Open Access

Behavioral video coding analysis of chronic morphine administration in rats

  • Authors:
    • Jie Yin
    • Tiecheng Zhang
    • Dan Li
    • Fan Xu
    • Huan Li
    • Xinyu Pan
    • Fang Liu
    • Yongqi Zhao
    • Xiechuan Weng
  • View Affiliations

  • Published online on: September 12, 2024     https://doi.org/10.3892/br.2024.1856
  • Article Number: 168
  • Copyright: © Yin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study assessed the behavior of morphine‑addicted rats using behavioral video coding technology, to evaluate effective methods for identifying morphine addiction. Rats were divided into a control group (n=15) and a morphine addiction group (n=15). The morphine addiction model was established with a 14‑day increasing dose scheme, confirmed using a conditional place preference (CPP) experiment. After successful modeling, the rats' behavior was recorded for 12 h, then coded and analyzed using Observer XT behavior analysis software. Compared with the control group, morphine‑addicted rats showed increased heat pain tolerance time (P=0.039) and spent more time in the white box during the CPP experiment (P<0.001). Video coding analysis revealed significant behavioral changes in morphine‑addicted rats compared to controls. In addition to being lighter, morphine‑addicted rats showed decreased water intake, reduced licking of forelimbs and hind limbs, and altered sleeping posture (sleeping curled up) during the day (all P<0.05). In conclusion, chronic morphine administration in rats leads to distinctive behavioral changes, including decreased licking frequency, reduced water intake and altered sleep posture. Video coding analysis, as a safe and non‑invasive method, may provide a convenient and efficient approach for studying morphine addiction in rats.

Introduction

Morphine, a typical opioid, is used in clinical settings to relieve cancer-related pain, especially in patients with bone cancer (1-4). However, addiction and dependence remain significant issues that affect its clinical use (5-8). In the context of social and public health, drug abuse and addiction are major public health concerns (9-12). Addiction is currently viewed as a chronic and relapsing disorder (13-15). The process of drug addiction engages reward-related learning and memory systems, showing synaptic plasticity within these systems (16-28). The mechanisms behind opioid addiction are closely related to the central dopamine reward neural pathway, mainly involving the prefrontal cortex (29), the nucleus accumbens (30-32), and the ventral tegmental area (33-38). Exploring the neurobiological mechanisms of drug addiction is crucial for clinical treatment.

Research on opioid addiction mechanisms often focuses on the morphine dependence model in rodents, usually rats or mice. The success of this model is assessed through conditioned place preference (CPP) or self-administration (SA) experiments (39-43). However, these methods are time-consuming, labor-intensive, require long-term behavioral training of animals and are subject to experimenter bias. CPP is an experimental tool for evaluating drug-seeking behavior or psychological craving in animals, requiring the association of a rewarding stimulus with a non-rewarding conditioned stimulus to confirm the presence of a rewarding stimulus. This process is tedious, time-consuming and requires a large sample size to avoid errors. SA experiments are the standard used for verifying drug addiction but need specific environments and equipment, and trained personnel. Additionally, issues such as hemorrhage, trauma, infection and death due to intravenous intubation in animals can affect the experimental results and process (44).

To the best of our knowledge, there have been no previous reports on the use of behavioral video coding analysis for morphine addiction. Behavioral video analysis technology may provide a convenient and efficient method for studying addictive behaviors. Recording videos of animals during the addiction modeling process and then coding these behaviors (45) allows for the identification of characteristic behavioral changes following morphine addiction in rats. Therefore, the present study assessed the behavior of morphine-addicted rats through behavioral video analysis and to evaluate this potentially efficient method to verify morphine addiction model in rats, offering a new approach for future studies on drug addiction behavior.

Materials and methods

Animals

Sprague-Dawley (SD) rats were purchased from SPF Biotechnology Co., Ltd. Rats were housed in a temperature-controlled environment (21-22˚C) within an animal holding room, following a 12 h light/dark cycle. Food and tap water were available ad libitum. The morphology, behavior, diet and water intake of the rats were observed at 8:00 a.m. and 8:00 p.m. each day. The body weight of the rats was measured every day before morphine injection. The experiment lasted a total of 22 days. Rats were acclimated to the laboratory for 1 week (day -6 to day 0) before starting administration. Chronic morphine administration and CPP assessment were performed in rats from day 1 to day 14, with video recording of rat behavior was carried out on day 15. The ethics committee of Beijing Institute of Basic Medical Sciences (Beijing, China; approval no. IACUC-DWZX-2022-715) approved the experimental protocol.

The experiment would be terminated in the event of any serious harm to animal welfare (including but not limited to): i) Loss of 15-20% of body weight; ii) infection in brain area; iii) other indicators such as pain, respiration (severe respiratory tract infection, dyspnea, cyanosis and other phenomena) and appearance (severe muscle atrophy and non-healing wounds). No rats met the criteria for early euthanasia during the course of the present study. After completing the experiment, the rats were placed into a non-precharged chamber and euthanized by CO2 inhalation (30% vol/min). Death was confirmed by cardiac and respiratory arrest and dilated pupils.

Drugs: Morphine hydrochloride injection (10 mg/ml, Shenyang First Pharmaceutical Factory, Shenyang) was diluted with sterile saline to achieve a 2.5 mg/ml working solution.

Apparatus

A Hot and Cold Plate Plantar Analgesia Instrument (cat. no. 28-0010; Shenzhen Huayang Biotech Co., Ltd.) was used to assess the thermal pain response of rats. The device included a transparent plastic chamber (25 cm diameter, 60 cm height) and a heating plate. Rats were placed in the plastic chamber, and the heating plate temperature was kept at 40±1˚C using a thermal probe and electronic feedback circuit. The timer started when the rats were placed in the chamber and was stopped manually when the rats showed behaviors such as lifting their hind limbs or licking their forelimbs. If no thermal response was observed within 3 min, the timer was stopped and the rats were promptly removed.

A Xiaomi Smart Video Camera PTZ Version 2K (cat. no. MJSXJ09CM; Xiaomi Inc.) was placed in the rat cage for video recording. A wooden board with a 5 cm diameter circular hole was positioned between the camera and the rat.

For the CPP experiment, a CPP apparatus was used, consisting of two compartments (30x60x30 cm; Zhongshi Technology). The different compartments had distinct floors and walls, separated by a removable plastic board. One compartment had black and white striped walls and a white floor, while the other had black and white checkered walls and a black floor. The test chamber was set up under 40 lux dim lighting and shielded from white noise (46).

The Observer XT (Noldus Information Technology BV) software was used for coding and analyzing recorded rat videos, focusing on typical rat behaviors (45).

Morphine administration

The morphine dependence model was established in 6-week-old (weight, 180-200 g) SD rats through a 14-day continuous dose escalation protocol. Male (n=15) and female (n=15) rats were randomly divided into two groups (47-52). The morphine group received intraperitoneal morphine at a dose of 5 mg/kg on day 1, while the control group received an equivalent volume of normal saline. The morphine dose increased gradually, reaching 100 mg/kg on day 14 for the morphine-addicted rats, while the control rats continued to receive an equivalent volume of normal saline (53-57). The initial dose of morphine group was 5 mg/kg on the first day, and the dose gradient was 5 mg/kg per day from day 2 to day 7, 10 mg/kg per day from day 8 to day 14, and reached 100 mg/kg on day 14 (53-65). The half-life of morphine in rats is generally 3-4 h, the drug effect of morphine lasts for 4-6 h and morphine is largely metabolized in 8-10 h (66-69). Morphine was administered daily at 8 am and 8 pm, ensuring an interval of about 12 h between doses (70).

Plantar heat tolerance test

Prior to each intraperitoneal injection of morphine, the time from the beginning of exposure to the hot plate at 40˚C to the lifting or licking of the hind foot was measured. The maximum measurement time was limited to 3 min. The same measurement was taken 1.5 h after the intraperitoneal injection of morphine under the same conditions, recording the time to foot lifting or licking.

CPP Test

The CPP experiment was divided into two phases, the pre-experimental phase and the test phase (46). The pre-experimental phase lasted four consecutive days, with two sessions conducted daily. Rats (n=10) were randomly selected from each of the morphine and control groups, resulting in a total of 20 rats. Each session lasted 15 min. Conditioned reflexes were established by confining the rats to a white drug delivery box (referred to as the white box) for 15 min, during which they received injections of either morphine (with dosage according to the aforementioned protocol) for the morphine group or an equivalent volume of saline for the control group, administered intraperitoneally. On day 4, during the retention experiment, the rats were released from the central section of the CPP apparatus and allowed to freely explore the two chambers for 15 min.

In the final testing phase, the same procedure was repeated for four consecutive days. CPP training was performed twice a day for 15 min each for 3 days and the CPP test was performed on the fourth day. The rats were again confined to the white box for 15 min, receiving the corresponding injections (morphine for the morphine group, with the final dose reaching 100 mg/kg, and an equivalent volume of saline for the control group). On day 14, during the test experiment, the rats were released from the central part of the CPP apparatus and allowed to explore the two chambers for 15 min.

Video behavior coding

During the 14-day morphine addiction model, rats were randomly selected from each of the two experiment groups (n=8 per group), and placed in a cage with one rat per cage. On day 15, video recordings were made and subsequently coded and analyzed using Observer XT software to quantify behavioral differences between the two groups.

The observed rats were divided into two batches of 8 (each with 4 addicted rats and 4 control rats), with one batch observed per day. Video recordings were made from 8:00 a.m. to 8:00 p.m. each day, with one rat per cage. A Xiaomi cloud platform video camera was temporarily placed in each rat cage, isolated from the rats by a wooden board, this setup allowed continuous recording of the rats' activity and behavior for 12 h, with the recorded video retained for subsequent analysis. Video coding and analysis were conducted using Observer XT software to examine differences in classical behaviors (such as eating, drinking, licking forelimbs, licking hind limbs, sleeping, walking and scratching) between the morphine group rats post-morphine addiction and the control group rats (45).

Statistical analysis

Data analysis was performed using GraphPad Prism (version 9; Dotmatics). The Wilcoxon rank-sum test and two-way ANOVA with Bonferroni correction were used to assess both experimental data and behavioral video coding recordings. P<0.05 was considered to indicate a statistically significant difference. Data are presented as mean ± SEM.

Results

Effect of continuous morphine administration on rat body weight

Following 14 days of escalating morphine doses, the weight difference between the morphine group and the control group showed a gradual and significant increase (day 14, z=2.717, P=0.0057, Fig. 1). After 14 days, compared with the control group, the weight growth rate of morphine group rats was lower. The final average body weight on day 14 was 206.2±3.4 g (maximum individual body weight, 222.0 g) for the morphine group. In contrast, the body weight of rats in the control group showed a steady increase, with a stable growth rate. The final average body weight on day 14 for the control group was 233.6±8.0 g (maximum individual body weight, 287.7 g). A difference between body weight in the two groups was evident from the first day, which increased over time. On day 14, the average body weight of control rats exceeded that of morphine-rats by 27.4 g with the greatest body weight difference between the two groups being 70.3 g.

Effect of morphine injection on thermal pain tolerance in rats

At 1.5 h post-intraperitoneal morphine injection, the morphine group exhibited a significantly extended duration of thermal pain tolerance compared with the same group pre-injection. There was no significant difference in thermal pain tolerance following saline injection in the control group. Ultimately, morphine-addicted rats demonstrated longer thermal tolerance than control rats (U=74.4, P=0.039, Fig. 2).

CPP experiments in morphine-addicted rats

CPP experiments were conducted on days 1-4 and days 11-14 for both the morphine and control groups. The analysis of CPP results involved recording the time spent by rats in the white box. The results before morphine addiction (day 4), demonstrated no statistically significant difference between the morphine and control groups (F=6.1., P=0.64, Fig. 3), indicating equivalent white box time for both groups.

In the experiment after morphine addiction (day 14), a significant difference was demonstrated between the morphine and control groups regarding time spent in the white box (F=5.7, P<0.001, Fig. 3), which demonstrated that morphine administration increased the time spent in the white box compared to saline administration, which validated the rat addiction model. Additionally, there was a significant difference in the white box time of morphine group rats before and after addiction (F=11.5, P<0.001, Fig. 3), further verifying the success of the morphine addiction model in rats.

Video coding analysis of the effects of morphine dependence on rat behavior

To record and statistically analyze the characteristic behaviors of rats after addiction, a 12-h video of rats in the morphine group and rats in the control group was recorded. The video was then coded and analyzed using Observer XT software, and the data was exported for analysis using the Mann-Whitney test to compare the statistical differences in the behaviors of the two groups. Compared with the control rats, the morphine group rats showed significant differences in drinking and licking the fore and hind limbs. They also exhibited characteristic behaviors such as an increased number ball sleeping, increased scratching, increased fur licking and changes in walking time at specific times of the day.

Regarding feeding, rats in the morphine group did not show significant differences compared with control rats (U=65.5, P=0.98, Fig. 4A). However, in terms of drinking, the morphine group drank significantly less water than the control group (U=17, P=0.0014, Fig. 4B). For the licking of the forelimbs, the morphine group licked significantly less compared with the control group (U=27, P=0.014, Fig. 4C). In contrast, for the licking of the hindlimbs, the morphine group licked significantly less than the control group (U=32, P=0.035, Fig. 4D). There was no significant difference between the two groups in terms of lifting the hindlimbs (U=62, P=0.082>0.05, Fig. 4E).

In the present study, ‘marten sleeping’ means sleeping curled up in the lateral arch of the rat with a clear view of the entire head. ‘Ball sleeping’ is when the rat is resting with its head curled up on its abdomen and its eyes closed. In the 12-h observation period, morphine-addicted rats displayed significantly more ‘ball sleeping’ behavior compared with control rats during the 2:30-3:00 p.m. period (z=-2.412, P=0.0159, Fig. 5A). There was no significant difference between the groups for ‘marten sleeping’ at any recorded time period. The frequency of scratching showed a statistically significant difference only during the 1:00-1:30 p.m. period (z=-1.964, P=0.0495; Fig. 5C). The frequency of licking showed a significant difference between the groups during the 3:30-4:00 p.m. period (z=-2.29, P=0.022, Fig. 5D). Additionally, the morphine group walked significantly less than the control group during the 12:00 a.m.-12:30 p.m. and 6:00-6:30 p.m. periods (z=-2.093, P=0.036; z=-2.039, P=0.0415; Fig. 5E). These results indicate that rats chronically injected with morphine exhibit characteristic behaviors such as increased ball sleeping, increased scratching, increased hair licking, and changes in walking behaviorat specific times of the day.

Discussion

Morphine addiction and dependence are recognized as neuropsychiatric diseases, involving various behavioral, neurobiological and molecular changes (55,71-75). In animal models, especially in rats, studying behavioral manifestations after morphine addiction is crucial for understanding the neural mechanisms of addiction and finding treatments. Video coding and analysis technology record animal behavior under specific conditions, using computer software to encode, identify and analyze the behavioral data. This technology enables comprehensive recording and accurate analysis of animal behavior, which is safe and non-invasive, and does not interfere with the animals' activities. Thus, it reveals the behavioral characteristics of rats after morphine addiction more accurately than traditional behavioral observation.

At present, the mechanism of morphine addiction has been studied and evaluated using various animal models, among which the rat model of chronic morphine administration is commonly used (48,49,51,76). In studies of rat or mouse behavior, most analyses use experiments to verify behavior, such as the rotarod, open field test, elevated plus maze test, cliff hanging, passive avoidance test, Morris water maze, light/dark box or light spot tests. The study of daily behaviors in rats mainly focuses on neuropathic pain and other diseases, using video coding analysis for assessment of daily walking gait (77-81). Research on addictive behavior primarily involves self-administration and CPP tests, with little use of daily behavior video coding analysis (82,83). However, after morphine administration, whether rats are addicted or not is usually judged by CPP test and self-administration experiment, and these methods have certain limitations and shortcomings, such as time-consuming, labor-intensive, requiring long-term behavioral training of animals and risk of infection and death due to invasive procedures. Therefore, in the present study, video coding was used to analyze the characteristic behavior of chronic morphine administration in rats, which was expected to provide a simple and quick method to analyze and evaluate morphine addiction through the behavioral changes before and after morphine addiction. Video behavior observation and analysis has many applications in studying animal behaviors such as pain, depression and anxiety. For example, Braw et al (84) demonstrated the depression and anxiety behavior of rats with different genetic models, finding differential expression of anxiety in pre-pubertal rats belonging to the ‘depressed’ strains, suggesting that these strains may be suitable for modelling different sub-groups of depression at young ages. Medvedev et al (85) demonstrated MK-801 and memantine acted against tactile allodynia induced by sciatic nerve ligatio. Yuan and Devine (86) demonstrated the self-injury behavior of rats induced by anxiety drugs, and finding the rats given anxiety-inducing drugs showed stronger self-injurious behavior. To the best of our knowledge, no previous studies have performed video behavioral coding analysis of morphine addiction behavior in rats. Therefore, using video coding analysis to study addiction behavior characteristics introduces innovative changes and has potential value for the researching of addiction mechanisms and treatments.

In the present study, weight observation, heat tolerance test, CPP test and behavioral video coding analysis were performed using a morphine addiction model in rats. The results indicated that morphine-addicted rats had increased heat pain tolerance and reduced weight gain. The present study used the method of video behavior coding analysis for the first time to find that rats with chronic morphine administration exhibited slower weight gain, smaller body size, and fragile fur (87). Behavioral changes were mainly characterized by decreased water intake, decreased toe licking and increased daytime sleep in a spherical posture. The present study analyzed various behavioral changes in rats before and after morphine addiction and provided a new method for verifying the morphine addiction rat model through behavioral video analysis (46-49,51,52).

Behavioral video analysis of morphine-addicted rats revealed significant differences in drinking and licking of the fore and hind limbs, as well as in sleeping postures and scratching during the middle of the day. According to Kon et al (88), morphine increases the expression of aquaporin-3 water channels in the colon by increasing the secretion of serotonin, which enhances water absorption from the lumen to the vasculature of the colon. Deroche et al (89) reported that opioids bind to MOP receptors in enteric neurons, delaying gastrointestinal transit time, and stimulating non-propulsive GI motility and the pylorus and ileocecal sphincters. Consequently, chronic morphine administration results in diminished thirst and reduced water excretion, leading to decreased water intake. Morphine group rats exhibited more frequent ball sleeping, especially during the 2:00-2:30 p.m. timeframe. The altered sleep posture suggests that long-term morphine exposure may lead to functional or structural changes in the limbic system and motor cortex, which are involved in mediating instinctive and emotional behaviors through Papez circuits (90). It is speculated that morphine affects the neurons and neural circuits in the limbic system or cortical nuclei, resulting in changes in sleep posture.

The present study analyzed the behavioral characteristics of morphine-addicted rats using the minimum sample size necessary to achieve statistical significance. Increasing the sample size could provide more precise behavior analysis and more convincing experimental conclusions. Additionally, the 14-day morphine addiction model is currently complex; future optimizations could make the model more efficient and reduce animal suffering.

In summary, high-resolution video equipment was used to record the behavior of morphine-addicted rats, and behavioral video coding and identification analysis were performed to systematically study the characteristics of morphine addiction behavior. By analyzing the behavioral characteristics of morphine-addicted rats, we can better understand the occurrence and development of addiction, reveal the impact of addiction on rat behavior and its potential neural mechanisms, and provide important theoretical and methodological support for further research on the neural circuit mechanisms of addiction and the development of related treatment strategies.

Acknowledgements

Not applicable.

Funding

Funding: This study was supported by the National Natural Science Foundation of China (grant no. 82073833) and the National Key R&D Plan (grant nos. 2022YFC3600500 and 2022YFC3600502).

Availability of data and materials

The datasets used and/or analyzed during the study are available from the corresponding author upon reasonable request.

Authors' contributions

JY performed multiple experiments, acquired data and wrote the first draft of the manuscript. JY, TZ, DL, HL, XP, and FL performed data analysis. YZ, FX, and XW contributed to the study design and participated in revising the manuscript. YZ, FX, and XW confirmed the authenticity of all raw data and all authors agreed to be accountable for all aspects of the study. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Experiments were conducted in compliance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals. The experimental procedures were approved by the ethics committee of Beijing Institute of Basic Medical Sciences (approval no. IACUC-DWZX-2022-715; Beijing, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Häuser W, Welsch P, Radbruch L, Fisher E, Bell RF and Moore RA: Cannabis-based medicines and medical cannabis for adults with cancer pain. Cochrane Database Syst Rev. 6(Cd014915)2023.PubMed/NCBI View Article : Google Scholar

2 

Luger NM, Mach DB, Sevcik MA and Mantyh PW: Bone cancer pain: From model to mechanism to therapy. J Pain Symptom Manage. 29 (5 Suppl):S32–S46. 2005.PubMed/NCBI View Article : Google Scholar

3 

Mercadante S: Intravenous morphine for management of cancer pain. Lancet Oncol. 11:484–489. 2010.PubMed/NCBI View Article : Google Scholar

4 

Nguyen QN, Chun SG, Chow E, Komaki R, Liao Z, Zacharia R, Szeto BK, Welsh JW, Hahn SM, Fuller CD, et al: Single-Fraction stereotactic vs conventional multifraction radiotherapy for pain relief in patients with predominantly nonspine bone metastases: A Randomized phase 2 trial. JAMA Oncol. 5:872–878. 2019.PubMed/NCBI View Article : Google Scholar

5 

Mantsch JR, Baker DA, Funk D, Lê AD and Shaham Y: Stress-Induced reinstatement of drug seeking: 20 years of progress. Neuropsychopharmacology. 41:335–356. 2016.PubMed/NCBI View Article : Google Scholar

6 

Reiner DJ, Fredriksson I, Lofaro OM, Bossert JM and Shaham Y: Relapse to opioid seeking in rat models: Behavior, pharmacology and circuits. Neuropsychopharmacology. 44:465–477. 2019.PubMed/NCBI View Article : Google Scholar

7 

Shalev U, Grimm JW and Shaham Y: Neurobiology of relapse to heroin and cocaine seeking: A review. Pharmacol Rev. 54:1–42. 2002.PubMed/NCBI View Article : Google Scholar

8 

Trujillo KA and Akil H: Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science. 251:85–87. 1991.PubMed/NCBI View Article : Google Scholar

9 

Garofoli M: Adolescent Substance Abuse. Prim Care. 47:383–394. 2020.PubMed/NCBI View Article : Google Scholar

10 

Lewis DC: Drug overdose, addiction and binge drinking: Medical problems with public health consequences. R I Med J (2013). 97:18–19. 2014.PubMed/NCBI

11 

McCarty D, Argeriou M, Huebner RB and Lubran B: Alcoholism, drug abuse, and the homeless. Am Psychol. 46:1139–1148. 1991.PubMed/NCBI View Article : Google Scholar

12 

Lu L and Wang X: Drug addiction in China. Ann N Y Acad Sci. 1141:304–317. 2008.PubMed/NCBI View Article : Google Scholar

13 

Leshner AI: Addiction is a brain disease, and it matters. Science. 278:45–47. 1997.PubMed/NCBI View Article : Google Scholar

14 

Liu JF and Li JX: Drug addiction: A curable mental disorder? Acta Pharmacol Sin. 39:1823–1829. 2018.PubMed/NCBI View Article : Google Scholar

15 

Nestler EJ: Molecular basis of long-term plasticity underlying addiction. Nat Rev Neurosci. 2:119–128. 2001.PubMed/NCBI View Article : Google Scholar

16 

Chiamulera C, Piva A and Abraham WC: Glutamate receptors and metaplasticity in addiction. Curr Opin Pharmacol. 56:39–45. 2021.PubMed/NCBI View Article : Google Scholar

17 

Fourgeaud L, Mato S, Bouchet D, Hémar A, Worley PF and Manzoni OJ: A single in vivo exposure to cocaine abolishes endocannabinoid-mediated long-term depression in the nucleus accumbens. J Neurosci. 24:6939–6945. 2004.PubMed/NCBI View Article : Google Scholar

18 

Gipson CD, Kupchik YM and Kalivas PW: Rapid, transient synaptic plasticity in addiction. Neuropharmacology. 76 Pt B:276–286. 2014.PubMed/NCBI View Article : Google Scholar

19 

Hafenbreidel M, Rafa Todd C and Mueller D: Infralimbic GluN2A-Containing NMDA receptors modulate reconsolidation of cocaine self-administration memory. Neuropsychopharmacology. 42:1113–1125. 2017.PubMed/NCBI View Article : Google Scholar

20 

Hyman SE: Addiction: A disease of learning and memory. Am J Psychiatry. 162:1414–1422. 2005.PubMed/NCBI View Article : Google Scholar

21 

Kauer JA and Malenka RC: Synaptic plasticity and addiction. Nat Rev Neurosci. 8:844–858. 2007.PubMed/NCBI View Article : Google Scholar

22 

Keralapurath MM, Briggs SB and Wagner JJ: Cocaine self-administration induces changes in synaptic transmission and plasticity in ventral hippocampus. Addict Biol. 22:446–456. 2017.PubMed/NCBI View Article : Google Scholar

23 

Lüscher C and Malenka RC: Drug-evoked synaptic plasticity in addiction: From molecular changes to circuit remodeling. Neuron. 69:650–663. 2011.PubMed/NCBI View Article : Google Scholar

24 

Mameli M, Bellone C, Brown MT and Lüscher C: Cocaine inverts rules for synaptic plasticity of glutamate transmission in the ventral tegmental area. Nat Neurosci. 14:414–416. 2011.PubMed/NCBI View Article : Google Scholar

25 

Tronson NC and Taylor JR: Molecular mechanisms of memory reconsolidation. Nat Rev Neurosci. 8:262–275. 2007.PubMed/NCBI View Article : Google Scholar

26 

Tzschentke TM and Schmidt WJ: Glutamatergic mechanisms in addiction. Mol Psychiatry. 8:373–382. 2003.PubMed/NCBI View Article : Google Scholar

27 

van Huijstee AN and Mansvelder HD: Glutamatergic synaptic plasticity in the mesocorticolimbic system in addiction. Front Cell Neurosci. 8(466)2014.PubMed/NCBI View Article : Google Scholar

28 

Ossipov MH, Lai J, King T, Vanderah TW, Malan TP Jr, Hruby VJ and Porreca F: Antinociceptive and nociceptive actions of opioids. J Neurobiol. 61:126–148. 2004.PubMed/NCBI View Article : Google Scholar

29 

Mickiewicz AL and Napier TC: Repeated exposure to morphine alters surface expression of AMPA receptors in the rat medial prefrontal cortex. Eur J Neurosci. 33:259–265. 2011.PubMed/NCBI View Article : Google Scholar

30 

Boudreau AC and Wolf ME: Behavioral sensitization to cocaine is associated with increased AMPA receptor surface expression in the nucleus accumbens. J Neurosci. 25:9144–9151. 2005.PubMed/NCBI View Article : Google Scholar

31 

Hemby SE, Tang W, Muly EC, Kuhar MJ, Howell L and Mash DC: Cocaine-induced alterations in nucleus accumbens ionotropic glutamate receptor subunits in human and non-human primates. J Neurochem. 95:1785–1793. 2005.PubMed/NCBI View Article : Google Scholar

32 

Sutton MA, Schmidt EF, Choi KH, Schad CA, Whisler K, Simmons D, Karanian DA, Monteggia LM, Neve RL and Self DW: Extinction-induced upregulation in AMPA receptors reduces cocaine-seeking behaviour. Nature. 421:70–75. 2003.PubMed/NCBI View Article : Google Scholar

33 

Bachtell RK, Choi KH, Simmons DL, Falcon E, Monteggia LM, Neve RL and Self DW: Role of GluR1 expression in nucleus accumbens neurons in cocaine sensitization and cocaine-seeking behavior. Eur J Neurosci. 27:2229–2240. 2008.PubMed/NCBI View Article : Google Scholar

34 

Conrad KL, Tseng KY, Uejima JL, Reimers JM, Heng LJ, Shaham Y, Marinelli M and Wolf ME: Formation of accumbens GluR2-lacking AMPA receptors mediates incubation of cocaine craving. Nature. 454:118–121. 2008.PubMed/NCBI View Article : Google Scholar

35 

Kasanetz F, Deroche-Gamonet V, Berson N, Balado E, Lafourcade M, Manzoni O and Piazza PV: Transition to addiction is associated with a persistent impairment in synaptic plasticity. Science. 328:1709–1712. 2010.PubMed/NCBI View Article : Google Scholar

36 

LaLumiere RT and Kalivas PW: Glutamate release in the nucleus accumbens core is necessary for heroin seeking. J Neurosci. 28:3170–3177. 2008.PubMed/NCBI View Article : Google Scholar

37 

Degoulet M, Stelly CE, Ahn KC and Morikawa H: L-type Ca²+ channel blockade with antihypertensive medication disrupts VTA synaptic plasticity and drug-associated contextual memory. Mol Psychiatry. 21:394–402. 2016.PubMed/NCBI View Article : Google Scholar

38 

Lane DA, Lessard AA, Chan J, Colago EE, Zhou Y, Schlussman SD, Kreek MJ and Pickel VM: Region-specific changes in the subcellular distribution of AMPA receptor GluR1 subunit in the rat ventral tegmental area after acute or chronic morphine administration. J Neurosci. 28:9670–9681. 2008.PubMed/NCBI View Article : Google Scholar

39 

Billa SK, Sinha N, Rudrabhatla SR and Morón JA: Extinction of morphine-dependent conditioned behavior is associated with increased phosphorylation of the GluR1 subunit of AMPA receptors at hippocampal synapses. Eur J Neurosci. 29:55–64. 2009.PubMed/NCBI View Article : Google Scholar

40 

Cai YQ, Wang W, Hou YY, Zhang Z, Xie J and Pan ZZ: Central amygdala GluA1 facilitates associative learning of opioid reward. J Neurosci. 33:1577–1588. 2013.PubMed/NCBI View Article : Google Scholar

41 

Sun Y, Chen G, Zhou K and Zhu Y: A conditioned place preference protocol for measuring incubation of craving in rats. J Vis Exp. 2018.PubMed/NCBI View Article : Google Scholar

42 

Tzschentke TM: Measuring reward with the conditioned place preference (CPP) paradigm: Update of the last decade. Addict Biol. 12:227–462. 2007.PubMed/NCBI View Article : Google Scholar

43 

Lin XJ, Zhang JJ and Yu LC: GluR2-3Y inhibits the acquisition and reinstatement of morphine-induced conditioned place preference in rats. Neurosci Bull. 32:177–182. 2016.PubMed/NCBI View Article : Google Scholar

44 

Tzschentke TM: Measuring reward with the conditioned place preference paradigm: A comprehensive review of drug effects, recent progress and new issues. Prog Neurobiol. 56:613–672. 1998.PubMed/NCBI View Article : Google Scholar

45 

Wang X, Bey AL, Katz BM, Badea A, Kim N, David LK, Duffney LJ, Kumar S, Mague SD, Hulbert SW, et al: Altered mGluR5-Homer scaffolds and corticostriatal connectivity in a Shank3 complete knockout model of autism. Nat Commun. 7(11459)2016.PubMed/NCBI View Article : Google Scholar

46 

Domínguez-Salazar E, Naser HF and Velázquez-Moctezuma J: D1-like antagonist blocks conditioned place preference induced by ejaculation in male rats. Behav Brain Res. 269:15–19. 2014.PubMed/NCBI View Article : Google Scholar

47 

Chang SL, Moldow RL, House SD and Zadina JE: Morphine affects the brain-immune axis by modulating an interleukin-1 beta dependent pathway. Adv Exp Med Biol. 402:35–42. 1996.PubMed/NCBI View Article : Google Scholar

48 

Graf JA, Patel JA and Chang SL: Chronic exposure to morphine, but not ethanol, attenuates the expression of interleukin-1 beta converting enzyme in rat spleen. Immunol Lett. 58:153–157. 1997.PubMed/NCBI View Article : Google Scholar

49 

House SD, Mao X, Wu G, Espinelli D, Li WX and Chang SL: Chronic morphine potentiates the inflammatory response by disrupting interleukin-1beta modulation of the hypothalamic-pituitary-adrenal axis. J Neuroimmunol. 118:277–285. 2001.PubMed/NCBI View Article : Google Scholar

50 

Lobo MK, Covington HE III, Chaudhury D, Friedman AK, Sun H, Damez-Werno D, Dietz DM, Zaman S, Koo JW, Kennedy PJ, et al: Cell type-specific loss of BDNF signaling mimics optogenetic control of cocaine reward. Science. 330:385–390. 2010.PubMed/NCBI View Article : Google Scholar

51 

Ocasio FM, Jiang Y, House SD and Chang SL: Chronic morphine accelerates the progression of lipopolysaccharide-induced sepsis to septic shock. J Neuroimmunol. 149:90–100. 2004.PubMed/NCBI View Article : Google Scholar

52 

Zadina JE, Kastin AJ, Harrison LM, Ge LJ and Chang SL: Opiate receptor changes after chronic exposure to agonists and antagonists. Ann N Y Acad Sci. 757:353–361. 1995.PubMed/NCBI View Article : Google Scholar

53 

Askari N, Mousavi A and Vaez-Mahdavi MR: Maternal deprivation effect on morphine-induced CPP is related to changes in opioid receptors in selected rat brain regions (hippocampus, prefrontal cortex, and nucleus accumbens). Behav Processes. 197(104607)2022.PubMed/NCBI View Article : Google Scholar

54 

Rodgers HM, Lim SA, Yow J, Dinkins ML, Patton R, Clemens S and Brewer KL: Dopamine D1 or D3 receptor modulators prevent morphine tolerance and reduce opioid withdrawal symptoms. Pharmacol Biochem Behav. 194(172935)2020.PubMed/NCBI View Article : Google Scholar

55 

Liu LW, Lu J, Wang XH, Fu SK, Li Q and Lin FQ: Neuronal apoptosis in morphine addiction and its molecular mechanism. Int J Clin Exp Med. 6:540–545. 2013.PubMed/NCBI

56 

Papaleo F and Contarino A: Gender- and morphine dose-linked expression of spontaneous somatic opiate withdrawal in mice. Behav Brain Res. 170:110–118. 2006.PubMed/NCBI View Article : Google Scholar

57 

Rahmati B and Beik A: Prevention of morphine dependence and tolerance by Nepeta menthoides was accompanied by attenuation of Nitric oxide overproduction in male mice. J Ethnopharmacol. 199:39–51. 2017.PubMed/NCBI View Article : Google Scholar

58 

Koek W: Morphine-induced conditioned place preference and effects of morphine pre-exposure in adolescent and adult male C57BL/6J mice. Psychopharmacology (Berl). 233:2015–2024. 2016.PubMed/NCBI View Article : Google Scholar

59 

Li X, Kshatriya D and Bello NT: Weight-gain propensity and morphine withdrawal alters locomotor behavior and regional norepinephrine-related gene expression in male and female mice. Pharmacol Biochem Behav. 213(173329)2022.PubMed/NCBI View Article : Google Scholar

60 

Madayag AC, Gomez D, Anderson EM, Ingebretson AE, Thomas MJ and Hearing MC: Cell-type and region-specific nucleus accumbens AMPAR plasticity associated with morphine reward, reinstatement, and spontaneous withdrawal. Brain Struct Funct. 224:2311–2324. 2019.PubMed/NCBI View Article : Google Scholar

61 

McDevitt DS, McKendrick G and Graziane NM: Anterior cingulate cortex is necessary for spontaneous opioid withdrawal and withdrawal-induced hyperalgesia in male mice. Neuropsychopharmacology. 46:1990–1999. 2021.PubMed/NCBI View Article : Google Scholar

62 

Nakamura A, Ono H, Ando A, Hinata M, Niidome K, Omachi S, Sakaguchi G and Shinohara S: Suppression of the acute upregulation of phosphorylated-extracellular regulated kinase in ventral tegmental area by a µ-opioid receptor agonist is related to resistance to rewarding effects in a mouse model of bone cancer. J Pharmacol Sci. 133:9–17. 2017.PubMed/NCBI View Article : Google Scholar

63 

Piccin A and Contarino A: Long-lasting pseudo-social aggressive behavior in opiate-withdrawn mice. Prog Neuropsychopharmacol Biol Psychiatry. 97(109780)2020.PubMed/NCBI View Article : Google Scholar

64 

Piccin A and Contarino A: The CRF(1) receptor mediates social behavior deficits induced by opiate withdrawal. J Neurosci Res. 100:309–321. 2022.PubMed/NCBI View Article : Google Scholar

65 

Varshneya NB, Walentiny DM, Stevens DL, Walker TD, Akinfiresoye LR and Beardsley PM: Structurally diverse fentanyl analogs yield differential locomotor activities in mice. Pharmacol Biochem Behav. 222(173496)2023.PubMed/NCBI View Article : Google Scholar

66 

Kalvass JC, Olson ER, Cassidy MP, Selley DE and Pollack GM: Pharmacokinetics and pharmacodynamics of seven opioids in P-glycoprotein-competent mice: Assessment of unbound brain EC50,u and correlation of in vitro, preclinical, and clinical data. J Pharmacol Exp Ther. 323:346–355. 2007.PubMed/NCBI View Article : Google Scholar

67 

Melzacka M: Pharmacokinetic aspects of some behavioral effects of psychotropic drugs. Pol J Pharmacol Pharm. 36:117–136. 1984.PubMed/NCBI

68 

Regenthal R, Krueger M, Koeppel C and Preiss R: Drug levels: Therapeutic and toxic serum/plasma concentrations of common drugs. J Clin Monit Comput. 15:529–544. 1999.PubMed/NCBI View Article : Google Scholar

69 

Sulimai NH, Ko JC, Jones-Hall YL, Weng HY, Deng M, Breur GJ and Knipp GT: Evaluation of 25% poloxamer as a slow release carrier for morphine in a rat model. Front Vet Sci. 5(19)2018.PubMed/NCBI View Article : Google Scholar

70 

Santos-Vera B, Vaquer-Alicea ADC, Maria-Rios CE, Montiel-Ramos A, Ramos-Cardona A, Vázquez-Torres R, Sanabria P and Jiménez-Rivera CA: Protein and surface expression of HCN2 and HCN4 subunits in mesocorticolimbic areas after cocaine sensitization. Neurochem Int. 125:91–98. 2019.PubMed/NCBI View Article : Google Scholar

71 

Cao DN, Song R, Zhang SZ, Wu N and Li J: Nucleus accumbens hyperpolarization-activated cyclic nucleotide-gated channels modulate methamphetamine self-administration in rats. Psychopharmacology (Berl). 233:3017–3029. 2016.PubMed/NCBI View Article : Google Scholar

72 

Caruso Brown AE: Treating addiction as a terminal disease. N Engl J Med. 382:207–209. 2020.PubMed/NCBI View Article : Google Scholar

73 

Mu L, Liu X, Yu H, Vickstrom CR, Friedman V, Kelly TJ, Hu Y, Su W, Liu S, Mantsch JR and Liu QS: cAMP-mediated upregulation of HCN channels in VTA dopamine neurons promotes cocaine reinforcement. Mol Psychiatry. 28:3930–3942. 2023.PubMed/NCBI View Article : Google Scholar

74 

Settley C: The physical and psychological wellbeing of caregivers of individuals suffering from substance addiction. Arch Psychiatr Nurs. 34:107–109. 2020.PubMed/NCBI View Article : Google Scholar

75 

Xiao ZW, Cao CY, Wang ZX, Li JX, Liao HY and Zhang XX: Changes of dopamine transporter function in striatum during acute morphine addiction and its abstinence in rhesus monkey. Chin Med J (Engl). 119:1802–1807. 2006.PubMed/NCBI

76 

Aramjoo H, Riahi-Zanjani B, Farkhondeh T, Forouzanfar F and Sadeghi M: Modulatory effect of opioid administration on the activity of cholinesterase enzyme: A systematic review of mice/rat models. Environ Sci Pollut Res Int. 28:52675–52688. 2021.PubMed/NCBI View Article : Google Scholar

77 

Djabirska I, Delaval L, Tromme A, Blomet J, Desmecht D and Van Laere AS: Longitudinal quantitative assessment of TMEV-IDD-induced MS phenotypes in two inbred mouse strains using automated video tracking technology. Exp Neurol. 379(114851)2024.PubMed/NCBI View Article : Google Scholar

78 

Kulbeth HJ, Fukuda S and Brents LK: Automated quantification of opioid withdrawal in neonatal rat pups using Ethovision® XT software. Neurotoxicol Teratol. 84(106959)2021.PubMed/NCBI View Article : Google Scholar

79 

Richmond-Hacham B, Tseitlin L, Bikovski L and Pick CG: Investigation of mild traumatic brain injury home cage behavior: The home cage assay advantages. J Neurotrauma. 41:e1780–e1792. 2024.PubMed/NCBI View Article : Google Scholar

80 

Timotius IK, Roelofs RF, Richmond-Hacham B, Noldus LPJJ, von Hörsten S and Bikovski L: CatWalk XT gait parameters: A review of reported parameters in pre-clinical studies of multiple central nervous system and peripheral nervous system disease models. Front Behav Neurosci. 17(1147784)2023.PubMed/NCBI View Article : Google Scholar

81 

Watkins J, Ghosh A, Keerie AFA, Alix JJP, Mead RJ and Sreedharan J: Female sex mitigates motor and behavioural phenotypes in TDP-43Q331K knock-in mice. Sci Rep. 10(19220)2020.PubMed/NCBI View Article : Google Scholar

82 

Lebedev IV, Pleskacheva MG and Anokhin KV: C57BL/6 mice open field behaviour qualitatively depends on arena size. Zh Vyssh Nerv Deiat Im I P Pavlova. 62:485–496. 2012.PubMed/NCBI(In Russian).

83 

Novati A, Manfré G, Flunkert S, Van der Harst JE, Homberg JR, Wronski R and Nguyen HP: Validation of behavioral phenotypes in the BACHD rat model. Behav Brain Res. 393(112783)2020.PubMed/NCBI View Article : Google Scholar

84 

Braw Y, Malkesman O, Dagan M, Bercovich A, Lavi-Avnon Y, Schroeder M, Overstreet DH and Weller A: Anxiety-like behaviors in pre-pubertal rats of the Flinders Sensitive Line (FSL) and Wistar-Kyoto (WKY) animal models of depression. Behav Brain Res. 167:261–269. 2006.PubMed/NCBI View Article : Google Scholar

85 

Medvedev IO, Malyshkin AA, Belozertseva IV, Sukhotina IA, Sevostianova NY, Aliev K, Zvartau EE, Parsons CG, Danysz W and Bespalov AY: Effects of low-affinity NMDA receptor channel blockers in two rat models of chronic pain. Neuropharmacology. 47:175–183. 2004.PubMed/NCBI View Article : Google Scholar

86 

Yuan X and Devine DP: The role of anxiety in vulnerability for self-injurious behaviour: studies in a rodent model. Behav Brain Res. 311:201–209. 2016.PubMed/NCBI View Article : Google Scholar

87 

Le T, Xia M, Jia M, Sarkar N, Chen J, Li H, Wynn GH, Ursano RJ and Choi KH: Association between initial morphine intake and body weight change, acoustic startle reflex and drug seeking in rats. Psychopharmacology (Berl). 231:4569–4577. 2014.PubMed/NCBI View Article : Google Scholar

88 

Kon R, Ikarashi N, Hayakawa A, Haga Y, Fueki A, Kusunoki Y, Tajima M, Ochiai W, Machida Y and Sugiyama K: Morphine-induced constipation develops with increased aquaporin-3 expression in the colon via increased serotonin secretion. Toxicol Sci. 145:337–347. 2015.PubMed/NCBI View Article : Google Scholar

89 

Deroche V, Piazza PV, Casolini P, Maccari S, Le Moal M and Simon H: Stress-induced sensitization to amphetamine and morphine psychomotor effects depend on stress-induced corticosterone secretion. Brain Res. 598:343–348. 1992.PubMed/NCBI View Article : Google Scholar

90 

Yunusa S, Müller CP and Hassan Z: Mitragynine (Kratom)-Withdrawal behaviour and cognitive impairments can be ameliorated by an epigenetic mechanism. Br J Pharmacol. 181:2070–2084. 2024.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2024
Volume 21 Issue 5

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yin J, Zhang T, Li D, Xu F, Li H, Pan X, Liu F, Zhao Y and Weng X: Behavioral video coding analysis of chronic morphine administration in rats. Biomed Rep 21: 168, 2024.
APA
Yin, J., Zhang, T., Li, D., Xu, F., Li, H., Pan, X. ... Weng, X. (2024). Behavioral video coding analysis of chronic morphine administration in rats. Biomedical Reports, 21, 168. https://doi.org/10.3892/br.2024.1856
MLA
Yin, J., Zhang, T., Li, D., Xu, F., Li, H., Pan, X., Liu, F., Zhao, Y., Weng, X."Behavioral video coding analysis of chronic morphine administration in rats". Biomedical Reports 21.5 (2024): 168.
Chicago
Yin, J., Zhang, T., Li, D., Xu, F., Li, H., Pan, X., Liu, F., Zhao, Y., Weng, X."Behavioral video coding analysis of chronic morphine administration in rats". Biomedical Reports 21, no. 5 (2024): 168. https://doi.org/10.3892/br.2024.1856