05 versus Vehicle, *P < 0.05 versus STZ, #P < 0.05 versus FK506 (0.5 mg/kg). FK, FK506; ICV, intracerebroventricular; STZ, streptozotocin.

Discussion The present study demonstrated a neuroprotective role of FK506, a specific CaN inhibitor, against STZ-induced behavioral, biochemical, and histological toxicity in rats. In line with earlier reports, ICV-STZ was found to produce significant impairment in spatial cognitive functions and increase oxidative–nitrosative stress (Sharma and Gupta, 2001; Sonkusare et al., 2005; Ishrat et al., 2006; Salkovic-Petrisic and Hoyer, 2007; SalkovicPetrisic, 2008). FK506 dose-dependently (0.5 and 1 mg/kg postoperatively) protected against the effect of STZ treatment in the MWM and passive avoidance task (Fig. 2 and Fig. 3a, b) without affecting locomotor activity (Fig. 4). Changes in locomotor activity have been suggested to modulate learning and memory in the passive avoidance and MWM paradigms (Sharma and

Gupta, 2003; Deshmukh et al., 2009). However, no significant difference in spontaneous locomotor activity was observed in any of the experimental groups in the present study. The cholinergic system plays an important role in memory formation and retrieval (Olton, 1990; Fibiger, 1991; Fibiger et al., 1991; Blokland, 1995). In the present study, STZ-induced cognitive deficit was also associated with an increase in cholinesterase activity (Fig. 5), which is in accordance with the findings of earlier studies (Sonkusare et al., 2005; Ishrat et al., 2006; Deshmukh et al., 2009). In the present study, FK506 dose-dependently protected against the effect of STZ treatment in the MWM and passive avoidance task and restored cholinesterase activity without modifying basal memory and cholinesterase

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

586 Behavioural Pharmacology 2013, Vol 24 No 7

Fig. 4

Fig. 6

500 LDH levels (IU/mg protein)

Activity counts/10 min

300

200

100

0

@

400 300

∗ ∗#

200 100 0

Vehicle

FK per se

STZ

FK 0.5 mg

FK 1 mg

Effect of FK506 on spontaneous locomotor activity in ICV-STZ-treated rats. Values are expressed as mean±SD. FK, FK506; ICV, intracerebroventricular; STZ, streptozotocin.

Vehicle

FK per se

STZ

FK 0.5 mg

FK 1 mg

Effect of FK506 on brain lactate dehydrogenase in ICV-STZ-treated rats. Values are expressed as mean±SD; @P < 0.05 versus vehicle; *P < 0.05 versus STZ; #P < 0.05 versus FK506 (0.5 mg/kg). FK, FK506; ICV, intracerebroventricular; STZ, streptozotocin.

Fig. 5

Acetylcholinesterase activity (nmol/mg protein)

500

@

400 ∗ 300

∗#

200 100 0 Vehicle

FK per se

STZ

FK (0.5 mg) FK (1 mg)

Effect of FK506 on brain acetylcholinesterase activity in ICV-STZtreated rats. Values are expressed as mean±SD; @P < 0.05 versus vehicle; *P < 0.05 versus STZ; #P < 0.05 versus FK506 (0.5 mg/kg). FK, FK506; ICV, Intracerebroventricular; LDH, lactate dehydrogenase; STZ, streptozotocin.

Table 1

Effect of FK506 on biochemical parameters Biochemical parameters

Groups Vehicle FK per se STZ FK (0.5 mg) FK (1 mg)

MDA (nmol/mg protein)

GSH (mmol/mg protein)

Nitrite (mmol/mg protein)

168.6±16.57 163.2±27.44 411.2±40.71@ 278.4±27.75* 213.6±23.32*#

1.84±0.23 1.95±0.28 0.18±0.08@ 0.91±0.18* 1.31±0.21*#

0.43±0.25 0.42±0.18 3.47±0.54@ 2.20±0.33* 1.55±0.28*#

Values are expressed as mean±SD. FK, FK506; GSH, reduced glutathione; MDA, malondialdehyde; STZ, streptozotocin. @ P < 0.05 versus vehicle. *P < 0.05 versus STZ. # P < 0.05 versus FK506 (0.5 mg/kg).

activity. CaN is a Ca2 + /calmodulin-dependent protein serine–threonine phosphatase 2B and the most abundant phosphatase in the central nervous system (CNS)

(Mansuy, 2003; Liu et al., 2007). Impairment of insulin signaling and glucose and energy metabolism following administration of STZ into the CNS has been suggested to cause Ca2 + overactivation (De la Monte and Wands, 2005; Sonkusare et al., 2005), which may lead to further imbalance between kinases and phosphatases (SalkovicPetrisic and Hoyer, 2007; Salkovic-Petrisic, 2008). Moreover, it has been reported that CaN is the first among the postsynaptic phosphatases to be activated in response to Ca2 + influx (Liu, 2009). Indeed, ICV-STZ administration has been reported to increase intracellular Ca2 + concentration (Muller et al., 1998), and blockade of the L-type calcium channel by lercanidipine has been reported to improve cholinergic function and cognitive behavior in STZ-treated rats (Sonkusare et al., 2005). In the present study, STZinduced cognitive deficits and the increase in AChE levels were significantly attenuated by FK506. It can be speculated that the increase in intracellular Ca2 + concentration following STZ administration may subsequently lead to CaN overactivation and contributes to cognitive deficit. Indeed, overactivation of CaN has been reported to negatively regulate learning and memory (Mansuy, 2003; Baumgartel et al., 2008). In addition, acute inhibition of CaN by FK506 has been reported to restore associative learning and memory in Tg2576 APP mice (Dineley et al., 2007; Taglialatela et al., 2009). Oxidative stress has been implicated in the pathogenesis of AD in humans (Pratico, 2008; Mangialasche et al., 2009). Oxyradical-induced damage to macromolecules (lipids, proteins, and nucleic acids, etc.) is considered an important factor in the acceleration of aging and agerelated neurodegenerative disorders such as AD (Liu et al., 2001; Wickens, 2001). An STZ-induced impairment in cerebral energy metabolism and mitochondrial disturbances may be responsible for increased intracellular Ca2 + influx and generation of reactive oxygen species,

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

FK506 against STZ-induced neurotoxicity Arora et al. 587

Fig. 7

(a, c, e, g, i) Cresyl violet-stained sections of the dentate gyrus and (b, d, f, h, j) the CA1 region of the hippocampus following treatment with (a, b) vehicle, (c, d) FK506 (1 mg/kg), (e, f) ICV-STZ, and chronic treatment with (g, h) 0.5 mg FK506 and (i, j) 1 mg FK506 following ICV-STZ treatment. (d, f, h, j) Pictures taken at a magnification of  400. FK, FK506; ICV, intracerebroventricular; STZ, streptozotocin.

leading to oxidative stress (Muller et al., 1998; Shoham et al., 2007; Deshmukh et al., 2009). Further, ICV administration of STZ has been reported to cause Ca2 + -dependent production of NO, a precursor for free radicals, which reacts with superoxide anions to form peroxynitrite, causing nitrosative stress (Shoham et al., 2007; Kumar and Kumar, 2009). Consistent with previous studies, in the present study, ICV-STZ treatment significantly increased MDA and nitrite levels and decreased the levels of endogenous antioxidant enzymes, indicating increased oxidative–nitrosative stress (Deshmukh et al., 2009). Chronic treatment of STZ-treated rats with FK506 dose-dependently attenuated the STZ-induced increase in oxidative–nitrosative stress. These results suggest that FK506 may have direct antioxidant potential

or may decrease free radical generation by inhibiting CaN activity. It has been reported that oxidative stress regulates Ca2 + dependent serine–threonine phosphatase, that is, CaN (Manik et al., 2003). In addition, mitochondrial dysfunction and oxidative stress have been observed in CaN transgenic mice, indicating that the upregulation of CaN signaling may lead to oxidative burden (Sayen et al., 2003). In addition, CaN has been reported to dephosphorylate nitric oxide synthase, resulting in an increase in NO production (Knirsch et al., 2001). Indeed, FK506, by CaN inhibition, has been reported to negatively regulate Ca2 + -channel activity and inhibit free radical generation and nitric oxide synthase activity

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

588 Behavioural Pharmacology 2013, Vol 24 No 7

(Victor et al., 1995; Phillis et al., 2002; Sasaki et al., 2004). In addition, FK506 has also been reported to possess direct free radical scavenging properties and to improve antioxidant defense mechanisms (Maziere et al., 2005). Therefore, FK506 may produce its antioxidant effect directly through free radical scavenging or indirectly through CaN inhibition. In the brain, STZ has been reported to cause neurodegeneration accompanied by an increase in LDH activity (Hoyer and Lannert, 2007). Consistent with these findings, in the present study, ICV-STZ-treated animals showed a marked increase in LDH levels, indicating cell damage. However, chronic administration of FK506 significantly attenuated the STZ-induced elevation in LDH levels. In line with the biochemical alterations, histological sections of the dentate gyrus and the CA1 region of the hippocampus of STZ-treated rats showed decreased neuronal density and necrotic cell death, as evidenced by pyknotic nuclear changes. Chronic administration of FK506 dose-dependently attenuated STZ-induced neurotoxicity and was able to restore neuronal density and a normal morphological pattern (Fig. 7), suggesting a neuroprotective potential of FK506. Ca2 + mobilization following stimulation of N-methyl-Daspartate receptors by glutamate has been implicated in cell death in the CNS. The Ca2 + sensitivity of nitric oxide synthase makes the Ca2 + -dependent phosphatase, CaN, an obvious candidate that affects neuronal cell death (Dawson et al., 1993), and therefore, CaN may be a material factor in Ca2 + -activated neuronal cell death (Shibasaki and Mckeon, 1995; Norris et al., 2005). In addition, overactivation of CaN has also been reported to increase LDH levels in the rat brain (Wei et al., 2002). Indeed, it is well documented that the neuroprotective effects of FK506 in experimental CNS diseases are associated with reduced neuronal loss, release of antiapoptotic proteins, and promotion of neurite outgrowth (Klettner and Herdegen, 2003; Morita et al., 2008). These findings potentially link CaN to cognitive dysfunction, impairment of cellular oxidant/antioxidant defenses, neurotransmitter deficits, and neuronal cell loss following ICV-STZ administration. FK506 attenuates the learning and memory deficits, cholinergic hypofunction, and oxidative–nitrosative stress induced by ICV-STZ infusion in rats. Conclusion

The observed cognitive enhancement following FK506 treatment in STZ-infused rats may be because of its antioxidant actions and neuromodulatory role over cholinergic neurons, which probably help restore acetylcholine levels. On the basis of the above results, it can be suggested that FK506 may prove to be a useful candidate drug molecule for the management of cognitive disorders such as AD.

Acknowledgements The authors are thankful to Mr Parveen Garg, Chairman, ISF College of Pharmacy, Moga (Punjab) for his praiseworthy inspiration and support for this study.

Conflicts of interest

There are no conflicts of interest.

References Balbo SL, Bonfleur ML, Carneiro EM, Amaral ME, Filiputti E, Mathias PC (2002). Parasympathetic activity changes insulin response to glucose and neurotransmitters. Diabetes Metab 28:3S13–3S17. Baumgartel K, Genoux D, Welzl H, Tweedie-Cullen RY, Koshibu K, LivingstoneZatchej M, et al. (2008). Control of the establishment of aversive memory by calcineurin and Zif268. Nat Neurosci 11:572–578. Blokland A (1995). Acetylcholine: a neurotransmitter for learning and memory? Brain Res Brain Res Rev 21:285–300. Butcher SP, Henshall DC, Teramura Y, Iwasaki K, Sharkey J (1997). Neuroprotective actions of FK506 in experimental stroke: in vivo evidence against an antiexcitotoxic mechanism. J Neurosci 17:6939–6946. Dawson TM, Steiner JP, Dawson VL, Dinerman JL, Urh GR, Soloman H (1993). Immunosuppressant FK506 enhances phosphorylation of nitric oxide synthase and protects against glutamate neurotoxicity. Proc Natl Acad Sci USA 90:9808–9812. De la Monte SM, Wands JR (2005). Review of insulin and insulin-like growth factor expression, signaling, and malfunction in the central nervous system: relevance to Alzheimer’s disease. J Alzheimers Dis 7:45–61. De la Torre JC (2008). Pathophysiology of neuronal energy crisis in Alzheimer’s disease. Neurodegener Dis 5:126–132. Deshmukh R, Sharma V, Mehan S, Sharma N, Bedi KL (2009). Ameleoration of intracerebroventricular streptozotocin induced cognitive dysfunction and oxidation stress by vinpocetine- a PDE-1 inhibitor. Eur J Pharmacol 620: 49–56. Dineley KT, Hogan D, Zhang WR, Taglialatela G (2007). Acute inhibition of calcineurin restores associative learning and memory in Tg2576 APP transgenic mice. Neurobiol Learn Mem 88:217–224. Dudek NL, Dai Y, Muma NA (2010). Neuroprotective effects of calmodulin peptide 76-121aa: disruption of calmodulin binding to mutant huntingtin. Brain Pathol 20:176–189, . Ellman GL (1959). Tissue sulfhydryl groups. Arch Biochem Biophys 82:70–77. Ellman GL, Courtney KD, Andres V, Featherston RMA (1961). New and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 7:88–91. Eto R, Abe M, Hayakawa N, Kato H, Araki T (2008). Age-related changes of calcineurin and Akt1/protein kinase Balpha (Akt1/PKBalpha) immunoreactivity in the mouse hippocampal CA1 sector: an immunohistochemical study. Metab Brain Dis 23:399–409. Fibiger HC (1991). Cholinergic mechanisms in learning, memory and dementia: a review of recent evidence. Trends Neurosci 14:220–223. Fibiger HC, Damsma G, Day JC (1991). Behavioral pharmacology and biochemistry of central cholinergic neurotransmission. Adv Exp Med Biol 295:399–414. Foster TC, Sharrow KM, Masse JR, Norris CM, Kumar A (2001). Calcineurin links Ca2 + dysregulation with brain aging. J Neurosci 21:4066–4073. Gold BG, Voda J, Yu X, McKeon G, Bourdette DN (2004). FK506 and a nonimmunosuppressant derivative reduce axonal and myelin damage in experimental autoimmune encephalomyelitis: neuroimmunophilin ligandmediated neuroprotection in a model of multiple sclerosis. J Neurosci Res 77:367–377. Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannebaum SR (1982). Analysis of nitrate, nitrite, and [15N] nitrate in biological fluids. Ann Biochem 126:131–138. Groetzner J, Reichart B, Roemer U, Reichel S, Kozlik-Feldmann R, Tiete A, et al. (2005). Cardiac transplantation in pediatric patients: fifteen-year experience of a single center. Ann Thorac Surg 79:53–60. Hoyer S, Lannert H (2007). Long-term abnormalities in brain glucose-energy metabolism after inhibition of the neuronal insulin receptor: implication of tauprotein. J Neural Transm 72:195–202. Ishrat T, Khan MB, Hoda MN, Yousuf S, Ahmad M, Ansari MA, et al. (2006). Coenzyme Q10 modulates cognitive impairment against intracerebroventricular injection of streptozotocin in rats. Behav Brain Res 171:9–16.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

FK506 against STZ-induced neurotoxicity Arora et al. 589

Ishrat T, Hoda MN, Khan MB, Yousuf S, Ahmad M, Khan MM, et al. (2009). Amelioration of cognitive deficits and neurodegeneration by curcumin in rat model of sporadic dementia of Alzheimer’s type (SDAT). Eur Neuropsychopharmacol 21:1–12. Klettner A, Herdegen T (2003). The immunophilin-ligands FK506 and V-10,367 mediate neuroprotection by the heat shock response. Br J Pharmacol 138:1004–1012. Knirsch U, Sturm S, Reuter A, Bachus R, Gosztonyi G, Voelkel H, Ludolph AC (2001). Calcineurin A and calbindin immunoreactivity in the spinal cord of G93A superoxide dismutase transgenic mice. Brain Res 889:234–238. Kumar P, Kumar A (2009). Neuroprotective effect of cyclosporine and FK506 against 3-nitropropionic acid induced cognitive dysfunction and glutathione redox in rat: possible role of nitric oxide. Neurosci Res 63:302–314. Liu D, McIlvain HB, Fennell M, Dunlop J, Wood A, Zaleska MM, et al. (2007). Screening of immunophilin ligands by quantitative analysis of neurofilament expression and neurite outgrowth in cultured neurons and cells. J Neurosci Methods 163:310–320. Liu JO (2009). Calmodulin-dependent phosphatase, kinase, and transcriptional corepressors involved in T-cell activation. Immunol Rev 228:184–198. Liu R, Liu IY, Bi X, Thompson RF, Doctrow SR, Malfroy B, Baudry M (2001). Reversal of age-related learning deficits and brain oxidative stress in mice with superoxide dismutase/catalase mimetics. Proc Natl Acad Sci USA 100:8526–8531. Lowry HO, Rosebrough JN, Farr L, Randall JR (1951). Protein measurement with the folin phenol reagent. J Biol Chem 193:265–275. Mangialasche F, Polidori MC, Monastero R, Ercolani S, Camarda C, Cecchetti R, Mecocci P (2009). Biomarkers of oxidative and nitrosative damage in Alzheimer’s disease and mild cognitive impairment. Ageing Res Rev 8:285–305. Manik C, Ghosh-Xuton W, Li S, Klee C (2003). Regulation of calcineurin by oxidative stress. Methods Enzymol 366:289–304. Mansuy MI (2003). Calcineurin in memory and bidirectional plasticity. Biochem Biophys Res Commun 311:1195–1208. Maziere C, Morlie`re P, Massy Z, Kamel S, Louandre C, Conte MA, Mazie`re JC (2005). Oxidized low-density lipoprotein elicits an intracellular calcium rise and increases the binding activity of the transcription factor NFAT. Free Radic Biol Med 38:472–480. Morita M, Imai H, Liu Y, Xu X, Sadamatsu M, Nakagami R, et al. (2008). FK506protective effects against trimethyltin neurotoxicity in rats: hippocampal expression analyses reveal the involvement of periarterial osteopontin. Neuroscience 153:1135–1145. Morris RGM (1984). Development of water –maze procedure for studying spatial learning in the rats. J Neurosci Methods 11:47–60. Muller D, Nitsch RM, Wurtman RJ, Hoyer S (1998). Streptozotocin increases free fatty acids and decreases phospholipids in rat brain. J Neural Transm 105:1271–1281. Nakamura TY, Iwata Y, Arai Y, Komamura K, Wakabayashi S (2008). Activation of Na + /H + exchanges is sufficient to genetic Ca + signals that induce cardiac hypertrophy and heart failure. Circ Res 103:891–899. Norris CM, Kadish I, Blalock EM, Chen KC, Thibault V, Porter NM, et al. (2005). Calcineurin triggers reactive/inflammatory processes in astrocytes and is upregulated in aging and Alzheimer’s models. J Neurosci 25:4649–4658. Olton DS (1990). Dementia: animal models of the cognitive impairments following damage to the basal forebrain cholinergic system. Brain Res Bull 25:499–502. Paxinos G, Watson C (1986). The rat brain in stereotaxic coordinates. 2nd ed. San Diego: Academic press. Phillis JW, Diaz FG, O’Regan MH, Pilitsis JG (2002). Effects of immunosuppressants, calcineurin inhibition, and blockade of endoplasmic reticulum calcium channels on free fatty acid efflux from the ischemic/reperfused rat cerebral cortex. Brain Res 957:12–24. Pratico D (2008). Evidence of oxidative stress in Alzheimer’s disease brain and antioxidant therapy: lights and shadows. Ann N Y Acad Sci 1147:70–78. Prickaerts J, Blokland A, Honig W, Meng F, Jolles J (1995). Spatial discrimination learning and choline acetyltransferase activity in streptozotocin-treated rats: effects of chronic treatment with acetyl-L-carnitine. Brain Res 674:142–146. Reed TT, Pierce WM, Maresbery WR, Butterfield DA (2009). Proteomic Identification of HNE bound proteins in early Alzheimer disease: Insight into the role of lipid peroxidation in the progression of AD. Brain Res 1274:66–76. Sakai K, Date I, Yoshimoto Y, Arisawa T, Nakashima H, Furuta T, et al. (1991). The effect of a new immunosuppressive agent, FK-506, on xenogeneic neural transplantation in rodents. Brain Res 565:167–170.

Salkovic-Petrisic M, Hoyer S (2007). Central insulin resistance as a trigger for Sporadic Alzheimer-like pathology: an experimental approach. J Neural Transm 72:217–233. Salkovic-Petrisic M (2008). Amyloid cascade hypothesis: is it true for sporadic Alzheimer’s disease. Periodicum Biologrum 110:17–25. Sasaki T, Hamada J, Shibata M, Gotoh J, Araki N, Fukuuchi Y (2004). FK506 abrogates delayed neuronal death via suppression of nitric oxide production in rats. Brain Res 1009:34–39. Sayen MR, Gustafsson AB, Sussman MA, Molkentin JD, Gottlieb RA (2003). Calcineurin transgenic mice have mitochondrial dysfunction and elevated superoxide production. Am J Physiol Cell Physiol 284:C562–C570. Sharma M, Gupta YK (2001). Intracerebroventricular injection of streptozotocin in rats produce both oxidative stress in the brain and cognitive impairment. Life Sci 68:1021–1029. Sharma M, Gupta YK (2002). Chronic treatment with trans resveratrol prevents intracerebroventricular streptozotocin induced cognitive impairment and oxidative stress in rats. Life Sci 71:2489–2498. Sharma M, Gupta YK (2003). Effect of alpha lipoic acid on intracerebroventricular streptozotocin model of cognitive impairment in rats. Eur Neuropsychopharmacol 13:241–247. Shibasaki F, Mckeon F (1995). Calcineurin functions in Ca(2 +)-activated cell death in mammalian cells. J Cell Biol 131:735–743. Shoham S, Bejar C, Kovalev E, Weinstock M (2003). Intracerebroventricular injection of streptozotocin causes neurotoxicity to myelin that contributes to spatial memory deficits in rats. Exp Neurol 184:1043–1052. Shoham S, Bejar C, Kovalev E, Schorer-Apelbaum D, Weinstock M (2007). Ladostigil prevents gliosis, oxidative-nitrative stress and memory deficits induced by intracerebroventricular injection of streptozotocin in rats. Neuropharmacology 52:836–843. Singh A, Kumar G, Naidu SP, Kulkarni KS (2003a). Protective effect of FK-506 (tacrolimus) in pentylenetetrazol-induced kindling in mice. Pharmacol Biochem Behav 75:853–860. Sonkusare S, Srinivasan K, Kaul C, Ramarao P (2005). Effect of donepezil and lercanidipine on memory impairment induced by intracerebroventricular streptozotocin in rats. Life Sci 77:1–14. Steen E, Terry BM, Rivera EJ, Neely TR, Tavares R, Xu XJ, et al. (2005). Impaired insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease-is this type 3 diabetes? J Alzheimers Dis 7:63–80. Taglialatela G, Hogan D, Zhang WR, Dineley KT (2009). Intermediate and long term recognition memory deficits in Tg2576 mice are reversed with acute calcineurin inhibition. Behav Brain Res 200:95–99. Van Rossum HH, Romijn FP, Smit NP, de Fijter JW, Van Pelt J (2009). Everolimus and sirolimus antagonize tacrolimus based calcineurin inhibition via competition for FK-binding protein 12. Biochem Pharmacol 77:1206–1212. Vazquez-Lopez A, Sierra-Paredes G, Sierra-Marcun˜o G (2006). Anticonvulsant effect of the calcineurin inhibitor ascomycin on seizures induced by picrotoxin microperfusion in the rat hippocampus. Pharmacol Biochem Behav 84:511–516. Victor RG, Thomas GD, Marbant E, Rourket OB (1995). Presynaptic modulation of cortical synaptic activity by calcineurin. Proc Natl Acad Sci USA 92:6269–6273. Voda J, Yamaji T, Gold BG (2005). Neuroimmunophilin ligands improve functional recovery and increase axonal growth after spinal cord hemisection in rats. J Neurotrauma 22:1150–1161. Watson GS, Craft S (2006). Insulin resistance, inflammation, and cognition in Alzheimer’s disease: lessons for multiple sclerosis. J Neurosci 245:21–33. Wei Q, Holzer M, Brueckner MK, Liu Y, Arendt T (2002). Dephosphorylation of tau protein by calcineurin triturated into neural living cells. Cell Mol Neurobiol 22:13–24. Wickens AP (2001). Ageing and the free radical theory. Respir Physiol 28:379–391. Wills ED (1966). Mechanism of lipid peroxide formation in animal tissues. Biochem J 99:667–676. Wright AK, Miller C, Williams M, Arbuthnott G (2008). Microglial activation is not prevented by tacrolimus but dopamine neuron damage is reduced in a rat model of Parkinson’s disease progression. Brain Res 1216:78–86. Xifiro X, Garcia-Martinez MJ, Toro-del D, Alberch J, Navarro-Perez E (2008). Calcineurin is involved in the early activation of NMDA-mediated cell death in mutant huntingtins knock-in striatal cells. J Neurochem 105:1596–1612.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

FK506 attenuates intracerebroventricular streptozotocin-induced neurotoxicity in rats.

Upregulation in calcineurin (CaN) signaling has been implicated in various neurodegenerative disorders. In the present study, we have investigated the...
778KB Sizes 0 Downloads 12 Views