Hormones & Behavior.pdf

0 downloads 0 Views 2MB Size Report
tion and induces type 2 diabetes like state (Ghaisas et al., 2009; Jatwa. 55 ... (MAPK) have been observed in brain regions during depressive like be- ..... taining protease inhibitor cocktail (MP Biomedicals) and then centri- ...... duces the variations of 11β-hydroxysteroid dehydrogenase type 1 and .... Curcumin produces anti-.
Dear Author, Please, note that changes made to the HTML content will be added to the article before publication, but are not reflected in this PDF. Note also that this file should not be used for submitting corrections.

YHBEH-03993; No. of pages: 11; 4C: Hormones and Behavior xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Hormones and Behavior

3Q2

Sita Sharan Patel a,c, Vineet Mehta a, Harish Changotra b, Udayabanu Malairaman a,⁎

4 5 6 7

a

8

a r t i c l e

9 10 11 12 13 14 33 34 35 36 37 38

Article history: Received 11 June 2015 Revised 5 September 2015 Accepted 25 November 2015 Available online xxxx

i n f o

R O

Department of Pharmacy, Jaypee University of Information Technology, Waknaghat, Himachal Pradesh, India Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Himachal Pradesh, India Department of Pharmacology, Lakshmi Narain College of Pharmacy, Bhopal, Madhya Pradesh, India

a b s t r a c t

Comorbidity of depression and diabetes is a serious risk factor worsening the complications such as cognitive function and locomotion. Treatment under this condition becomes extremely complicated. Insulin signaling and autophagy pathways are involved in modulation of learning and memory. Rosiglitazone (ROSI) ameliorate cognitive deficit associated with depression and insulin resistance. In the present study, we investigated the effect of ROSI against chronic unpredictable stress (CUS) induced depression as a risk factor for diabetes and behavioral dysfunctions. Adult male Swiss albino mice were exposed to CUS alongside ROSI (5 mg/kg/day) treatment for 21 days. Thereafter, animals were subjected to different behavioral studies to assess depressive like behavior, cognition and locomotion. The effect of ROSI on insulin signaling, autophagy and apoptosis were evaluated in the hippocampus. CUS resulted in depressive like behavior, cognitive impairment and hypolocomotion associated with oxidative stress, impaired glucose tolerance and hypercorticosteronemia. CUS significantly impaired hippocampal insulin signaling, membrane translocation of glucose transporter type 4 (GLUT4) as well as decreased the expression of autophagy5, autophagy7, B-cell lymphoma 2 and apoptosis inhibitory protein 2. ROSI significantly reduced depressive like behavior, postprandial blood glucose, hypercorticosteronemia, oxidative and inflammatory stress, and apoptosis in stressed mice. Moreover, ROSI treatment effectively improved hippocampal insulin signaling, GLUT4 membrane translocation and cognitive performance in depressed mice. ROSI administration might prove to be effective for neurological disorders associated with depressive like behavior and impaired glucose tolerance. © 2015 Published by Elsevier Inc.

P

c

D

b

O

2

Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone

1Q1

F

journal homepage: www.elsevier.com/locate/yhbeh

R

E

C

T

E

Keywords: Chronic stress Cognition Depression Glucose tolerance Rosiglitazone

42 40 39

O R

41

Introduction

44 45

The chronic unpredictable stress (CUS) model is widely used for the induction of depressive like behavior in rodents, which consists of repeated exposure to an array of unpredictable stressors over a sustained period of time (Katz, 1982; Kessler et al., 1985). Repeated stressors are associated with hyperactivation of hypothalamic–pituitary–adrenal axis (HPA axis) which is known to induce neurodegeneration,

48 49

U

46 47

N C

43 Q3

Abbreviations: AIP2, apoptosis inhibitory protein 2; BCL2, B-cell lymphoma 2; CUS, chronic unpredictable stress; FST, forced swim test; GLP1, glucagon like peptide 1; GLUT4, glucose transporter type 4; HPA axis, hypothalamic–pituitary–adrenal axis; INSG1, insulin induced gene 1; ILGF 1r, insulin like growth factor 1 receptor; IR, insulin receptor; IRS1, insulin receptor substrate 1; IRS2, insulin receptor substrate 2; MAPK1, mitogen activated protein kinase 1; MWM, Morris water maze; NO, nitric oxide; OGTT, oral glucose tolerance test; PA task, passive avoidance step-through task; PPARγ, peroxisome proliferator activated receptor gamma; PI3K, phosphoinositide 3-kinase; PKB, protein kinase B; ROSI, rosiglitazone; STL, step through latency; TST, tail suspension test; TBARS, thiobarbituric acid reactive substances. ⁎ Corresponding author at: Jaypee University of Information Technology, Waknaghat, Himachal Pradesh 173234, India. E-mail address: [email protected] (U. Malairaman).

depression and cognitive dysfunction (Rossetti et al., 2014; Sousa et al., 2008). Chronic activation of HPA axis mediates hypercorticosteronemia, inhibits insulin secretion from pancreatic βcells, reduces glucose uptake and utilization, stimulates glucagon secretion and induces type 2 diabetes like state (Ghaisas et al., 2009; Jatwa et al., 2007) as well as impairs neuronal plasticity in hippocampus (Grillo et al., 2009; Piroli et al., 2007). Depressed diabetics show impaired cognitive performance in attention and information processing (Watari et al., 2006). Hypercorticosteronemia induces neuronal oxidative stress and inflammation resulting in cognitive impairment (Pariante and Miller, 2001; Peng et al., 2012; Suwanjang et al., 2013). Reactive nitrogen species such as nitric oxide (NO) has been implicated in stress mediated inflammation and cognitive deficit (Peng et al., 2012). In addition, NO impairs autophagy in rat cortical neurons, which is known to modulate neuronal health (Sarkar et al., 2011). Dysfunctioning of neuronal peroxisome proliferator-activated receptor-γ (PPARγ), insulin receptor (IR), brain derived neurotrophic factor and mitogen activated protein kinase (MAPK) have been observed in brain regions during depressive like behavior and cognitive impairment (Gottschalk et al., 1999;

http://dx.doi.org/10.1016/j.yhbeh.2015.11.010 0018-506X/© 2015 Published by Elsevier Inc.

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32

50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69

2

105 106

Behavioral assessment

111

Forced swim test (FST) The animals were individually forced to swim in a cylinder with radius 24 cm and height 25 cm filled with water (26 ± 2 °C) up to a height of 18 cm. An animal was considered immobile whenever it remained floating passively in the water in a slightly hunched but upright position and its nose just above the water surface. The total immobility period of each animal during the 6 min test was recorded (Kulkarni and Mehta, 1985).

112 113

87

Materials and methods

120 121

88

Animals

89 90

94

Experimental design

95 96

Animals were divided into four groups: group I received 0.3% carboxymethyl-cellulose (0.3% CMC, p.o.) and served as control; group II was exposed to CUS and received vehicle (0.3% CMC, p.o.); group III was subjected to CUS and received ROSI (5 mg/kg, p.o.) and group IV received ROSI (5 mg/kg, p.o.). Dose of ROSI was selected from previous study (Lee et al., 2006) and administered once daily for 21 days. The animals were subjected to CUS paradigm as described previously (Bhutani et al., 2009; Katz, 1981) with modifications. Animals underwent stress paradigm once a day over a period of 21 days (Fig. 1). After 3 weeks of CUS and drug treatment, animals were

Morris water maze task (MWM) Spatial memory was assessed using MWM, which consisted of a white circular pool of 1 m diameter, filled with water at room temperature and had a submerged transparent escape platform kept 1 cm below the water surface. The pool was made opaque with addition of nontoxic water-soluble white paint, which makes the submerged platform invisible to the mice. The pool was divided into four hypothetical quadrants. Each mouse was individually allowed to swim freely (habituation trial) in the maze for 5 min (without platform) on day 21. During training trial (days 22–25) the platform was positioned in the centre of a quadrant and each mouse was released facing toward the wall of the pool in the randomly selected quadrant. The mice were allowed to search the platform spontaneously within 60 s. Mice that failed to find the submerged platform within 60 s were placed onto the platform by the experimenter and allowed to remain on the platform for 5 s (learning trial). Each

126

93

Male Swiss albino mice weighing 25–30 g were housed under a 12 h light/dark cycle at 26 ± 2 °C. The animals had access to food and water ad libitum. All animal experiments were carried out in accordance with CPCSEA guidelines and Institutional Animal Ethical Committee. All efforts were made to minimise pain or discomfort.

Tail suspension test (TST) The animals were individually suspended on the edge of a shelf by adhesive tape placed approximately 1 cm from the tip of the tail. Animals were considered immobile when they hang passively and motionless. The duration of immobility was recorded for the periods of 6 min during the test (Steru et al., 1985).

97 98 99 100 101 102 103 104

O

R O

P

D

E

91 92

T

83 84

C

81 82

E

79 80

R R

77 78

O

76

C

74 75

N

72 73

F

subjected to different behavioral studies to assess depressive like behavior, cognition and locomotion. On day 25, immediately after behavioral studies (3–5 pm) blood was collected via retro-orbital puncture. Serum and plasma were separated for biochemical estimation. Animals were sacrificed by cervical dislocation and the hippocampus was dissected for further studies.

85 86

Greene-Schloesser et al., 2014; Grillo et al., 2011; Sadaghiani et al., 2011; Zhang et al., 2012). Rosiglitazone (ROSI) is a selective agonist for nuclear PPARγ receptor, known to increase glucose influx in adipose tissue and muscle by enhancing synthesis and translocation of glucose transporters (Hardman and Limberd, 2001; Tripathi, 2013). ROSI improves cognitive performance, β-cell functions, attenuates insulin resistance and inflammation in diabetics (Abbatecola et al., 2010; Awara et al., 2005; Hanley et al., 2010; Hsu et al., 2005). ROSI attenuates depression associated insulin resistance (Rasgon et al., 2010) and impaired hippocampal neurogenesis (Cheng et al., 2015). The present study was designed to investigate the effect of CUS induced depression as a risk factor for diabetes and associated behavioral dysfunction. We investigated the involvement of insulin signaling and autophagy in the hippocampus region of brain during chronic stress. Further, we studied the neuromodulatory role of ROSI, an antidiabetic drug, against CUS induced depression.

U

70 71

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

Fig. 1. CUS procedure and experimental design: C — cold swim (8 °C, 3 min); T — tail pinch (1 min); F — food and water deprivation (24 h); I1 — immobilization (3 h); O — overnight illumination; FS — foot shock (20 trials, 0.5 mA, 5.0 s maximum duration, 1 min intervals); T1 — tail pinch (2 min); C1 — cold swim (10 °C, 5 min); FS1 — foot shock (20 trials, 0.5 mA, 5.0 s maximum duration, 30 s intervals); I2 — immobilization (4 h); T2 — tail pinch (3 min); O1 — overnight illumination with wet cage; C2 — cold swim (6 °C, 3 min); I3 — immobilization (5 h); OT — overnight illumination with tilted cage.

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

107 108 109 110

114 115 116 117 118 119

122 123 124 125

127 128 129 130 131 132 133 134 135 136 137 138 139 140

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

Locomotor activity

162

165 166

The locomotor activity of mice was assessed by using digital actophotometer (Inco Ambala, India). Animals were individually placed at the centre of the square arena (35 cm × 35 cm) of apparatus. After an initial familiarization period (3 min), the digital locomotor score was recorded for the next 10 min (Mahesh et al., 2012).

167

Real-time quantitative reverse transcription PCR

168 169

179 180

After behavioral studies, the animals were sacrificed, hippocampus was dissected and total RNA was isolated using TRIzol reagent (Invitrogen). The integrity of RNA was checked on 2% agarose gel and quantified using nano-drop (ND-2000C, Thermo Scientific). The reverse transcription of 3 μg of total RNA was performed using First strand cDNA synthesis kit (Fermentas life-sciences). qPCR amplifications were performed in a CFX96™ Real-Time PCR Detection System (Bio-Rad) using the iQ™ SYBR green supermix (Bio-Rad). Reactions were carried out in total volumes of 12.5 μl, included 2.5 pM of each primer (Table 1) and 1 μl of cDNA template containing 100 ng cDNA. The thermal cycler conditions for cDNA amplification were as follows: Step 1, 95 °C for 3:00 min; Step 2, 95 °C for 10 s, 52–58 °C for 30 s and 72 °C for 2:20 s (35 cycles). GAPDH was used as an internal control and thermal cycler

t1:1 t1:2

Table 1 Sequence of oligonucleotides used for qRT-PCR.

171 172 173 174 175 176 177 178

C

E

170

R

163 164

O R

157 158

t1:3

S no.

t1:4 t1:5 t1:6 t1:7 t1:8 t1:9 t1:10 t1:11 t1:12 t1:13 t1:14 t1:15 t1:16 t1:17 t1:18 t1:19

1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16.

N C

155 156

U

153 154

F

161

151 152

184 185

O

159 160

PA task was used to evaluate the associative memory, which consisted of a 25 cm long box partitioned into light (10 cm × 14 cm × 16 cm) and dark (10 cm × 10 cm × 16 cm) compartments. The light compartment was illuminated with 60 W bulb, kept 60 cm above the apparatus. Initially, all mice were given one habituation trial to explore both compartments for 120 s. On day 1 of acquisition trial the animals were allowed to explore the light chamber for 5 s. The guillotine door was opened and time taken by mouse to enter the dark chamber was recorded as step through latency (STL). Each animal received an inescapable electric shock for 2 s on a grid floor in the dark chamber. On day 2 of acquisition, memory retention was tested for each mouse in the same manner, but the shock was not delivered (Udayabanu et al., 2012).

149 150

Hippocampus was homogenized in 400 μl ice-cold RIPA buffer containing protease inhibitor cocktail (MP Biomedicals) and then centrifuged at 4 °C 1000 × g for 10 min. The supernatant was collected, centrifuged at 16,000 ×g for 15 min to remove cytosolic fraction. The pellet was resuspended in 500 μl cell lysis RIPA buffer containing 1% Triton X-100, centrifuged at 4 °C 16,000 ×g for 15 min and the supernatant was collected as a crude membrane fraction (Potapenko et al., 2012) for GLUT4 expression. Immunoprecipitation of GLUT4 from crude membrane fraction was then performed with modifications (Mayat et al., 1995). Briefly, 50 μl of Protein A/G PLUS-Agarose (Santa Cruz Biotechnology) was added to 500 μl of membrane lysate and left to incubate for 1 h in rotor. The mixture was then centrifuged (3000 × g) for 3 min at 4 °C. Supernatant was then transferred into fresh tubes containing 10 μg of GLUT4 antibody (Santa Cruz Biotechnology) and subjected to overnight incubation on a rotor. Thereafter, 50 μl of Protein A/G PLUSAgarose slurry was added and incubated in rotor for 1 h. The complex was pelleted by centrifugation 3000 ×g for 10 min at 4 °C and washed five times with 500 μl of ice cold PBS. The washed-immune complex was then resuspended in 50 μl of 1 × Laemmli buffer and boiled for 3 min. After centrifugation, the supernatant was subjected to 10% sodium dodecyl sulphate-polyacrylamide gel electrophoresis. The electrophoretically separated GLUT4 protein was analyzed by Coomassie staining and band density analysis was performed using densitometer (GS-800 Calibrated densitometer, BioRad).

R O

148

183

P

Passive avoidance step-through (PA) task

Glucose transporter type 4 (GLUT4) translocation

D

147

143 144

condition for GAPDH was as follows: Step 1, 95 °C for 3:00 min; Step 181 2, 95 °C for 10 s, 57.6 °C for 30 s and 72 °C for 2:20 s (35 cycles). 182

T

145 146

mouse received four learning session per day with 10 min interval between each trial. In each trial, the time taken by the mouse to find the hidden platform was recorded as escape latency. Probe trial test was conducted on day 25 to evaluate the index of memory in which the number of crossings across the platform area was recorded (Udayabanu et al., 2012).

E

141 142

3

186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207

Biochemical estimation

208

Blood glucose levels were measured in blood samples collected from the tail vein using Accu-check (Roach Diagnostics) blood glucose monitoring system. Oral glucose tolerance test (OGTT) was performed on 16 h fasted mice using 2 g glucose/kg body weight. Blood was collected from the animals by tail snipping at 0, 0.5, 1.0, 1.5 and 2.0 h after glucose load. The level of serum insulin was determined by chemiluminescent immunoassay using a commercially available kit (AccuLite CLIA Microwells, Monobind Inc., USA). An HPLC-UV system was used for quantification of plasma corticosterone using dexamethasone as an internal standard. Briefly, 500 μl of plasma containing known quantity of dexamethasone was extracted with 5 ml of dichloromethane (DCM). The DCM extract was evaporated to dryness and dissolved in 100 μl of mobile phase. 20 μl of extract was injected into HPLC system for quantification. Mobile phase was composed of methanol:water (70:30) and flow was adjusted at 1.2

209 210

Gene

Forward primer 5′ → 3′

Reverse primer 5′ → 3′

PPARγ IR ILGF 1r GLP1 IRS1 IRS2 PI3K PKB GLUT4 INSG1 MAPK1 BCL2 AIP2 Autophagy5 Autophagy7 GAPDH

AGG GCG ATC TTG ACA GGA AA TTT GTC ATG GAT GGA GGC TA GTG GGG GCT GCT CGT GTT TCT C TCA GAG ACG GTG CAG AAA TGG CGA TGG CTT CTC AGA CGT G CTG CGT CCT CTC CCA AAG TG CGA GAG TGT CGT CAC AGT GTC TGC CCA CAC GCT TAC TGA GA GAT GGG CTT TCT CCG TCC CAC GAC CAC GTC TGG AAC TAT CCC TTA GAC ACT GTG ACG GT GGC TGA GCA CTA CCT TCA GTA AGC TTG GTG TCT GTT CTC TGT CTC GCT AGA TGG AAC CAC TGG CTG CTA CTT CTG CAA TGA TGT TTC ACC ACC ATG GAG AAG GC

CGA AAC TGG CAC CCT TGA AA CCT CAT CTT GGG GTT GAA CT GAT CAC CGT GCA GTT TTC CA ATC AAA GGT CCG GTT GCA GAA CAG CCC GCT TGT TGA TGT TG GGG GTC ATG GGC ATG TAG C TGT TCG CTT CCA CAA ACA CAG CAA AGC AGA GGC GGT CGT GTG TGG CAA GAG TTC AGT GG TGA GAA GAG CAC TAG GCT CCG CAC AGT CCC AAA GCC ACA AA TGG CGG TAT CTA TGG ATT CCA C TGG AGG GAA GAT AGG TCC CAC AGT GGT CCT GTG TGT CTC CAG GAC AGA GAC CAT CAG CTC CAC GGC ATG GAC TGT GGT CAT GA

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

211 212 213 214 215 216 217 218 219 220 221 222 223

245

All data were analyzed using GraphPad prism 5 software. Two-way ANOVA with Bonferroni post hoc test was used to analyze MWM and OGTT data. The statistical significance of other data was assessed by one-way ANOVA followed by Tukey's post hoc test for differences among the groups. Further, two-way ANOVA for main effect and interaction was performed for all data. A confidence level of p b 0.05 was considered as significant. Eta squared effect sizes (η2) for ANOVA results and Cohen's d effect size estimates for pair-wise comparisons were performed.

246

Results

247

Behavioral assessment

248

FST

249 250

A two-way ANOVA of depressive behavior in FST did not demonstrate a significant CUS/ROSI interaction [F (1,20) = 2.59, p N 0.05, η2 = 0.05], a main effect of CUS [F (1,20) = 21.18, p b 0.001, η2 = 0.41 and d = 2.12] and ROSI [F (1,20) = 7.87, p b 0.05, η2 = 0.15 and d = 3.03]. Post hoc comparisons demonstrated that CUS significantly increased the duration of immobility whereas chronic ROSI administration significantly improved the mobility period in stressed mice (Fig. 2A).

255 256 257 258 259 260 261

PA task A one-way ANOVA revealed that chronically stressed mice showed no significant alteration in STL [F (7,40) = 41.32, p N 0.05, R2 = 0.87] on day 23 (memory retention trial) when compared with their respective day 22 (acquisition trial) STL. A two-way ANOVA of associative memory performance during memory retention trial revealed a significant CUS/ROSI interaction [F (1,20) = 24.8, p b 0.001, η2 = 0.26] and a main effect of CUS [F (1,20) = 27.49, p b 0.001, η2 = 0.29 and d = 4.38] and ROSI [F (1,20) = 20.17, p b 0.001, η2 = 0.21 and d = 4.24]. Post hoc comparisons during memory retention trial demonstrated that CUS significantly decreased STL, and the administration of ROSI prevented the decrease of STL (Fig. 3C).

296

Locomotor activity A two-way ANOVA of locomotor activity in the actophotometer did not reveal a significant CUS/ROSI interaction [F (1,20) = 0.87, p N 0.05, η2 = 0.01], a main effect of CUS [F (1,20) = 52.23, p b 0.001, η2 = 0.71 and d = 3.18] and ROSI [F (1,20) = 0.32, p N 0.05, η2 = 0.004 and d = 0.21]. Post hoc comparisons revealed that CUS exposure significantly reduced the locomotor activity, and the administration of ROSI did not reverse hypolocomotion (Fig. 4).

308 309

Determination of blood glucose level

316

Fasting blood glucose A two-way ANOVA of fasting blood glucose levels in mice did not reveal a significant CUS/ROSI interaction [F (1,20) = 2.12, p N 0.05, η2 = 0.06], a main effect of CUS [F (1,20) = 4.30, p N 0.05, η2 = 0.13 and d = 1.37] and ROSI [F (1,20) = 6.01, p b 0.05, η2 = 0.18 and d =

317 318

T

C

253 254

E

251 252

R R

243 244

TST

A two-way ANOVA of depressive behavior in TST revealed a significant CUS/ROSI interaction [F (1,20) = 5.40, p b 0.05, η2 = 0.14] and a main effect of CUS [F (1,20) = 6.30, p b 0.05, η2 = 0.16 and d = 1.84] and ROSI [F (1,20) = 6.64, p b 0.05, η2 = 0.17 and d = 2.68]. Post hoc

O

241 242

C

239 240

N

233 234

U

231 232

265

F

237 238

230

MWM task A two-way ANOVA of spatial memory in MWM task did not reveal a significant CUS/ROSI interaction [F (1,20) = 3.10, p N 0.05, η2 = 0.13], a main effect of CUS [F (1,20) = 0.04, p N 0.05, η2 = 0.001 and d = 0.62] and ROSI [F (1,20) = 0.45, p N 0.05, η2 = 0.01 and d = 0.45] on day 1 of training. On day 2, there was a significant CUS/ROSI interaction [F (1,20) = 14.49, p b 0.01, η2 = 0.24] and a main effect of CUS [F (1,20) = 23.57, p b 0.001, η2 = 0.40 and d = 4.0] and ROSI [F (1,20) = 0.27, p N 0.05, η2 = 0.004 and d = 1.20]. On day 3, there was a significant CUS/ROSI interaction [F (1,20) = 27.3, p b 0.001, η2 = 0.36] and a main effect of CUS [F (1,20) = 26.57, p b 0.001, η2 = 0.35 and d = 3.41] and ROSI [F (1,20) = 0.34, p N 0.05, η2 = 0.004 and d = 1.96]. On day 4, there was a significant CUS/ROSI interaction [F (1,20) = 25.77, p b 0.001, η2 = 0.22] and a main effect of CUS [F (1,20) = 36.73, p b 0.001, η2 = 0.31 and d = 3.67] and ROSI [F (1,20) = 32.94, p b 0.001, η2 = 0.28 and d = 3.26]. Post hoc comparisons revealed that there was no significant alteration in the learning pattern between the groups on day 1 of training (day 22). During training trial, CUS significantly increased the escape latency on day 2, day 3 and day 4. Chronic ROSI treatment significantly decreased the escape latency on day 3 and day 4 in stressed animals. Control animals treated with ROSI showed a significant increase in escape latency on day 2 and day 3 but not on day 4 (Fig. 3A). A two-way ANOVA of probe trial test demonstrated a significant CUS/ROSI interaction [F (1,20) = 8.88, p b 0.01, η2 = 0.17] and a main effect CUS [F (1,20) = 8.88, p b 0.01, η2 = 0.17 and d = 2.76] and ROSI [F (1,20) = 13.89, p b 0.01, η2 = 0.26 and d = 3.22]. Post hoc comparisons revealed that the number of crossings across the platform area was significantly decreased in stressed mice. Chronic ROSI treatment significantly increased the number of crossings across the platform area in stressed mice (Fig. 3B).

O

Statistical analysis

228 229

R O

236

226 227

comparisons revealed that CUS significantly increased the duration of 262 immobility, and the administration of ROSI prevented the increase of 263 immobility period caused by CUS (Fig. 2B). 264

D

235

ml/min. Corticosterone was detected at 250 nm using photodiode array detector (Sheikh et al., 2007). Lipid peroxidation level in plasma was measured by the quantification of thiobarbituric acid-reactive substances (TBARS) according to the method of Ohkawa et al. (1979). Plasma NO level was estimated as reported earlier (Patel and Udayabanu, 2014). Briefly, the nitrates were reduced to nitrite by using 2% ammonium molybdate and 4% ferrous ammonium sulphate and quantified by using Griess reagent at 540 nm. Catalase level was measured by the method of Kuhad and Chopra (2007). Briefly, the rate of decomposition of H2O2 was measured spectrophotometrically at 240 nm. The level of total thiol in plasma was determined as per More et al. (2012) using 10 mM DTNB and measured at 412 nm.

E

224 225

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

P

4

Fig. 2. Effect of ROSI on CUS-induced behavioral alterations in FST (A) and TST (B). Data were mean ± SEM values (n = 6). Significant differences: #CTRL vs. CUS; *CUS vs. CUS + ROSI. *p b 0.05, ***p b 0.001. CTRL = control; CUS = chronic unpredictable stress; ROSI = rosiglitazone; FST = forced swim test; TST = tail suspension test.

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295

297 298 299 300 301 302 303 304 305 306 307

310 311 312 313 314 315

319 320 321

5

R O

O

F

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

Fig. 4. Effect of ROSI on CUS-induced alteration in locomotor activity. Data were mean ± SEM values (n = 6). Significant differences: #CTRL vs. CUS. ###p b 0.001. CTRL = control; CUS = chronic unpredictable stress; ROSI = rosiglitazone.

P

1.47]. Post hoc comparisons revealed that the fasting blood glucose level 322 did not change significantly in all groups (Fig. 5A). 323 324 325

Determination of corticosterone level A two-way ANOVA of corticosterone levels in the plasma of mice revealed a significant CUS/ROSI interaction [F (1,12) = 47.4, p b 0.001, η2 = 0.13] and a main effect CUS [F (1,12) = 278.3, p b 0.001, η2 = 0.77 and d = 12.50] and ROSI [F (1,12) = 21.64, p b 0.001, η2 = 0.06 and d = 5.51]. Post hoc comparisons revealed that ROSI significantly reversed the hypercorticosteronemia in mice caused by CUS exposure (Fig. 5C).

345

Determination of serum insulin level A two-way ANOVA of insulin levels in the serum of mice did not demonstrate a significant CUS/ROSI interaction [F (1,12) = 0.40, p N 0.05, η2 = 0.03], a main effect of CUS [F (1,12) = 0.93, p N 0.05, η2 = 0.07 and d = 1.41] and ROSI [F (1,12) = 0.03, p N 0.05, η2 = 0.002 and d = 0.17]. Post hoc comparisons revealed that the serum insulin level did not change significantly in all groups (Fig. 5D).

353 354

U

N C

O R

R

E

C

T

E

D

OGTT A two-way ANOVA of blood glucose levels in the OGTT did not reveal a significant CUS/ROSI interaction [F (1,16) = 1.96, p N 0.05, η2 = 0.09], a main effect of CUS [F (1,16) = 2.15, p N 0.05, η2 = 0.09 and d = 1.14] and ROSI [F (1,16) = 1.45, p N 0.05, η2 = 0.06 and d = 1.03] at 0 h. There was a significant CUS/ROSI interaction at 0.5 h [F (1,16) = 12.65, p b 0.01, η2 = 0.20], 1 h [F (1,16) = 62.04, p b 0.001, η2 = 0.25], 1.5 h [F (1,16) = 10.97, p b 0.01, η2 = 0.19] and 2 h [F (1,16) = 15, p b 0.01, η2 = 0.34]. The main effect of CUS at 0.5 h, 1 h, 1.5 h and 2 h was observed as [F (1,16) = 23.33, p b 0.001, η2 = 0.37 and d = 3.30], [F (1,16) = 73.23, p b 0.001, η2 = 0.29 and d = 5.89], [F (1,16) = 20.45, p b 0.001, η2 = 0.36 and d = 3.19] and [F (1,16) = 8.86, p b 0.01, η2 = 0.20 and d = 2.33], respectively. The main effect of ROSI at 0.5 h, 1 h, 1.5 h and 2 h was observed as [F (1,16) = 9.70, p b 0.01, η2 = 0.15 and d = 3.34], [F (1,16) = 95.49, p b 0.001, η2 = 0.38 and d = 9.45], [F (1,16) = 9.16, p b 0.01, η2 = 0.16 and d = 2.65] and [F (1,16) = 3.14, p N 0.05, η2 = 0.07 and d = 2.03], respectively. Post hoc comparisons revealed that CUS significantly increased the level of blood glucose from 0.5 to 2 h, and chronic ROSI administration significantly reversed the glucose load caused by CUS exposure (Fig. 5B).

Fig. 3. Effect of ROSI on CUS-induced behavioral alterations in Morris water maze task (A), probe trial (number of crossings) (B) and passive avoidance step through task (C). Data were mean ± SEM values (n = 6). Significant differences: #CTRL vs. CUS; *CUS vs. CUS + ROSI; + CTRL vs. CTRL + ROSI. *p b 0.05, **p b 0.01, ***p b 0.001. CTRL = control; CUS = chronic unpredictable stress; ROSI = rosiglitazone.

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344

346 347 348 349 350 351 352

355 356 357 358 359

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

P

R O

O

F

6

369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392

C

E

R R

367 368

O

365 366

C

363 364

Insulin signaling A two-way ANOVA of PPARγ mRNA expression in the hippocampus of mice demonstrated a significant CUS/ROSI interaction [F (1,20) = 5.44, p b 0.05, η2 = 0.14] and a main effect of CUS [F (1,20) = 6.05, p b 0.05, η2 = 0.15 and d = 1.56] and ROSI [F (1,20) = 6.62, p b 0.05, η2 = 0.17 and d = 2.03]. Post hoc comparisons revealed that CUS exposure significantly downregulated the PPARγ expression and the administration of ROSI prevented the downregulation of PPARγ expression in the hippocampus of mice (Fig. 6A). A two-way ANOVA of IR expression in the hippocampus of mice revealed a significant CUS/ROSI interaction [F (1,12) = 9.93, p b 0.01, η2 = 0.17] and a main effect of CUS [F (1,12) = 17.92, p b 0.01, η2 = 0.31 and d = 2.96] and ROSI [F (1,12) = 17.76, p b 0.01, η2 = 0.30 and d = 3.15]. Post hoc comparisons demonstrated that CUS exposure significantly downregulated the IR expression and ROSI administration prevented the downregulation in the expression of IR (Fig. 6B). A two-way ANOVA of insulin like growth factor 1 receptor (ILGF 1r) expression in the hippocampus of mice did not reveal a significant CUS/ ROSI interaction [F (1,12) = 0.87, p N 0.05, η2 = 0.06], a main effect of CUS [F (1,12) = 1.15, p N 0.05, η2 = 0.08 and d = 1.04] and ROSI [F (1,12) = 0.16, p N 0.05, η2 = 0.01 and d = 0.22]. Post hoc comparisons revealed that the hippocampal ILGF 1r expression did not change significantly in all groups (Fig. 6C). A two-way ANOVA of glucagon like peptide 1 (GLP1) expression in the hippocampus of mice did not reveal a significant CUS/ROSI interaction [F (1,12) = 2.04, p N 0.05, η2 = 0.10], a main effect of CUS [F (1,12) = 5.98, p b 0.05, η2 = 0.29 and d = 0.52] and ROSI [F (1,12) = 0.06, p N 0.05, η2 = 0.003 and d = 0.58]. Post hoc comparisons demonstrated that the hippocampal GLP1 expression did not change significantly in all groups (Fig. 6D). A two-way ANOVA of insulin receptor substrate 1 (IRS1) expression in the hippocampus of mice demonstrated a significant CUS/ROSI interaction [F (1,16) = 19.95, p b 0.001, η2 = 0.30] and a main effect of CUS

N

361 362

U

360

T

E

D

Fig. 5. Effect of ROSI on CUS-induced alterations in the level of fasting blood glucose (A), OGTT (B), plasma corticosterone (C) and serum insulin (D). Data were mean ± SEM values (n = 4– 6). Significant differences: #CTRL vs. CUS; *CUS vs. CUS + ROSI. **p b 0.01, ***p b 0.001. CTRL = control; CUS = chronic unpredictable stress; OGTT = oral glucose tolerance test; ROSI = rosiglitazone.

[F (1,16) = 13.15, p b 0.01, η2 = 0.19 and d = 4.51] and ROSI [F (1,16) = 17.28, p b 0.001, η2 = 0.26 and d = 4.66]. Post hoc comparisons demonstrated that CUS exposure significantly downregulated the IRS1 expression and ROSI administration prevented the downregulation in the expression of IRS1 (Fig. 6E). A two-way ANOVA of insulin receptor substrate 2 (IRS2) expression in the hippocampus of mice revealed a significant CUS/ROSI interaction [F (1,12) = 9.80, p b 0.01, η2 = 0.17] and a main effect of CUS [F (1,12) = 17.11, p b 0.01, η2 = 0.29 and d = 3.58] and ROSI [F (1,12) = 18.46, p b 0.01, η2 = 0.32 and d = 4.16]. Post hoc comparisons demonstrated that CUS exposure significantly downregulated the IRS2 expression and chronic ROSI administration significantly reversed the downregulation of hippocampal IRS2 expression in stressed mice (Fig. 6F). A two-way ANOVA of phosphoinositide 3-kinase (PI3K) expression in the hippocampus of mice demonstrated a significant CUS/ROSI interaction [F (1,12) = 18.06, p b 0.01, η2 = 0.11] and a main effect of CUS [F (1,12) = 77.6, p b 0.001, η2 = 0.49 and d = 9.38] and ROSI [F (1,12) = 49.14, p b 0.001, η2 = 0.31 and d = 4.68]. Post hoc comparisons revealed that ROSI significantly reversed the downregulation of PI3K expression on the hippocampus of mice caused by CUS exposure (Fig. 6G). A two-way ANOVA of protein kinase B (PKB) expression in the hippocampus of mice revealed a significant CUS/ROSI interaction [F (1,12) = 6.42, p b 0.05, η2 = 0.20] and a main effect of CUS [F (1,12) = 6.42, p b 0.05, η2 = 0.20 and d = 2.13] and ROSI [F (1,12) = 6.79, p b 0.05, η2 = 0.21 and d = 2.23]. Post hoc comparisons demonstrated that CUS exposure significantly downregulated the PKB expression and ROSI administration prevented the downregulation in the expression of PKB (Fig. 6H). A two-way ANOVA of GLUT4 expression in the hippocampus of mice demonstrated a significant CUS/ROSI interaction [F (1,12) = 6.12, p b 0.05, η2 = 0.17] and a main effect of CUS [F (1,12) = 7.71, p b 0.05, η2 = 0.21 and d = 4.56] and ROSI [F (1,12) = 9.86, p b 0.01,

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

R O

O

F

A two-way ANOVA of insulin induced gene 1 (INSG1) expression in the hippocampus of mice revealed a significant CUS/ROSI interaction [F (1,12) = 19.79, p b 0.001, η2 = 0.46] and a main effect of CUS [F (1,12) = 6.34, p b 0.05, η2 = 0.14 and d = 4.25] and ROSI [F (1,12) = 4.61, p N 0.05, η2 = 0.10 and d = 3.07]. Post hoc comparisons demonstrated that CUS exposure significantly downregulated the INSG1 expression and chronic ROSI administration significantly reversed the downregulation of hippocampal INSG1 expression in stressed mice (Fig. 6J). A two-way ANOVA of MAPK1 expression in the hippocampus of mice revealed a significant CUS/ROSI interaction [F (1,12) = 7.26, p b 0.05, η2 = 0.20] and a main effect of CUS [F (1,12) = 10.16, p b 0.01, η2 = 0.28 and d = 5.68] and ROSI [F (1,12) = 6.06, p b 0.05, η2 = 0.17 and d = 2.40]. Post hoc comparisons demonstrated that CUS exposure significantly downregulated the MAPK1 expression and chronic ROSI administration significantly reversed the downregulation of hippocampal MAPK1 expression in stressed mice (Fig. 6K).

O R

R

E

C

T

E

D

P

GLUT4 translocation A two-way ANOVA of GLUT4 protein expression in the hippocampus of mice revealed a significant CUS/ROSI interaction [F (1,12) = 220.6, p b 0.001, η2 = 0.35] and a main effect of CUS [F (1,12) = 103.4, p b 0.001, η2 = 0.16 and d = 30.12] and ROSI [F (1,12) = 294.2, p b 0.001, η2 = 0.46 and d = 14.23]. Post hoc comparisons demonstrated that GLUT4 membrane protein was significantly downregulated in the hippocampus of chronically stressed mice and chronic ROSI treatment significantly reversed the downregulation of GLUT4 membrane protein in stressed mice (Fig. 7).

N C

428

η2 = 0.27 and d = 4.92]. Post hoc comparisons revealed that ROSI significantly reversed the downregulation of GLUT4 mRNA expression on the hippocampus of mice caused by CUS exposure (Fig. 6I).

U

426 427

7

Apoptosis and neuronal survival A two-way ANOVA of B-cell lymphoma 2 (BCL2) expression in the hippocampus of mice revealed a significant CUS/ROSI interaction [F (1,12) = 7.62, p b 0.05, η2 = 0.19] and a main effect of CUS [F (1,12) = 13.77, p b 0.01, η2 = 0.36 and d = 3.70] and ROSI [F (1,12) = 4.83, p b 0.05, η2 = 0.12 and d = 3.16]. Post hoc comparisons demonstrated that CUS exposure significantly downregulated the BCL2 expression and ROSI administration prevented the downregulation in the expression of BCL2 (Fig. 8A). A two-way ANOVA of apoptosis inhibitory protein 2 (AIP2) expression in the hippocampus of mice did not demonstrate a significant CUS/ROSI interaction [F (1,12) = 0.99, p N 0.05, η2 = 0.01], a main effect of CUS [F (1,12) = 50.43, p b 0.001, η2 = 0.74 and d = 2.73] and ROSI [F (1,12) = 3.85, p N 0.05, η2 = 0.05 and d = 1.32]. Post hoc comparisons demonstrated that CUS exposure significantly downregulated the AIP2 expression and ROSI administration did not modulate the expression of AIP2 in stressed mice (Fig. 8B). A two-way ANOVA of autophagy5 expression in the hippocampus of mice did not reveal a significant CUS/ROSI interaction [F (1,12) = 1.98, p N 0.05, η2 = 0.06], a main effect of CUS [F (1,12) = 17.98, p b 0.01, η2 = 0.55 and d = 4.35] and ROSI [F (1,12) = 0.61, p N 0.05, η2 = 0.01 and d = 1.50]. Post hoc comparisons revealed that ROSI administration did not reverse the downregulation of autophagy5 expression on the hippocampus of mice caused by CUS exposure (Fig. 8C). A two-way ANOVA of autophagy7 expression in the hippocampus of mice did not demonstrate a significant CUS/ROSI interaction [F

Fig. 6. Effect of ROSI on CUS-induced alterations in the mRNA expression of hippocampal PPARγ (A), IR (B), ILGF 1r (C), GLP1 (D), IRS1 (E), IRS2 (F), PI3K (G), PKB (H), GLUT4 (I), INSG1 (J) and MAPK1 (K). Data were mean ± SEM values (n = 4). Significant differences: # CTRL vs. CUS; *CUS vs. CUS + ROSI. *p b 0.05, **p b 0.01, ***p b 0.001. CTRL = control; CUS = chronic unpredictable stress; ROSI = rosiglitazone; PPARγ = peroxisome proliferator activated receptor gamma; IR = insulin receptor; ILGF 1r = insulin like growth factor 1 receptor; GLP1 = glucagon like peptide 1; IRS1 = insulin receptor substrate 1; IRS2 = insulin receptor substrate 2; PI3K = phosphoinositide 3-kinase; PKB = protein kinase B; GLUT4 = glucose transporter type 4; INSG1 = insulin induced gene 1; MAPK1 = mitogen activated protein kinase 1.

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481

8

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

a main effect of CUS [F (1,20) = 11.89, p b 0.01, η2 = 0.25 and d = 3.41] and ROSI [F (1,20) = 8.56, p b 0.01, η2 = 0.18 and d = 1.83]. Post hoc comparisons revealed that ROSI significantly reversed the increase of TBARS level on the plasma of mice caused by CUS exposure (Fig. 9A).

492 493

NO

496 497

F

A two-way ANOVA of NO levels in the plasma of mice revealed a significant CUS/ROSI interaction [F (1,20) = 12.92, p b 0.01, η2 = 0.13] and a main effect of CUS [F (1,20) = 55.04, p b 0.001, η2 = 0.58 and d = 4.37] and ROSI [F (1,20) = 5.50, p b 0.05, η2 = 0.05 and d = 2.77]. Post hoc comparisons revealed that ROSI significantly reversed the increase of NO level on the plasma of mice caused by CUS exposure (Fig. 9B).

Oxidative stress and inflammation

489

TBARS A two-way ANOVA of TBARS levels in the plasma of mice revealed a significant CUS/ROSI interaction [F (1,20) = 5.6, p b 0.05, η2 = 0.12] and

C

E

R R

O C N U

490 491

Fig. 8. Effect of ROSI on CUS-induced alterations in the mRNA expression of hippocampal BCL2 (A), AIP2 (B), autophagy5 (C) and autophagy7 (D). Data were mean ± SEM values (n = 4). Significant differences: #CTRL vs. CUS; *CUS vs. CUS + ROSI. *p b 0.05, **p b 0.01, ***p b 0.001. CTRL = control; CUS = chronic unpredictable stress; ROSI = rosiglitazone; BCL2 = B-cell lymphoma 2; AIP2 = apoptosis inhibitory protein 2.

502 503 504 505 506 507 508 509 510 511 512

Discussion

522

Chronic stress often precipitates depression and affects the normal physiological state of body, interfering with the emotional, cognitive and physical aspects of health. Clinically, depression has been reported to induce neurocognitive dysfunctions (Shimizu et al., 2013). Chronic stress has been reported to induce disruption of spatial and associative memory (Bisaz et al., 2011; Gumuslu et al., 2013) as well as motor function (Strekalova et al., 2004). In the present study, exposure of mice to CUS resulted in increased duration of immobility in FST and TST, depicting depressive like behavior. We observed that CUS increased the escape latency in MWM task, a reliable index of spatial memory dysfunction. In addition, CUS lead to associative memory dysfunction in mice as evident from decreased STL in PA task. The depressive like behavior in stressed mice was attenuated by ROSI administration. ROSI treatment significantly reversed spatial and associative memory dysfunction associated with depressive like behavior. ROSI is known to induce neurogenesis and attenuate depressive like behavior (Cheng et al., 2015) as well as improve learning and memory (O'Reilly and Lynch, 2012; Pipatpiboon et al., 2011). Further, CUS induced hypolocomotion was not altered by chronic ROSI administration. The major neuro-endocrine responses to chronic stress involve the release of glucocorticoids leading to depressogenic-like effect, hyperglycemia and cognitive impairment (Detka et al., 2013; Haynes et al., 2001; Patel and Udayabanu, 2014; Wróbel et al., 2014). CUS exhibited insulin resistance, hypoinsulinemia and hyperglycemia in animal models (Lin et al., 2005; Pan et al., 2013). In the present study, CUS significantly increased the level of plasma corticosterone, impaired glucose tolerance but did not alter the level of fasting blood glucose and serum insulin. It has been reported that fasting glucose level is often normal but with glucose load hyperglycemia occurs depicting insulin resistance during type 2 diabetes (O'Rahilly et al., 1994). Chronic ROSI administration

523

D

488

484 485

500 501

513 514

T

486 487

(1,12) = 0.17, p N 0.05, η2 = 0.002], a main effect of CUS [F (1,12) = 50.65, p b 0.001, η2 = 0.75 and d = 3.80] and ROSI [F (1,12) = 4.07, p N 0.05, η2 = 0.06 and d = 1.18]. Post hoc comparisons revealed that ROSI administration did not reverse the downregulation of autophagy7 expression on the hippocampus of mice caused by CUS exposure (Fig. 8D).

482 483

498 499

Total thiol A two-way ANOVA of total thiol levels in the plasma of mice revealed a significant CUS/ROSI interaction [F (1,20) = 7.97, p b 0.05, η2 = 0.18] and a main effect of CUS [F (1,20) = 10.13, p b 0.01, η2 = 0.23 and d = 2.13] and ROSI [F (1,20) = 4.44, p b 0.05, η2 = 0.10 and d = 1.69]. Post hoc comparisons demonstrated that CUS exposure significantly decreased the total thiol level and chronic ROSI administration significantly reversed the decrease of plasma total thiol in stressed mice (Fig. 9D).

E

Fig. 7. Effect of ROSI on CUS-induced alteration in the level of hippocampal GLUT4 membrane protein. Data were mean ± SEM values (n = 4). Significant differences: #CTRL vs. CUS; *CUS vs. CUS + ROSI. ***p b 0.001. CTRL = control; CUS = chronic unpredictable stress; ROSI = rosiglitazone; GLUT4 = glucose transporter type 4.

P

R O

O

Catalase A two-way ANOVA of catalase levels in the plasma of mice did not demonstrate a significant CUS/ROSI interaction [F (1,20) = 4.33, p N 0.05, η2 = 0.08], a main effect of CUS [F (1,20) = 15.52, p b 0.001, η2 = 0.29 and d = 2.85] and ROSI [F (1,20) = 12.64, p b 0.01, η2 = 0.24 and d = 2.06]. Post hoc comparisons demonstrated that CUS exposure significantly decreased the catalase level and chronic ROSI administration significantly reversed the decrease of plasma catalase in stressed mice (Fig. 9C).

494 495

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

515 516 517 518 519 520 521

524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552

9

T

E

D

P

R O

O

F

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582

E

R

557 558

O R

556

significantly reversed hypercorticosteronemia and impaired glucose tolerance in stressed mice. PPARγ agonist is known to induce insulin sensitizing action (Kavak et al., 2008), improve glucose uptake and reduce hypercorticosteronemia (Yang et al., 2007). Insulin signaling in the hippocampus is involved in synaptic plasticity, energy metabolism, neuroprotection and cognitive function (Datusalia and Sharma, 2014; Mielke and Wang, 2011). Alterations in insulin signaling mechanism have been observed in the hippocampus of depressed mice (Detka et al., 2013; Hölscher, 2011; Patel and Udayabanu, 2014). We observed that, CUS induced depressed mice have impaired glucose tolerance which might be due to hypercorticosteronemia or downregulation of IR. Hypercorticosteronemia or downregulation of IR leads to dysregulation of insulin signaling components like PPARγ, IRS1, IRS2, PI3K, PKB, MAPK1, INSG1 and GLUT4, resulting in impaired memory performance and depressive like behavior (Agrawal et al., 2011; Hölscher, 2011; Patel and Udayabanu, 2014; Qi et al., 2012; Vannucci et al., 1998). CUS induced depression significantly impaired insulin signaling and GLUT4 membrane translocation in the hippocampus, which was revered by ROSI. We did not observe any significant alteration in the levels of ILGF 1r and GLP1 in hippocampus. ROSI treatment increases IR expression via PPARγ gene transcription resulting in improved insulin sensitivity and GLUT4 membrane translocation (Hernandez et al., 2003; Tripathi, 2013). During neuronal activity, IR → Src → MAPK pathway activates gene transcription essential for glucose homeostasis, synapse growth and cell repair (Hölscher, 2011). Induction of INSG1 via MAPK pathway plays a role in cellular glucose homeostasis (Krapivner et al., 2007). Activation of IR induces GLUT4 membrane translocation and suppresses apoptosis through stimulation of IRS1/2 binding to PI3K, and activation of PI3K and Akt/PKB (Hölscher, 2011; Schubert et al.,

N C

554 555

U

553

C

Fig. 9. Effect of ROSI on CUS-induced alterations in TBARS level (A), NO level (B), catalase level (C) and total thiol level (D) in plasma. Data were mean ± SEM values (n = 6). Significant differences: #CTRL vs. CUS; *CUS vs. CUS + ROSI. *p b 0.05, **p b 0.01, ***p b 0.001. CTRL = control; CUS = chronic unpredictable stress; ROSI = rosiglitazone; TBARS = thiobarbituric acid reactive substances; NO = nitric oxide.

2003). Further, GLUT4 membrane translocation in hippocampal neurons is associated with neurocognitive improvement (Piroli et al., 2007). It has been well accepted that, exposure to stress increases the number of apoptotic cells in the hippocampus resulting in cognitive impairment (Lucassen et al., 2001; Rinwa and Kumar, 2014; Zhang et al., 2007), whereas antidepressants upregulate BCl2 expression and improve neural plasticity (Manji et al., 2001). The ability of AIP2 to inhibit caspase-2 indicates that it occupies a unique position in programmed cell death (Cheung et al., 2006). In the present study, CUS significantly downregulated the level of hippocampal BCl2 and AIP2. Chronic ROSI treatment significantly upregulated hippocampal BCL2 but did not modulate AIP2 in stressed mice. Earlier study demonstrated that, ROSI protect the neurons against advanced glycation end products-induced injury via its antiapoptotic property, mediated by PPARγ activation (Wang et al., 2011). In contrast to apoptosis, autophagy has a protective role against the pathogenesis of a number of neurodegenerative diseases. In the present study, ROSI treatment did not modulate the CUS mediated downregulation of autophagy5 and autophagy7 in hippocampus. Dysfunction in autophagy results in neuronal damage accompanied by ubiquitinated protein aggregates, suggesting that basal levels of autophagy are essential for neuronal health (Cherra and Chu, 2008). CUS plays an important role in induction of various clinical disorders by inducing oxidative stress and inflammation (Ahmad et al., 2010; Rinwa and Kumar, 2014). In the present study, CUS resulted in elevation in the level of oxidative and nitrative stress markers as well as depletion of antioxidants such as catalase and total thiol. Chronic treatment with ROSI reversed CUS induced alteration in oxidative and nitrative stress, which is in line with a previous report (García-Bueno et al., 2005). In conclusion, CUS induces depression and diabetic like state associated with cognitive impairment, dysregulation of neuronal insulin

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612

619

Acknowledgement

620

623 624

Authors would like to thank Council of Scientific & Industrial Research for financial assistance as senior research fellowship (09/957 (0002)/2012-EMR-I) and Defence Research and Development Organisation (DLS/81/48222/LSRB-175/FSB/2008) (Ministry of Defence, Govt. of India) for funding.

625

References

626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687

Abbatecola, A.M., Lattanzio, F., Molinari, A.M., Cioffi, M., Mansi, L., Rambaldi, P., DiCioccio, L., Cacciapuoti, F., Canonico, R., Paolisso, G., 2010. Rosiglitazone and cognitive stability in older individuals with type 2 diabetes and mild cognitive impairment. Diabetes Care 33, 1706–1711. Agrawal, R., Tyagi, E., Shukla, R., Nath, C., 2011. Insulin receptor signaling in rat hippocampus: a study in STZ (ICV) induced memory deficit model. Eur. Neuropsychopharmacol. 21, 261–273. Ahmad, A., Rasheed, N., Banu, N., Palit, G., 2010. Alterations in monoamine levels and oxidative systems in frontal cortex, striatum, and hippocampus of the rat brain during chronic unpredictable stress. Stress 13, 356–365. Awara, W.M., El-Sisi, A.E., El-Refaei, M., El-Naa, M.M., El-Desoky, K., 2005. Insulinotropic and anti-inflammatory effects of rosiglitazone in experimental autoimmune diabetes. Rev. Diabet. Stud. 2, 146–156. Bhutani, M.K., Bishnoi, M., Kulkarni, S.K., 2009. Anti-depressant like effect of curcumin and its combination with piperine in unpredictable chronic stress-induced behavioral, biochemical and neurochemical changes. Pharmacol. Biochem. Behav. 92, 39–43. Bisaz, R., Schachner, M., Sandi, C., 2011. Causal evidence for the involvement of the neural cell adhesion molecule, NCAM, in chronic stress-induced cognitive impairments. Hippocampus 21, 56–71. Cheng, Y., Rodriguiz, R., Murthy, S., Senatorov, V., Thouennon, E., Cawley, N.X., Aryal, D.K., Ahn, S., Lecka-Czernik, B., Wetsel, W.C., Loh, Y.P., 2015. Neurotrophic factor-α1 prevents stress-induced depression through enhancement of neurogenesis and is activated by rosiglitazone. Mol. Psychiatry 20, 744–754. Cherra, S.J., Chu, C.T., 2008. Autophagy in neuroprotection and neurodegeneration: a question of balance. Future Neurol. 3, 309–323. Cheung, H.H., Kelly, N.L., Liston, P., Korneluk, R.G., 2006. Involvement of caspase-2 and caspase-9 in endoplasmic reticulum stress-induced apoptosis: a role for the IAPs. Exp. Cell Res. 312, 2347–2357. Datusalia, A.K., Sharma, S.S., 2014. Amelioration of diabetes-induced cognitive deficits by GSK-3β inhibition is attributed to modulation of neurotransmitters and neuroinflammation. Mol. Neurobiol. 50, 390–405. Detka, J., Kurek, A., Basta-Kaim, A., Kubera, M., Lasoń, W., Budziszewska, B., 2013. Neuroendocrine link between stress, depression and diabetes. Pharmacol. Rep. 65, 1591–1600. García-Bueno, B., Madrigal, J.L., Lizasoain, I., Moro, M.A., Lorenzo, P., Leza, J.C., 2005. Peroxisome proliferator-activated receptor gamma activation decreases neuroinflammation in brain after stress in rats. Biol. Psychiatry 57, 885–894. Ghaisas, M., Navghare, V., Takawale, A., Zope, V., Tanwar, M., Deshpande, A., 2009. Effect of Tectona grandis Linn. On dexamethasone-induced insulin resistance in mice. J. Ethnopharmacol. 122, 304–307. Gottschalk, W.A., Jiang, H., Tartaglia, N., Feng, L., Figurov, A., Lu, B., 1999. Signaling mechanisms mediating BDNF modulation of synaptic plasticity in the hippocampus. Learn. Mem. 6, 243–256. Greene-Schloesser, D., Payne, V., Peiffer, A.M., Hsu, F.C., Riddle, D.R., Zhao, W., Chan, M.D., Metheny-Barlow, L., Robbins, M.E., 2014. The peroxisomal proliferator-activated receptor (PPAR) α agonist, fenofibrate, prevents fractionated whole-brain irradiationinduced cognitive impairment. Radiat. Res. 181, 33–44. Grillo, C., Piroli, G., Hendry, R., Reagan, L., 2009. Insulin-stimulated translocation of GLUT4 to the plasma membrane in rat hippocampus is PI3-kinase dependent. Brain Res. 1296, 35–45. Grillo, C.A., Piroli, G.G., Kaigler, K.F., Wilson, S.P., Wilson, M.A., Reagan, L.P., 2011. Downregulation of hypothalamic insulin receptor expression elicits depressive-like behaviors in rats. Behav. Brain Res. 222, 230–235. Gumuslu, E., Mutlu, O., Sunnetci, D., Ulak, G., Celikyurt, I.K., Cine, N., Akar, F., 2013. The effects of tianeptine, olanzapine and fluoxetine on the cognitive behaviors of unpredictable chronic mild stress-exposed mice. Drug Res. (Stuttg) 63, 532–539. Hanley, A.J., Zinman, B., Sheridan, P., Yusuf, S., Gerstein, H.C., 2010. Effect of rosiglitazone and ramipril on β-cell function in people with impaired glucose tolerance or impaired fasting glucose: the DREAM trial. Diabetes Care 33, 608–613. Hardman, J., Limberd, L., 2001. Goodman and Gilman's: The Pharmacological Basis of Therapeutics. tenth ed. McGraw-Hill, New York.

R O

P

D

T

C

E

R R

O

C

N

621 622

U

615 616

Haynes, L., Griffiths, M., Hyde, R., Barber, D., Mitchell, I., 2001. Dexamethasone induces limited apoptosis and extensive sublethal damage to specific subregions of the striatum and hippocampus: implications for mood disorders. Neuroscience 104, 57–69. Hernandez, R., Teruel, T., Lorenzo, M., 2003. Rosiglitazone produces insulin sensitisation by increasing expression of the insulin receptor and its tyrosine kinase activity in brown adipocytes. Diabetologia 46, 1618–1628. Hölscher, C., 2011. Diabetes as a risk factor for Alzheimer's disease: insulin signalling impairment in the brain as an alternative model of Alzheimer's disease. Biochem. Soc. Trans. 39, 891–897. Hsu, B.S., Fu, S.H., Ku, K.W., Yang, T.Y., Juang, J.H., Chen, S.T., 2005. Enhancing islet engraftment with rosiglitazone. Transplant. Proc. 37, 245–247. Jatwa, R., Parmar, H.S., Panda, S., Kar, A., 2007. Amelioration of corticosteroid-induced type 2 diabetes mellitus by rosiglitazone is possibly mediated through stimulation of thyroid function and inhibition of tissue lipid peroxidation in mice. Basic Clin. Pharmacol. Toxicol. 101, 177–180. Katz, R.J., 1981. Animal models and human depressive disorders. Neurosci. Biobehav. Rev. 5, 231–246. Katz, R.J., 1982. Animal model of depression: pharmacological sensitivity of a hedonic deficit. Pharmacol. Biochem. Behav. 16, 965–968. Kavak, S., Ayaz, L., Emre, M., Inal, T., Tamer, L., Günay, I., 2008. The effects of rosiglitazone on oxidative stress and lipid profile in left ventricular muscles of diabetic rats. Cell Biochem. Funct. 26, 478–485. Kessler, R.C., Price, R.H., Wortman, C.B., 1985. Social factors in psychopathology: stress, social support, and coping processes. Annu. Rev. Psychol. 36, 531–572. Krapivner, S., Chernogubova, E., Ericsson, M., Ahlbeck-Glader, C., Hamsten, A., van't Hooft, F.M., 2007. Human evidence for the involvement of insulin-induced gene 1 in the regulation of plasma glucose concentration. Diabetologia 50, 94–102. Kuhad, A., Chopra, K., 2007. Curcumin attenuates diabetic encephalopathy in rats: behavioral and biochemical evidences. Eur. J. Pharmacol. 576, 34–42. Kulkarni, S., Mehta, A.K., 1985. Purine nucleoside-mediated immobility in mice: reversal by antidepressants. Psychopharmacology 85, 460–463. Lee, K.S., Park, S.J., Kim, S.R., Min, K.H., Jin, S.M., Lee, H.K., Lee, Y.C., 2006. Modulation of airway remodeling and airway inflammation by peroxisome proliferator-activated receptor γ in a murine model of toluene diisocyanate-induced asthma. J. Immunol. 177, 5248–5257. Lin, Y.H., Liu, A.H., Xu, Y., Tie, L., Yu, H.M., Li, X.J., 2005. Effect of chronic unpredictable mild stress on brain-pancreas relative protein in rat brain and pancreas. Behav. Brain Res. 165, 63–71. Lucassen, P.J., Vollmann-Honsdorf, G.K., Gleisberg, M., Czéh, B., De Kloet, E.R., Fuchs, E., 2001. Chronic psychosocial stress differentially affects apoptosis in hippocampal subregions and cortex of the adult tree shrew. Eur. J. Neurosci. 14, 161–166. Mahesh, R., Kumar, B., Jindal, A., Bhatt, S., Devadoss, T., Pandey, D.K., 2012. Antidepressant-like activity of (4-phenylpiperazin-1-yl)(quinoxalin-2-yl) methanone (4a), a novel 5-HT3 receptor antagonist: an investigation in behaviorbased rodent models of depression. Indian J. Pharmacol. 44, 560–565. Manji, H.K., Drevets, W.C., Charney, D.S., 2001. The cellular neurobiology of depression. Nat. Med. 7, 541–547. Mayat, E., Petralia, R.S., Wang, Y.X., Wenthold, R.J., 1995. Immunoprecipitation, immunoblotting, and immunocytochemistry studies suggest that glutamate receptor delta subunits form novel postsynaptic receptor complexes. J. Neurosci. 15, 2533–2546. Mielke, J.G., Wang, Y.T., 2011. Insulin, synaptic function, and opportunities for neuroprotection. Prog. Mol. Biol. Transl. Sci. 98, 133–186. More, A.S., Kumari, R.R., Gupta, G., Kathirvel, K., Lonare, M.K., Dhayagude, R.S., Kumar, D., Kumar, D., Sharma, A.K., Tandan, S.K., 2012. Effect of S-methylisothiourea in acetaminophen-induced hepatotoxicity in rat. Naunyn Schmiedeberg's Arch. Pharmacol. 385, 1127–1139. Ohkawa, H., Ohishi, N., Yagi, K., 1979. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal. Biochem. 95, 351–358. O'Rahilly, S., Hattersley, A., Vaag, A., Gray, H., 1994. Insulin resistance as the major cause of impaired glucose tolerance: a self-fulfilling prophesy? Lancet 344, 585–589. O'Reilly, J.A., Lynch, M., 2012. Rosiglitazone improves spatial memory and decreases insoluble Aβ1-42 in APP/PS1 mice. J. NeuroImmune Pharmacol. 7, 140–144. Pan, Y., Hong, Y., Zhang, Q.Y., Kong, L.D., 2013. Impaired hypothalamic insulin signaling in CUMS rats: restored by icariin and fluoxetine through inhibiting CRF system. Psychoneuroendocrinology 38, 122–134. Pariante, C.M., Miller, A.H., 2001. Glucocorticoid receptors in major depression: relevance to pathophysiology and treatment. Biol. Psychiatry 49, 391–404. Patel, S.S., Udayabanu, M., 2014. Urtica dioica extract attenuates depressive like behavior and associative memory dysfunction in dexamethasone induced diabetic mice. Metab. Brain Dis. 29, 121–130. Peng, Y.L., Liu, Y.N., Liu, L., Wang, X., Jiang, C.L., Wang, Y.X., 2012. Inducible nitric oxide synthase is involved in the modulation of depressive behaviors induced by unpredictable chronic mild stress. J. Neuroinflammation 9, 75. Pipatpiboon, N., Pratchayasakul, W., Chattipakorn, N., Chattipakorn, S.C., 2011. PPARγ agonist improves neuronal insulin receptor function in hippocampus and brain mitochondria function in rats with insulin resistance induced by long term high-fat diets. Endocrinology 153, 329–338. Piroli, G.G., Grillo, C.A., Reznikov, L.R., Adams, S., McEwen, B.S., Charron, M.J., Reagan, L.P., 2007. Corticosterone impairs insulin-stimulated translocation of GLUT4 in the rat hippocampus. Neuroendocrinology 85, 71–80. Potapenko, E.S., Biancardi, V.C., Zhou, Y., Stern, J.E., 2012. Altered astrocyte glutamate transporter regulation of hypothalamic neurosecretory neurons in heart failure rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 303, R291–R300. Qi, W.W., Zhong, L.Y., Li, X.R., Li, G., Liu, Z.X., Hu, J.F., Chen, N.H., 2012. Hyperglycemia induces the variations of 11β-hydroxysteroid dehydrogenase type 1 and peroxisome

O

617 618

signaling, GLUT4 membrane translocation and autophagy. ROSI administration ameliorated insulin signaling and GLUT4 translocation in hippocampus along with attenuation of oxidative and inflammatory stress. These results suggest that ROSI administration might prove to be effective for neurological disorders associated with depressive like behavior.

E

613 614

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

F

10

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772

S.S. Patel et al. / Hormones and Behavior xxx (2015) xxx–xxx

Strekalova, T., Spanagel, R., Bartsch, D., Henn, F.A., Gass, P., 2004. Stress-induced anhedonia in mice is associated with deficits in forced swimming and exploration. Neuropsychopharmacology 29, 2007–2017. Suwanjang, W., Abramov, A.Y., Govitrapong, P., Chetsawang, B., 2013. Melatonin attenuates dexamethasone toxicity-induced oxidative stress, calpain and caspase activation in human neuroblastoma SH-SY5Y cells. The J. Steroid Biochem. Mol. Biol. 138, 116–122. Tripathi, K.D., 2013. Essentials of Medical Pharmacology. seventh ed. Jaypee Brothers Medical Publishers Pvt. Ltd., New Delhi. Udayabanu, M., Kumaran, D., Katyal, A., 2012. Free chelatable zinc modulates the cholinergic function during hypobaric hypoxia-induced neuronal damage: an in vivo study. Neuroscience 202, 434–445. Vannucci, S.J., Koehler-Stec, E.M., Li, K., Reynolds, T.H., Clark, R., Simpson, I.A., 1998. GLUT4 glucose transporter expression in rodent brain: effect of diabetes. Brain Res. 797, 1–11. Wang, L., Yu, C.J., Liu, W., Cheng, L.Y., Zhang, Y.N., 2011. Rosiglitazone protects neuroblastoma cells against advanced glycation end products-induced injury. Acta Pharmacol. Sin. 32, 991–998. Watari, K., Letamendi, A., Elderkin-Thompson, V., Haroon, E., Miller, J., Darwin, C., Kumar, A., 2006. Cognitive function in adults with type 2 diabetes and major depression. Arch. Clin. Neuropsychol. 21, 787–796. Wróbel, A., Serefko, A., Wlaź, P., Poleszak, E., 2014. The depressogenic-like effect of acute and chronic treatment with dexamethasone and its influence on the activity of antidepressant drugs in the forced swim test in adult mice. Prog. NeuroPsychopharmacol. Biol. Psychiatry 54, 243–248. Yang, G.Z., Gao, X.P., Yan, J.F., Ou, K.Q., Zhao, J., 2007. Mechanism of rosiglitasone to improve glucose-uptake of 3T3-L1 adipocyte with insulin resistance induced by dexamethasone and insulin. Sichuan Da Xue Xue Bao Yi Xue Ban 38, 816–818. Zhang, X., Dong, Y.L., Yang, N., Liu, Y.Y., Gao, R.F., Zuo, P.P., 2007. Effects of ning shen ling granule and dehydroepiandrosterone on cognitive function in mice undergoing chronic mild stress. Chin. J. Integr. Med. 13, 46–49. Zhang, L., Xu, T., Wang, S., Yu, L., Liu, D., Zhan, R., Yu, S.Y., 2012. Curcumin produces antidepressant effects via activating MAPK/ERK-dependent brain-derived neurotrophic factor expression in the amygdala of mice. Behav. Brain Res. 235, 67–72.

N C

O R

R

E

C

T

E

D

P

R O

O

F

proliferator-activated receptor-γ expression in hippocampus and hypothalamus of diabetic rats. Exp. Diabetes Res. 2012, 107130. Rasgon, N.L., Kenna, H.A., Williams, K.E., Powers, B., Wroolie, T., Schatzberg, A.F., 2010. Rosiglitazone add-on in treatment of depressed patients with insulin resistance: a pilot study. Sci. World J. 10, 321–328. Rinwa, P., Kumar, A., 2014. Modulation of nitrergic signalling pathway by American ginseng attenuates chronic unpredictable stress-induced cognitive impairment, neuroinflammation, and biochemical alterations. Naunyn Schmiedeberg's Arch. Pharmacol. 387, 129–141. Rossetti, C., Halfon, O., Boutrel, B., 2014. Controversies about a common etiology for eating and mood disorders. Front. Psychol. 5, 1205. Sadaghiani, M.S., Javadi-Paydar, M., Gharedaghi, M.H., Fard, Y.Y., Dehpour, A.R., 2011. Antidepressant-like effect of pioglitazone in the forced swimming test in mice: the role of PPAR-gamma receptor and nitric oxide pathway. Behav. Brain Res. 224, 336–343. Sarkar, S., Korolchuk, V.I., Renna, M., Imarisio, S., Fleming, A., Williams, A., GarciaArencibia, M., Rose, C., Luo, S., Underwood, B.R., Kroemer, G., O'Kane, C.J., Rubinsztein, D.C., 2011. Complex inhibitory effects of nitric oxide on autophagy. Mol. Cell 43, 19–32. Schubert, M., Brazil, D.P., Burks, D.J., Kushner, J.A., Ye, J., Flint, C.L., Farhang-Fallah, J., Dikkes, P., Warot, X.M., Rio, C., Corfas, G., White, M.F., 2003. Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J. Neurosci. 23, 7084–7092. Sheikh, N., Ahmad, A., Siripurapu, K.B., Kuchibhotla, V.K., Singh, S., Palit, G., 2007. Effect of Bacopa monniera on stress induced changes in plasma corticosterone and brain monoamines in rats. J. Ethnopharmacol. 111, 671–676. Shimizu, Y., Kitagawa, N., Mitsui, N., Fujii, Y., Toyomaki, A., Hashimoto, N., Kako, Y., Tanaka, T., Asakura, S., Kusumi, I., 2013. Neurocognitive impairments and quality of life in unemployed patients with remitted major depressive disorder. Psychiatry Res. 210, 913–918. Sousa, N., Cerqueira, J.J., Almeida, O.F., 2008. Corticosteroid receptors and neuroplasticity. Brain Res. Rev. 57, 561–570. Steru, L., Chermat, R., Thierry, B., Simon, P., 1985. The tail suspension test: a new method for screening antidepressants in mice. Psychopharmacology 85, 367–370.

U

773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806

11

Please cite this article as: Patel, S.S., et al., Depression mediates impaired glucose tolerance and cognitive dysfunction: A neuromodulatory role of rosiglitazone, Horm. Behav. (2015), http://dx.doi.org/10.1016/j.yhbeh.2015.11.010

807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840