Behavioral tests for evaluating the characteristics of brain diseases in rodent models: Optimal choices for improved outcomes (Review)

  • Authors:
    • Lingyi Huang
    • Dongqiong Xiao
    • Hao Sun
    • Yi Qu
    • Xiaojuan Su
  • View Affiliations

  • Published online on: March 28, 2022     https://doi.org/10.3892/mmr.2022.12699
  • Article Number: 183
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Behavioral assessment is the dominant approach for evaluating whether animal models of brain diseases can successfully mimic the clinical characteristics of diseases. At present, most research regarding brain diseases involves the use of rodent models. While studies have reported numerous methods of behavioral assessments in rodent models of brain diseases, each with different principles, procedures, and assessment criteria, only few reviews have focused on characterizing and differentiating these methods based on applications for which they are most appropriate. Therefore, in the present review, the representative behavioral tests in rodent models of brain diseases were compared and differentiated, aiming to provide convenience for researchers in selecting the optimal methods for their studies.

Introduction

The term ‘brain disease’ encompasses various conditions, including brain injuries [e.g., stroke, white matter injury (WMI), and traumatic brain injury] (1), neurodegenerative diseases (e.g., Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis) (2), and affective disorders (e.g., depression and anxiety) (3), with the associated lesions mainly localized in the cortex, hippocampus, corpus callosum, and nerve nuclei (4). Patients with brain injuries and neurodegenerative diseases typically exhibit movement disorders and cognitive impairment (5,6), whereas those with affective disorders typically exhibit working memory deficits and impaired emotional processing (7). These conditions severely affect the quality of life of patients; however, the pathogenetic mechanisms underlying numerous brain diseases remain to be fully elucidated, and effective strategies for clinical treatment of such diseases are often lacking. Therefore, investigation of the pathogenesis and treatment of human brain diseases is of considerable clinical value. However, due to ethical and methodological limitations of experimentation involving human participants, the dominant approach for studying the nature, prevention, and treatment of human brain diseases involves the use of animal models.

Human brain diseases are mainly modeled in mice and rats, and considerable advancements have been made based on the data derived using these models (8,9). Despite such advancements, various testing methods with different principles, operational procedures, and assessment criteria have been used in animal research, and a specific optimal approach has not been generally accepted, to date. Selecting the optimal methods for investigating specific diseases will help in improving outcomes in both research and clinical settings. The methods used to assess brain disease in animal models can be generally categorized into pathological observation, specific marker identification, and behavioral performance assessment (10). Typically, behavioral tests are used to determine whether movement, cognition, working memory, and emotion have been affected, and such tests appear to be the most effective approach for evaluating whether animal models mimic the clinical characteristics of specific diseases (10,11).

The aim of the present review was to evaluate behavioral assessment methods for investigating the effects of brain diseases and relevant treatment strategies in animal models. First, the typical behavioral tests used in rodent models of brain diseases, including the Morris water maze (MWM) test, novel object recognition test, balance beam walking test, rotarod test, open field test (OFT), elevated plus-maze (EPM) test, tail suspension test (TST), and forced swimming test (FST), were summarized and reviewed. Then, the advantages and limitations of each approach were compared and recommendations that can aid researchers in selecting the optimal methods for their investigations were provided.

The present study was designed as a narrative review. It was performed by searching for the key words in databases (PubMed and Web of science) including ‘behavioral tests’, ‘brain diseases’, ‘rodent models’, and ‘behavioral assessments’. The studies searched were covered between 1947 and 2021. After reading the abstract, the studies that met with the scope of the present study were included and finally 104 studies were cited.

Principles, procedures, assessment, and application of different behavioral tests

MWM test

The MWM test was originally developed in 1982 by Morris et al, who sought to take advantage of the congenital abilities for spatial navigation and swimming in rodents. This test also relies on the innate drive of the animals to escape the water by locating and reaching the standing platform, which has been considered to reflect motivation for learning and memorization. Following its development, this test was immediately adopted as the standard method for investigating cognitive function based on the spatial memory and navigation abilities of the animal (12). Morris et al developed the test based on prior electrophysiological study showing that some cells in the hippocampus responded during the spatial learning and exploration phase, whereas other cells exhibited electrophysiological activity only when rodents entered a familiar environment (usually a specific and restricted area) (12). Moreover, damage to the hippocampus or decreases in the number of hippocampal synapses can lead to deficits in spatial learning and memory (12). Several recent studies have aimed to verify the theory that the hippocampus functions as a dynamic central hub for the hippocampal-cortical network, whose activation is considered to occur during episodic memory acquisition and retrieval in both humans and rodents (13,14). The capacity for episodic memory acquisition and retrieval of an animal is usually considered to reflect their ability to perceive spatial factors or cues, which are processed and consolidated afterward and are finally used to locate the standing platform in the MWM test (15). However, a previous study suggested that the spatial learning and navigation aspects of the MWM test performance do not solely rely on hippocampal activity but require significant involvement of cortical and subcortical regions (16). In addition, in another previous study it was reported that focal injuries to the medial thalamus impair the ability to adopt search strategies and swimming behavior without impacting spatial mapping and navigation performance (17). Furthermore, another study examined several novel variables and measures including a spatial learning index, which has greatly enhanced the ability to assess subtle differences in the MWM test performance (15). This index has considerably facilitated comparisons among groups and has aided correlation analyses with neurobiological markers or other behavioral measures (15). Moreover, one study demonstrated that the spatial learning index was sensitive enough to detect delicate behavioral alterations among aged individuals (15). Therefore, findings of studies using this spatial learning index have improved our understanding of age-related cognitive decline and cognitive function maintenance in aged individuals.

The equipment for the MWM test comprises three main elements: A large water tank (150 cm in diameter), an escape platform (15 cm in diameter), and a video monitor placed above the tank. The MWM test involves a navigation training stage, followed by a spatial exploration test to assess cognitive abilities (18). Adult animals are trained during the first 5–6 days. During training, the rodents are placed in the tank and allowed to search for the platform (typically 2 min for rats and 1 min for mice), and escape latency (i.e., the time required to find the platform) is recorded. The mean escape latency during the training stage is then used as a measure of the capacity of the animal to understand spatial information. After 5–6 days of training, animals undergo the spatial navigation test (18). First, the rodents are placed in the third quadrant and allowed to swim freely in the tank (without the platform) for 1 or 2 min, and the number of times they cross the position of the removed platform is recorded for further analysis (19) (Fig. 1).

In the MWM test, the average escape latency and the number of platform crossings are used to evaluate learning and memory ability (20). To improve the assessment, researchers have developed a novel parameter known as ‘proximity’, which is calculated as the frequency at which the rodent comes near the platform in 1 sec. This measure generates two additional variables, cumulative search error and average proximity, which are more sensitive in detecting group differences in behavior. In addition to their sensitivity, proximity measures require a small number of experimental animals and can increase the throughput of behavioral characterization facilities (21). Although proximity measures allow for improved quantification of navigation ability in the MWM task, other measures are still necessary. Accordingly, researchers have proposed the ‘learning index’ that can be used to associate spatial learning ability with other behavioral or neurobiological measures. The rodents are subjected to four trials, and the average proximity of the four probe trials is finally calculated as the learning index (22). In summary, the evaluation indices for the MWM test include the average escape latency (sec), number of platform crossings, cumulative search error, average proximity, and learning index (Fig. 1).

The MWM test is primarily designed to assess spatial learning and memory function, as these processes are considered to be similar in rodents and humans, particularly in terms of episodic memory ability. Therefore, the MWM test has been widely used and is well-recognized as a method for evaluating cognitive ability in experimental models of brain injuries such as WMI, stroke, and traumatic brain injury (2325). Moreover, as the ‘visuospatial navigation’ aspect of rodent performance is also reflected in ‘everyday cognitive’ processes in humans, the MWM test can be used to study neurodegenerative diseases characterized by impaired cognition, such as Alzheimer's disease or Parkinson's disease (26,27). Additional studies have shown that reversal learning and other aspects of cognitive flexibility rely on the prefrontal cortex in both humans and rodents (21,28). Therefore, the MWM test has also been used to assess the therapeutic effects of potential treatments on cognitive deficits in experimental models, which can provide critical information for clinical studies (Table I; 23-52).

Table I.

Applicable conditions for behavioral tests in rodent models of brain diseases.

Table I.

Applicable conditions for behavioral tests in rodent models of brain diseases.

Authors, yearBehavioral testsApplicable conditions(Refs.)
Kim H et al%, 2020Morris water maze testWhite matter injury(23)
Tucker LB et al%, 2018 Stroke(24)
Zhong JY et al%, 2017 Traumatic brain injury(25)
Schneider CB et al%, 2017 Alzheimer's disease(26)
Deng-Bryant et al%, 2016 Parkinson's disease(27)
Zhang R et al%, 2012Novel object recognition testAlzheimer's disease, aging, traumatic brain injury, schizophrenia(29,30)
Sadegzadeh F et al%, 2020
Chen W et al%, 2019Balance beam walking testWhite matter injury(31)
Uematsu A et al%, 2018 Age-related motor deficits(32,33)
Gyengesi E et al%, 2019 Central nervous system lesions
Mychasiuk R et al%, 2014 Huntington's disease(34)
El-Sahar AE et al%, 2020 Parkinson's disease(35)
Sun J et al%, 2021 Anxiety(36)
Marques-Carneiro JE et al%, 2014 Stroke(37)
Bohr A et al%, 2020 Multiple sclerosis(38)
Mitra NK et al%, 2020 (39)
Dong W et al%, 2020Rotarod testAmyotrophic lateral sclerosis(40)
Hayashi T et al%, 2017 Cerebellar ataxia(41,42)
Main SL et al%, 2017 Traumatic brain injury
Park G et al%, 2021 Stroke(43)
Owfard M et al%, 2021 (44)
Chkhartishvili E et al%, 2011Open field testDepressive disorder(45)
Lecorps B et al%, 2016 Anxiety-like behavior(46)
Su X et al%, 2020 White matter injury(47)
Shoi H et al%, 2021Elevated plus-maze testAnxiety-like behavior(48)
Ren C et al%, 2021Tail suspension testAnxiety(49)
Castagné V et al%, 2011 Depression(50)
Wang W et al%, 2021 White matter injury(51)
Ráez A et al%, 2020Forced swim testDepression(52)
Anxiety
Novel object recognition test

The novel object recognition test is another behavioral assessment method that is primarily associated with cognitive ability. It was originally developed by examining the natural tendency of rodents to explore novel objects (53). This test is unique in that it does not follow strict rules: The rodents only need to be familiar with the arena prior to testing, and the procedure is flexible and easy to follow (54,55).

The novel object recognition test comprises three stages: Habituation, training, and testing. On the 1st day (habituation stage), rodents are placed in a plastic chamber (35 cm in length ×35 cm in width ×35 cm in height) for 10 min for familiarization with the arena. On the 2nd day (training stage), two objects are placed symmetrically along the central line of the arena, and the rodents are allowed to examine the objects for 3 min. The duration of exploration is recorded for each object as an index of exploratory behavior. On the 3rd day (testing stage), the rodents are returned to their home cages for 3 min, and one of the objects is relocated to another adjacent quadrant, following which the rodents are allowed to explore the objects again. The time spent exploring the novel object is recorded and recorded as the recognition index (56,57) (Fig. 2).

At present, there are two widely accepted indices used to assess exploratory behavior during the test session. One is the novel object preference ratio, which is calculated by dividing the exploration time for the novel object by the exploration time for the total objects. A value of >0.5 indicates preference for the novel object, whereas a value of <0.5 indicates preference for the familiar object. The exploration time for each object is also used as an index of exploratory activity, such that the duration for which the nose is within 1 cm of the object in the novel location is recorded as the recognition index (58,59). Moreover, this test can be used to evaluate memory ability based on the time required to identify the novel objects (60) (Fig. 2).

Currently, this method is extensively used in studies investigating conditions associated with memory deficits, such as Alzheimer's disease, aging, traumatic brain injury, and schizophrenia, as it can help in evaluating the neurobiology of non-spatial memory in rodents (29,30) (Table I).

Balance beam walking test

It is widely accepted that rodents exhibit innate abilities for coordination and balance. Researchers have taken advantage of this characteristic to develop the balance beam walking test, which is used to assess motor balance and coordination ability in rodents with damage to the motor cortex (61,62). This test is advantageous in that it is easier to set up and is less expensive than the rotarod test (63).

The modified beam walking test equipment comprises a beam (80 cm in length, 0.5 cm in width, and 50 cm above the floor), with a lamp on one end and a box (non-transparent) on the other end and video-capturing equipment hanging above. First, for training, the beam equipment is placed in a dark and enclosed room, and the rodents are placed at the end of the beam containing the lamp. During the training phase, the rodents are allowed to walk 30, 50, and 70 cm for a maximum time of 60 sec. For each rodent, three trials per day are performed for 3 consecutive days. Then, during testing, the rodents are allowed to walk along the beam, similar to that in the training phase, and the following three metrics are used to evaluate performance: The frequency of hind limb slippage, the time spent on the beam, and the number of falls when walking the full distance (64,65) (Fig. 3).

The balance beam walking test performance is a useful measure of fine coordination and balance (31). The results are typically used to determine a beam walking performance index, which is calculated using the frequency of hind limb slippage, the time spent walking along the beam, the number of falls when walking the full distance of the beam, the distance traveled within the set time, the number of left and right turns, and the number of left and right paw slips (66,67) (Fig. 3).

Since its development and widespread acceptance, the balance beam walking test has been primarily used in studies of age-related motor deficits (32,33), central nervous system lesions (34), and genetic and pharmacological manipulations (68). The test has also been used to assess models of WMI (31), Huntington's disease (35), Parkinson's disease (36), anxiety (37), stroke (38), and multiple sclerosis (39) (Table I).

Rotarod test

The rotarod test represents another widely accepted and utilized method for evaluating motor coordination and balance in rodents, and both the balance beam walking test and the rotarod test share nearly identical principles (69). The rotarod test is unique, in that it is useful in evaluating endurance in rodents and is especially sensitive to cerebellar disorders (70). Researchers have primarily taken advantage of the ability of this test to assess motor coordination to investigate the sedative properties of novel drugs and determine their clinical value (71). However, researchers have also begun to realize that the test is associated with certain shortcomings. First, drug efficacy can differ between animal experiments and clinical settings, with some drugs exhibiting high sensitivity in rodents but insufficient sensitivity in humans (72). For example, administration of benzodiazepines or bretazenil exerts nearly no effect on mouse rotarod performance, although it can lead to excessive sedation in humans (63). Second, since most young adult mice can maintain balance during the testing interval (60 sec) even at a high speed (e.g., 44 rpm), researchers have argued that the rotarod test should not be used to evaluate whether motor coordination or balance ability has improved (73).

The rotarod is an automated apparatus comprising a cylindrical rotating beam connected to a computer. Before the trial, the rotarod is switched on with a starting rate of 4 rpm, and the computer is checked to ensure that it is properly connected for data recording. During the trial (generally lasting for 5 min), rodents are allowed to walk on the rotating rod (4 rpm) so that they can learn the motor coordination skills required for the activity. The rotation speed is then slowly and incrementally increased up to 40 rpm. The trial comes to an end when the tested rodent touches the magnetized pressure sensor upon falling from the rod. Three trials are conducted for each rodent, and the best trial result of each rodent is recorded as the score for that day (69,70) (Fig. 4).

The rotarod test is the dominant method for evaluating balancing ability in rodents (70). The test includes two stages: A constant speed stage and an accelerating speed stage. The constant speed stage is used to estimate muscle strength, whereas the accelerating speed stage is used to assess coordination, endurance, and muscular power (74). The rotarod test not only measures the maximum rotation speed at which the animal can maintain balance for a given running duration (e.g., 30 sec) but also calculates the latency to fall from the rod at different speeds and distances traveled. These are recorded as indices of motor coordination and balance performance, respectively (70). However, the surface and diameter of the rod as well as the rodents' body weight and physiological (e.g., fatigue) and biochemical parameters should be considered because they may influence the test results (69) (Fig. 4).

Given the extensive evidence accumulated thus far, researchers generally agree that the test can be used to assess sensorimotor abilities in several animal models, including those of amyotrophic lateral sclerosis (40), cerebellar ataxia disorders (41,42), traumatic brain injury (43), and stroke (44) (Table I).

OFT

While the balance beam walking test and rotarod test are widely used to evaluate motor coordination, the OFT is specifically used to assess overall locomotor function in rodents. Although it was initially developed to assess ‘timidity’ in mice based on defecation (75,76), the test takes advantage of the ability of the rodent to perceive new surroundings. The test may seem contradictory, given that rodents can exhibit two types of behaviors when entering a new setting (i.e., exploration of new settings and escaping from the bright/exposed area due to fear) (77). However, rodent responses are assessed by monitoring movement parameters when the rodent enters a new open field, which can help in determining the general pattern of locomotor activity (78), the exploratory ability (79), and the level of fear (80).

The OFT requires an open field and a video computer system. The field is a box (70 cm in width ×70 cm in length ×46 cm in height), wherein rodents are allowed to stay for 15 min for familiarization with the surroundings before starting the test. During the test, the rodents are placed in the center of the field and are allowed to move for 5 min. The distance traveled, the time spent in the center of the field, the level of spontaneous activity, and the number of entries into the central area are recorded. This test can be conducted either in dark or light settings (81,82) (Fig. 5).

Performance indices for the OFT include the distance traveled, the time spent moving, and the alterations in activity over time, which are integrated to determine the exploratory capacity of the animal (83). Additionally, the OFT can be used to assess the emotional state of a rodent by measuring the duration for which the animal remains stationary, the number of ‘depression-like’ episodes, and the escape activity. These variables are then integrated to determine the level of anxiety (84) (Fig. 5).

At present, the OFT is widely used to evaluate animal models of depressive disorders (45) and anxiety-like behavior (46). The test can also be used to assess animal models of WMI by measuring the distance traveled and the amount of time spent moving or being immobile in the central area or the periphery (47) (Table I).

EPM test

Although both the EPM test and the OFT are used to assess anxiety-like behavior, the principle of the EPM test differs from that of the OFT to some degree. The EPM test was originally devised based on the natural fondness of the rodents for dark and enclosed spaces, their fear of open areas and heights, and their desire to explore unfamiliar environments (48).

The EPM apparatus comprises two open arms (40×8 cm) positioned at right angles and two closed arms of the same size that are surrounded by black walls with a height of 30 cm. Before the test, rodents are subjected to single-frequency ultrasonic stimulation for 6 min to induce anxiety-like conditions. Following stimulation, each rodent is placed in the central platform of the maze facing an open arm and is allowed to freely explore the EPM for 5 min (85,86) (Fig. 6).

Performance indices in the EPM test include the number of arm entries and the duration spent in the open and closed arms. Entry into an arm is defined as the animal placing its two front paws inside an arm. Two additional measures can be derived as indices of anxiety: The percentage of open-arm entries and the percentage of the open-arm time. The percentage of time spent is calculated as follows: (Time spent in an arm/300 sec) × 100 (48) (Fig. 6). A higher percentage of open-arm time or open-arm entries indicates a lower level of anxiety (78).

Currently, the EPM test is primarily used to assess anxiety-like behaviors and anxiolytic drug properties in rodents (78,87) (Table I).

TST

Apart from the OFT and the EPM test, dominant methods for assessment of affective disorders include the TST and FST. The TST was developed based on the innate ability of rodents to respond to an external stimulus via a set of affective alterations; it is usually used to investigate depression-like behaviors in rodents and screen the effects of psychotropic drugs (e.g., antidepressants) (8891). Moreover, the TST has been used to investigate motor changes (i.e., motor coordination) in models of Parkinson's disease (92) and other metrics such as stress responses (93), helplessness (94), and anxiety (49).

The tail suspension apparatus mainly comprises a rectangular box (60 cm in total length × 40–55 cm in height × 15 cm in width) without external cues. During the test, rodents are suspended by their tails in each compartment for ~5 min, and the duration of immobility is calculated (95) (Fig. 7).

The TST performance is usually assessed over two periods: An escape stage and a stationary stage. Escape activity includes movements of the hind/forelimbs, movements of the head, and the number of attempts made per minute to reach the tail by bending the body and crawling upward. Immobility activity is defined as a lack of attempts made to rescue oneself. Both escape and immobility behaviors are recorded as the typical indices of the TST performance. Measurements of body temperature, including hyperthermia assessment, are also used as an index of emotional stress (96,97) (Fig. 7).

The TST was originally developed to investigate depression-like behavior (50). Currently, the TST is extensively used to assess animal models of depression and anxiety (49), and it has recently been applied to assess behavioral performance in models of WMI (51) (Table I).

FST

The FST is a dominant approach for investigating affective disorders and was originally developed based on the immobility response of rodents to external stimuli, which is considered to indicate ‘behavioral despair’ and a state of ‘depression’ (98). Researchers have demonstrated that immobility in the FST is achieved gradually through repeated failures, indicating that memory consolidation has occurred, which is associated with the role of the left dorsolateral striatum (99).

The FST apparatus comprises a small container with a visible escape platform and a video capture system, and the procedure includes two forced swimming sessions. On day 1, all the rodents are exposed to a 15-min pretest. On day 2, the rodents are placed in containers filled with water and are forced to swim for 6 min; the first 2 min represent the adaptation period (excluded from analysis), whereas the remaining 4 min are used to calculate the immobility time. Finally, behaviors (such as clawing at the edges of the container, aggressive swimming, and diving) and the number of escape attempts are recorded in the immobility condition (100) (Fig. 8).

The FST performance is typically determined based on the amount of time the animal spends in being immobile, swimming, drifting, diving, and sinking. Other measurements, such as the number of paw strokes, the swimming speed, the number of platform crossings, and uncoordinated swimming movements, are also used to evaluate locomotor ability (52,101) (Fig. 8).

Initially, this test was primarily used to assess depression-like behavior based on behavioral despair and motor behavior (52). However, the results can be influenced by changes in motor activity, thus producing false-positive results in drug screenings (102). Therefore, the original paradigm and its analysis have been modified for the screening of potential antidepressant drugs, and researchers have reached an agreement that the FST can be used in animal models of depression and anxiety (102) (Table I).

Discussion

In this review, the principles, procedures, evaluation, and applications of representative methods for behavioral assessment in rodent models of brain diseases were comprehensively analyzed. As illustrated above, all these methods were developed by taking advantage of the innate features of rodents, with each method based on different principles and performance indices. For example, the MWM test, novel object recognition test, balance beam walking test, and rotarod test are preferred for assessing brain injuries and neurodegenerative diseases, whereas the OFT, EPM test, TST, and FST are preferred for assessing affective disorders. These preferences are based on the expected alterations in cognitive function, motor coordination, and emotional states associated with the disease being modeled. For example, the MWM test is usually preferred when assessing models of WMI because the pathological changes associated with WMI occur in brain areas related to the principles of the MWM test (Fig. S1) (Table II).

Table II.

Behavioral tests used in rodent models of brain diseases.

Table II.

Behavioral tests used in rodent models of brain diseases.

Author, yearRodent modelsBehavioral tests(Refs.)
Kim H et al%, 2020White matter injuryMorris water maze test(23)
Chen W et al%, 2019 Balance beam walking test(31)
Su X et al%, 2020 Open field test(47)
Wang W et al%, 2021 Tail suspension test(51)
Tucker LB et al%, 2018StrokeMorris water maze test(24)
Bohr A et al%, 2020 Balance beam walking test(38)
Owfard M et al%, 2021 Rotarod test(44)
Zhong JY et al%, 2017Traumatic brain injuryMorris water maze test(25)
Zhang R et al%, 2012 Novel object recognition test(29,30)
Sadegzadeh F et al%, 2020 Rotarod test
Park G et al%, 2021 (43)
Schneider CB et al%, 2017Alzheimer's diseaseMorris water maze test(26)
Zhang R et al%, 2012 Novel object recognition test(29,30)
Sadegzadeh F et al%, 2020
Deng-Bryant et al%, 2016Parkinson's diseaseMorris water maze test(27)
Sun J et al%, 2021 Balance beam walking test(36)
Zhang R et al%, 2012SchizophreniaNovel object recognition test(29,30)
Sadegzadeh F et al%, 2020
Marques-Carneiro JE et al, 2014AnxietyBalance beam walking test(37)
Lecorps B et al%, 2016 Open field test(46)
Shoi H et al%, 2021 Elevated plus-maze test(48)
Ren C et al%, 2021 Tail suspension test(49)
Ráez A et al%, 2020 Forced swim test(52)
Mitra NK et al%, 2020Multiple sclerosisBalance beam walking test(39)
Dong W et al%, 2020 Rotarod test(40)
Chkhartishvili E et al%, 2011Depressive disorderOpen field test(45)
Castagné V et al%, 2011 Tail suspension test(50)
Ráez A et al%, 2020 Forced swim test(52)

Importantly, each test has both advantages and limitations. For example, the MWM test is a versatile tool that can be used to evaluate cognitive deficits associated with brain injuries and neurodegenerative diseases; however, it requires extensive setup, strict procedures, and long experimental duration (7 days) compared with the novel object recognition test, which is more flexible and easier to follow. Moreover, although both these tests are used to assess cognitive ability, the MWM test is considered to be more reflective of spatial learning and memory (e.g., WMI), whereas the novel object recognition test is considered to be more reflective of non-spatial memory (e.g., Alzheimer's disease). Furthermore, while the balance beam walking test is easier to set up and lower in cost than the rotarod test, it requires a longer training period (2 days). Moreover, although both these tests are used to evaluate motor coordination and balance ability, the balance beam walking test exhibits improved sensitivity for detecting motor coordination deficits compared with the rotarod test. Nonetheless, the rotarod test is more effective in evaluating endurance and disorders that affect the cerebellum. In terms of affective disorders, although both the OFT and EPM test are used to assess anxiety-like behavior, a previous study has indicated that the walls in the EPM test form visual barriers that may affect the performance results (103). Furthermore, while the OFT, TST, and FST are used for screening depression-like behavior, the OFT is more reflective of ‘exploratory fear’ behavior, whereas the TST is more reflective of depression induced by ‘stress reactivity’. The TST is also simpler, more drug sensitive, and more reliable than the FST, particularly in response to selective serotonin reuptake inhibitors (104). Importantly, the FST cannot be used for antidepressant drug screening, given that it can be influenced by changes in motor activity that lead to false-positive results (102) (Fig. S1) (Table II).

Conclusion

After making a comprehensive comparison of these behavioral tests, it was found that each test could be used to evaluate more than one kind of disease animal models. However, using only a single test might not precisely reflect the characteristic of a specific disease. Therefore, at present, lack of the specific behavioral approaches for assessing specific disease animal models is a key problem that needs to be solved. Thus, developing new behavioral tests or modifying the available tests which will concisely reflect the specific animal models would be a future research direction.

In summary, our review of the preferred settings, advantages, and limitations of various behavioral assessment methods may aid researchers in selecting the optimal strategies based on their research aims, which will in turn help in improving the reliability of their experimental results.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (grant nos. 81971428 and 81771634).

Availability of data and materials

Data sharing is not applicable to this article, as no data sets were generated or analyzed during the current study.

Authors' contributions

XS and LH contributed to the conception and design of the review and drafted the manuscript. DX and HS critically reviewed the article for important intellectual content. YQ gave important suggestions for the writing of the review. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sun P, Liu DZ, Jickling GC, Sharp FR and Yin KJ: MicroRNA-based therapeutics in central nervous system injuries. J Cereb Blood Flow Metab. 38:1125–1148. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Garofalo M, Pandini C, Bordoni M, Pansarasa O, Rey F, Costa A, Minafra B, Diamanti L, Zucca S, Carelli S, et al: Alzheimer's, Parkinson's disease and amyotrophic lateral sclerosis gene expression patterns divergence reveals different grade of RNA metabolism involvement. Int J Mol Sci. 21:95002020. View Article : Google Scholar : PubMed/NCBI

3 

Bednarova A, Cizmarikova M, Habalova V and Jarcuskova D: Evaluation of 5-HTTLPR (insertion/deletion) and BDNF (rs6265) genetic variations in the Slovakian individuals suffering from affective disorders. General Physiol Biophys. 40:365–376. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Velosky AG, Tucker LB, Fu AH, Liu J and McCabe JT: Cognitive performance of male and female C57BL/6J mice after repetitive concussive brain injuries. Behav Brain Res. 324:115–124. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Zlokovic BV, Gottesman RF, Bernstein KE, Seshadri S, McKee A, Snyder H, Greenberg SM, Yaffe K, Schaffer CB, Yuan C, et al: Vascular contributions to cognitive impairment and dementia (VCID): A report from the 2018 national heart, lung, and blood institute and national institute of neurological disorders and stroke workshop. Alzheimers Dement. 16:1714–1733. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Yakovleva OV, Poluektov MG, Lyashenko EA and Levin OS: Sleep and cognitive impairment in neurodegenerative diseases. Zh Nevrol Psikhiatr Im SS Korsakova. 119:89–98. 2019.(In Russian). View Article : Google Scholar : PubMed/NCBI

7 

Dehn LB and Beblo T: Depressed, biased, forgetful: The interaction of emotional and cognitive dysfunctions in depression. Neuropsychiatr. 33:123–130. 2019.(In German). View Article : Google Scholar : PubMed/NCBI

8 

Hamdy N, Eide S, Sun HS and Feng ZP: Animal models for neonatal brain injury induced by hypoxic ischemic conditions in rodents. Exp Neurol. 334:1134572020. View Article : Google Scholar : PubMed/NCBI

9 

Wintler T, Schoch H, Frank MG and Peixoto L: Sleep, brain development, and autism spectrum disorders: Insights from animal models. J Neurosci Res. 98:1137–1149. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Leader RW and Padgett GA: The genesis and validation of animal models. Am J Pathol. 101 (Suppl 3):S11–S16. 1980.PubMed/NCBI

11 

Cox TC: Utility and limitations of animal models for the functional validation of human sequence variants. Mol Genet Genomic Med. 3:375–382. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Morris RG, Garrud P, Rawlins JN and O'Keefe J: Place navigation impaired in rats with hippocampal lesions. Nature. 297:681–683. 1982. View Article : Google Scholar : PubMed/NCBI

13 

Chao OY, de Souza Silva MA, Yang YM and Huston JP: The medial prefrontal cortex-hippocampus circuit that integrates information of object, place and time to construct episodic memory in rodents: Behavioral, anatomical and neurochemical properties. Neurosci Biobehav Rev. 113:373–407. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Sabariego M, Tabrizi NS, Marshall GJ, McLagan AN, Jawad S and Hales JB: In the temporal organization of episodic memory, the hippocampus supports the experience of elapsed time. Hippocampus. 31:46–55. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Pereira IT and Burwell RD: Using the spatial learning index to evaluate performance on the water maze. Behav Neurosci. 129:533–539. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Urbach A, Baum E, Braun F and Witte OW: Cortical spreading depolarization increases adult neurogenesis, and alters behavior and hippocampus-dependent memory in mice. J Cereb Blood Flow Metab. 37:1776–1790. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Holmberg P, Liljequist S and Wägner A: Secondary brain injuries in thalamus and hippocampus after focal ischemia caused by mild, transient extradural compression of the somatosensori cortex in the rat. Curr Neurovasc Res. 6:1–11. 2009. View Article : Google Scholar : PubMed/NCBI

18 

D'Hooge R and De Deyn PP: Applications of the Morris water maze in the study of learning and memory. Brain Res Brain Res Rev. 36:60–90. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Mulder GB and Pritchett K: The Morris water maze. Contemp Top Lab Anim Sci. 42:49–50. 2003.

20 

Barry DN and Commins S: A novel control condition for spatial learning in the Morris water maze. J Neurosci Methods. 318:1–5. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Shah D, Verhoye M, Van der Linden A and D'Hooge R: Acquisition of spatial search strategies and reversal learning in the Morris water maze depend on disparate brain functional connectivity in mice. Cereb Cortex. 29:4519–4529. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Yuan Z, Zhou H, Zhou N, Dong D, Chu Y, Shen J, Han Y, Chu XP and Zhu K: Dynamic evaluation indices in spatial learning and memory of rat vascular dementia in the Morris water maze. Sci Rep. 9:72242019. View Article : Google Scholar : PubMed/NCBI

23 

Kim H, Seo JS, Lee SY, Ha KT, Choi BT, Shin YI, Yun YJ and Shin HK: AIM2 inflammasome contributes to brain injury and chronic post-stroke cognitive impairment in mice. Brain Behav Immun. 87:765–776. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Tucker LB, Velosky AG and McCabe JT: Applications of the Morris water maze in translational traumatic brain injury research. Neurosci Biobehav Rev. 88:187–200. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Zhong JY, Magnusson KR, Swarts ME, Clendinen CA, Reynolds NC and Moffat SD: The application of a rodent-based morris water maze (MWM) protocol to an investigation of age-related differences in human spatial learning. Behav Neurosci. 131:470–482. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Schneider CB, Linse K, Schönfeld R, Brown S, Koch R, Reichmann H, Leplow B and Storch A: Spatial learning deficits in Parkinson's disease with and without mild cognitive impairment. Parkinsonism Relat Disord. 36:83–88. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Deng-Bryant Y, Leung LY, Caudle K, Tortella F and Shear D: Cognitive evaluation using Morris water maze in neurotrauma. Methods Mol Biol. 1462:539–551. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Britten RA, Duncan VD, Fesshaye A, Rudobeck E, Nelson GA and Vlkolinsky R: Altered cognitive flexibility and synaptic plasticity in the rat prefrontal cortex after exposure to low (≤15 cGy) doses of 28Si radiation. Radiat Res. 193:223–235. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Zhang R, Xue G, Wang S, Zhang L, Shi C and Xie X: Novel object recognition as a facile behavior test for evaluating drug effects in AβPP/PS1 Alzheimer's disease mouse model. J Alzheimers Dis. 31:801–812. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Sadegzadeh F, Sakhaie N, Dehghany R, Adak O and Saadati H: Effects of adolescent administration of fluoxetine on novel object recognition memory, anxiety-like behaviors, and hippocampal brain-derived neurotrophic factor level. Life Sci. 260:1183382020. View Article : Google Scholar : PubMed/NCBI

31 

Chen W, Xia M, Guo C, Jia Z, Wang J, Li C, Li M, Tang X, Hu R, Chen Y, et al: Modified behavioural tests to detect white matter injury-induced motor deficits after intracerebral haemorrhage in mice. Sci Rep. 9:169582019. View Article : Google Scholar : PubMed/NCBI

32 

Uematsu A, Tsuchiya K, Suzuki S and Hortobágyi T: Cognitive dual-tasking augments age-differences in dynamic balance quantified by beam walking distance: A pilot study. Exp Gerontol. 114:27–31. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Gyengesi E, Rangel A, Ullah F, Liang H, Niedermayer G, Asgarov R, Venigalla M, Gunawardena D, Karl T and Münch G: Chronic microglial activation in the GFAP-IL6 mouse contributes to age-dependent cerebellar volume loss and impairment in motor function. Front Neurosci. 13:3032019. View Article : Google Scholar : PubMed/NCBI

34 

Mychasiuk R, Farran A and Esser MJ: Assessment of an experimental rodent model of pediatric mild traumatic brain injury. J Neurotrauma. 31:749–757. 2014. View Article : Google Scholar : PubMed/NCBI

35 

El-Sahar AE, Rastanawi AA, El-Yamany MF and Saad MA: Dapagliflozin improves behavioral dysfunction of Huntington's disease in rats via inhibiting apoptosis-related glycolysis. Life Sci. 257:1180762020. View Article : Google Scholar : PubMed/NCBI

36 

Sun J, Li H, Jin Y, Yu J, Mao S, Su KP, Ling Z and Liu J: Probiotic Clostridium butyricum ameliorated motor deficits in a mouse model of Parkinson's disease via gut microbiota-GLP-1 pathway. Brain Behav Immun. 91:703–715. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Marques-Carneiro JE, Faure JB, Cosquer B, Koning E, Ferrandon A, de Vasconcelos AP, Cassel JC and Nehlig A: Anxiety and locomotion in genetic absence epilepsy rats from strasbourg (GAERS): Inclusion of Wistar rats as a second control. Epilepsia. 55:1460–1468. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Bohr A, Schuhmann MK, Papp L, Volkmann J and Fluri F: Deep brain stimulation for stroke: Continuous stimulation of the pedunculopontine tegmental nucleus has no impact on skilled walking in rats after photothrombotic stroke. Curr Neurovasc Res. 17:636–643. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Mitra NK, Xuan KY, Teo CC, Xian-Zhuang N, Singh A and Chellian J: Evaluation of neuroprotective effects of alpha-tocopherol in cuprizone-induced demyelination model of multiple sclerosis. Res Pharm Sci. 15:602–611. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Dong W and Zhang L, Sun C, Gao X, Guan F, Li J, Chen W, Ma Y and Zhang L: Knock in of a hexanucleotide repeat expansion in the C9orf72 gene induces ALS in rats. Animal Model Exp Med. 3:237–244. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Hayashi T, Onozato T, Wanajo I, Hayashi M, Takeda H and Fujimori Y: Longitudinal analysis of motor symptoms and histopathology in woozy mice, a model of cerebellar ataxia. Neuroreport. 28:779–787. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Main SL and Kulesza RJ: Repeated prenatal exposure to valproic acid results in cerebellar hypoplasia and ataxia. Neuroscience. 340:34–47. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Park G, Suh JH and Han SJ: Transcranial direct current stimulation for balance and gait in repetitive mild traumatic brain injury in rats. BMC Neurosci. 22:262021. View Article : Google Scholar : PubMed/NCBI

44 

Owfard M, Bigdeli MR, Safari A, Haghani M and Namavar MR: Effect of dimethyl fumarate on the motor function and spatial arrangement of primary motor cortical neurons in the sub-acute phase of stroke in a rat model. J Stroke Cerebrovasc Dis. 30:1056302021. View Article : Google Scholar : PubMed/NCBI

45 

Chkhartishvili E, Maglakelidze N, Babilodze M, Chijavadze E and Nachkebia N: Changes of open field behavior in animal model of depression. Georgian Med News. 11:107–112. 2011.PubMed/NCBI

46 

Lecorps B, Rödel HG and Féron C: Assessment of anxiety in open field and elevated plus maze using infrared thermography. Physiol Behav. 157:209–216. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Su X, Yuan H, Bai Y, Chen J, Sui M, Zhang X, Liang Y, Feng W, Dou Z and Zhu H: Clobetasol attenuates white matter injury by promoting oligodendrocyte precursor cell differentiation. Pediatr Neurosurg. 55:188–196. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Shoji H and Miyakawa T: Effects of test experience, closed-arm wall color, and illumination level on behavior and plasma corticosterone response in an elevated plus maze in male C57BL/6J mice: A challenge against conventional interpretation of the test. Mol Brain. 14:342021. View Article : Google Scholar : PubMed/NCBI

49 

Ren C, Li LX, Dong AQ, Zhang YT, Hu H, Mao CJ, Wang F and Liu CF: Depression induced by chronic unpredictable mild stress increases susceptibility to Parkinson's disease in mice via neuroinflammation mediated by P2X7 receptor. ACS Chem Neurosci. 12:1262–1272. 2021. View Article : Google Scholar : PubMed/NCBI

50 

Castagné V, Moser P, Roux S and Porsolt RD: Rodent models of depression: Forced swim and tail suspension behavioral despair tests in rats and mice. Curr Protoc Neurosci. 8:Unit 8.10A. 2011.PubMed/NCBI

51 

Wang W, Wang J, Tang Q, Zhu X, Zhu R, Cui D, Wei C, Liu X, Liu X, Ran S, et al: CX3CR1 deficiency aggravates brain white matter injury and affects expression of the CD36/15LO/NR4A1 signal. Biochem Biophys Res Commun. 549:47–53. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Ráez A, Oliveras I, Río-Álamos C, Díaz-Morán S, Cañete T, Blázquez G, Tobeña A and Fernández-Teruel A: A missing link between depression models: Forced swimming test, helplessness and passive coping in genetically heterogeneous NIH-HS rats. Behav Processes. 177:1041422020. View Article : Google Scholar : PubMed/NCBI

53 

Richler JJ, Wilmer JB and Gauthier I: General object recognition is specific: Evidence from novel and familiar objects. Cognition. 166:42–55. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Ishikawa H, Yamada K, Pavlides C and Ichitani Y: Sleep deprivation impairs spontaneous object-place but not novel-object recognition in rats. Neurosci Lett. 580:114–118. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Miedel CJ, Patton JM, Miedel AN, Miedel ES and Levenson JM: Assessment of spontaneous alternation, novel object recognition and limb clasping in transgenic mouse models of amyloid-β and tau neuropathology. J Vis Exp. 28:555232017.

56 

Antunes M and Biala G: The novel object recognition memory: Neurobiology, test procedure, and its modifications. Cogn Process. 13:93–110. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Grayson B, Leger M, Piercy C, Adamson L, Harte M and Neill JC: Assessment of disease-related cognitive impairments using the novel object recognition (NOR) task in rodents. Behav Brain Res. 285:176–193. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Cohen SJ and Stackman RW Jr: Assessing rodent hippocampal involvement in the novel object recognition task. A review. Behav Brain Res. 285:105–117. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Da Cruz JFO, Gomis-Gonzalez M, Maldonado R, Marsicano G, Ozaita A and Busquets-Garcia A: An alternative maze to assess novel object recognition in mice. Bio Protoc. 10:e36512020.PubMed/NCBI

60 

Lueptow LM: Novel object recognition test for the investigation of learning and memory in mice. J Vis Exp. 126:557182017.PubMed/NCBI

61 

Luong TN, Carlisle HJ, Southwell A and Patterson PH: Assessment of motor balance and coordination in mice using the balance beam. J Vis Exp. 49:23762011.

62 

Hortobágyi T, Uematsu A, Sanders L, Kliegl R, Tollár J, Moraes R and Granacher U: Beam walking to assess dynamic balance in health and disease: A protocol for the ‘BEAM’ multicenter observational study. Gerontology. 65:332–339. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Stanley JL, Lincoln RJ, Brown TA, McDonald LM, Dawson GR and Reynolds DS: The mouse beam walking assay offers improved sensitivity over the mouse rotarod in determining motor coordination deficits induced by benzodiazepines. J Psychopharmacol. 19:221–227. 2005. View Article : Google Scholar : PubMed/NCBI

64 

Seashore HG: The development of a beam-walking test and its use in measuring development of balance in children. Res Q. 18:246–259. 1947.PubMed/NCBI

65 

Sawers A and Hafner B: Validation of the narrowing beam walking test in lower limb prosthesis users. Arch Phys Med Rehabil. 99:1491–1498.e1. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Castillo JT, Welch GW and Sarver CM: Walking a high beam: The balance between employment stability, workplace flexibility, and nonresident father involvement. Am J Mens Health. 6:120–131. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Sawers A and Ting LH: Beam walking can detect differences in walking balance proficiency across a range of sensorimotor abilities. Gait Posture. 41:619–623. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Kaur H, Kumar A, Jaggi AS and Singh N: Pharmacologic investigations on the role of Sirt-1 in neuroprotective mechanism of postconditioning in mice. J Surg Res. 197:191–200. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Monville C, Torres EM and Dunnett SB: Comparison of incremental and accelerating protocols of the rotarod test for the assessment of motor deficits in the 6-OHDA model. J Neurosci Methods. 158:219–223. 2006. View Article : Google Scholar : PubMed/NCBI

70 

Shiotsuki H, Yoshimi K, Shimo Y, Funayama M, Takamatsu Y, Ikeda K, Takahashi R, Kitazawa S and Hattori N: A rotarod test for evaluation of motor skill learning. J Neurosci Methods. 189:180–185. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Matias M, Silvestre S, Falcão A and Alves G: Considerations and pitfalls in selecting the drug vehicles for evaluation of new drug candidates: Focus on in vivo pharmaco-toxicological assays based on the rotarod performance test. J Pharm Pharm Sci. 21:110–118. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Kirschbaum KM, Hiemke C and Schmitt U: Rotarod impairment: Catalepsy-like screening test for antipsychotic side effects. Int J Neurosci. 119:1509–1522. 2009. View Article : Google Scholar : PubMed/NCBI

73 

Tkáč I, Benneyworth MA, Nichols-Meade T, Steuer EL, Larson SN, Metzger GJ and Uğurbil K: Long-term behavioral effects observed in mice chronically exposed to static ultra-high magnetic fields. Magn Reson Med. 86:1544–1559. 2021. View Article : Google Scholar : PubMed/NCBI

74 

Toklu HZ, Yang Z, Ersahin M and Wang KKW: Neurological exam in rats following stroke and traumatic brain injury. Methods Mol Biol. 2011:371–381. 2019. View Article : Google Scholar : PubMed/NCBI

75 

Jeffery ND, Brakel K, Aceves M, Hook MA and Jeffery UB: Variability in open-field locomotor scoring following force-defined spinal cord injury in rats: Quantification and implications. Front Neurol. 11:6502020. View Article : Google Scholar : PubMed/NCBI

76 

Miller CK, Halbing AA, Patisaul HB and Meitzen J: Interactions of the estrous cycle, novelty, and light on female and male rat open field locomotor and anxiety-related behaviors. Physiol Behav. 228:1132032021. View Article : Google Scholar : PubMed/NCBI

77 

Sturman O, Germain PL and Bohacek J: Exploratory rearing: A context- and stress-sensitive behavior recorded in the open-field test. Stress. 21:443–452. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Kraeuter AK, Guest PC and Sarnyai Z: The open field test for measuring locomotor activity and anxiety-like behavior. Methods Mol Biol. 1916:99–103. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Tartar JL, Ward CP, Cordeira JW, Legare SL, Blanchette AJ, McCarley RW and Strecker RE: Experimental sleep fragmentation and sleep deprivation in rats increases exploration in an open field test of anxiety while increasing plasma corticosterone levels. Behav Brain Res. 197:450–453. 2009. View Article : Google Scholar : PubMed/NCBI

80 

Walz N, Mühlberger A and Pauli P: A human open field test reveals thigmotaxis related to agoraphobic fear. Biol Psychiatry. 80:390–397. 2016. View Article : Google Scholar : PubMed/NCBI

81 

Walsh RN and Cummins RA: The open-field test: A critical review. Psychol Bull. 83:482–504. 1976. View Article : Google Scholar : PubMed/NCBI

82 

Knight P, Chellian R, Wilson R, Behnood-Rod A, Panunzio S and Bruijnzeel AW: Sex differences in the elevated plus-maze test and large open field test in adult Wistar rats. Pharmacol Biochem Behav. 204:1731682021. View Article : Google Scholar : PubMed/NCBI

83 

Justel N, Salguero A, Marengo L, Psyrdellis M and Pautassi RM: Open field exposure facilitates the expression of a spatial, recognition memory. Neurosci Lett. 757:1359972021. View Article : Google Scholar : PubMed/NCBI

84 

Snyder CN, Brown AR and Buffalari D: Similar tests of anxiety-like behavior yield different results: Comparison of the open field and free exploratory rodent procedures. Physiol Behav. 230:1132462021. View Article : Google Scholar : PubMed/NCBI

85 

Hogg S: A review of the validity and variability of the elevated plus-maze as an animal model of anxiety. Pharmacol Biochem Behav. 54:21–30. 1996. View Article : Google Scholar : PubMed/NCBI

86 

Carobrez AP and Bertoglio LJ: Ethological and temporal analyses of anxiety-like behavior: The elevated plus-maze model 20 years on. Neurosci Biobehav Rev. 29:1193–1205. 2005. View Article : Google Scholar : PubMed/NCBI

87 

Bespalov A and Steckler T: Pharmacology of anxiety or pharmacology of elevated plus maze? Biol Psychiatry. 89:e732021. View Article : Google Scholar : PubMed/NCBI

88 

Holmes A, Li Q, Koenig EA, Gold E, Stephenson D, Yang RJ, Dreiling J, Sullivan T and Crawley JN: Phenotypic assessment of galanin overexpressing and galanin receptor R1 knockout mice in the tail suspension test for depression-related behavior. Psychopharmacology (Berl). 178:276–285. 2005. View Article : Google Scholar : PubMed/NCBI

89 

Shao S, Cui Y, Chen ZB, Zhang B, Huang SM and Liu XW: Androgen deficit changes the response to antidepressant drugs in tail suspension test in mice. Aging Male. 23:1259–1265. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Iyer KA, Alix K, Eltit JM, Solis E Jr, Pan X, Argade MD, Khatri S, Felice LJD, Sweet DH, Schulte MK and Dukat M: Multi-modal antidepressant-like action of 6- and 7-chloro-2-aminodihydroquinazolines in the mouse tail suspension test. Psychopharmacology (Berl). 236:2093–2104. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Poleszak E, Szopa A, Bogatko K, Wyska E, Wośko S, Świader K, Doboszewska U, Wlaź A, Wróbel A, Wlaź P and Serefko A: Antidepressant-like activity of typical antidepressant drugs in the forced swim test and tail suspension test in mice is augmented by DMPX, an adenosine A2A receptor antagonist. Neurotox Res. 35:344–352. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Pelloux Y, Hagues G, Costentin J and Duterte-Boucher D: Helplessness in the tail suspension test is associated with an increase in ethanol intake and its rewarding effect in female mice. Alcohol Clin Exp Res. 29:378–388. 2005. View Article : Google Scholar : PubMed/NCBI

93 

Kale PP, Addepalli V and Ghadawale SR: Impact of pre-exposure of tail suspension on behavioural parameters like locomotion, exploration, and anxiety in mice. Indian J Exp Biol. 51:732–738. 2013.PubMed/NCBI

94 

Reis-Silva TM, Sandini TM, Calefi AS, Orlando BCG, Moreira N, Lima APN, Florio JC, Queiroz-Hazarbassanov NGT and Bernardi MM: Stress resilience evidenced by grooming behaviour and dopamine levels in male mice selected for high and low immobility using the tail suspension test. Eur J Neurosci. 50:2942–2954. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Nomura S, Naruse R and Okada H: The tail suspension test: Its theory and practical application. Yakubutsu Seishin Kodo. 12:207–213. 1992.(In Japanese). PubMed/NCBI

96 

Rosa I, Di Censo D, Ranieri B, Di Giovanni G, Scarnati E, Alecci M, Galante A and Florio TM: Comparison between tail suspension swing test and standard rotation test in revealing early motor behavioral changes and neurodegeneration in 6-OHDA hemiparkinsonian rats. Int J Mol Sci. 21:28742020. View Article : Google Scholar : PubMed/NCBI

97 

Stukalin Y, Lan A and Einat H: Revisiting the validity of the mouse tail suspension test: Systematic review and meta-analysis of the effects of prototypic antidepressants. Neurosci Biobehav Rev. 112:39–47. 2020. View Article : Google Scholar : PubMed/NCBI

98 

Vieira C, De Lima TC, de Pádua Carobrez A and Lino-de-Oliveira C: Frequency of climbing behavior as a predictor of altered motor activity in rat forced swimming test. Neurosci Lett. 445:170–173. 2008. View Article : Google Scholar : PubMed/NCBI

99 

Flores-Serrano AG, Zaldivar-Rae J, Salgado H and Pineda JC: Immobility time during the forced swimming test predicts sensitivity to amitriptyline, whereas traveled distance in the circular corridor indicates resistance to treatment in female Wistar rats. Neuroreport. 26:233–238. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Petit-Demouliere B, Chenu F and Bourin M: Forced swimming test in mice: A review of antidepressant activity. Psychopharmacology (Berl). 177:245–255. 2005. View Article : Google Scholar : PubMed/NCBI

101 

Herbst LS, Gaigher T, Siqueira AA, Joca SRL, Sampaio KN and Beijamini V: New evidence for refinement of anesthetic choice in procedures preceding the forced swimming test and the elevated plus-maze. Behav Brain Res. 368:1118972019. View Article : Google Scholar : PubMed/NCBI

102 

Dang H, Chen Y, Liu X, Wang Q, Wang L, Jia W and Wang Y: Antidepressant effects of ginseng total saponins in the forced swimming test and chronic mild stress models of depression. Prog Neuropsychopharmacol Biol Psychiatry. 33:1417–1424. 2009. View Article : Google Scholar : PubMed/NCBI

103 

Rebolledo-Solleiro D, Crespo-Ramírez M, Roldán-Roldán G, Hiriart M and de la Mora M: Role of thirst and visual barriers in the differential behavior displayed by streptozotocin-treated rats in the elevated plus-maze and the open field test. Physiol Behav. 120:130–135. 2013. View Article : Google Scholar : PubMed/NCBI

104 

Lucki I, Dalvi A and Mayorga AJ: Sensitivity to the effects of pharmacologically selective antidepressants in different strains of mice. Psychopharmacology (Berl). 155:315–322. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2022
Volume 25 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang L, Xiao D, Sun H, Qu Y and Su X: Behavioral tests for evaluating the characteristics of brain diseases in rodent models: Optimal choices for improved outcomes (Review). Mol Med Rep 25: 183, 2022
APA
Huang, L., Xiao, D., Sun, H., Qu, Y., & Su, X. (2022). Behavioral tests for evaluating the characteristics of brain diseases in rodent models: Optimal choices for improved outcomes (Review). Molecular Medicine Reports, 25, 183. https://doi.org/10.3892/mmr.2022.12699
MLA
Huang, L., Xiao, D., Sun, H., Qu, Y., Su, X."Behavioral tests for evaluating the characteristics of brain diseases in rodent models: Optimal choices for improved outcomes (Review)". Molecular Medicine Reports 25.5 (2022): 183.
Chicago
Huang, L., Xiao, D., Sun, H., Qu, Y., Su, X."Behavioral tests for evaluating the characteristics of brain diseases in rodent models: Optimal choices for improved outcomes (Review)". Molecular Medicine Reports 25, no. 5 (2022): 183. https://doi.org/10.3892/mmr.2022.12699