You are on page 1of 11

Biomedicine & Pharmacotherapy 140 (2021) 111734

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


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

Original article

Pioglitazone rescues high-fat diet-induced depression-like phenotypes and


hippocampal astrocytic deficits in mice
Ying-Yiu Lam a, 1, Sheng-Feng Tsai b, c, 1, Pei-Chun Chen b, d, Yu-Min Kuo b, c, Yun-Wen Chen a, *
a
Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
b
Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
c
Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
d
Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan

A R T I C L E I N F O A B S T R A C T

Keywords: The prevalence of diabetes is rapidly increasing worldwide and is highly associated with the incidence of
Astrocyte depression. Pioglitazone, a Peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, is widely used
Antidepressant for treating patients with type 2 diabetes. However, whether pioglitazone alleviates metabolic disorder-related
Diabetes
depression and astrocytic deficits remains unclear. Here we showed that 12 weeks of high-fat diet (HFD)
Obesity
Thiazolidinedione
feeding (from 8- to 20-week-old) induced not only obesity and insulin resistance, but also depression-like be­
haviors in mice. Astrocytic activation, a sign closely associated with depression, was also evident in the ventral
hippocampus. Four weeks of pioglitazone (10 or 20 mg/kg, daily, from 20- to 24-week-old) treatment alleviated
the HFD-induced glucose-metabolic dysfunctions, upregulation of ventral hippocampal GFAP, reduction of the
total process lengths and the number of branch points of the ventral hippocampal CA1 GFAP-immunoreactive
astrocytes and depressive phenotypes but had no effect on anxiety-like behaviors or hippocampus-related
learning and memory in mice. These findings suggest that pioglitazone could be a potential therapeutic agent
for metabolic disorders and associated depression.

1. Introduction sclerosis, and depression [9–11]. PPAR-γ agonizts have been hypothe­
sized to influence mood status through inhibiting the expression of in­
Diabetes mellitus is a chronic disease characterized by hyperglyce­ flammatory genes and improving insulin sensitivity. PIO improves
mia with disturbances of carbohydrate, protein, and fat metabolism [1]. learning and memory and reduces neuroinflammation [12,13].
Major depressive disorder (MDD) is a mental illness characterized by at Recently, it has been reported that PIO mediated the exhibition of
least 2 weeks of low mood or anhedonia [2]. MDD displays high rates of anti-depressant-like behaviors in mice [14]. Moreover, clinical studies
comorbidity with diabetes. Patients with MDD have a higher incidence suggest that PIO could be a potential therapy for treating depression
of type 2 diabetes mellitus (T2DM) when compared to the general [15]. However, we still know very little about the mechanisms under­
populations [3,4]. Patients with depression have a 32–41% increased lying the antidepressant-like effects of PIO.
risk of diabetes and 8–15% of diabetic patients are diagnosed with Astrocytes are abundant throughout the brain and involved in many
depressive disorders [5]. Several animal studies have shown that animal fundamental processes, such as synaptic transmission, neurovascular
models of diabetes and/or obesity are associated with an exhibition of coupling, and blood-brain barrier maintenance [16]. Astrocytes also
depression-like and/or anxiety-like behaviors [6–8]. But the patho­ play an important role in the control of neurons activity and the con­
physiological mechanisms underlying the concurrence of T2DM and nections between synapses [17]. Astrocytes modulate both short-term
MDD are unclear. and long-term synaptic plasticity, maintain stability of synaptic scaling
Pioglitazone (PIO) is a thiazolidinedione antidiabetic drug that acts and regulate hippocampal neurotransmission [18]. Reactive astrocytes
as a PPARγ agonist. PPARγ gonists have been known to improve varied show signs of hypertrophy and increase of glial fibrillary acidic protein
neurological disorders, including Alzheimer’s disease, multiple (GFAP) expression under different pathological situations [19].

* Correspondence to: Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
E-mail address: yunwen_chen@mail.ncku.edu.tw (Y.-W. Chen).
1
These authors contributed equally.

https://doi.org/10.1016/j.biopha.2021.111734
Received 15 February 2021; Received in revised form 7 May 2021; Accepted 11 May 2021
Available online 19 May 2021
0753-3322/© 2021 The Author(s). Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

Fig. 1. Details of number of animals used in each experiment. (a) Experimental procedures and number of animals used in each experiment of HFD study. (b)
Experimental procedures and number of animals used in each experiment of HFD-PIO study.

Numerous studies have demonstrated that altered changes in 2. Materials and methods
morphology and number of astrocytes are closely associated with severe
depression [20,21]. Hippocampal astrocyte atrophy was observed in a 2.1. Animals
mouse depression model induced by corticosterone [22]. Previous
studies have demonstrated that metabolic disorders disturb the structure The 8-week-old male C57BL/6N mice were purchased from National
and function of astrocytes in the hypothalamus. Short-term high-fat diet Cheng Kung University Laboratory Animal Center. Under the care of
(HFD) feeding triggers an increase number of astrocytes [23]. Long-term National Cheng Kung University Institutional Animal Care, mice were
HFD feeding induces astrocyte shrinkage and subsequently impairs housed (4–5 per cage) in a temperature- (temperature: 25 ± 2 ◦ C; hu­
GABA reuptake capacity of these astrocytes [24]. In line with these re­ midity: around 60–80%) and light-control environment under a 12:12 h
sults, we recently reported that HFD induces exhibition of light-dark cycle (lights on at 6:00 AM). All experimental procedures of
depression-like behaviors accompanied with an astrocytic activation animal studies were approved by the Institutional Animal Care and Use
and a reduced astrocytic process arborization in the hippocampus of Committees (IACUC) of National Cheng Kung University (IACUC
mice [8]. Nevertheless, whether PIO is able to alleviate the HFD-induced approval number: 104135). Mice randomly assigned to normal diet (ND)
behavioral and cellular abnormalities remains unclear. and high-fat diet (HFD) feeding groups. The ND mice were fed with a
In the present study, we sought to examine the effects of PIO on the standard diet (Cat. #: 5010, LabDiet, St. Louis, MO, USA), and the HFD
metabolic disorder-related depression and the ventral hippocampal mice were fed with a diet containing 60% kcal fat (Cat. #: 58Y1, Test­
astrocytic changes in mice. We fed the mice with a 12-week HFD Diet, St. Louis, MO, USA). PIO (Cat. #: 047903, Matrix Scientific,
regimen to induce metabolic dysregulations. Body weights and systemic Columbia, SC, USA) at doses of 10 mg/kg or 20 mg/kg or vehicle (0.25%
insulin resistance were used as indicators of metabolic impairments. PIO carboxymethylcellulose, CMC; Cat. #: C5678, Sigma-Aldrich, St. Louis,
was daily exposed to mice via oral gavage during 4 weeks after 12-week MO, USA) was administered to ND and HFD mice by oral gavage once
feeding period. The effects of HFD and PIO on the exhibition of daily for 4 weeks (from 20 to 24 weeks old). The mice were grouped
depression-like behaviors and the morphological properties of ventral during experimentation. The experimental timelines could be found in
hippocampal astrocytes were determined. Since the hippocampus is the figures. The details of number of mice used in each experiment were
involved in regulating anxiety-like behaviors and memory processes, we list in Fig. 1.
also examined the HFD and PIO effects on the performances of these
behaviors.

2
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

2.2. Intraperitoneal glucose tolerance test 2.8. Tail suspension test

Intraperitoneal glucose tolerance test (IPGTT) was performed as The tail suspension test (TST) was performed as described [27]. Mice
previously described [25]. Mice were intraperitoneally injected with were suspended 50 cm above the floor of a white plastic chamber. The
40% sterile glucose solution dissolved in saline at a dose of 2 g/kg after a behavior was recorded over a 6–min test period but only the last 4-min
12-h fasting. Blood samples were obtained from tail of mice at 0, 30, 60, data were analyzed.
90, 120 min after glucose injection. Glucose level were examined by
using OneTouch® Ultra test strips and OneTouch® UltraEasy blood
glucose meter (LifeScan, Milpitas, CA, USA). 2.9. Tissue preparations

One day after the conclusion of experiments, the anesthetized mice


2.3. Intraperitoneal insulin tolerance test (pentobarbital, 1 g/kg, i.p.) were perfused from the left ventricle with
chilled saline, and their brains were removed and divided into two
Intraperitoneal insulin tolerance test (IPITT) was performed as pre­ hemispheres used for immunoblotting and immunohistochemistry,
viously described [25]. Mice were intraperitoneally injected with insulin respectively. For immunoblotting, the ventral hippocampus was quickly
(0.75 U/kg) after 4-h fasting. Blood samples were collected from tails of dissected out, frozen in liquid nitrogen, and stored at 80 ◦ C until use. For
mice at 0, 30, 60, 90 and 120 min after insulin injection. Glucose levels immunohistochemistry, the brain hemispheres were fixed in 4% para­
were measured by using OneTouch® Ultra test strips and OneTouch® formaldehyde in 0.1 M phosphate buffer for 2 days at 4 ◦ C. The brain
UltraEasy blood glucose meter (LifeScan). specimens were then dehydrated in graded sucrose solutions (10%, 20%,
30%, and 35%, dissolved in 0.1 M phosphate buffer), and embedded
2.4. Measuring fasting plasma levels of glucose and insulin with frozen section media (Cat. #: 3801480, Leica Biosystems, Wetzlar,
Hessen, Germany). The brains were coronally sliced into 25-μm sections
After a 12-h fasting, the mice were anesthetized with pentobarbital and stored in cryoprotectant at − 20 ◦ C.
(1 g/kg, i.p.) and blood samples were collected from the cardiac punc­
ture with heparinized capillary tubes. Plasma was collected after
2.10. Immunoblotting
centrifuging the blood at 3000g for 10 min. Plasma glucose levels were
determined by using a commercial glucose-oxidase kit (Cat. #: 11538,
The ventral hippocampal tissue specimens of mice were lysed with
BioSystems, Barcelona, Spain). Plasma insulin levels were determined
ice-cold commercial tissue protein extraction reagent (Cat. #: 78510,
by using a commercial mouse insulin ELISA kit (Cat. #: 10-1247-01,
Thermo Fisher Scientific, Waltham, MA, USA) containing complete
Mercodia, Uppsala, Sweden).
protease inhibitor cocktail tablets (Cat. #: 04693132001, Roche, Basel,
Switzerland). Protein concentrations were determined by a Bio-Rad
2.5. Calculating homeostasis model assessment insulin resistance index protein assay. Equal amount of protein lysates was separated by SDS-
PAGE, and then transferred to nitrocellulose membrane (Cat. #:
The homeostasis model assessment insulin resistance (HOMA-IR) S80209, Pall Life Science, Port Washington, NY, USA). Membranes were
index was calculated as followed formula: fasting glucose (mM) × blocked in blocking buffer, and then incubated with rabbit anti-GFAP
fasting insulin (mU/L)/22.5 [26]. antibodies (1:1000, Cat. #: Z0334, Dako-Agilent, Santa Clara, CA,
USA) and mouse anti-β-actin (1:10,000, Cat. #: A5441, Sigma-Aldrich),
2.6. Sucrose preference test washed, and incubated with the corresponding horseradish peroxidase-
conjugated secondary antibodies (Cat. #: 115-035-003 and 111-035-
The sucrose preference test (SPT) was performed as described [27]. 003, Jackson ImmunoResearch, West Grove, PA, USA), and visualized
To avoid the confounding effects resulted from isolation housing and with western blotting luminol reagent (Cat. #: sc-2048, Santa Cruz
switching cagemates, the SPT was performed by the cage. Total 4 cages Biotechnology, Inc.). Bands in the immunoblots were quantified by
of mice (18 mice) were subjected. Two identical water bottles were using ImageQuant LAS 4000 (GE Healthcare, Chicago, IL, USA).
provided to mice for 24 h. One is filled with distilled drinking water and
the other is filled with a palatable 1% sucrose solution. To minimize the
2.11. Immunohistochemistry
side preference, these 2 bottles were switched at 12 h. Consumptions of
sucrose solution and water were examined by measuring the changes in
The ventral hippocampal sections were collected in the range, ste­
weight of fluid consumed. Sucrose preference was defined as the ratio of
reotaxic reference point, from 2.30 mm to 3.88 mm posterior to the
the consumed weight of sucrose solution to the total consumed weight of
Bregma. The collected brain sections were washed with phosphate-
sucrose solution and pure water during 24 h.
buffered saline containing 0.3% Triton X-100 to remove the embed­
ding frozen section media, immersed in 3% H2O2 to abolish endogenous
2.7. Forced swimming test peroxidase activity, and blocked with 3% normal goat serum for 1 h at
room temperature. The free-floating brain sections were stained using
The forced swimming test (FST) was performed as described [28]. rabbit anti-GFAP (1:1000, Cat. #: Z0334, Dako-Agilent) antibody in 4 ◦ C
Mice were placed in a cylindrical transparent tank (height 30 cm, overnight., washed, and then incubated with secondary antibodies (Cat.
diameter 20 cm and depth 15 cm) filled with temperature-controlled #: 111-035-003, Jackson ImmunoResearch) for 2 h at room tempera­
water (23–25 ◦ C). The scoring of active (swimming and climbing) or ture. The brain sections were washed and applied for 3,3´-dia­
passive (immobility) behavior was recorded over a 6-min test period but minobenzidine kit (SK-4100, Vector Laboratories, Burlingame, CA,
only the last 4-min data were analyzed. USA).

3
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

Fig. 2. Effects of HFD on body weight, systemic glucose metabolism and exhibition of depression-like behaviors in mice. (a) Experimental timeline. (b) Quantitative
results of body weight of mice during the feeding period. (c) Quantitative results of body weight of mice after the end of regimen (d) Quantitative results of energy
intake of mice. (e) Alterations of blood glucose levels of mice in the IPGTT were shown in left panel and the quantitative results of area under curves (AUC) were
shown in the right panel. (f) Alterations of blood glucose levels of mice in the IPITT were shown in left panel and the quantitative results of AUC were shown in the
right panel. (g) Quantitative results of fasting plasma levels of glucose in mice. (h) Quantitative results of fasting plasma levels of insulin in mice. (i) Quantitative
results of HOMA-IR index. (j) Quantitative results of SPT. (k) Quantitative results of FST. (l) Quantitative results of TST. *p < 0.05, **p < 0.01, ***p < 0.001,
****p < 0.0001, vs. ND, unpaired Student’s t-test in scatter plots and repeated measure two-way ANOVA in line graphs. Numbers given in the parentheses indicates
the sample sizes. Values represent mean ± S.E.M. from three independent experiments.

4
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

Fig. 3. Effects of PIO on body weight and systemic glucose metabolism in HFD-induce obese mice. (a) Experimental timeline. (b) Quantitative results of body weight
of mice during the feeding period. (c) Quantitative results of body weight of mice after the end of regimen. (d) Quantitative results of energy intake of mice. (e)
Alterations of blood glucose levels of mice in the IPGTT were shown in left panel and the quantitative results of AUC were shown in the right panel. (f) Alterations of
blood glucose levels of mice in the IPITT were shown in left panel and the quantitative results of AUC were shown in the right panel. (g) Quantitative results of fasting
plasma levels of glucose in mice. (h) Quantitative results of fasting plasma levels of insulin in mice. (i) Quantitative results of HOMA-IR index. *p < 0.05, **p < 0.01,
***p < 0.001, ****p < 0.0001, vs. respective ND group. #p < 0.05, ##p < 0.01, ###p < 0.001, ####p < 0.0001, vs. respective CMC group, ordinary two-way
ANOVA in scatter plots and repeated measure two-way ANOVA in line graphs. Numbers given in the parentheses indicates the sample sizes. Values represent
mean ± S.E.M. from three independent experiments.

5
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

Fig. 4. Effects of PIO on the plasma levels of lipids in HFD-induced obese mice. (a) Quantitative results of plasma levels of triglyceride in mice. (b) Quantitative
results of plasma levels of free fatty acid in mice. (c) Quantitative results of plasma levels of LDL/VLDL cholesterol in mice. *p < 0.05, **p < 0.01, ***p < 0.001,
****p < 0.0001, vs. respective ND group. #p < 0.05, ##p < 0.01, ###p < 0.001, ####p < 0.0001vs. respective CMC group, ordinary two-way ANOVA. Values
represent mean ± S.E.M. from three independent experiments.

2.12. Analysis of astrocytic morphology 2.14. Open field test

Photomicrographs were taken using a digital camera connected to a The open field test (OFT) was performed as previously described
computer equipped with Axiovision 4.8 software (Carl Zeiss, Oberko­ [30]. Each mouse was placed in a white plastic box
chen, Germany). To minimize the batch-to-batch variation, each set of (50 cm × 40 cm × 40 cm) for 15 min. The time spent in the central
immunohistochemical experiment was performed at the same time. The area (25% of surface area), the total travel distance and the locomotor
GFAP-immunoreactive (GFAP+) signals were identified by the pixels immobility were measured and analyzed using the EthoVision video
display higher intensity than the background threshold using the ImageJ tracking system (Noldus, Wageningen, GE, Netherlands).
Fiji software. The background intensity threshold was fixed and applied
to all sections. The ImageJ plugin, NeuronJ, was used to analyze the 2.15. Elevated plus maze test
process arbors (Sholl analysis), process lengths and branch points of the
GFAP+ astrocytes located in the CA1 subregion of the ventral hippo­ The elevated plus maze test (EPM) was conducted as previously
campus. Total 25 astrocytes (5 astrocytes/brain section, 5 brain sec­ described [31]. A plus-shaped apparatus that included 2 open and 2
tions) obtained from each mouse were measured. The averaged value of enclosed arms (50 cm × 5 cm × 40 cm) was used in this test. The mice
measured parameters obtained from the 25 selected astrocytes was were first placed in the central zone of the maze with open access to any
presented as one data point. Five mice in each group were analyzed. The arm and allowed to freely explore for 10 min. The frequency and
experimenter who analyzed the morphologies of the hippocampal as­ duration of entering the open arms and enclosed arms within 10 min
trocytes was blinded from the experimental grouping. were recorded by using the EthoVision video tracking system (Noldus).

2.13. Object recognition test


2.16. Statistical analysis
The object recognition test (ORT) was used to determine the
hippocampus-related non-spatial learning and memory in mice as pre­ Total number (N) of observations in each group was indicated in
viously described [29]. Each mouse was placed an open field Figures. All data were plotted and reported as mean ± standard devia­
(40 cm × 50 cm × 50 cm brown plywood box) and allowed free tion. Significance was set at p < 0.05. Student’s t-test was adopted to
exploration for 5 min every day for 3 consecutive days (habituation analyze the data sets with a single factor (HFD effect). The body weight
phase). On the fourth day, the mouse was introduced in the same box and energy intake of mice were analyzed using repeated measured two-
consisting of two identical objects (A1, A2, towers of Lego bricks with way ANOVA. The process arborization of individual astrocytes (Sholl
diameter 8 cm and height 9.5 cm) positioned separately at 10 cm from a analysis) was analyzed using mixed-model two-way ANOVA with
wall for 5 min (acquisition phase). Short-term memory test was per­ concentric rings and HFD as the two main effects. Ordinary two-way
formed after 120 min. The mouse was placed in the same box with one of ANOVAs were used to analyze the results with two factors (HFD and
the old object A1 that had been replaced with a new object B (trans­ PIO). Sidak’s post-hoc test was used to perform multiple comparison
parent glass bottles with diameter 5 cm and height 9.5 cm) for 5 min. analysis after the two-way ANOVAs. Statistical analyses were performed
Long-term memory test was performed after 24 h. The mouse was with GraphPad Prism 6.0 (GraphPad Software, Inc., San Diego, CA).
introduced in the same box with object B that had been replaced with a
new object C (white plastic box 3 cm × 7.5 cm × 8 cm) for 5 min. The 3. Results
time spent to explore each object was recorded. The exploring behavior
was defined as “mice touching the object with the nose or sniffing to­ 3.1. HFD concurrently induces metabolic disorders and exhibition of
ward the object within a distance of 1 cm.” The ratio of “new object depression-like behaviors in mice
exploring time divided by total exploring time” to represent the memory
function. We fed male C57BL/6N mice with HFD with 60% of kcals from fat at
the age of 8 weeks for 12 weeks to generate obesity mouse model
(Fig. 2a), which is a widely used animal model of T2DM, showing insulin

6
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

Fig. 5. Effects of PIO and HFD on exhibition of depression-like behaviors in mice. (a) Quantitative results of SPT. (b) Quantitative results of FST. (c) Quantitative
results of TST. ****p < 0.0001, vs. respective ND group. ###p < 0.001, ####p < 0.0001, vs. respective CMC group, ordinary two-way ANOVA. Values represent
mean ± S.E.M. from three independent experiments.

resistance [32]. We compared the body weight gain and energy intake of 3.3. Pioglitazone improves HFD-induced depressive phenotypes in mice
the ND and HFD mice and found that HFD gradually increased body
weight gain (Fig. 2b). At the end of the 12-week feeding, the body We further evaluated whether PIO improves depression-like pheno­
weights of the HFD group increased 50% more than those of the chow types in mice. Results showed that HFD induced the exhibition of
group (Fig. 2c). There was no significant difference on energy con­ depression-like behaviors in the CMC mice by lowering sucrose con­
sumption of mice fed with HFD when compared with mice fed with sumption in the SPT (Fig. 5a) and increasing the time fractions of
chow (Fig. 2d). We next analyzed the capacity of the mice to break down immobility in the FST (Fig. 5b) and TST (Fig. 5c). A 4-week 20 mg/kg
glucose by IPGTT and examined the glucose-lowering effects of insulin PIO treatment reversed the HFD-induced reduction of sucrose con­
by IPITT. As a result, HFD mice displayed glucose intolerance (Fig. 2e) sumption in the SPT (Fig. 5a) and HFD-induced elevations of time
and impaired insulin sensitivity (Fig. 2f). The HFD feeding induced in­ fractions of immobility in the FST (Fig. 5b), TST (Fig. 5c). Our results
sulin resistance in mice, by increasing the levels of fasting plasma showed that the 20 mg/kg PIO treatment exerts potential therapeutic
glucose (Fig. 2g) and insulin (Fig. 2h), and the HOMA-IR index (Fig. 2i). efficacy on the HFD-induced exhibition of depression-like behaviors in
A growing number of studies indicate an association existing be­ mice.
tween metabolic disorders and depression [33,34]. Hence, we further
evaluated whether these obese mice exhibit depression-like behaviors
3.4. Pioglitazone reverses the HFD-induced astrocyte activation and
by performing SPT, FST, and TST. Results showed that HFD induced
remodeling of astrocytic morphology in the hippocampi of mice
depression-like phenotypes in mice by decreasing sucrose consumption
in SPT (Fig. 2j) and increasing time fractions of immobility in FST
Recently, we demonstrated that HFD induces not only exhibition of
(Fig. 2k) and TST (Fig. 2l). These data indicated that a 12-week HFD
depression-like behaviors but also astrocyte activation and remodeling
induces obesity, systemic insulin resistance, as well as exhibition of
of the astrocytic morphology in the hippocampi of mice [8]. Here, we
depression-like behaviors in mice.
examined the effects of PIO treatment on the HFD-induced astrocytic
alterations. In line with our previous findings [8], HFD upregulated the
3.2. Pioglitazone ameliorates HFD-induced metabolic dysfunctions in
ventral hippocampal GFAP expression in the CMC mice, suggesting HFD
mice
induced astrocyte activation (Fig. 6a and b). PIO (20 mg/kg) abolished
the HFD-induced increase of ventral hippocampal GFAP expression
Next, we examined whether PIO improves HFD-induced impair­
(Fig. 6a and b). Activation of astrocytes is known to be associated with
ments of glucose metabolism. PIO (10 or 20 mg/kg) was treated to mice
morphological changes, including hypertrophy and process remodeling,
during 4 weeks after the 12-week feeding period. The HFD regimen was
so we further examined the effects of PIO on the fine structure of the
continued within the period of 4-week PIO treatment (Fig. 3a). The mice
astrocytes. Results showed that the total process lengths, the number of
administrated with the equal volume of vehicle, CMC, served as control
branch points, and the number of process intersections with concentric
groups. Results showed that both doses of PIO did not affect the body
rings in the Sholl analysis were significantly decreased in the GFAP+
weight gain as well as energy intake in the ND and HFD mice (Fig. 3b–d).
astrocytes located in the ventral hippocampal CA1 of HFD mice of CMC
In the IPGTT, the HFD mice treated with either 10 mg/kg or 20 mg/kg
groups (Fig. 6c–g). HFD-induced process remodeling of the ventral
PIO cleared glucose faster than the HFD mice treated with CMC, indi­
hippocampal CA1 astrocytes were reversed by the PIO treatment
cating PIO improved glucose intolerance in the HFD mice (Fig. 3e)
(Fig. 6c–g), suggesting that PIO increases astrocytic process arborization
Consistent with this, IPITT showed that glucose-lowering effects of in­
in HFD mice.
sulin were improved in the HFD mice treated with 20 mg/kg PIO
compared with the HFD mice supplemented with CMC (Fig. 3f).
Furthermore, 20 mg/kg PIO reduced plasma glucose levels (Fig. 3g), 3.5. Pioglitazone has no effect on the exhibition of anxiety-like behaviors
fasting plasma insulin levels (Fig. 3h), as well as HOMA-IR index and the hippocampus-related learning and memory in mice
(Fig. 3i). Additionally, 20 mg/kg PIO reduced the HFD-induced eleva­
tions of plasma levels of triglyceride (Fig. 4a), free fatty acid (Fig. 4b) Since the hippocampus also mediates anxious phenotypes and
and LDL/VLDL cholesterol (Fig. 4c). Our results showed that 20 mg/kg memory functions, we further assessed the effects of PIO treatment on
PIO treatment improves the HFD-induced impairments of glucose and the performances of these behaviors. The OFT and EPM tests were
lipid metabolism in mice. Therefore, we focused on 20 mg/kg PIO adopted to estimate the exhibition of anxiety-like depression. In the
treatment for the rest of the studies. OFT, both HFD and PIO did not affect the measured parameters,
including the total travel distances, time fraction of immobility, time
spend in the center, number of entries into the center, and travel

7
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

(caption on next page)

8
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

Fig. 6. Effects of PIO and HFD on GFAP expression and astrocytic fine structures in the hippocampus of mice. (a) Representative immunoblotting micrographs of
GFAP in the hippocampus of mice. (b) Quantitative results of GFAP levels in the hippocampus of mice. (c) Representative immunomicrographs of the GFAP+ as­
trocytes in the hippocampus (upper four panels). Representative process tracing graphs of selected GFAP+ astrocyte (red frames in the upper four panels) are shown
in the respective lower four panels. (d) Quantitative results of total process length. (e) Quantitative results of number of branch points. (f) Quantitative results of Sholl
analysis. (g) Quantitative results of total number of intersections obtained from the Sholl analysis. *p < 0.05, **p < 0.01, ****p < 0.0001, vs. respective ND group.
###p < 0.001, ####p < 0.0001, vs. respective CMC group, ordinary two-way ANOVA in scatter plots and mix-model repeated measure two-way ANOVA in line
graphs. Numbers given in the parentheses indicates the sample sizes. Values represent mean ± S.E.M. from three independent experiments. (For interpretation of the
references to color in this figure legend, the reader is referred to the web version of this article.)

distance within the center (Fig. 7a–e). Similarly, in the EPM, HFD and shown), suggesting that PPAR-γ may be involved in the PIO-induced
PIO also exerted no effects on the travel distances and the time spent in antidepressant-like effects. Whether PPAR-γ antagonists abolish the
the center, open arms, and closed arms (Fig. 7f–k). PIO-induced antidepressant effects in the HFD mice deserves further
To determine the effects of HFD and PIO on the performances in the investigations in the future.
hippocampus-related learning and memory task, the ORT was carried Disrupted neuroplasticity is a major cause of hippocampus-related
out. Results revealed that neither HFD nor PIO altered the formations of behavioral impairments, including depression, anxiety and memory
short-term and long-term memory in the ORT (Fig. 7l and m). Taken loss. Astrocytes composed approximately 30% of all glial cells in the
together, our results suggested that both HFD and PIO have no effect on central nervous system support the processes of neuroplasticity. In line
the exhibition of anxiety-like behaviors and the hippocampus-related with our results, it has been reported that HFD induces astrocyte acti­
learning and memory in mice. vation. Upregulated GFAP expression and increased area of GFAP+ as­
trocytes were observed in the cerebellum but not the cerebral cortex
4. Discussion after 16-week HFD feeding [38]. The number of astrocytic processes is
reduced in the hippocampal CA1 region of HFD fed rats [39]. Herein, we
This study highlights the therapeutic effects of PIO on both T2DM demonstrated that PIO could reverse the altered astrocytic morphology
and its concurrent depression. Our findings suggested that PIO rescues remodeling by increasing the total process length and the number of
the HFD-induced depressive-like phenotypes and ventral hippocampal branch points. Hence, it is possible that PIO counteracts the
astrocytic deficits probably via improving the HFD-induced metabolic HFD-induced depression-like phenotypes by correcting the
dysregulations. This conclusion is supported by following evidence: (1) astrocyte-associated deficits in the ventral hippocampus of mice. A
PIO improved HFD-induced metabolic disorders; (2) PIO increased su­ previous study by Zhang et al. reported that shortened processes of
crose preference in the SPT and reduced immobility time in the FST and hypothalamic astrocyte in HFD-fed mice and by moderate-level
TST; (3) PIO decreased HFD-induced GFAP expression in the ventral IKKβ/NFkB upregulation [40]. Zhang et al. revealed that IKKβ/NFkB
hippocampus; (4) PIO restored the process arborizations of the GFAP+ activation resulted in astrocytic process reduction while IKKβ/NFkB
astrocytes in the ventral hippocampal CA1 of HFD mice. inhibition reversed this process defect in obese mice by using genetic
An accumulating body of evidence links T2DM to the development of models, suggesting IKKβ/NFkB pathway is very important for regulation
MDD [33]. However, the mechanisms underlying the interactions be­ of astrocytic process [40]. In this study, we demonstrated that HFD led
tween insulin resistance and neuropsychiatric disorders are still unclear. to hippocampal astrocytic deficits while PIO reversed this process defect
PIO, a widely used insulin sensitizer, has been known to relieve in obese mice. Whether IKKβ/NFkB pathway participate in HFD-induced
depressive symptoms. The improvements in insulin sensitivity and the hippocampal astrocytic deficits, and how? Whether PIO reverses the
antidepressant-like effects were reported among patients with severe altered astrocytic morphology remodeling by inhibition of IKKβ/NFkB
depression receiving PIO therapy [35]. In current study, we showed that pathway? While the answers to these are waiting for further delineation.
long-term HFD feeding induced metabolic disorders accompanied with It’s worth mentioning that the present study employs male mice
depression-like phenotype in obese mice, which concurs with a previous only, whereas in the clinic, T2DM-associated MDD is reported for both
study [8]. We showed that a 12-week HFD feeding induced metabolic males and females with some studies showing a higher prevalence in
disorders in mice, including obesity and systemic insulin resistance, and women [41]. This obviously poses some limitations when concluding on
dysregulated lipid metabolism. Our plasma biochemical examinations the present findings, but it is very interesting and worth of further
showed elevated levels of plasma glucose, total cholesterol and tri­ investigation.
glycerides in the HFD obese mice (Figs. 1f and 4). PIO reversed these
metabolic dysregulations through increasing the peripheral glucose 5. Conclusions
utilization (Figs. 2 and 4). We also found that HFD induced the devel­
opment of depression-like behaviors which was counteracted by PIO In conclusion, herein, we demonstrated that chronic treatment of
treatment. It suggested that the PIO exerts antidepressant effect at least PPARγ agonist, PIO, reverses the HFD-induced metabolic, behavioral,
in part through enhancing insulin sensitization and ameliorating lipid and ventral hippocampal astrocytic impairments in mice. PIO could be a
distribution. promising therapeutic drug for the patients suffering from concurrent
PPAR-γ is present in various brain regions, including the hippo­ metabolic disorders and depression.
campus, frontal cortex, and hypothalamus [36]. PIO is described to cross
the blood-brain barrier and mediate the depressive behaviors through
CRediT authorship contribution statement
PPAR-γ [37]. The activation of PPAR-γ in the nervous system decreases
the expression of inflammatory cytokines, represses glutamate toxicity
Y.Y.L. executed experiments and analyzed the results. S.F.T., P.C.C.
and endoplasmic reticulum stress, and promotes neurogenesis and
and Y.M.K. interpreted the results and edited manuscript. Y.W.C. su­
neural plasticity [37]. Our results showed that PIO significantly
pervised this project, interpreted the results, and prepared the manu­
increased PPAR-γ expression in the hippocampus of mice (data not
script. All authors have read and approved this manuscript.

9
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

Fig. 7. Effects of PIO and HFD on exhibition anxiety-like behaviors and performances in the hippocampus-related memory test. (a) Quantitative results of total travel
distance in OFT. (b) Quantitative results of time fraction of immobility in OFT. (c) Quantitative results of time spent in center in OFT. (d) Quantitative results of
number of entries into center in OFT. (e) Quantitative results of travel distance within center in OFT. (f) Quantitative results of total travel distance in EPM. (g)
Quantitative results of time spent in open arm in EPM. (h) Quantitative results of time spent in center in EPM. (i) Quantitative results of travel distance in open arm in
EPM. (j) Quantitative results of travel distance in center in EPM. (k) Quantitative results of travel distance in closed arm in EPM. (l) Quantitative results of per­
formances of short-term memory in ORT. (m) Quantitative results of performances of long-term memory in ORT. Ordinary two-way ANOVA. OFT: open field test,
EPM: elevated plus maze, ORT: object recognition test. Values represent mean ± S.E.M. from three independent experiments.

10
Y.-Y. Lam et al. Biomedicine & Pharmacotherapy 140 (2021) 111734

Conflict of interest statement randomized double-blind placebo-controlled trial, Neuropsychopharmacology 37


(9) (2012) 2093–2100.
[16] M.V. Sofroniew, H.V. Vinters, Astrocytes: biology and pathology, Acta
The authors have no conflict of interest to declare. Neuropathol. 119 (1) (2010) 7–35.
[17] A. Araque, G. Carmignoto, P.G. Haydon, Dynamic signaling between astrocytes and
Acknowledgements neurons, Annu. Rev. Physiol. 63 (2001) 795–813.
[18] Y. Ota, A.T. Zanetti, R.M. Hallock, The role of astrocytes in the regulation of
synaptic plasticity and memory formation, Neural Plast. 2013 (2013), 185463.
We thank Miss. Yi-Ying Kuo for her skilled technical assistance. This [19] M. Pekny, U. Wilhelmsson, M. Pekna, The dual role of astrocyte activation and
work was supported by Ministry of Science and Technology, Taiwan reactive gliosis, Neurosci. Lett. 565 (2014) 30–38.
[20] J.A. Cobb, K. O’Neill, J. Milner, G.J. Mahajan, T.J. Lawrence, W.L. May WL,
(MOST 106-2320-B-006-058-MY3 and MOST 109-2320-B-006-041- J. Miguel-Hidalgo, G. Rajkowska, C.A. Stockmeier, Density of GFAP-
MY3). immunoreactive astrocytes is decreased in left hippocampi in major depressive
disorder, Neuroscience 316 (2016) 209–220.
[21] M.J. Rubinow, G. Mahajan, W. May, J.C. Overholser, G.J. Jurjus, L. Dieter,
Appendix A. Supporting information N. Herbst, D.C. Steffens, J.J. Miguel-Hidalgo, G. Rajkowska, C.A. Stockmeier,
Basolateral amygdala volume and cell numbers in major depressive disorder: a
Supplementary data associated with this article can be found in the postmortem stereological study, Brain Struct. Funct. 221 (1) (2016) 171–184.
[22] Y. Zhao, Z. Lin, L. Chen, L. Ouyang, L. Gu, F. Chen, Q. Zhang, Hippocampal
online version at doi:10.1016/j.biopha.2021.111734. astrocyte atrophy in a mouse depression model induced by corticosterone is
reversed by fluoxetine instead of benzodiazepine diazepam, Prog.
References Neuropsychopharmacol. Biol. Psychiatry 83 (2018) 99–109.
[23] E. Balland, M.A. Cowley, Short-term high-fat diet increases the presence of
astrocytes in the hypothalamus of C57BL6 mice without altering leptin sensitivity,
[1] J.C. Mbanya, J. Sandow, W. Landgraf, D.R. Owens, Recombinant human insulin in
J. Neuroendocr. 29 (10) (2017).
global diabetes management - focus on clinical efficacy, Eur. Endocrinol. 13 (1)
[24] A. Sharif, V. Prevot, When size matters: how astrocytic processes shape
(2017) 21–25.
metabolism, Cell Metab. 25 (5) (2017) 995–996.
[2] D.S. Hasin, A.L. Sarvet, J.L. Meyers, T.D. Saha, W.J. Ruan, M. Stohl, B.F. Grant,
[25] H.T. Wu, H.Y. Ou, H.C. Hung, Y.C. Su, F.H. Lu, J.S. Wu, Y.C. Yang, C.L. Wu, C.
Epidemiology of adult DSM-5 major depressive disorder and its specifiers in the
J. Chang, A novel hepatokine, HFREP1, plays a crucial role in the development of
United States, JAMA Psychiatry 75 (4) (2018) 336–346.
insulin resistance and type 2 diabetes, Diabetologia 59 (8) (2016) 732–742.
[3] W.W. Eaton, H. Armenian, J. Gallo, L. Pratt, D.E. Ford, Depression and risk for
[26] D.R. Matthews, J.P. Hosker, A.S. Rudenski, B.A. Naylor, D.F. Treacher, R.C. Turner,
onset of type II diabetes. A prospective population-based study, Diabetes Care 19
Homeostasis model assessment: insulin resistance and beta-cell function from
(10) (1996) 1097–1102.
fasting plasma glucose and insulin concentrations in man, Diabetologia 28 (7)
[4] D. Luppens, C. Piette, R.P. Radermecker, G. Scantamburlo, M. Ansseau, W. Pitchot,
(1985) 412–419.
Depression and type 2 diabetes: etiopathogenic analysis of a frequent comorbidity,
[27] T.R. Powell, C. Fernandes, L.C. Schalkwyk, Depression-related behavioral tests,
Rev. Med. Liege 69 (11) (2014) 611–617.
Curr. Protoc. Mouse Biol. 2 (2) (2012) 119–127.
[5] S.V. Badescu, C. Tataru, L. Kobylinska, E.L. Georgescu, D.M. Zahiu, A.M. Zagrean,
[28] A. Can, D.T. Dao, M. Arad, C.E. Terrillion, S.C. Piantadosi, T.D. Gould, The mouse
L. Zăgrean, The association between Diabetes mellitus and depression, J. Med. Life
forced swim test, J. Vis. Exp. (59) (2012), e3638.
9 (2) (2016) 120–125.
[29] T.W. Lin, Y.H. Shih, S.J. Chen, C.H. Lien, C.Y. Chang, T.Y. Huang, S.H. Chen, C.
[6] M. Collin, M.L. Hakansson-Ovesjo, I. Misane, S.O. Ogren, B. Meister, Decreased 5-
J. Jen, Y.M. Kuo, Running exercise delays neurodegeneration in amygdala and
HT transporter mRNA in neurons of the dorsal raphe nucleus and behavioral
hippocampus of Alzheimer’s disease (APP/PS1) transgenic mice, Neurobiol. Learn.
depression in the obese leptin-deficient ob/ob mouse, Brain Res. Mol. Brain Res. 81
Mem. 118 (2015) 189–197.
(1–2) (2000) 51–61.
[30] K.S. Tatem, J.L. Quinn, A. Phadke, Q. Yu, H. Gordish-Dressman, K. Nagaraju K,
[7] J.E. Johansen, C. Broberger, C. Lavebratt, C. Johansson, M.J. Kuhar, T. Hokfelt,
Behavioral and locomotor measurements using an open field activity monitoring
M. Schalling, Hypothalamic CART and serum leptin levels are reduced in the
system for skeletal muscle diseases, J. Vis. Exp. 9 (2014) 51785.
anorectic (anx/anx) mouse, Brain Res. Mol. Brain Res. 84 (1–2) (2000) 97–105.
[31] A.A. Walf, C.A. Frye, The use of the elevated plus maze as an assay of anxiety-
[8] S.F. Tsai, H.T. Wu, P.C. Chen, Y.W. Chen, M. Yu, T.F. Wang, S.Y. Wu, S.F. Tzeng, Y.
related behavior in rodents, Nat. Protoc. 2 (2) (2007) 322–328.
M. Kuo, High-fat diet suppresses the astrocytic process arborization and
[32] K. Srinivasan, P. Ramarao, Animal models in type 2 diabetes research: an overview,
downregulates the glial glutamate transporters in the hippocampus of mice, Brain
Indian J. Med. Res. 125 (3) (2007) 451–472.
Res. 1700 (2018) 66–77.
[33] M.S. Faith, P.E. Matz, M.A. Jorge, Obesity-depression associations in the
[9] S. Sundararajan, J.L. Gamboa, N.A. Victor, E.W. Wanderi, W.D. Lust, G.E. Landreth
population, J. Psychosom. Res. 53 (4) (2002) 935–942.
GE, Peroxisome proliferator-activated receptor-gamma ligands reduce
[34] C. Hryhorczuk, S. Sharma, S.E. Fulton, Metabolic disturbances connecting obesity
inflammation and infarction size in transient focal ischemia, Neuroscience 130 (3)
and depression, Front. Neurosci. 7 (2013) 177.
(2005) 685–696.
[35] D.E. Kemp, M. Schinagle, K. Gao, C. Conroy, S.J. Ganocy, F. Ismail-Beigi, J.
[10] D.L. Feinstein, E. Galea, V. Gavrilyuk, C.F. Brosnan, C.C. Whitacre, L. Dumitrescu-
R. Calabrese, PPAR-γ agonism as a modulator of mood: proof-of-concept for
Ozimek, G.E. Landreth, H.A. Pershadsingh, G. Weinberg, M.T. Heneka, Peroxisome
pioglitazone in bipolar depression, CNS Drugs 28 (6) (2014) 571–581.
proliferator-activated receptor-gamma agonists prevent experimental autoimmune
[36] P.D. Drew, J. Xu, P.D. Storer, J.A. Chavis, M.K. Racke, Peroxisome proliferator-
encephalomyelitis, Ann. Neurol. 51 (6) (2002) 694–702.
activated receptor agonist regulation of glial activation: relevance to CNS
[11] A. Shahsavarian, S. Javadi, S. Jahanabadi, M. Khoshnoodi, J. Shamsaee,
inflammatory disorders, Neurochem. Int. 49 (2) (2006) 183–189.
H. Shafaroodi, S.E. Mehr, A. Dehpour, Antidepressant-like effect of atorvastatin in
[37] P.W. Gold, J. Licinio, M.G. Pavlatou, Pathological parainflammation and
the forced swimming test in mice: the role of PPAR-gamma receptor and nitric
endoplasmic reticulum stress in depression: potential translational targets through
oxide pathway, Eur. J. Pharmacol. 745 (2014) 52–58.
the CNS insulin, klotho and PPAR-gamma systems, Mol. Psychiatry 18 (2) (2013)
[12] M.T. Heneka, M. Sastre, L. Dumitrescu-Ozimek, A. Hanke, I. Dewachter, C. Kuiperi,
154–165.
K. O’Banion, T. Klockgether, F. Van Leuven, G.E. Landreth, Acute treatment with
[38] O. Guillemot-Legris, J. Masquelier, A. Everard, P.D. Cani, M. Alhouayek, G.
the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation
G. Muccioli, High-fat diet feeding differentially affects the development of
and Abeta1-42 levels in APPV717I transgenic mice, Brain 128 (2005) 1442–1453.
inflammation in the central nervous system, J. Neuroinflamm. 13 (1) (2016) 206.
[13] H. Seok, M. Lee, E. Shin, M.R. Yun, Y.H. Lee, J.H. Moon, E. Kim, P.H. Lee, B.
[39] K. Gzielo, M. Kielbinski, J. Ploszaj, K. Janeczko, S.P. Gazdzinski, Z. Setkowicz,
Wan Lee, E.S. Kang, H.C. Lee, B.S. Cha, Low-dose pioglitazone can ameliorate
Long-term consumption of high-fat diet in rats: effects on microglial and astrocytic
learning and memory impairment in a mouse model of dementia by increasing
morphology and neuronal nitric oxide synthase expression, Cell. Mol. Neurobiol.
LRP1 expression in the hippocampus, Sci. Rep. 9 (1) (2019) 4414.
37 (5) (2017) 783–789.
[14] M. Salehi-Sadaghiani, M. Javadi-Paydar, M.H. Gharedaghi, A. Zandieh,
[40] Y. Zhang, J.M. Reichel, C. Han, J.P. Zuniga-Hertz, D. Cai, Astrocytic process
P. Heydarpour, Y. Yousefzadeh-Fard, A.R. Dehpour, NMDA receptor involvement
plasticity and IKKβ/NF-κB in central control of blood glucose, blood pressure, and
in antidepressant-like effect of pioglitazone in the forced swimming test in mice,
body weight, Cell Metab. 25 (5) (2017) 1091–1102.e4.
Psychopharmacology 223 (3) (2012) 345–355.
[41] P.R. Albert, Why is depression more prevalent in women? J. Psychiatry Neurosci.
[15] K. Sepanjnia, A. Modabbernia, M. Ashrafi, M.J. Modabbernia, S. Akhondzadeh,
40 (4) (2015) 219–221.
Pioglitazone adjunctive therapy for moderate-to-severe major depressive disorder:

11

You might also like