Skip to main content
Animal Models and Experimental Medicine logoLink to Animal Models and Experimental Medicine
. 2020 Mar 31;3(1):69–78. doi: 10.1002/ame2.12107

Optimization of food deprivation and sucrose preference test in SD rat model undergoing chronic unpredictable mild stress

Li‐Wen He 1, Li Zeng 1, Na Tian 2, Yi Li 1, Tong He 1, Dong‐Mei Tan 1, Qian Zhang 1, Yi Tan 1,
PMCID: PMC7167236  PMID: 32318662

Abstract

Background

The chronic unpredictable mild stress (CUMS) model has long been considered the best model for exploring the pathophysiological mechanisms underlying depression. However, there are no widely recognised standards for strategies for modeling and for behavioral testing. The present study aimed to optimize the protocols for food deprivation and the sucrose preference test (SPT) for the CUMS model.

Methods

We first evaluated the effects of different long periods of food deprivation on the body weight of Sprague Dawley (SD) rats by testing food deprivation for 24 hours (8:00‐8:00+), food deprivation for 12 hours during the daytime (8:00‐20:00) and food deprivation for 12 hours at night (20:00‐8:00+). Next, we established a SD rat CUMS model with 15 different stimulations, and used body weight measurement, SPT, forced swim test (FST), open field test (OFT) and Morris water maze (MWM) test to verify the success of the modeling. In the SPT, consumption of sucrose and pure water within 1 and 12 hours was measured.

Results

Twelve hours of food deprivation during the daytime (8:00‐20:00) had no effect on body weight, while 12 hours of food deprivation at night (20:00‐8:00+) and 24 hours of food deprivation (8:00‐8:00+) significantly reduced the mean body weight of the SD rats. When SPT was used to verify the successful establishment of the CUMS rat model, sucrose consumption measured within 12 hours was less variable than that measured within 1 hour.

Conclusions

Twelve hours of food deprivation in the daytime (8:00‐20:00) may be considered a mild stimulus for the establishment of a CUMS rat model. Measuring sucrose consumption over 12 hours is recommended for SPT.

Keywords: chronic unpredictable mild stress, forced swim test, Morris water maze, open field test, sucrose preference test, weight body

1. INTRODUCTION

Depression is a chronic heterogeneous mental disease associated with high prevalence, recurrence, and mortality, with serious implications for human physical and mental health. 1 , 2 , 3 , 4 The World Health Organization has predicted that by 2020 it will become the second leading cause of disability worldwide and by 2030 major depressive disorders (MDDs) will represent the main cost in healthcare spending. 5 , 6 Although progress has been made in the research and treatment of depression in recent years, the exact pathogenesis of this disease is still not clear, and experimental results from animal models are not in good agreement with those of clinical trials. 7 Therefore, the establishment of appropriate animal models of depression is of great importance.

Current animal models of depression are mainly divided into three categories: stress models, surgical models, and chemical drug‐induced models. 8 , 9 , 10 The most commonly used animal model is the chronic unpredictable mild stress (CUMS) model, which is the only model which manifests high surface, structural, and predictive validity. 11 , 12 However, the CUMS model has been questioned for its poor repeatability, most likely due to variation in stimulus modes, intensity and frequency. 13 Establishment of the CUMS model generally consists of two stages. The first stage is the exposure of the animals to noxious stimuli such as a tilted cage or food/water deprivation for long periods; and the second stage is the use of behavioral tests to screen the animals exhibiting depression‐like behavior. 14 , 15 The use of food deprivation as one of the stimuli for inducing depression has been questioned in previous reports because it results in significant weight loss independently of other stimuli used in the CUMS model. 16 , 17 One of the behavioral tests, the sucrose preference test (SPT), often differs in its duration, and this can affect the experimental results. Hence, identifying the best time frame for the SPT, that will not impact negatively on the experimental outcome, is very important.

In SPT, the consumption of sucrose and pure water within 1 hour, immediately after 24 hours of food and water deprivation, is calculated. In order to rapidly quench their thirst after 24 hours of food and water deprivation, rats usually consume whatever liquid they come across, regardless of whether it contains sucrose or is pure water. Thus, a 1 hour test may be too short to record any decline in sucrose preference in rats in a depression‐like state.

In this study, we compared the effects of 24 and 12 hours of food deprivation on the body weight of Sprague Dawley (SD) rats. In addition, given the distinctive circadian rhythm of rats, we further analyzed the results of 12 hours of food deprivation during the daytime (8:00‐20:00) and at night (20:00‐8:00+). For the SPT, consumption of sucrose and pure water within 1 and 12 hours was measured.

2. MATERIALS AND METHODS

2.1. Animals and treatment

Twenty adult male SD rats (230‐250 g) were used to investigate the effect of food deprivation on body weight. Another thirty adult male SD rats (230‐250 g) were used to establish a CUMS rat model and to optimize the SPT method. All rats were purchased from the Laboratory Animal Center of Chongqing Medical University (SCXK (Yu) 2018‐0003), housed in a single cage with a 12 hour light/12 hour dark cycle, and fed with food and water ad libitum (SYXK (Yu) 2018‐0003). All animals were housed in the laboratory for 1 week prior to experiments to acclimatize to the environment. Experimental manipulations were approved by the Ethics Committee of Chongqing Medical University.

2.2. CUMS paradigm

The schedule for establishing CUMS was conducted according to previously published methods, 18 but with minor modifications, and is presented in Table 1. The stress stimuli included tilting of the cage, food/water deprivation, a wet cage, crowding, hot water swimming, cold water swimming, no bedding in the cage, restraint, tail clamping, inverted light/dark cycle, hot water in the cage box, strobe lighting, forced swimming, and alternating periods of light and darkness. SD rats in the CUMS group were subjected to stimulation twice a day for 4 weeks. The rats were not exposed to the same stressor for two consecutive days.

TABLE 1.

Chronic unpredictable mild stress protocols conducted in this study

Stressor Details
Food deprivation Rats were subjected to 12 h of food deprivation starting from 8:00 am
Water deprivation Rats were subjected to 12 h of water deprivation starting from 8:00 am
Restraint Rats were individually restrained for 4 h inside plastic cylinders
Crowding Rats were placed in a 29 cm × 18 cm × 16 cm cage, five/cage
Strobe light Rats were subjected to 12 h of strobe light stress starting from 8:00 pm
Wet cage Rats were immersed in 200 mL water in 100 g sawdust bedding for 24 h
Inversion of light/dark cycle Rats were subjected to 24 h of reversed light/dark cycle (8:00 am lights off, 8:00 pm lights on)
No padding Rats were placed in cages without padding for 24 h
Hot water into the cage box Rats were exposed to 40℃ water at 15 cm depth for 10 min
Tilted cage Rats were subjected to cage tilting (45°) along the vertical axis for 24 h
Cold water swimming Rats were placed in a cylindrical clear plastic tank filled with 4℃ water for 5 min
Hot water swimming Rats were placed in a cylindrical clear plastic tank filled with 45℃ water for 5 min
Tail clamping Rats were endured tail pinch 1cm apart from the end of the tail for 1 min
Alternate light and dark Rats were subjected to light on and off every 1 h for 12 h from 8:00 am
Forced swimming Rats were placed in a cylindrical clear plastic tank filled with 20℃ water for 5 min

2.3. Scheme of food deprivation

Twenty SD rats were randomly divided into four groups of five rats each, comprising a normal control group, a 24 hour food deprivation group (8:00‐8:00+), a 12 hour daytime food deprivation group (8:00‐20:00) and 12 hour nighttime food deprivation group (20:00‐8:00+). The rats were made to fast every Tuesday for 5 weeks.

2.4. Body weight measurement

The body weight of each rat was measured with an electronic balance at 9:00 am every Monday. Weight gain was calculated as the ratio of the weekly weight gain to the initial body weight.

2.5. Sucrose preference test

Sucrose preference is defined as follows: sucrose preference percentage (%) = sucrose solution consumption (g)/(sucrose solution consumption [g] + water consumption [g]) × 100%. All rats underwent adaptive training from day 1 to day 4, with two bottles of pure water available on days 1 and 2, two bottles of 1% sucrose on day 3, and one bottle of pure water and one bottle of 1% sucrose on day 4. Next, after 12 hours of food and water deprivation, each rat was given 200 mL of pure water and 200 mL of 1% sucrose solution. The quantities of pure water and sucrose consumed were recorded after 1 hour and again after 12 hours. The SPT manipulation procedure is shown in Figure 1.

FIGURE 1.

FIGURE 1

Time schedule for sucrose preference test. ①, ②, ③, and ④ represent different durations of food and water deprivation and time points at which sucrose preference was performed

2.6. Behavioral experiments

2.6.1. Forced swim test

Rats were put individually in a transparent plexiglass cylinder (20 cm diameter × 50 cm high) filled with water (23‐25℃) to a depth of 30 cm. Immobility time was measured over a period of 5 minutes after 1 minute of adaptable swimming. Rats that floated without swimming to keep their heads above the water were judged to be immobile.

2.6.2. Open field test

After being placed in the center of a 100 cm × 100 cm × 40 cm black square cage, the rats freely explored the environment for 5.5 minutes, with the first 30 seconds used to adapt to the environment. Between each test, the inner wall and the bottom surface of the square box were cleaned with 75% alcohol to eliminate the odor from the previous rat. The movements of the rats were recorded by a camera mounted above the center of the field. Smart 2.0 software was used to analyze the time spent in the center of the open field box and the total distance moved during the 5.5 minute test. Open field test (OFT) was performed on days 0 and 28 to assess the impact of CUMS on locomotor activity.

2.6.3. Morris water maze test

A Morris water maze (MWM) is a black open circular pool with a diameter of 180 cm and a height of 60 cm. The pool was filled with 24 ± 1°C water, and an escape platform was placed 1.5 cm below the surface of the water during training. Black ink was then added to the water and stirred in order to obscure the platform. The pool was divided into four quadrants with the platform being placed in the center of the third quadrant.

Learning trials to test the rats' ability to navigate the maze were conducted over 7 days. On the first day (day 1), rats were dropped from any quadrant into a platformless pool for 60 seconds to adapt to the environment. From days 2 to 6, each rat was placed into the water and expected to find the platform within 60 seconds. When a rat failed to find the hidden platform within 60 seconds, it was guided gently onto the platform and made to stay on it for 15 seconds. Each rat was trained 4 times daily with 30 minute intervals between successive training sessions. On day 7, the platform was removed and each rat was put into the water on the side opposite the original platform quadrant for a free 60 second probe. The escape latency, ie the time taken for the rat to find the platform (days 2‐6), was recorded by a tracking‐system as a measure of spatial learning and memory ability.

2.7. Statistical analysis

Statistical analysis was performed with SPSS18.0 software. Data are presented as mean ± SE. Differences between the control and CUMS groups in body weight, and SPT and MWM data were assessed using repeated measures ANOVA; for other parameters, differences were assessed using a two‐sample Student's t test or a non‐parametric Mann‐Whitney test. All tests were two‐tailed. A P‐value of less than .05 was considered statistically significant. All analyses and graph generation were performed with GraphPad Prism 8.0 software.

3. RESULTS

3.1. The effects of food deprivation and CUMS on body weight in SD rats

As shown in Figure 2A, after 5 weeks, the mean body weight of the 12 hour daytime food deprivation (8:00‐20:00) group of rats was not significantly different from that of the control group (F(1,8) = 0.372, P = .559). However, the body weights of the 12 hour nighttime food deprivation (20:00‐8:00+) group and of the 24 hour food deprivation (8:00‐8:00+) group were significantly lower than the control group (F(1,8) = 6.322, P = .036; F(1,8) = 5.373, P = .049). Therefore, the 12 hour daytime food deprivation (8:00‐20:00) group was selected to establish the CUMS rat model. As shown in Figure 2B, after being exposed to stressors for 4 weeks, the results of repeated measures ANOVA showed that body weight gain in the CUMS group was persistently significantly lower than in the control group (F(1,14) = 70.490, P < .0005). This first became evident by the end of week 2.

FIGURE 2.

FIGURE 2

Effects of food deprivation and chronic unpredictable mild stress (CUMS) on rat body weight. A, Effects of 12 and 24 h of food deprivation on body weight in rats. 12 h of food deprivation in the daytime (8:00‐20:00) had no effect on body weight (P > .05), while 24 and 12 h of food deprivation at night (20:00‐8:00+) for 5 wk significantly decreased body weight (P < .05). B, Effects of CUMS on body weight in rats. Results are presented as the mean ± SEM (n = 8), *P < .05, **P < .01, ***P < .001, CUMS group compared with the control group

3.2. Behaviorial tests

After conducting sucrose preference and OFTs at the baseline stage, two rats with low activity and intolerance to sucrose were eliminated, and the remaining 28 normal rats were randomly divided into two groups: a control group (n = 8) and a CUMS group (n = 20) for the subsequent experiments. After 4 weeks of stress, 8 out of 20 rats with depression‐like tendencies were selected for the final behavioral comparison with the control group (n = 8). The corresponding results are summarized below.

3.2.1. The effect of CUMS on anhedonia in the SD rats

As shown in Figure 3A, when the sucrose preference percentages of normal rats tested after 1 hour or after 12 hours were compared, greater data deviation was seen after 1 hour. This suggested that the accuracy and reproducibility of the sucrose preference percentage value was higher after 12 hours than that after 1 hour. In further tests with the CUMS rat model (Figure 3B), tested weekly from week 0 to week 4, repeated measures ANOVA showed that rats in the CUMS group had significantly lower sucrose preference percentages than the rats in the control group (F(1,14) = 7.583, P = .016).

FIGURE 3.

FIGURE 3

Effect of chronic unpredictable mild stress (CUMS) on the sucrose preference percentage in rats. A, Sucrose preference measured within 1 h (blue) and within 12 h (red). B, Sucrose consumption within 12 h was examined in rats after 4 wk of treatment with CUMS. CUMS exposure decreased the sucrose preference in rats. Results are presented as the mean ± SEM (n = 8), *P < .05, CUMS group compared with the control group

3.2.2. The effect of CUMS on the despair state of SD rats

As shown in Figure 4A, after 4 weeks of stimulation, the immobility time of the CUMS group in the forced swim test (FST) significantly increased compared with the control group (t = 2.151, P = .0494).

FIGURE 4.

FIGURE 4

Forced swim test (FST), open field test (OFT) and Morris water maze (MWM) tests in chronic unpredictable mild stress (CUMS)‐induced rat. A, The immobility time in FST. B, The total distance moved in OFT. C, The central distance percentage in OFT. D, The time to find the platform (indicated as escape latency time) in the MWM test probe trial. CUMS exposure decreased the total distance moved and central zone distance percentage in OFT and increased immobility and escape latency times in FST and MWM, respectively. Results are presented as the mean ± SEM (n = 8), *P < .05, **P < .01, ***P < .001, CUMS group compared with the control group

3.2.3. The effect of CUMS on the locomotor activity of SD rats

After 4 weeks of stimulation, the total distance moved and the central distance percentage in OFT was significantly reduced in the CUMS group compared with the control group (z = −2.209, P = .027; Figure 4B) (t = −3.385, P = .0044; Figure 4C).

3.2.4. The effect of CUMS on learning and memory in SD rats

Figure 4D shows that the escape latency of both groups declined gradually over 5 consecutive days of the MWM test. The CUMS rats seemed to spend slightly more time finding the hidden platform than the control rats, and repeated measures ANOVA confirmed that the rats in the CUMS group had a longer escape latency than the control group (F(1,14) = 6.733, P = .021).

4. DISCUSSION

Research into dynamic changes during depression in human beings is constrained by methodological and ethical issues. Hence, establishing relevant animal models has become a common investigation strategy. 19 The most appropriate depression model of CUMS, first proposed by Willner et al in 1987, is a stress rat/mouse model in which the reward reflex activity is damaged. 20 Obvious anxiety, motility agitation, slow response, decreased learning and memory ability are clinically observed in patients with depression, with each episode lasting for at least two weeks. These symptoms also manifest in the CUMS animal models.

Decreased appetite is one of common symptoms of depressed animals so that weight loss is often used as one of the auxiliary indicators of depression. However, many reports have argued that it may not be a wholly satisfactory indicator, because weight loss could be the consequence of food deprivation alone rather than the combined effect of various stimuli during the CUMS modeling process. Any one of a variety of chronic mild stimuli may contribute, more or less, to weight loss in depressed animals. Out of 91 related reports in the literature (Table 2), 70 employed 24 hours of food deprivation. Our results show that 24 hours (8:00‐8:00+) of food deprivation significantly reduced the body weight of the rats. Furthermore, 11 out of the 91 reports listed in Table 2 recommended 12 hours of food deprivation. However, no specific circadian period (daytime or night) for the 12 hour deprivation period was clearly indicated. In this study, we observed that 12 hours of food deprivation during the daytime (8:00‐20:00) did not affect the weight of the rats, but food deprivation for 12 hours at night (20:00‐8:00+) and for 24 hours significantly decreased their body weight. This is probably due to the distinctive circadian rhythm of the rats which makes them more active, and therefore more likely to eat, at night.

TABLE 2.

Durations of food deprivation from 91 published reports in the past 3 y

In this study, 15 types of mild stressors were selected to induce depression in the SD rats. A classic test for anhedonia (the core symptom of depression) is the SPT. 21 Of the 74 reports in the literature that described methodologies involving SPT, most of the researchers measured sucrose and pure water consumption within 1 hour immediately after 24 hours of food and water deprivation (Table 3). Under normal physiological conditions, the water intake of rats is 10‐12 mL/100 g/d, which means that the quantity water drunk in each hour is small. Rats experiencing 24 hours of food and water deprivation would be in a state of desperate thirst, and so would drink whichever of sucrose solution or pure water they found first. Since rats eat and drink more at night, as mentioned above, measuring the consumption of sucrose and pure water ad libitum between 20:00 and 8:00+ could more accurately reveal the mood of rats. In the CUMS verification protocol, SPT is usually performed immediately after food and water deprivation. In this study, 12 hours of food and water deprivation (8:00‐20:00) were followed by SPT measured over 12 hours from 20:00 to 8:00+.

TABLE 3.

Variation in SPT protocols from 74 reports published in the past 3 y (the default concentration of sucrose is 1%)

Duration of food and water deprivation (h) Duration of exposure to sucrose and pure water after deprivation (h) No. reports
24 1 50 , 51 , 52 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , 60 , 61 , 62 , 63 , 64 , 110 , 116 , 117 , 118 19
24 65 , 66 , 67 , 119 4
3 22 , 68 , 120 , 121 , 122 5
2 69 , 104 2
12 70 1
23 1 71 , 72 , 73 , 74 , 75 , 76 , 77 , 78 , 79 , 80 , 81 , 100 , 123 , 124 14
20 2 111 1
1 82 , 83 2
24 99 1
18 1 6 , 125 , 126 , 127 4
15 2 103 1
12 1 84 , 85 , 112 , 128 , 129 5
3 19 , 86 , 87 3
12 88 , 113 , 114 3
2 89 1
24 90 1
4 91 , 92 , 115 3
6 93 , 94 2
4 1 130 , 131 2

Abbreviation: SPT, sucrose preference test.

The immobility time in the FST is thought to reflect the behavioral despair state of rats, so it is often used to determine whether the rats are depressed. OFT is used to assess locomotor activity and spontaneous exploration in a novel environment. 22 The MWM test, an experiment in which rats or mice are forced to swim and learn to seek hidden platforms in the water, is mainly used to test the learning and memory ability of experimental animals for spatial location and sense of direction. 23 , 24 , 25 In this study, the immobility time of the CUMS group was longer than that of the control group, indicating that CUMS induced a depression‐like state in the rats. Moreover, on OFT, the total distance moved and the percentage of distance moved in the central area were decreased in the CUMS group, indicating that the curiosity and preference for spontaneous activities of the rats were decreased. In addition, the time spent by the CUMS group in finding the platform in the MWM test was longer than the time spent by the control group, indicating that exposure to stressful stimuli damaged the cognitive ability of the rats. Overall, the 15 mild stimuli in the present study induced a behavioral despair state (immobility) as well as a significant reduction in body weight, exploration ability, learning and memory ability and the sucrose preference rate of the rats, which collectively are similar to the anhedonia in clinical patients with MDD. Our results indicate that a rat model of depression was successfully established in the current study.

There are about 20 stressors that are used in the CUMS model. However, this study only focused on food deprivation and SPT since they are frequently questioned or challenged in the literature. Future research will test other CUMS stressors, such as a safe and effective intensity of tail clamping.

5. CONCLUSION

In summary, we suggest that 12 hours of food deprivation during the daytime (8:00‐20:00) is a mild stimulus for the establishment of CUMS rat model, because it did not directly affect body weight, which is considered to be an indicator of the success of the depression animal model. Sucrose consumption over 12 hours is recommended for SPT.

CONFLICTS OF INTEREST

None.

AUTHOR CONTRIBUTIONS

YT and QZ conceived the idea and designed the experiments. LWH, LZ, YL participated in the main experiments. LWH and DMT co‐wrote the main manuscript. NT and QZ analyzed data. TH took care of animals. YT revised the manuscript. All authors read and approved the manuscript.

ACKNOWLEDGMENTS

This work was supported by Social livelihood projects of Chongqing Science and Technology Bureau (cstc2017shms‐zdyfX0048, csts2017shmsA00007).

He LW, Zeng L, Tian N, et al. Optimization of food deprivation and sucrose preference test in SD rat model undergoing chronic unpredictable mild stress. Animal Model Exp Med. 2020;3:69–78. 10.1002/ame2.12107

REFERENCES

  • 1. Ryan PJ, Büchler E, Shabanpoor F, et al. Central relaxin‐3 receptor (RXFP3) activation decreases anxiety‐ and depressive‐like behaviours in the rat. Behav Brain Res. 2013;244:142‐151. [DOI] [PubMed] [Google Scholar]
  • 2. Scully C, Chaudhry SI. Aspects of human disease. 21. Anxiety, stress and depression. Dent Update. 2008;35(3):213. [DOI] [PubMed] [Google Scholar]
  • 3. Kessler RC, Berglund P, Demler O, et al. The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS‐R). JAMA. 2003;289(23):3095‐3105. [DOI] [PubMed] [Google Scholar]
  • 4. Czeh B, Fuchs E, Wiborg O, Simon M. Animal models of major depression and their clinical implications. Prog Neuropsychopharmacol Biol Psychiatry. 2016;64:293‐310. [DOI] [PubMed] [Google Scholar]
  • 5. Zu X, Zhang M, Li W, et al. Antidepressant‐like effect of bacopaside I in mice exposed to chronic unpredictable mild stress by modulating the hypothalamic‐pituitary‐adrenal axis function and activating BDNF signaling pathway. Neurochem Res. 2017;42(11):3233‐3244. [DOI] [PubMed] [Google Scholar]
  • 6. Xia Z, Zhang C, Du Y, et al. The effect of traditional Chinese medicine Zhike‐Houpu herbal pair on depressive behaviors and hippocampal serotonin 1A receptors in rats after chronic unpredictable mild stress. Psychosom Med. 2019;81(1):100‐109. [DOI] [PubMed] [Google Scholar]
  • 7. Agid Y, Buzsáki G, Diamond DM, et al. How can drug discovery for psychiatric disorders be improved? Nat Rev Drug Discov. 2007;6(3):189‐201. [DOI] [PubMed] [Google Scholar]
  • 8. Yin X, Guven N, Dietis N. Stress‐based animal models of depression: do we actually know what we are doing? Brain Res. 2016;1652:30‐42. [DOI] [PubMed] [Google Scholar]
  • 9. Porsolt RD. Animal models of depression: utility for transgenic research. Rev Neurosci. 2000;11(1):53‐58. [DOI] [PubMed] [Google Scholar]
  • 10. Qiao H, Li MX, Xu C, Chen HB, An SC, Ma XM. Dendritic spines in depression: what we learned from animal models. Neural Plast. 2016;2016:8056370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Kato T, Kasahara T, Kubota‐Sakashita M, Kato TM, Nakajima K. Animal models of recurrent or bipolar depression. Neuroscience. 2016;321:189‐196. [DOI] [PubMed] [Google Scholar]
  • 12. Willner P. The validity of animal models of depression. Psychopharmacology. 1984;83(1):1‐16. [DOI] [PubMed] [Google Scholar]
  • 13. Willner P. Chronic mild stress (CMS) revisited: consistency and behavioural‐neurobiological concordance in the effects of CMS. Neuropsychobiology. 2005;52(2):90‐110. [DOI] [PubMed] [Google Scholar]
  • 14. Ayuob NN, Firgany AEL, El‐Mansy AA, Ali S. Can ocimum basilicum relieve chronic unpredictable mild stress‐induced depression in mice? Exp Mol Pathol. 2017;103(2):153‐161. [DOI] [PubMed] [Google Scholar]
  • 15. Banasr M, Duman RS. Glial loss in the prefrontal cortex is sufficient to induce depressive‐like behaviors. Biol Psychiatry. 2008;64(10):863‐870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Willner P. Validity, reliability and utility of the chronic mild stress model of depression: a 10‐year review and evaluation. Psychopharmacology. 1997;134(4):319‐329. [DOI] [PubMed] [Google Scholar]
  • 17. Benelli A, Filaferro M, Bertolini A, Genedani S. Influence of S‐adenosyl‐L‐methionine on chronic mild stress‐induced anhedonia in castrated rats. Br J Pharmacol. 1999;127(3):645‐654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Willner P, Towell A, Sampson D, Sophokleous S, Muscat R. Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology. 1987;93(3):358‐364. [DOI] [PubMed] [Google Scholar]
  • 19. Gupta GL, Fernandes J. Protective effect of Convolvulus pluricaulis against neuroinflammation associated depressive behavior induced by chronic unpredictable mild stress in rat. Biomed Pharmacother. 2019;109:1698‐1708. [DOI] [PubMed] [Google Scholar]
  • 20. Su WJ, Zhang YI, Chen Y, et al. NLRP3 gene knockout blocks NF‐κB and MAPK signaling pathway in CUMS‐induced depression mouse model. Behav Brain Res. 2017;322:1‐8. [DOI] [PubMed] [Google Scholar]
  • 21. Fawcett J, Clark DC, Scheftner WA, Gibbons RD. Assessing anhedonia in psychiatric patients. Arch Gen Psychiatry. 1983;40(1):79‐84. [DOI] [PubMed] [Google Scholar]
  • 22. Zhou M, Wang M, Wang X, et al. Abnormal expression of MicroRNAs induced by chronic unpredictable mild stress in rat hippocampal tissues. Mol Neurobiol. 2018;55(2):917‐935. [DOI] [PubMed] [Google Scholar]
  • 23. Svensson M, Hallin T, Broms J, Ekstrand J, Tingström A. Spatial memory impairment in Morris water maze after electroconvulsive seizures. Acta Neuropsychiatr. 2016;29(1):17‐26. [DOI] [PubMed] [Google Scholar]
  • 24. Dong Z, Bai Y, Wu X, et al. Hippocampal long‐term depression mediates spatial reversal learning in the Morris water maze. Neuropharmacology. 2013;64:65‐73. [DOI] [PubMed] [Google Scholar]
  • 25. Warner TA, Stafford NP, Rompala GR, Van Hoogenstyn AJ, Elgert E, Drugan RC. Intermittent swim stress causes Morris water maze performance deficits in a massed‐learning trial procedure that are exacerbated by reboxetine. Pharmacol Biochem Behav. 2013;113:12‐19. [DOI] [PubMed] [Google Scholar]
  • 26. Cao Y, Li Q. The variation of the 5‐hydroxytryptamine system between chronic unpredictable mild stress rats and chronic fatigue syndrome rats induced by forced treadmill running. NeuroReport. 2017;28(11):630‐637. [DOI] [PubMed] [Google Scholar]
  • 27. Chen B, Li J, Xie Y, et al. Cang‐ai volatile oil improves depressive‐like behaviors and regulates DA and 5‐HT metabolism in the brains of CUMS‐induced rats. J Ethnopharmacol. 2019;244:112088. [DOI] [PubMed] [Google Scholar]
  • 28. Fahim AT, Abd El‐Fattah AA, Sadik NAH, Ali BM. Resveratrol and dimethyl fumarate ameliorate testicular dysfunction caused by chronic unpredictable mild stress‐induced depression in rats. Arch Biochem Biophys. 2019;665:152‐165. [DOI] [PubMed] [Google Scholar]
  • 29. Fu XY, Chen HH, Zhang N, et al. Effects of chronic unpredictable mild stress on ovarian reserve in female rats: feasibility analysis of a rat model of premature ovarian failure. Mol Med Rep. 2018;18(1):532‐540. [DOI] [PubMed] [Google Scholar]
  • 30. Gea LP, Colombo R, Rosa EDD, et al. Anhedonic‐like behavior correlates with IFNgamma serum levels in a two‐hit model of depression. Behav Brain Res. 2019;373:112076. [DOI] [PubMed] [Google Scholar]
  • 31. Li J, Hou L, Wang C, Jia X, Qin X, Wu C. Short term intrarectal administration of sodium propionate induces antidepressant‐like effects in rats exposed to chronic unpredictable mild stress. Front Psychiatry. 2018;9:454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Li X, Wu T, Yu Z, et al. Apocynum venetum leaf extract reverses depressive‐like behaviors in chronically stressed rats by inhibiting oxidative stress and apoptosis. Biomed Pharmacother. 2018;100:394‐406. [DOI] [PubMed] [Google Scholar]
  • 33. Liu L, Zhou X, Zhang Y, et al. Hippocampal metabolic differences implicate distinctions between physical and psychological stress in four rat models of depression. Transl Psychiatry. 2018;8(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Liu X, Zheng X, Du G, Li Z, Qin X. Brain metabonomics study of the antidepressant‐like effect of Xiaoyaosan on the CUMS‐depression rats by 1H NMR analysis. J Ethnopharmacol. 2019;235:141‐154. [DOI] [PubMed] [Google Scholar]
  • 35. Lu J, Shao RH, Jin SY, Hu L, Tu Y, Guo JY. Acupuncture ameliorates inflammatory response in a chronic unpredictable stress rat model of depression. Brain Res Bull. 2017;128:106‐112. [DOI] [PubMed] [Google Scholar]
  • 36. Qiao H, An SC, Xu C, Ma XM. Role of proBDNF and BDNF in dendritic spine plasticity and depressive‐like behaviors induced by an animal model of depression. Brain Res. 2017;1663:29‐37. [DOI] [PubMed] [Google Scholar]
  • 37. Shen J, Qu C, Xu L, Sun H, Zhang J. Resveratrol exerts a protective effect in chronic unpredictable mild stress‐induced depressive‐like behavior: involvement of the AKT/GSK3beta signaling pathway in hippocampus. Psychopharmacology. 2019;236(2):591‐602. [DOI] [PubMed] [Google Scholar]
  • 38. Shen J, Xu L, Qu C, Sun H, Zhang J. Resveratrol prevents cognitive deficits induced by chronic unpredictable mild stress: Sirt1/miR‐134 signalling pathway regulates CREB/BDNF expression in hippocampus in vivo and in vitro. Behav Brain Res. 2018;349:1‐7. [DOI] [PubMed] [Google Scholar]
  • 39. Shen XF, Yuan HB, Wang GQ, Xue H, Liu YF, Zhang CX. Role of DNA hypomethylation in lateral habenular nucleus in the development of depressive‐like behavior in rats. J Affect Disord. 2019;252:373‐381. [DOI] [PubMed] [Google Scholar]
  • 40. Tayyab M, Farheen S, M MMP, Khanam N, Mobarak Hossain M, Shahi MH. Antidepressant and neuroprotective effects of naringenin via sonic hedgehog‐GLI1 cell signaling pathway in a rat model of chronic unpredictable mild stress. Neuromolecular Med. 2019;21(3):250‐261. [DOI] [PubMed] [Google Scholar]
  • 41. Tong J, Zhou Z, Qi W, et al. Antidepressant effect of helicid in chronic unpredictable mild stress model in rats. Int Immunopharmacol. 2019;67:13‐21. [DOI] [PubMed] [Google Scholar]
  • 42. Wei LE, Kan LY, Zeng HY, et al. BDNF/TrkB pathway mediates the antidepressant‐like role of H2S in CUMS‐exposed rats by inhibition of hippocampal ER stress. Neuromolecular Med. 2018;20(2):252‐261. [DOI] [PubMed] [Google Scholar]
  • 43. Xing H, Zhang X, Xing N, Qu H, Zhang K. Uncovering pharmacological mechanisms of Zhi‐Zi‐Hou‐Po decoction in chronic unpredictable mild stress induced rats through pharmacokinetics, monoamine neurotransmitter and neurogenesis. J Ethnopharmacol. 2019;243:112079. [DOI] [PubMed] [Google Scholar]
  • 44. Xu YH, Yu M, Wei H, et al. Fibroblast growth factor 22 is a novel modulator of depression through interleukin‐1beta. CNS Neurosci Ther. 2017;23(11):907‐916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Zhang WY, Wang KY, Li YJ, Li YR, Lu RZ. Chronic stress causes protein kinase C epsilon‐aldehyde dehydrogenase 2 signaling pathway perturbation in the rat hippocampus and prefrontal cortex, but not in the myocardium. Neural Regen Res. 2018;13(7):1225‐1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Zhang WY, Guo YJ, Han WX, et al. Curcumin relieves depressive‐like behaviors via inhibition of the NLRP3 inflammasome and kynurenine pathway in rats suffering from chronic unpredictable mild stress. Int Immunopharmacol. 2019;67:138‐144. [DOI] [PubMed] [Google Scholar]
  • 47. Zhang Y, Yuan S, Pu J, et al. Integrated metabolomics and proteomics analysis of hippocampus in a rat model of depression. Neuroscience. 2018;371:207‐220. [DOI] [PubMed] [Google Scholar]
  • 48. Huang D, Zhang LU, Yang JQ, et al. Evaluation on monoamine neurotransmitters changes in depression rats given with sertraline, meloxicam or/and caffeic acid. Genes Dis. 2018;6(2):167‐175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Kuang WH, Dong ZQ, Tian LT, Li J. IGF‐1 defends against chronic‐stress induced depression in rat models of chronic unpredictable mild stress through the PI3K/Akt/FoxO3a pathway. Kaohsiung J Med Sci. 2018;34(7):370‐376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Chen QI, Luo Y, Kuang S, et al. Cyclooxygenase‐2 signalling pathway in the cortex is involved in the pathophysiological mechanisms in the rat model of depression. Sci Rep. 2017;7(1):488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Dong XZ, Wang DX, Lu YP, Yuan S, Liu P, Hu Y. Antidepressant effects of Kai‐Xin‐San in fluoxetine‐resistant depression rats. Braz J Med Biol Res. 2017;50(10):e6161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Guo X, Qiu W, Liu Y, Zhang Y, Zhao H, Chen J. Effects of refined xiaoyaosan on depressive‐like behaviors in rats with chronic unpredictable mild stress through neurosteroids, their synthesis and metabolic enzymes. Molecules. 2017;22(8):1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Han XM, Qin YJ, Zhu Y, et al. Development of an underivatized LC‐MS/MS method for quantitation of 14 neurotransmitters in rat hippocampus, plasma and urine: application to CUMS induced depression rats. J Pharm Biomed Anal. 2019;174:683‐695. [DOI] [PubMed] [Google Scholar]
  • 54. Li Q, Qu FL, Gao Y, et al. Piper sarmentosum Roxb. produces antidepressant‐like effects in rodents, associated with activation of the CREB‐BDNF‐ERK signaling pathway and reversal of HPA axis hyperactivity. J Ethnopharmacol. 2017;199:9‐19. [DOI] [PubMed] [Google Scholar]
  • 55. Li Y, Zhu X, Ju S, et al. Detection of volume alterations in hippocampal subfields of rats under chronic unpredictable mild stress using 7T MRI: a follow‐up study. J Magn Reson Imaging. 2017;46(5):1456‐1463. [DOI] [PubMed] [Google Scholar]
  • 56. Lu Y, Ho CS, McIntyre RS, Wang W, Ho RC. Agomelatine‐induced modulation of brain‐derived neurotrophic factor (BDNF) in the rat hippocampus. Life Sci. 2018;210:177‐184. [DOI] [PubMed] [Google Scholar]
  • 57. Lu Y, Ho CS, McIntyre RS, Wang W, Ho RC. Effects of vortioxetine and fluoxetine on the level of Brain Derived Neurotrophic Factors (BDNF) in the hippocampus of chronic unpredictable mild stress‐induced depressive rats. Brain Res Bull. 2018;142:1‐7. [DOI] [PubMed] [Google Scholar]
  • 58. Mou Z, Huang Q, Chu SF, et al. Antidepressive effects of ginsenoside Rg1 via regulation of HPA and HPG axis. Biomed Pharmacother. 2017;92:962‐971. [DOI] [PubMed] [Google Scholar]
  • 59. Shen J, Li Y, Qu C, Xu L, Sun H, Zhang J. The enriched environment ameliorates chronic unpredictable mild stress‐induced depressive‐like behaviors and cognitive impairment by activating the SIRT1/miR‐134 signaling pathway in hippocampus. J Affect Disord. 2019;248:81‐90. [DOI] [PubMed] [Google Scholar]
  • 60. Wang R, Wang W, Xu J, Liu D, Jiang H, Pan F. Dynamic effects of early adolescent stress on depressive‐like behaviors and expression of cytokines and JMJD3 in the prefrontal cortex and hippocampus of rats. Front Psychiatry. 2018;9:471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Li XY, Qi WW, Zhang YX, et al. Helicid ameliorates learning and cognitive ability and activities cAMP/PKA/CREB signaling in chronic unpredictable mild stress rats. Biol Pharm Bull. 2019;42(7):1146‐1154. [DOI] [PubMed] [Google Scholar]
  • 62. Yang L, Zheng L, Wan Y, Chen Z, Li P, Wang Y. Metoprolol, N‐Acetylcysteine, and escitalopram prevents chronic unpredictable mild stress‐induced depression by inhibition of endoplasmic reticulum stress. Front Psychiatry. 2018;9:696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Zhang J, Zhang Z, Zhang J, et al. iTRAQ‐based protein profiling in CUMS rats provides insights into hippocampal ribosome lesion and ras protein changes underlying synaptic plasticity in depression. Neural Plast. 2019;2019:7492306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Zhao L, Ren H, Gu S, et al. rTMS ameliorated depressive‐like behaviors by restoring HPA axis balance and prohibiting hippocampal neuron apoptosis in a rat model of depression. Psychiatry Res. 2018;269:126‐133. [DOI] [PubMed] [Google Scholar]
  • 65. Gill M, Kinra M, Rai A, Chamallamudi MR, Kumar N. Evaluation of antidepressant activity of methanolic extract of Saraca asoca bark in a chronic unpredictable mild stress model. NeuroReport. 2018;29(2):134‐140. [DOI] [PubMed] [Google Scholar]
  • 66. Li Y, Yan J, Zhu X, et al. Dilated Virchow‐Robin spaces in the hippocampus impact behaviors and effects of anti‐depressant treatment in model of depressed rats. J Affect Disord. 2017;219:17‐24. [DOI] [PubMed] [Google Scholar]
  • 67. Xiang D, Xiao J, Fu L, et al. DNA methylation of the Tacr2 gene in a CUMS model of depression. Behav Brain Res. 2019;365:103‐109. [DOI] [PubMed] [Google Scholar]
  • 68. Fan C, Song Q, Wang P, Li Y, Yang M, Yu SY. Neuroprotective effects of ginsenoside‐Rg1 against depression‐like behaviors via suppressing glial activation, synaptic deficits, and neuronal apoptosis in rats. Front Immunol. 2018;9:2889. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 69. Chen WG, Zheng JX, Xu X, Hu YM, Ma YM. Hippocampal FXR plays a role in the pathogenesis of depression: a preliminary study based on lentiviral gene modulation. Psychiatry Res. 2018;264:374‐379. [DOI] [PubMed] [Google Scholar]
  • 70. Zhang CY, Zeng MJ, Zhou LP, et al. Baicalin exerts neuroprotective effects via inhibiting activation of GSK3beta/NF‐kappaB/NLRP3 signal pathway in a rat model of depression. Int Immunopharmacol. 2018;64:175‐182. [DOI] [PubMed] [Google Scholar]
  • 71. Du H, Zhao H, Lai X, et al. Metabolic profiles revealed synergistically antidepressant effects of lilies and Rhizoma Anemarrhenae in a rat model of depression. Biomed Chromatogr. 2017;31(7):e3923. [DOI] [PubMed] [Google Scholar]
  • 72. Jiang H, Zhang X, Lu J, et al. Antidepressant‐like effects of acupuncture‐insights from DNA methylation and histone modifications of brain‐derived neurotrophic factor. Front Psychiatry. 2018;9:102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Jiang H, Zhang X, Wang YU, et al. Mechanisms underlying the antidepressant response of acupuncture via PKA/CREB signaling pathway. Neural Plast. 2017;2017:4135164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Ning LN, Zhang T, Chu J, et al. Gender‐related hippocampal proteomics study from young rats after chronic unpredicted mild stress exposure. Mol Neurobiol. 2018;55(1):835‐850. [DOI] [PubMed] [Google Scholar]
  • 75. Shao QY, You F, Zhang YH, et al. CSF miR‐16 expression and its association with miR‐16 and serotonin transporter in the raphe of a rat model of depression. J Affect Disord. 2018;238:609‐614. [DOI] [PubMed] [Google Scholar]
  • 76. Tang M, Floyd S, Cai H, Zhang M, Yang R, Dang R. The status of ω‐3 PUFAs influence chronic unpredicted mild stress‐induced metabolic side effects in rats through INSIG/SREBP pathway. Food Funct. 2019;10(8):4649‐4660. [DOI] [PubMed] [Google Scholar]
  • 77. Xu J, Wang R, Liu Y, Liu D, Jiang H, Pan F. FKBP5 and specific microRNAs via glucocorticoid receptor in the basolateral amygdala involved in the susceptibility to depressive disorder in early adolescent stressed rats. J Psychiatr Res. 2017;95:102‐113. [DOI] [PubMed] [Google Scholar]
  • 78. Xu J, Wang R, Liu Y, et al. Short‐ and long‐term alterations of FKBP5‐GR and specific microRNAs in the prefrontal cortex and hippocampus of male rats induced by adolescent stress contribute to depression susceptibility. Psychoneuroendocrinology. 2019;101:204‐215. [DOI] [PubMed] [Google Scholar]
  • 79. Yang Y, Hu Z, Du X, Davies H, Huo X, Fang M. miR‐16 and fluoxetine both reverse autophagic and apoptotic change in chronic unpredictable mild stress model rats. Front Neurosci. 2017;11:428. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 80. Zhang F, Luo J, Zhu X. Ketamine ameliorates depressive‐like behaviors by tPA‐mediated conversion of proBDNF to mBDNF in the hippocampus of stressed rats. Psychiatry Res. 2018;269:646‐651. [DOI] [PubMed] [Google Scholar]
  • 81. Zhang J, Zhang Z, Zhang J, et al. Electroacupuncture Improves antidepressant effects in CUMS rats by protecting hippocampal synapse and mitochondrion: an ultrastructural and iTRAQ proteomic study. Evid Based Complement Alternat Med. 2019;2019:3424698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Ma H, Wang W, Xu S, Wang L, Wang X. Potassium 2‐(1‐hydroxypentyl)‐benzoate improves depressive‐like behaviors in rat model. Acta Pharm Sin B. 2018;8(6):881‐888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Liu X, Qu C, Yang H, et al. Chronic stimulation of the sigma‐1 receptor ameliorates autonomic nerve dysfunction and atrial fibrillation susceptibility in a rat model of depression. Am J Physiol Heart Circ Physiol. 2018;315(6):H1521‐H1531. [DOI] [PubMed] [Google Scholar]
  • 84. Li W, Zhu Y, Saud SM, et al. Electroacupuncture relieves depression‐like symptoms in rats exposed to chronic unpredictable mild stress by activating ERK signaling pathway. Neurosci Lett. 2017;642:43‐50. [DOI] [PubMed] [Google Scholar]
  • 85. Wu GF, Ren S, Tang RY, et al. Antidepressant effect of taurine in chronic unpredictable mild stress‐induced depressive rats. Sci Rep. 2017;7(1):4989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Fernandes J, Gupta GL. N‐acetylcysteine attenuates neuroinflammation associated depressive behavior induced by chronic unpredictable mild stress in rat. Behav Brain Res. 2019;364:356‐365. [DOI] [PubMed] [Google Scholar]
  • 87. Song Q, Fan C, Wang P, Li Y, Yang M, Yu SY. Hippocampal CA1 βCaMKII mediates neuroinflammatory responses via COX‐2/PGE2 signaling pathways in depression. J Neuroinflammation. 2018;15(1):338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Li H, Wang P, Huang L, Li P, Zhang D. Effects of regulating gut microbiota on the serotonin metabolism in the chronic unpredictable mild stress rat model. Neurogastroenterol Motil. 2019;31(10):e13677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Zhong X, Li G, Qiu F, Huang Z. Paeoniflorin ameliorates chronic stress‐induced depression‐like behaviors and neuronal damages in rats via activation of the ERK‐CREB pathway. Front Psychiatry. 2018;9:772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Gao L, Liu X, Yu L, Wu J, Xu M, Liu Y. Folic acid exerts antidepressant effects by upregulating brain‐derived neurotrophic factor and glutamate receptor 1 expression in brain. NeuroReport. 2017;28(16):1078‐1084. [DOI] [PubMed] [Google Scholar]
  • 91. Zhang Y, Ge JF, Wang FF, Liu F, Shi C, Li N. Crassifoside H improve the depressive‐like behavior of rats under chronic unpredictable mild stress: possible involved mechanisms. Brain Res Bull. 2017;135:77‐84. [DOI] [PubMed] [Google Scholar]
  • 92. Ke Q, Li R, Cai L, Wu SD, Li CM. Ro41‐5253, a selective antagonist of retinoic acid receptor alpha, ameliorates chronic unpredictable mild stress‐induced depressive‐like behaviors in rats: involvement of regulating HPA axis and improving hippocampal neuronal deficits. Brain Res Bull. 2019;146:302‐309. [DOI] [PubMed] [Google Scholar]
  • 93. Han YX, Tao C, Gao XR, et al. BDNF‐related imbalance of copine 6 and synaptic plasticity markers couples with depression‐like behavior and immune activation in CUMS Rats. Front Neurosci. 2018;12:731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Yang XH, Song SQ, Xu Y. Resveratrol ameliorates chronic unpredictable mild stress‐induced depression‐like behavior: involvement of the HPA axis, inflammatory markers, BDNF, and Wnt/beta‐catenin pathway in rats. Neuropsychiatr Dis Treat. 2017;13:2727‐2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Bakhtiarzadeh F, Nahavandi A, Goudarzi M, Shirvalilou S, Rakhshan K, Niknazar S. Axonal transport proteins and depressive like behavior, following Chronic Unpredictable Mild Stress in male rat. Physiol Behav. 2018;194:9‐14. [DOI] [PubMed] [Google Scholar]
  • 96. Jia HM, Yu M, Ma LY, Zhang HW, Zou ZM. Chaihu‐Shu‐Gan‐San regulates phospholipids and bile acid metabolism against hepatic injury induced by chronic unpredictable stress in rat. J Chromatogr B Analyt Technol Biomed Life Sci. 2017;1064:14‐21. [DOI] [PubMed] [Google Scholar]
  • 97. Van Laeken N, Pauwelyn G, Dockx R, et al. Regional alterations of cerebral [18F]FDG metabolism in the chronic unpredictable mild stress‐ and the repeated corticosterone depression model in rats. J Neural Transm. 2018;125(9):1381‐1393. [DOI] [PubMed] [Google Scholar]
  • 98. Yang H, Li W, Meng P, Liu Z, Liu J, Wang Y. Chronic unpredictable mild stress aggravates mood disorder, cognitive impairment, and brain insulin resistance in diabetic rat. Evid Based Complement Alternat Med. 2018;2018:2901863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Xu C, Sun Y, Cai X, et al. Medial habenula‐interpeduncular nucleus circuit contributes to anhedonia‐like behavior in a rat model of depression. Front Behav Neurosci. 2018;12:238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Liu X, Liu C. Baicalin ameliorates chronic unpredictable mild stress‐induced depressive behavior: involving the inhibition of NLRP3 inflammasome activation in rat prefrontal cortex. Int Immunopharmacol. 2017;48:30‐34. [DOI] [PubMed] [Google Scholar]
  • 101. Yu D, Cheng Z, Ali AI, et al. Chronic unexpected mild stress destroys synaptic plasticity of neurons through a glutamate transporter, GLT‐1, of astrocytes in the ischemic stroke rat. Neural Plast. 2019;2019:1615925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Stefanovic B, Spasojevic N, Jovanovic P, Dronjak S. Melatonin treatment affects changes in adrenal gene expression of catecholamine biosynthesizing enzymes and norepinephrine transporter in the rat model of chronic‐stress‐induced depression. Can J Physiol Pharmacol. 2019;97(7):685‐690. [DOI] [PubMed] [Google Scholar]
  • 103. Wang B, Wang Y, Wu Q, Huang HP, Li S. Effects of alpha2A adrenoceptors on norepinephrine secretion from the locus coeruleus during chronic stress‐induced depression. Front Neurosci. 2017;11:243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Yuan Q, Li Y, Deng X, et al. Effects of Xingpi Kaiyu Fang on ATP, Na/K‐ATPase, and respiratory chain complexes of hippocampus and gastrocnemius muscle in depressed rats. Evid Based Complement Alternat Med. 2019;2019:6054926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Hu MZ, Wang AR, Zhao ZY, Chen XY, Li YB, Liu B. Antidepressant‐like effects of paeoniflorin on post‐stroke depression in a rat model. Neurol Res. 2019;41(5):446‐455. [DOI] [PubMed] [Google Scholar]
  • 106. Li JG, Jia XY, Wang C, Wu CX, Qin XM. Altered gut metabolome contributes to depression‐like behaviors in rats exposed to chronic unpredictable mild stress. Transl Psychiatry. 2019;9(1):40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Liu C, Wang J, Xu S, et al. Paecilomyces tenuipes extract prevents depression‐like behaviors in chronic unpredictable mild stress‐induced rat model via modulation of neurotransmitters. Mol Med Rep. 2017;16(2):2172‐2178. [DOI] [PubMed] [Google Scholar]
  • 108. Wang GL, He ZM, Zhu HY, et al. Involvement of serotonergic, noradrenergic and dopaminergic systems in the antidepressant‐like effect of ginsenoside Rb1, a major active ingredient of Panax ginseng C.A. Meyer. J Ethnopharmacol. 2017;204:118‐124. [DOI] [PubMed] [Google Scholar]
  • 109. Wang J, Li X, He S, et al. Regulation of the kynurenine metabolism pathway by Xiaoyao San and the underlying effect in the hippocampus of the depressed rat. J Ethnopharmacol. 2018;214:13‐21. [DOI] [PubMed] [Google Scholar]
  • 110. Shen M, Yang YI, Wu Y, et al. L‐theanine ameliorate depressive‐like behavior in a chronic unpredictable mild stress rat model via modulating the monoamine levels in limbic‐cortical‐striatal‐pallidal‐thalamic‐circuit related brain regions. Phytother Res. 2019;33(2):412‐421. [DOI] [PubMed] [Google Scholar]
  • 111. Wang J, Liu Y, Li L, et al. Dopamine and serotonin contribute to Paecilomyces hepiali against chronic unpredictable mild stress induced depressive behavior in Sprague Dawley rats. Mol Med Rep. 2017;16(4):5675‐5682. [DOI] [PubMed] [Google Scholar]
  • 112. Song J, Wang X, Huang YU, et al. Antidepressant‐like effects of Marasmius androsaceus metabolic exopolysaccharides on chronic unpredictable mild stress‐induced rat model. Mol Med Rep. 2017;16(4):5043‐5049. [DOI] [PubMed] [Google Scholar]
  • 113. Zhang R, Guo L, Ji Z, et al. Radix scutellariae attenuates CUMS‐induced depressive‐like behavior by promoting neurogenesis via cAMP/PKA pathway. Neurochem Res. 2018;43(11):2111‐2120. [DOI] [PubMed] [Google Scholar]
  • 114. Song Y, Sun R, Ji Z, Li X, Fu Q, Ma S. Perilla aldehyde attenuates CUMS‐induced depressive‐like behaviors via regulating TXNIP/TRX/NLRP3 pathway in rats. Life Sci. 2018;206:117‐124. [DOI] [PubMed] [Google Scholar]
  • 115. Li J, Yang R, Xia K, et al. Effects of stress on behavior and resting‐state fMRI in rats and evaluation of Telmisartan therapy in a stress‐induced depression model. BMC Psychiatry. 2018;18(1):337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Hou XY, Hu ZL, Zhang D‐Z, et al. Rapid antidepressant effect of hydrogen sulfide: evidence for activation of mTORC1‐TrkB‐AMPA receptor pathways. Antioxid Redox Signal. 2017;27(8):472‐488. [DOI] [PubMed] [Google Scholar]
  • 117. Zhang C, Zhang YP, Li YY, et al. Minocycline ameliorates depressive behaviors and neuro‐immune dysfunction induced by chronic unpredictable mild stress in the rat. Behav Brain Res. 2019;356:348‐357. [DOI] [PubMed] [Google Scholar]
  • 118. Wei H, Zhou T, Tan B, et al. Impact of chronic unpredicted mild stress‐induced depression on repaglinide fate via glucocorticoid signaling pathway. Oncotarget. 2017;8(27):44351‐44365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Zhu XL, Chen JJ, Han F, et al. Novel antidepressant effects of Paeonol alleviate neuronal injury with concomitant alterations in BDNF, Rac1 and RhoA levels in chronic unpredictable mild stress rats. Psychopharmacology. 2018;235(7):2177‐2191. [DOI] [PubMed] [Google Scholar]
  • 120. Abuelezz SA, Hendawy N, Magdy Y. The potential benefit of combined versus monotherapy of coenzyme Q10 and fluoxetine on depressive‐like behaviors and intermediates coupled to Gsk‐3beta in rats. Toxicol Appl Pharmacol. 2018;340:39‐48. [DOI] [PubMed] [Google Scholar]
  • 121. Wan YQ, Feng JG, Li M, et al. Prefrontal cortex miR‐29b‐3p plays a key role in the antidepressant‐like effect of ketamine in rats. Exp Mol Med. 2018;50(10):140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Zhao Y, Wang S, Chu Z, Dang Y, Zhu J, Su X. MicroRNA‐101 in the ventrolateral orbital cortex (VLO) modulates depressive‐like behaviors in rats and targets dual‐specificity phosphatase 1 (DUSP1). Brain Res. 2017;1669:55‐62. [DOI] [PubMed] [Google Scholar]
  • 123. Wang JM, Pei LX, Zhang YY, et al. Ethanol extract of Rehmannia glutinosa exerts antidepressant‐like effects on a rat chronic unpredictable mild stress model by involving monoamines and BDNF. Metab Brain Dis. 2018;33(3):885‐892. [DOI] [PubMed] [Google Scholar]
  • 124. Aricioglu F, Ozkartal CS, Bastaskin T, et al. Antidepressant‐like effects induced by chronic blockade of the purinergic 2x7 receptor through inhibition of non‐like receptor protein 1 inflammasome in chronic unpredictable mild stress model of depression in rats. Clin Psychopharmacol Neurosci. 2019;17(2):261‐272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Jia KK, Ding H, Yu HW, Dong TJ, Pan Y, Kong LD. κHuanglian‐Wendan decoction inhibits NF‐B/NLRP3 inflammasome activation in liver and brain of rats exposed to chronic unpredictable mild stress. Mediators Inflamm. 2018;2018:3093516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Wu T, Li X, Li T, et al. Apocynum venetum leaf extract exerts antidepressant‐like effects and inhibits hippocampal and cortical apoptosis of rats exposed to chronic unpredictable mild stress. Evid Based Complement Alternat Med. 2018;2018:5916451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Jia KK, Zheng YJ, Zhang YX, et al. Banxia‐houpu decoction restores glucose intolerance in CUMS rats through improvement of insulin signaling and suppression of NLRP3 inflammasome activation in liver and brain. J Ethnopharmacol. 2017;209:219‐229. [DOI] [PubMed] [Google Scholar]
  • 128. Chen XQ, Li CF, Chen SJ, et al. The antidepressant‐like effects of Chaihu Shugan San: dependent on the hippocampal BDNF‐TrkB‐ERK/Akt signaling activation in perimenopausal depression‐like rats. Biomed Pharmacother. 2018;105:45‐52. [DOI] [PubMed] [Google Scholar]
  • 129. Chen XQ, Chen SJ, Liang WN, et al. Saikosaponin A attenuates perimenopausal depression‐like symptoms by chronic unpredictable mild stress. Neurosci Lett. 2018;662:283‐289. [DOI] [PubMed] [Google Scholar]
  • 130. Luo D, Ma R, Wu Y, et al. Mechanism underlying acupuncture‐ameliorated depressive behaviors by enhancing glial glutamate transporter in chronic unpredictable mild stress (CUMS) rats. Med Sci Monit. 2017;23:3080‐3087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Huang HJ, Zhu XC, Han QQ, et al. Ghrelin alleviates anxiety‐ and depression‐like behaviors induced by chronic unpredictable mild stress in rodents. Behav Brain Res. 2017;326:33‐43. [DOI] [PubMed] [Google Scholar]

Articles from Animal Models and Experimental Medicine are provided here courtesy of Wiley

RESOURCES