Q1.3.2分 Advancing Post-Stroke Depression Research Insights from
Q1.3.2分 Advancing Post-Stroke Depression Research Insights from
Q1.3.2分 Advancing Post-Stroke Depression Research Insights from
Review
Advancing Post-Stroke Depression Research: Insights from
Murine Models and Behavioral Analyses
Mădălina Iuliana Mus, at 1 , Bogdan Cătălin 1, * , Michael Hadjiargyrou 2, * , Aurel Popa-Wagner 1,3
and Andrei Gres, it, ă 1,4
1 Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of
Craiova, 200349 Craiova, Romania; madalina.musat3@gmail.com (M.I.M.);
aurel.popa-wagner@geriatrics-healthyageing.com (A.P.-W.)
2 Department of Biological and Chemical Sciences, New York Institute of Technology,
Old Westbury, NY 11568, USA
3 Department of Neurology, Vascular Neurology and Dementia, University of Medicine Essen,
45122 Essen, Germany
4 Department of Biomedical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
* Correspondence: bogdan.catalin@umfcv.ro (B.C.); mhadji@nyit.edu (M.H.)
and cognitive [19] dysfunction represent long-term consequences of ischemic injury, PSD
represents the most frequent and challenging neuropsychiatric complication [20,21].
Estimating the exact prevalence of PSD is challenging, particularly due to methodolog-
ical complexities, such as variations in the timing of patient evaluations after stroke onset
and differences in instruments and criteria utilized in experimental settings [21]. However,
recent findings suggest that PSD occurs in approximately 18–33% of cases [22,23], with
the greatest number of occurrences observed within the first year following the ischemic
insult [24]. Also, ~53% of individuals who were depressed within 3 months after stroke
experienced persistent depression [21]. These results are concerning, especially because
the mortality rate is higher among patients who suffer from PSD [22]. The risk of suici-
dal death is ~2 times higher for stroke patients compared to the general population [25],
and in addition to suicidal ideation [26], cognitive deficits [27], long-term disability [28],
and a substantially lower quality of life [29] are additional symptoms in patients who
develop PSD.
Thus, it is of paramount importance to understand the underlying mechanisms of
this disease and to accurately identify efficient screening tools and therapeutic modalities.
However, studying the pathopsychological mechanisms underlying PSD, such as affected
cellular plasticity [30], neuroinflammation, and neurodegeneration [31], as well as intrinsic
recovery pathways (neurogenesis) [32], requires a highly accurate and performant experi-
mental approach. But, establishing preclinical models can also pose challenges, particularly
given the subjective nature of the psychological and physiological PSD symptoms [33].
Researchers have endeavored to create rodent models that capture key aspects of PSD,
allowing for the evaluation of behavioral manifestations and underlying neurobiolog-
ical changes [34–36]. One such approach involves inducing focal cerebral ischemia in
rodents [37] and simulating the conditions of stroke observed in humans. Following
a stroke, protocols for inducing depressive behavior [38] and behavioral tests tailored
to assess depressive-like symptoms are used [39] and encompass a wide range of as-
says designed to evaluate various aspects of depressive behavior, including despair-like
behavior [40], anhedonia [41], and alterations in locomotor activity and exploration [42,43].
Despite recent progress in PSD-related research, challenges persist in accurately re-
producing the multifaceted nature of this condition in in vivo animal models. To advance
translational research and the development of new therapeutic treatments for PSD, it is cru-
cial to develop valid animal models that accurately replicate the complex symptomatology
of the condition. According to the International Classification of Diseases, 11th Revision
(ICD 11) criteria [44], psychiatrists diagnosing depression in patients typically confirm
the presence of five or more symptoms from the depressive spectrum (i.e., depressed
mood, anhedonia, appetite or weight changes, sleep disturbances, psychomotor agitation
or retardation, fatigue, reduced concentration, feelings of worthlessness or excessive guilt,
and recurrent thoughts of suicide or death) [45]. Accurate replication of the human con-
dition in these models is crucial for understanding the pathology and to effectively test
various therapeutic interventions. Variability in individual responses [46], differences in
genetic backgrounds [47–51], and the subjective nature of depressive symptoms [52,53]
all present challenges in interpreting the data effectively. Moreover, the environmental
and social factors that can influence the onset and progression of PSD in humans [54,55]
are difficult to replicate in murine models, limiting the validity and effectiveness of these
studies. The complexity of stroke-induced brain injury, which involves not just the neural
circuits traditionally associated with mood regulation [56,57] but also the broader neurolog-
ical disruptions [58], complicates the accuracy of modeling PSD. Additionally, the lack of
biomarkers or a valid genetic model for depression further contributes to the difficult task of
studying neuropsychiatric disorders, including PSD, in a preclinical setting [59]. These chal-
lenges highlight the need for continued refinement of animal models and methodologies to
enhance the translational potential of PSD preclinical research.
The purpose of this review was to conduct a thorough investigation of the current
literature on murine models for PSD, highlighting both the strengths and weaknesses of these
for depression further contributes to the difficult task of studying neuropsychiatric
disorders, including PSD, in a preclinical setting [59]. These challenges highlight the need
for continued refinement of animal models and methodologies to enhance the
translational potential of PSD preclinical research.
Life 2024, 14, 1110 The purpose of this review was to conduct a thorough investigation of the current
3 of 31
literature on murine models for PSD, highlighting both the strengths and weaknesses of
these models, as well as the behavioral assessments employed. Our goal is to identify
effective animal models and behavioral evaluations so that their inherent limitations can
models, as well as the behavioral assessments employed. Our goal is to identify effective
be overcome. Further, this review aims not only to underline the tools available for such
animal
studiesmodels
but alsoand behavioral
discuss evaluations
the challenges so that
faced their inherent
in translating theselimitations
preclinicalcan be overcome.
results into
Further, this
the clinic. review aims not only to underline the tools available for such studies but also
discuss the challenges faced in translating these preclinical results into the clinic.
2. Materials and Methods
2. Materials and Methods
We have used PubMed and ClinicalTrials.gov in order to identify a thorough range
We have
of relevant used PubMed
scientific manuscripts.and Importantly,
ClinicalTrials.gov in order
our search to identify
extended a thorough
to encompass animalrange
of relevant scientific manuscripts. Importantly, our search extended
models of PSD and various behavioral studies and provides a comprehensive overview to encompass animal
models of PSDused
of the methods and in
various
exploringbehavioral
PSD. Thestudies and provides
search terms employed a mostly
comprehensive overview
included “post-
of the methods used in exploring PSD. The search terms
stroke depression murine models” and “post-stroke depression murine behavioral employed mostly included
“post-stroke depression murine models” and “post-stroke depression
studies”. Also, to ensure thorough coverage, we used derived terms such as “vascular murine behavioral
studies”.
depression”, Also, to ensure tests’,
“behavioral thorough coverage,
“cognition in we used derived
rodents”, “social terms
activitysuch as “vascular
in rodents”,
depression”, “behavioral tests’, “cognition in rodents”, “social activity
“motor function in rodents”, “stroke in rodents”, “chronic mild stress”, “depressive-like in rodents”, “motor
function
behavior”, in and
rodents”,
“anxiety“stroke in rodents”,
and depression “chronic mild
assessments stress”,
in murine “depressive-like
models”. Additionally,behavior”,
we
and “anxiety
manually and depression
reviewed the referenceassessments
lists of allinsourced
murinearticles
models”. Additionally,
to uncover furtherwe manually
citations
reviewed the reference
that the initial databaselists of all
search sourced
might havearticles
missed.toAlthough
uncover additional
further citations that
searches the initial
yielded
database search of
a large number might have
results, wemissed.
focusedAlthough
on these additional
specific termssearches yieldedthe
to maintain a large number
relevance
of
andresults, we focused
manageability of on these specific
manuscripts. We terms to maintain
specifically chose the relevance
articles and manageability
published within the
of manuscripts.
past two decades,We specifically
spanning chose
from 2004 to articles
2024, thus published
ensuring within the past
the inclusion two decades,
of up-to-date
research findings.
spanning from 2004 A tolanguage
2024, thuscriterion wasthe
ensuring established
inclusiontoofonly considerresearch
up-to-date articles findings.
written A
in English. This review primarily adopts a narrative approach and
language criterion was established to only consider articles written in English. This does not adhere to the
review
Preferred Reporting Items for Systematic Reviews and Meta-Analysis
primarily adopts a narrative approach and does not adhere to the Preferred Reporting (PRISMA)
guidelines.
Items However, the
for Systematic article and
Reviews selection process was(PRISMA)
Meta-Analysis meticulously structured,However,
guidelines. ensuring the
that included articles were scrutinized by a panel of three independent
article selection process was meticulously structured, ensuring that included articles reviewers (Figurewere
1).
scrutinized by a panel of three independent reviewers (Figure 1).
Figure2.2. Diagram
Figure Diagramshowing
showingriskrisk
factors associated
factors with PSD.
associated withSerotonin transporter
PSD. Serotonin (SERT), brain-
transporter (SERT),
derived neurotrophic factor (BNF), apolipoprotein E (ApoE), methylenetetrahydrofolate reductase
brain-derived neurotrophic factor (BNF), apolipoprotein E (ApoE), methylenetetrahydrofolate reduc-
(MTHFR), and Protein Kinase C Eta Gene (PRKCH).
tase (MTHFR), and Protein Kinase C Eta Gene (PRKCH).
4.2. Demographic Factors
Demographic factors play a significant role in influencing the risk and occurrence of
PSD. Gender disparities exist in the prevalence of PSD, with women generally exhibiting
higher rates of depression following a stroke compared to men [63,72,73]. Hormonal
fluctuations, psychosocial factors, and differences in coping mechanisms may contribute
to these gender differences observed [72]. Socioeconomic factors, including education
level, income, and access to healthcare resources, also play a crucial role in the
Life 2024, 14, 1110 5 of 31
4.3. Age
Studies have shown that the incidence and severity of PSD increase with advancing age,
highlighting the importance of age-related considerations in its management [76,77]. The inci-
dence of PSD is especially high in elderly stroke survivors [78], with 34% in stroke patients vs.
13% in the matched general population [79]. As most candidate therapies for PSD are being
developed and studied on young animal models [34], age, which is proven to be detrimental
to recovery, may aid in ushering a certain degree of accuracy to existing murine models, thus
reducing the translational gap between preclinical and clinical studies [80]. Although a vast array
of risk factors such as diabetes [81], hypertension [82], obesity, or dyslipidemia [83] are linked to
ischemic brain injury, age represents a non-modifiable risk factor that is not only linked to an
increased susceptibility to stroke but also to significantly decreased functional recovery [76,77,84].
It is, therefore, considered that age is a key modulatory factor for both stroke and PSD.
programs [97], aquatic [98], music [99], cognitive behavioral therapy [100], repetitive transcra-
nial magnetic stimulation [101], pet therapy [102], and robotic devices [103], can all significantly
improve the quality of life and prevent or reduce PSD symptoms.
Days of
Stressors and Duration CUMS/Mice
Housing
Alterations of Food
Gaignier F., Illumination Soiled cage Paired 21 days
the Cage tilt deprivation Small cage
2018 at night overnight housing Individually
light-dark 1 h, 2 h, 15 h overnight 1 h, 2 h
[124] 15 h 15 h 2h housing
cycle 15 h
Food and Crowding:
Zhang M., Exposure to a Traffic noise Food Illumination Water 28 days
Cage tilt water ten mice per Level shaking
2023 stroboscope (70–90 dB) deprivation at night deprivation Individually
12 h deprivation cage 15 min
[125] 12 h 6h 12 h 12 h 12 h housing
24 h 12 h
Exposure to
Ice water Food Illumination Restraint Exposure to a Water 28 days
Yan W., 2021 Cage tilt an empty Soiled cage
swimming deprivation at night stress foreign object deprivation Group
[126] 5 min bottle 24 h
5 min 24 h 12 h 2h 24 h 24 h housing
1h
Ice water Foot electric Food Restraint Water 35 days
Wu J., 2021 Cage tilt Continuous illumination Wet bedding Tail-clamp Cage shaking
swimming shock deprivation stress deprivation Individually
[128] 24 h 24 h 24 h 90 s 15 min
5 min twice 24 h 2h 24 h housing
Wang Y.I., Ice water Food Restraint Water 35 days
Cage tilt Continuous illumination Wet bedding Cage shaking
2021 swimming deprivation stress deprivation Individually
24 h 24 h 24 h 30 min
[129] 5 min 24 h 6h 24 h housing
Plantar
Wang G., Ice water Food Water 35 days
Cage tilt electrical Continuous illumination White noise Soiled cage Tail nipping Exposure to a stroboscope
2019 swimming deprivation deprivation Group
12 h stimulation 36 h 12 h 24 h 2 min 2h
[130] 5 min 12 h 12 h housing
10 min
Exposure to a Food Restraint Water 42 days
Wen G., 2019 Cage tilt 120-dB noise Alterations of the light-dark Wet bedding
stroboscope deprivation stress deprivation Individually
[131] 4h overnight cycle 4h
overnight 24 h 4h 24 h housing
Alterations of Exposure to
Li M., Food White noise Exposure to a Water 42 days
the Cage tilt an empty Overnight Overhang Soiled cage Tail pinch Oscillation
2014 deprivation 1h foreign object deprivation Group
light-dark 12 h bottle illumination 10 min 24 h 1 min 5 min
[132] 24 h 12 h 24 h housing
cycle 10 min
Food Restraint Water 49 days
Xie M., 2022 No bedding Cage tilt Overnight illumination (twice Wet bedding Tail pinching Cage shaking
deprivation stress deprivation Individually
[133] 24 h 24 h per week) 24 h 5 min 15 min
24 h 6h 24 h housing
Switched
Wassouf Z., Food Illumination Restraint Rat Water 56 days
day/night- Cage tilt
2019 deprivation at night stress confrontation deprivation Individually
cycle 2h
[134] 16 h 12 h 1h 30 min 16 h housing
48 h
Wang Y., Food Restraint Damp Water 56 days
4 ◦ C exposure Cage tilt Cage shaking
2021 deprivation Day/night inversion stress bedding deprivation Group
1h 12 h 30 min
[135] 23 h 1h 24 h 23 h housing
Life 2024, 14, 1110 9 of 31
vascular occlusion [155]. One commonly used dye is Rose Bengal, which, when activated
by green light (560 nm) [156], generates reactive oxygen species, leading to clot formation
and vascular occlusion [157]. Another example is Erythrosin B, which operates in a similar
manner but is activated by a different wavelength (near 660 nm) [158] and offers flexibility
in experimental setups. These dyes are selected for their high quantum yield of singlet
oxygen production, a key factor in inducing rapid and targeted vascular occlusion [159].
The photothrombosis model provides exceptional precision in dictating both the location
and extent of ischemic lesions, a feature that has been instrumental in linking specific
brain areas to behavioral outcomes. This level of control has facilitated research demon-
strating that rodents subjected to ischemic lesions exhibit behavioral changes reminiscent
of depression [38].
Figure 4.
Figure 4. Behavioral
Behavioral assessment
assessment methods
methods for
for post-stroke
post-stroke depression
depression in
in rodent
rodent models.
models.
of anhedonia primarily relies on measuring the preference for sucrose, a simple yet ef-
fective indicator of pleasure-seeking behavior. SPT involves providing each mouse with
two identical bottles: one containing a sucrose solution and the other water. The animals
are then allowed to choose freely between them [163]. A marked decrease in sucrose
consumption is interpreted as an expression of anhedonia [164,165]. The test procedure
encompasses several variables, including housing during the habituation, concentration
of sucrose solution, period at which the experiments are performed (light/dark), type of
habituation to the SPT procedure, type of food/water deprivation during the test, and test
duration [166]. SPT has proven to have satisfactory results in various studies on differ-
ent animal models of PSD. For example, it revealed severe anhedonia after MCAO and
18 consecutive days of CUMS [123]. Also, MCAO and spatial restraint stress led to a de-
creased percentage of sucrose consumption compared to stroke alone [113]. Lastly, MCAO
followed by social isolation showed a significant increase in sucrose consumption for
post-stroke pair-housed mice compared to those that were socially isolated [137].
6.2. Depression-like Behavior Tests: Forced Swim Test and Tail Suspension Test
Depression encompasses a complex array of symptoms, including profound feelings
of hopelessness, persistent sadness, and thoughts of death or suicide [167]. The forced swim
and tail suspension tests have been employed as a means to gauge aspects of despair and
motivational withdrawal in rodents, which are considered analogs to the human experience
of hopelessness and passive resignation.
The Forced Swim Test (FST) is widely utilized for evaluating despair-like behavior in
animal models, quantifying the duration of immobility, except for the minimal movements
necessary to maintain the animal’s head above water. This behavior is interpreted as a sign
of behavioral despair, mirroring aspects of depression [168]. Studies have shown that both
MCAO and chronic mild stress can induce despair-like behavior in C57BL/6 mice [35], as
well as in albino mice subjected to BCCAO [149].
The Tail Suspension Test (TST) is also used, but only in mouse models. In this test,
animals are suspended by their tails, and the duration of immobility is recorded [169], with
longer immobility times indicating a higher level of despair. The TST has demonstrated
a significant increase in immobility time in C57BL/6 mice five days post-BCCAO [170],
NMRI mice 72 h after permanent double ligation of the right common carotid artery [171],
and ICR mice following MCAO and spatial restraint stress [172].
Anxiety levels in this context are analyzed from the animal’s exploration patterns, with a
preference for the periphery over the center indicating higher anxiety [176]. Experiments
using C57Bl/6 mice subjected to MCAO and 17 days of chronic mild stress revealed
significant anxiety with OFT, as demonstrated by reduced central area exploration [35].
Similarly, MCAO followed by periods of social isolation also led to a noticeable decrease in
the time these mice spent in the center [137].
The Novelty Suppressed Feeding Test (NSFT) is a behavioral assay designed to evalu-
ate anxiety and depression-related behaviors by measuring both the amount of food intake
and the delay before the animal engages with a new, highly palatable food item [177].
This test is predicated on the natural conflict between the fear of a novel environment
and the motivation to eat, with increased latency and reduced food consumption indi-
cating heightened anxiety or depressive states [177]. Studies involving C57BL/6 mice
demonstrated that after microinjection of ET-1, mice exhibited a decreased interest in food
and a significant delay before beginning to eat the novel food, suggesting an increase
in anxiety or depressive-like behavior [153]. Similarly, following an MCAO procedure,
mice showed increased latency to approaching and consuming the food pellet three weeks
post-ischemia [178].
The Light/Dark Test (L/D Test) measures an animal’s willingness to explore or avoid
new environments [179,180]. This test utilizes a chamber divided into illuminated and dark
sections and allows measurement of the time taken to enter the light compartment and the
number of transitions between compartments, reflecting the animal’s exploratory behavior
and its aversion to brightly lit areas, respectively [179]. A preference for spending more
time in the dark compartment is interpreted as an indication of anxiety. The L/D Test has
been effectively employed in studying PSD in murine models [153].
In the Marble Burying Test, animals are placed into a cage layered with bedding, in
which marbles or similar small objects are evenly distributed. Researchers then measure the
number of marbles the rodent buries within a specified period. A tendency to bury more
marbles is interpreted as an indication of heightened anxiety or compulsive tendencies,
providing a straightforward method for assessing these behaviors [181,182].
Nestlet Shredding and Nest Building Tests also serve as valuable tools for determining
stress levels in rodents [183]. These tests examine the natural nesting behavior, where
rodents are provided with materials, like cotton nestlets, to build nests. The extent and
quality of the nest constructed, along with the degree of shredding of the provided materials,
are indicative of the animal’s well-being, with poor nesting behavior suggesting elevated
stress or discomfort [183].
The Social Preference–Avoidance Test is used for both mice and rats and is designed to
measure the dynamics of social interaction, specifically the speed of approach or avoidance
displayed by the animal during the test [189,190]. This test provides insight into the social
tendencies of rodents, offering a nuanced view of how they navigate social spaces and
whether they show a propensity towards engaging with or avoiding other animals.
The Three Chamber Test is employed to assess sociability and social memory by eval-
uating a rodent’s preference for an unfamiliar conspecific or an inanimate object and its
preference for a new or a familiar conspecific [191]. This test effectively distinguishes be-
tween the animal’s interest in social interactions and its ability to recognize and differentiate
between familiar and unfamiliar individuals.
The Olfactory Habituation–Dishabituation Test, while initially utilized in evaluating
autistic behaviors in mice, serves a broader purpose in assessing the olfactory system,
which is vital for studying sensory processing in the brain [192]. Mice naturally exhibit a
preference for novel scents over familiar ones [193]. Assessment of the olfactory system
has proven useful for studying sensory processing in the brain [194] but also serves as
a valuable tool for assessing social interaction, memory, and anxiety [195]. Social inter-
action is conditioned by the level of anxiety, while anxious behavior is often associated
with depression [196].
The Resident–Intruder Test is another significant behavioral assay where a resident
rodent is confronted with an unfamiliar “intruder” in its environment. The resultant
behaviors, ranging from aggressive to affiliative, are observed and scored. This test is
instrumental in evaluating behaviors such as territorial aggression, social dominance, and
social recognition memory [197].
The Tube Dominance Test is primarily employed to measure social hierarchy and
dominance in mice [198]. After a day of habituation and training, two mice enter a narrow
tube from opposite sides and meet in the middle. The mouse that persuades the other to
retreat is deemed the winner, respectively dominant. The test was successfully used in a
mouse model of depression involving CUMS [199] and could provide additional insights
regarding PSD.
for observing short-term memory by analyzing the percentage of correct alternations made
by the rodent, reflecting its ability to remember previously visited arms [205]. Research
involving C57BL/6 mice post-MCAO surgery and subsequent individual housing revealed
a decrease in the percentage of correct alternations in the Y-maze, suggesting impairments
in spatial working memory as compared to pair-housed mice [137].
The Novel Object Recognition Test (NORT) serves as a behavioral assay for evaluating
memory capability, particularly recognition memory [206]. Initially, animals are allowed to
familiarize themselves with an arena containing two identical objects [207]. Subsequently,
one of the original objects is replaced with a novel object, and the animal’s interaction
with both objects is observed [208]. A preference for exploring the novel object over the
familiar one is typically indicative of healthy recognition memory, as it suggests the animal
remembers the original object and finds the new one more interesting [207,208]. Studies
employing NORT have demonstrated its utility in detecting memory impairment. For
example, socially isolated mice subjected to MCAO showed impairment in recognition
memory, as evidenced by their equal interest in exploring both novel and familiar objects.
This lack of preference for the novel object indicates a difficulty in recognizing or remem-
bering the previously encountered object, underscoring the impact of social isolation and
stroke on cognitive functions [137].
The Radial Arm Maze Test typically consists of a central platform with multiple arms
extending outward, resembling the spokes of a wheel [209]. At the end of each arm,
food rewards or other incentives are placed to motivate the rodent. The animal is placed
in the central area and must efficiently navigate through the maze to collect the rewards.
Successful navigation involves remembering which arms have already been visited to avoid
unnecessary revisits, thereby demonstrating the animal’s ability to learn and remember
spatial information [210].
The Passive Avoidance Test is a specific behavioral assay used to assess learning
and memory after stroke [211]. Rodents are typically placed into a two-compartment
apparatus, one illuminated and one darkened. Initially, the animal is allowed to explore
both compartments freely. After a predetermined period of time, usually during the
training phase, the animal receives a mild aversive stimulus (i.e., foot shock) upon entering
one of the compartments, typically the darkened compartment. This creates an association
between the aversive stimulus and the compartment. During the testing phase, the animal
is again placed in the apparatus and allowed to freely explore both compartments. The
latency to enter the aversive compartment is recorded. Animals with intact memory will
exhibit a longer latency to enter the aversive compartment due to their association with
the stimulus [212].
ability to navigate the staircase and retrieve the food rewards offers insight into their skilled
reaching and grasping capabilities and overall motor recovery [233].
The Corner Test also provides valuable insights into motor asymmetry and sensori-
motor function [43]. Following a stroke, the rodent is placed near a corner of a testing
apparatus, typically a rectangular or triangular enclosure with two converging walls form-
ing the corner. As the rodent approaches the corner, it tends to turn in the direction of its
more impaired side, leading to a higher frequency of turns toward the affected side com-
pared to the unaffected one. Researchers observe and record the direction of the turns as
well as any asymmetry in movement patterns [234]. This method allows for the assessment
of both the preference in turning direction and any motor deficits that might influence this
preference, offering a direct indication of unilateral sensorimotor impairment.
The Pasta Test provides valuable insights into the motor abilities and functional
recovery of rodents following neurological insults [235]. Animals are typically given a
piece of pasta to manipulate and eat. The researchers then observe and analyze the rodent’s
behavior, focusing on the symmetry and effectiveness of its forepaw movements during
the manipulation and consumption of the pasta. Impairments in fine motor skills or
asymmetrical use of the forepaws can indicate deficits in manual dexterity, indicative of
neurological damage or dysfunction [236].
Motor dysfunction and depressive symptoms frequently coexist in patients following
a stroke, and there is evidence to suggest that these motor deficits can influence the onset
and severity of depressive symptoms [237]. The DigiGait system provides detailed and
quantitative assessments of gait and offers a unique opportunity to study these motor
abnormalities in stroke models [43]. Although DigiGait has not been extensively used
specifically for PSD research, its ability to precisely measure changes in gait and coordi-
nation could be invaluable in understanding the relationship between motor deficits and
depressive behaviors post-stroke [238]. Incorporating DigiGait assessments in PSD studies
could enhance our understanding of how motor impairments contribute to or exacerbate
depressive symptoms. Future research may explore this intersection, potentially leading to
more comprehensive therapeutic strategies that address both the motor and psychological
aspects of post-stroke recovery.
outcomes following stroke, suggesting potential genetic markers for susceptibility to PSD
subtypes. Inflammation is a critical contributor, with elevated levels of pro-inflammatory
cytokines (IL-1β, IL-4, IL-8, TNF-α) [244,245] and the activation of pathways, such as
NLRP3, signifying an influence on PSD pathophysiology [246]. Neuroendocrine dysregu-
lation, especially concerning the HPA axis and resultant elevated cortisol levels, also con-
tributes significantly to PSD, highlighting the neuroendocrine system’s critical role in mood
regulation [247,248]. Poor post-stroke prognosis is linked to alterations in the HPA axis,
elevated levels of catecholamines and natriuretic peptides, and reduced levels of melatonin
and IGF-1 [249]. Moreover, neurotrophic factors, like brain-derived neurotrophic factor
(BDNF) and glial cell line-derived neurotrophic factor (GDNF), essential for neuronal
health and regeneration post-injury, are also linked to PSD development, with varia-
tions in their levels and methylation status closely associated with depressive outcomes
post-stroke [250,251]. Together, these mechanisms offer a comprehensive insight into the
intricate number of factors contributing to PSD [56].
innovative methodologies and testing that can bridge this divide, which are essential in
accelerating the development of effective clinical PSD treatments.
pression, typically exhibiting mild to moderate symptoms shortly after the stroke or
after several months [266]. However, the hypothesis that the location of the brain le-
sion influences the risk of PSD is contested, with several studies challenging the notion
that depression is more commonly associated with left-hemisphere strokes than with
right-hemisphere strokes [70,267,268].
Age and gender also represent crucial factors that significantly impact the study of
neurodegenerative diseases, including PSD. Research predominantly utilizes young an-
imal models, which may not accurately reproduce the age-related complexities of PSD
in humans [34,146–148]. Including older animals in these studies will enhance model
validity, improve our understanding of age as a critical factor in disease progression and
recovery [80], and hopefully reduce the translational gap in clinical applications. Moreover,
the prevalence of studies focusing on male rodents [35,113,123,137] may overlook key
sex-based dimorphic differences that could influence both the presentation and progression
of PSD. Males and females may respond differently to stroke, with potential variations
in motor function, mood, cognitive abilities, and memory tasks [269]. Notably, female
rodents are more prone to weight loss during chronic social stress and may exhibit height-
ened anxious behaviors [270]. These differences are modulated by sex hormones, like
estrogen and testosterone, which are also known to affect stroke responses and depres-
sive behaviors [271]. For example, estrogen was shown to alleviate depressive symptoms
post-stroke [272]; however, recent findings suggest that the estrous cycle in females does not
significantly impact behavior or neurogenesis under basal conditions [273,274], indicating
that sex differences might not drastically alter outcomes in commonly used behavioral
tests. However, the inflammatory response to stroke, which is integral to PSD pathology,
appears to vary between sexes, with females often showing a stronger anti-inflammatory
response [275]. Future research should continue to address these variables, providing
a deeper understanding of how age and sex influence the development, treatment, and
ultimately, recovery from PSD. Such insights are vital for designing tailored neuro- or
psychotherapeutic clinical approaches.
Behavioral tests play a crucial role in assessing depressive-like behaviors in animal
models that aid our understanding of the multifaceted nature of PSD. These tests evaluate
a range of symptoms, from mood disturbances and cognitive deficits to motor dysfunctions
and social behavior changes. Commonly employed tests include the sucrose preference test
for anhedonia, the forced swim and tail suspension tests for despair-like behavior, or social
interaction tests for assessing social withdrawal. Additionally, motor function tests also
help to gauge the physical aspects of depression, which are often impacted in post-stroke
conditions [39]. Each test is designed to measure specific symptoms associated with PSD,
providing a comprehensive view of the animal’s emotional and cognitive state post-stroke.
However, interpreting the results of these behavioral tests requires careful consideration
and interpretation. The inherent variability in rodent behavior, the subjective nature of
depressive symptoms, and the potential for human error in collecting data during the
experiment or subsequent analyses necessitate a cautious approach [33,252]. Variations in
test conditions, handling, and even the environment can influence the outcomes, potentially
affecting the accuracy of the data [166]. Given these challenges, there is a pressing need
for future research to focus on refining existing behavioral tests and also develop more
sophisticated and direct methods for assessing PSD. This involves enhancing the objec-
tivity, sensitivity, and specificity of behavioral assays to accurately capture the nuanced
manifestations of PSD in animal models. Improvements in test design, execution, and data
analysis can lead to more reliable and valid measurements of depressive-like behaviors,
facilitating the identification of effective treatments and interventions for PSD.
In conclusion, this review summarized the complex and multifaceted nature of PSD,
emphasizing the significant challenges involved in modeling and evaluating this condi-
tion in preclinical trials. Our exploration into the epidemiology, risk factors, underlying
mechanisms, and the development of animal models for PSD has underscored the crucial
need for advanced, nuanced approaches in preclinical research. Indeed, bridging the gap
Life 2024, 14, 1110 20 of 31
between animal studies and clinical applications requires focused efforts to refine and
develop animal models and sophisticated behavioral assessments that more accurately
mirror the human condition and behavior. Enhancing these models and assessments is
essential for improving the translation of research findings into the clinic, resulting in more
effective diagnostic tools and treatments for PSD. This includes addressing the variability
in rodent responses, the challenge of extending the results to human pathology, and the
integration of diverse biological, psychological, and social determinants of PSD. Addi-
tionally, considerations of age, gender differences, and strain variability among rodents
highlight the importance of a custom-tailored approach in understanding and treating PSD.
This approach will help ensure that the insights gained in the laboratory can be effectively
applied in the clinic, ultimately improving patient outcomes and accelerating recovery.
While most previous studies address these aspects individually, our aim was to integrate
all recent developments in PSD pathology within both clinical and experimental contexts
in order to provide a complete perspective, highlighting the gap between human clinical
data and preclinical research.
11. Summary
PSD stands as a significant barrier to recovery from stroke and is defined by its com-
plexity stemming from an interplay of physiological, psychological, and social factors.
PSD research has expanded our understanding, revealing that risk factors such as age,
gender, pre-stroke psychiatric history, and the physical location of the stroke significantly
influence the likelihood and severity of PSD. Animal models, particularly those involv-
ing rodents, are pivotal for understanding the pathophysiological underpinnings of PSD.
Models such as MCAO, BCAO, and various genetic models are utilized to mimic stroke
in rodents, enabling the study of depressive-like behaviors subsequent to cerebrovascular
insults. Techniques combining MCAO with CUMS or social isolation post-stroke have been
particularly insightful and highlight the role of PSD environmental and social stressors.
Further, behavioral assessments are crucial in measuring symptoms, like anhedonia, de-
spair, hopelessness, and motor dysfunctions, which help evaluate the efficacy of potential
treatments and the validity of the models themselves. Despite these advances, translating
findings from animal models to human patients remains challenging. Variability in rodent
responses, differences in stroke etiology, and the subjective nature of depressive symptoms
complicate the direct application of preclinical results to the clinic. Moreover, the outcomes
of these studies underline the need for a personalized approach to treatment, considering
individual risk factors such as age, sex, comorbidities, social network, etc. While animal
models and behavioral studies have greatly contributed to our understanding of PSD,
the relatively limited number of clinical studies compared to the many preclinical studies
underscores the complexity of this condition and the existing translational gap. Advance-
ments in these areas are vital for developing targeted psychiatric, neuro-, or psychological
interventions, ultimately improving the quality of life and recovery outcomes for stroke
survivors worldwide.
Author Contributions: M.I.M., A.G., B.C., M.H. and A.P.-W.: methodology. B.C., M.H., A.G. and
A.P.-W.: validation. M.I.M. and A.G.: formal analysis. M.I.M. and A.G.: investigation. A.G. and
M.I.M.: resources. M.I.M. and A.G.: data curation. M.I.M. and A.G.: original draft. B.C., M.H.
and A.P.-W.: final manuscript. B.C., M.H. and A.P.-W.: supervision. All authors contributed to
editorial changes in the manuscript. All authors have read and agreed to the published version of
the manuscript.
Funding: The Article Processing Charges were funded by the University of Medicine and Pharmacy
of Craiova, Romania.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Life 2024, 14, 1110 21 of 31
Data Availability Statement: No new data were created or analyzed in this study. Data sharing is
not applicable to this article.
Conflicts of Interest: The authors declare no conflicts of interest. The funders had no role in the design
of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or
in the decision to publish the results.
References
1. Donkor, E.S. Stroke in the 21(st) Century: A Snapshot of the Burden, Epidemiology, and Quality of Life. Stroke Res. Treat. 2018,
2018, 3238165. [CrossRef] [PubMed]
2. Fekadu, G.; Adola, B.; Mosisa, G.; Shibiru, T.; Chelkeba, L. Clinical characteristics and treatment outcomes among stroke patients
hospitalized to Nekemte referral hospital, western Ethiopia. J. Clin. Neurosci. 2020, 71, 170–176. [CrossRef]
3. Boehme, A.K.; Esenwa, C.; Elkind, M.S. Stroke Risk Factors, Genetics, and Prevention. Circ. Res. 2017, 120, 472–495. [CrossRef]
[PubMed]
4. Arboix, A. Cardiovascular risk factors for acute stroke: Risk profiles in the different subtypes of ischemic stroke. World J. Clin.
Cases 2015, 3, 418–429. [CrossRef] [PubMed]
5. Kernan, W.N.; Dearborn, J.L. Obesity increases stroke risk in young adults: Opportunity for prevention. Stroke 2015, 46, 1435–1436.
[CrossRef]
6. Kotl˛ega, D.; Gołab-Janowska,
˛ M.; Masztalewicz, M.; Ciećwież, S.; Nowacki, P. The emotional stress and risk of ischemic stroke.
Neurol. Neurochir. Pol. 2016, 50, 265–270. [CrossRef]
7. Kelly-Hayes, M. Influence of age and health behaviors on stroke risk: Lessons from longitudinal studies. J. Am. Geriatr. Soc. 2010,
58 (Suppl. 2), S325–S328. [CrossRef]
8. Platz, T. Evidence-Based Guidelines and Clinical Pathways in Stroke Rehabilitation-An International Perspective. Front. Neurol.
2019, 10, 200. [CrossRef]
9. Inatomi, Y.; Nakajima, M.; Yonehara, T.; Ando, Y. Ipsilateral hemiparesis in ischemic stroke patients. Acta Neurol. Scand. 2017,
136, 31–40. [CrossRef] [PubMed]
10. Yanohara, R.; Teranishi, T.; Tomita, Y.; Tanino, G.; Ueno, Y.; Sonoda, S. Recovery process of standing postural control in hemiplegia
after stroke. J. Phys. Ther. Sci. 2014, 26, 1761–1765. [CrossRef]
11. Kim, Y.W. Update on Stroke Rehabilitation in Motor Impairment. Brain Neurorehabil. 2022, 15, e12. [CrossRef] [PubMed]
12. Chohan, S.A.; Venkatesh, P.K.; How, C.H. Long-term complications of stroke and secondary prevention: An overview for primary
care physicians. Singap. Med. J. 2019, 60, 616–620. [CrossRef] [PubMed]
13. Rost, N.S.; Brodtmann, A.; Pase, M.P.; van Veluw, S.J.; Biffi, A.; Duering, M.; Hinman, J.D.; Dichgans, M. Post-Stroke Cognitive
Impairment and Dementia. Circ. Res. 2022, 130, 1252–1271. [CrossRef]
14. Gaete, J.M.; Bogousslavsky, J. Post-stroke depression. Expert. Rev. Neurother. 2008, 8, 75–92. [CrossRef]
15. Li, W.; Xiao, W.M.; Chen, Y.K.; Qu, J.F.; Liu, Y.L.; Fang, X.W.; Weng, H.Y.; Luo, G.P. Anxiety in Patients With Acute Ischemic
Stroke: Risk Factors and Effects on Functional Status. Front. Psychiatry 2019, 10, 257. [CrossRef]
16. Acciarresi, M.; Bogousslavsky, J.; Paciaroni, M. Post-stroke fatigue: Epidemiology, clinical characteristics and treatment.
Eur. Neurol. 2014, 72, 255–261. [CrossRef] [PubMed]
17. Cai, H.; Wang, X.P.; Yang, G.Y. Sleep Disorders in Stroke: An Update on Management. Aging Dis. 2021, 12, 570–585. [CrossRef]
18. Guo, J.; Wang, J.; Sun, W.; Liu, X. The advances of post-stroke depression: 2021 update. J. Neurol. 2022, 269, 1236–1249. [CrossRef]
19. Al-Qazzaz, N.K.; Ali, S.H.; Ahmad, S.A.; Islam, S.; Mohamad, K. Cognitive impairment and memory dysfunction after a stroke
diagnosis: A post-stroke memory assessment. Neuropsychiatr. Dis. Treat. 2014, 10, 1677–1691. [CrossRef]
20. Lee, C.H.; Jeon, S.H.; Kim, M.J.; Ra, G.D.; Lee, Y.H.; Hong, S.H.; Shin, B.S.; Kang, H.G. Factors Affecting Post-Stroke Depression in
Acute Ischemic Stroke Patients after 3 Months. J. Pers. Med. 2021, 11, 1178. [CrossRef]
21. Liu, L.; Xu, M.; Marshall, I.J.; Wolfe, C.D.; Wang, Y.; O’Connell, M.D. Prevalence and natural history of depression after stroke:
A systematic review and meta-analysis of observational studies. PLoS Med. 2023, 20, e1004200. [CrossRef]
22. Cai, W.; Mueller, C.; Li, Y.-J.; Shen, W.-D.; Stewart, R. Post stroke depression and risk of stroke recurrence and mortality:
A systematic review and meta-analysis. Ageing Res. Rev. 2019, 50, 102–109. [CrossRef] [PubMed]
23. Shi, Y.; Xiang, Y.; Yang, Y.; Zhang, N.; Wang, S.; Ungvari, G.S.; Chiu, H.F.K.; Tang, W.K.; Wang, Y.; Zhao, X.; et al. Depression after
minor stroke: Prevalence and predictors. J. Psychosom. Res. 2015, 79, 143–147. [CrossRef] [PubMed]
24. Bour, A.; Rasquin, S.; Aben, I.; Boreas, A.; Limburg, M.; Verhey, F. A one-year follow-up study into the course of depression after
stroke. J. Nutr. Health Aging 2010, 14, 488–493. [CrossRef]
25. Hong, J.P.; Park, S.; Ahn, S.-H.; Kim, J.S. Factors associated with post-stroke suicidal death. J. Psychiatr. Res. 2018, 96, 135–137.
[CrossRef] [PubMed]
26. Bartoli, F.; Pompili, M.; Lillia, N.; Crocamo, C.; Salemi, G.; Clerici, M.; Carrà, G. Rates and correlates of suicidal ideation among
stroke survivors: A meta-analysis. J. Neurol. Neurosurg. Psychiatry 2017, 88, 498–504. [CrossRef]
27. Kauhanen, M.; Korpelainen, J.T.; Hiltunen, P.; Brusin, E.; Mononen, H.; Määttä, R.; Nieminen, P.; Sotaniemi, K.A.; Myllylä, V.V.
Poststroke depression correlates with cognitive impairment and neurological deficits. Stroke 1999, 30, 1875–1880. [CrossRef]
Life 2024, 14, 1110 22 of 31
28. Srivastava, A.; Taly, A.B.; Gupta, A.; Murali, T. Post-stroke depression: Prevalence and relationship with disability in chronic
stroke survivors. Ann. Indian. Acad. Neurol. 2010, 13, 123–127. [CrossRef]
29. Kim, E.S.; Kim, J.W.; Kang, H.J.; Bae, K.Y.; Kim, S.W.; Kim, J.T.; Park, M.S.; Cho, K.H.; Kim, J.M. Longitudinal Impact of Depression
on Quality of Life in Stroke Patients. Psychiatry Investig. 2018, 15, 141–146. [CrossRef]
30. Paparella, I.; Vandewalle, G.; Stagg, C.J.; Maquet, P. An integrated measure of GABA to characterize post-stroke plasticity.
NeuroImage Clin. 2023, 39, 103463. [CrossRef]
31. Nagy, E.E.; Frigy, A.; Szász, J.A.; Horváth, E. Neuroinflammation and microglia/macrophage phenotype modulate the molecular
background of post-stroke depression: A literature review. Exp. Ther. Med. 2020, 20, 2510–2523. [CrossRef]
32. Loubinoux, I.; Kronenberg, G.; Endres, M.; Schumann-Bard, P.; Freret, T.; Filipkowski, R.K.; Kaczmarek, L.; Popa-Wagner, A.
Post-stroke depression: Mechanisms, translation and therapy. J. Cell Mol. Med. 2012, 16, 1961–1969. [CrossRef] [PubMed]
33. Zirk, M.; Storm, V. Subjective Stroke Impact and Depressive Symptoms: Indications for a Moderating Role of Health-Related
Locus of Control. Front. Psychiatry 2019, 10, 918. [CrossRef] [PubMed]
34. Kronenberg, G.; Gertz, K.; Heinz, A.; Endres, M. Of mice and men: Modelling post-stroke depression experimentally.
Br. J. Pharmacol. 2014, 171, 4673–4689. [CrossRef] [PubMed]
35. Kim, Y.R.; Kim, H.N.; Pak, M.E.; Ahn, S.M.; Hong, K.H.; Shin, H.K.; Choi, B.T. Studies on the animal model of post-stroke
depression and application of antipsychotic aripiprazole. Behav. Brain Res. 2015, 287, 294–303. [CrossRef]
36. Wu, C.; Zhang, J.; Chen, Y. Study on the behavioral changes of a post-stroke depression rat model. Exp. Ther. Med. 2015,
10, 159–163. [CrossRef]
37. Braeuninger, S.; Kleinschnitz, C. Rodent models of focal cerebral ischemia: Procedural pitfalls and translational problems.
Exp. Transl. Stroke Med. 2009, 1, 8. [CrossRef]
38. Tao, X.; Yang, W.; Zhu, S.; Que, R.; Liu, C.; Fan, T.; Wang, J.; Mo, D.; Zhang, Z.; Tan, J.; et al. Models of poststroke depression and
assessments of core depressive symptoms in rodents: How to choose? Exp. Neurol. 2019, 322, 113060. [CrossRef]
39. Chen, D.; Wang, J.; Xing, Y.; Jia, P.; Zhang, Y.; Wang, J.; Ren, H.; Le, A.; Chen, X.; Wang, J. Behavioral assessment of post-stroke
depression and anxiety in rodents. Brain Hemorrhages 2020, 1, 105–111. [CrossRef]
40. Can, A.; Dao, D.T.; Arad, M.; Terrillion, C.E.; Piantadosi, S.C.; Gould, T.D. The mouse forced swim test. J. Vis. Exp. 2012, 59, e3638.
[CrossRef]
41. Liu, M.Y.; Yin, C.Y.; Zhu, L.J.; Zhu, X.H.; Xu, C.; Luo, C.X.; Chen, H.; Zhu, D.Y.; Zhou, Q.G. Sucrose preference test for
measurement of stress-induced anhedonia in mice. Nat. Protoc. 2018, 13, 1686–1698. [CrossRef]
42. Kraeuter, A.K.; Guest, P.C.; Sarnyai, Z. The Open Field Test for Measuring Locomotor Activity and Anxiety-Like Behavior.
Methods Mol. Biol. 2019, 1916, 99–103. [CrossRef] [PubMed]
43. Balkaya, M.; Kröber, J.M.; Rex, A.; Endres, M. Assessing post-stroke behavior in mouse models of focal ischemia. J. Cereb. Blood
Flow Metab. 2013, 33, 330–338. [CrossRef] [PubMed]
44. Harrison, J.E.; Weber, S.; Jakob, R.; Chute, C.G. ICD-11: An international classification of diseases for the twenty-first century.
BMC Med. Inform. Decis. Mak. 2021, 21, 206. [CrossRef] [PubMed]
45. Tolentino, J.C.; Schmidt, S.L. DSM-5 Criteria and Depression Severity: Implications for Clinical Practice. Front. Psychiatry 2018,
9, 450. [CrossRef] [PubMed]
46. Bos, E.H.; de Jonge, P.; Cox, R.F.A. Affective variability in depression: Revisiting the inertia-instability paradox. Br. J. Psychol.
2019, 110, 814–827. [CrossRef]
47. Liang, J.; Yue, Y.; Jiang, H.; Geng, D.; Wang, J.; Lu, J.; Li, S.; Zhang, K.; Wu, A.; Yuan, Y. Genetic variations in the p11/tPA/BDNF
pathway are associated with post stroke depression. J. Affect. Disord. 2018, 226, 313–325. [CrossRef]
48. Zhou, Z.; Ding, X.; Yang, Q.; Hu, J.; Shang, X.; Huang, X.; Ge, L.; Zhou, T. Association between Single-Nucleotide Polymorphisms
of the Tyrosine Kinase Receptor B (TrkB) and Post-Stroke Depression in China. PLoS ONE 2015, 10, e0144301. [CrossRef]
49. Zhao, F.; Yue, Y.; Jiang, H.; Yuan, Y. Shared genetic risk factors for depression and stroke. Prog. Neuro-Psychopharmacol. Biol.
Psychiatry 2019, 93, 55–70. [CrossRef]
50. Okbay, A.; Baselmans, B.M.; De Neve, J.E.; Turley, P.; Nivard, M.G.; Fontana, M.A.; Meddens, S.F.; Linnér, R.K.; Rietveld, C.A.;
Derringer, J.; et al. Genetic variants associated with subjective well-being, depressive symptoms, and neuroticism identified
through genome-wide analyses. Nat. Genet. 2016, 48, 624–633. [CrossRef]
51. Wang, Z.; Shi, Y.; Liu, F.; Jia, N.; Gao, J.; Pang, X.; Deng, F. Diversiform Etiologies for Post-stroke Depression. Front. Psychiatry
2018, 9, 761. [CrossRef] [PubMed]
52. Caplan, S.; Alvidrez, J.; Paris, M.; Escobar, J.I.; Dixon, J.K.; Desai, M.M.; Whittemore, R.; Scahill, L.D. Subjective versus objective:
An exploratory analysis of latino primary care patients with self-perceived depression who do not fulfill primary care evaluation
of mental disorders patient health questionnaire criteria for depression. Prim. Care Companion J. Clin. Psychiatry 2010, 12, 26886.
[CrossRef] [PubMed]
53. Perna, L.; Zhang, Y.; Matias-Garcia, P.R.; Ladwig, K.-H.; Wiechmann, T.; Wild, B.; Waldenberger, M.; Schöttker, B.; Mons, U.;
Ihle, A.; et al. Subjective mental health, incidence of depressive symptoms in later life, and the role of epigenetics: Results from
two longitudinal cohort studies. Transl. Psychiatry 2020, 10, 323. [CrossRef] [PubMed]
54. Dabrowska-Bender,
˛ M.; Milewska, M.; Gołabek,˛ A.; Duda-Zalewska, A.; Staniszewska, A. The Impact of Ischemic Cerebral
Stroke on the Quality of Life of Patients Based on Clinical, Social, and Psychoemotional Factors. J. Stroke Cerebrovasc. Dis. 2017,
26, 101–107. [CrossRef]
Life 2024, 14, 1110 23 of 31
55. Paprocka-Borowicz, M.; Wiatr, M.; Ciałowicz, M.; Borowicz, W.; Kaczmarek, A.; Marques, A.; Murawska-Ciałowicz, E. Influence
of Physical Activity and Socio-Economic Status on Depression and Anxiety Symptoms in Patients after Stroke. Int. J. Environ. Res.
Public Health 2021, 18, 8058. [CrossRef]
56. Zhan, Q.; Kong, F. Mechanisms associated with post-stroke depression and pharmacologic therapy. Front. Neurol. 2023,
14, 1274709. [CrossRef]
57. Naghavi, F.S.; Koffman, E.E.; Lin, B.; Du, J. Post-stroke neuronal circuits and mental illnesses. Int. J. Physiol. Pathophysiol.
Pharmacol. 2019, 11, 1–11.
58. Balami, J.S.; Chen, R.L.; Grunwald, I.Q.; Buchan, A.M. Neurological complications of acute ischaemic stroke. Lancet Neurol. 2011,
10, 357–371. [CrossRef]
59. Strawbridge, R.; Young, A.H.; Cleare, A.J. Biomarkers for depression: Recent insights, current challenges and future prospects.
Neuropsychiatr. Dis. Treat. 2017, 13, 1245–1262. [CrossRef]
60. Ayerbe, L.; Ayis, S.; Wolfe, C.D.; Rudd, A.G. Natural history, predictors and outcomes of depression after stroke: Systematic
review and meta-analysis. Br. J. Psychiatry 2013, 202, 14–21. [CrossRef]
61. Hackett, M.L.; Pickles, K. Part I: Frequency of depression after stroke: An updated systematic review and meta-analysis of
observational studies. Int. J. Stroke 2014, 9, 1017–1025. [CrossRef]
62. Dong, L.; Sánchez, B.N.; Skolarus, L.E.; Stulberg, E.; Morgenstern, L.B.; Lisabeth, L.D. Sex difference in prevalence of depression
after stroke. Neurology 2020, 94, e1973–e1983. [CrossRef] [PubMed]
63. Mayman, N.A.; Tuhrim, S.; Jette, N.; Dhamoon, M.S.; Stein, L.K. Sex Differences in Post-Stroke Depression in the Elderly. J. Stroke
Cerebrovasc. Dis. 2021, 30, 105948. [CrossRef]
64. Driga, M.P.; Catalin, B.; Olaru, D.G.; Slowik, A.; Plesnila, N.; Hermann, D.M.; Popa-Wagner, A. The Need for New Biomarkers to
Assist with Stroke Prevention and Prediction of Post-Stroke Therapy Based on Plasma-Derived Extracellular Vesicles. Biomedicines
2021, 9, 1226. [CrossRef] [PubMed]
65. Kim, J.S. Post-stroke Mood and Emotional Disturbances: Pharmacological Therapy Based on Mechanisms. J. Stroke 2016,
18, 244–255. [CrossRef] [PubMed]
66. Rupprechter, S.; Romaniuk, L.; Series, P.; Hirose, Y.; Hawkins, E.; Sandu, A.L.; Waiter, G.D.; McNeil, C.J.; Shen, X.; Harris, M.A.; et al. Blunted
medial prefrontal cortico-limbic reward-related effective connectivity and depression. Brain 2020, 143, 1946–1956. [CrossRef]
67. Li, G.; Liu, Y.; Zheng, Y.; Wu, Y.; Li, D.; Liang, X.; Chen, Y.; Cui, Y.; Yap, P.T.; Qiu, S.; et al. Multiscale neural modeling of
resting-state fMRI reveals executive-limbic malfunction as a core mechanism in major depressive disorder. Neuroimage Clin. 2021,
31, 102758. [CrossRef]
68. Soares, J.C.; Mann, J.J. The anatomy of mood disorders--review of structural neuroimaging studies. Biol. Psychiatry 1997,
41, 86–106. [CrossRef]
69. Barker-Collo, S.L. Depression and anxiety 3 months post stroke: Prevalence and correlates. Arch. Clin. Neuropsychol. 2007,
22, 519–531. [CrossRef]
70. Wei, N.; Yong, W.; Li, X.; Zhou, Y.; Deng, M.; Zhu, H.; Jin, H. Post-stroke depression and lesion location: A systematic review.
J. Neurol. 2015, 262, 81–90. [CrossRef]
71. Klingbeil, J.; Brandt, M.L.; Wawrzyniak, M.; Stockert, A.; Schneider, H.R.; Baum, P.; Hoffmann, K.T.; Saur, D. Association of
Lesion Location and Depressive Symptoms Poststroke. Stroke 2022, 53, e467–e471. [CrossRef]
72. Volz, M.; Ladwig, S.; Werheid, K. Gender differences in post-stroke depression: A longitudinal analysis of prevalence, persistence
and predictive value of known risk factors. Neuropsychol. Rehabil. 2021, 31, 1–17. [CrossRef] [PubMed]
73. Poynter, B.; Shuman Hon, M.; Diaz-Granados, N.; Kapral, M.; Grace, S.L.; Stewart, D.E. Sex Differences in the Prevalence of
Post-Stroke Depression: A Systematic Review. Psychosomatics 2009, 50, 563–569. [CrossRef]
74. Mirolovics, Á.; Bokor, M.; Dobi, B.; Zsuga, J.; Bereczki, D. Socioeconomic Factors Predicting Depression Differ in the Acute Stage
and at 1 year After Ischemic Stroke or TIA. J. Stroke Cerebrovasc. Dis. 2020, 29, 105241. [CrossRef]
75. Gloria, M.U.; Jonah, O.E.; Olusanjo, A.C.; Chiebuka, O.E.; Nene, J.J.; Nwakego, A.U.; Chinyere, A.C. Post-Stroke Depression and
Suicidal Ideations: Relationship with Gender and Marital Status: A Cross Sectional Study. J. Prim. Care Community Health 2024,
15, 21501319241233172. [CrossRef] [PubMed]
76. Tento, T.; Kume, A.; Kumaso, S. Risk factors for stroke-related functional disability and mortality at Felege Hiwot Referral
Hospital, Ethiopia. BMC Neurol. 2023, 23, 393. [CrossRef]
77. Yoo, J.W.; Hong, B.Y.; Jo, L.; Kim, J.S.; Park, J.G.; Shin, B.K.; Lim, S.H. Effects of Age on Long-Term Functional Recovery in Patients
with Stroke. Medicina 2020, 56, 451. [CrossRef] [PubMed]
78. Lökk, J.; Delbari, A. Management of depression in elderly stroke patients. Neuropsychiatr. Dis. Treat. 2010, 6, 539–549. [CrossRef]
79. Lindén, T.; Blomstrand, C.; Skoog, I. Depressive disorders after 20 months in elderly stroke patients: A case-control study. Stroke
2007, 38, 1860–1863. [CrossRef]
80. Wolf, V.L.; Ergul, A. Progress and challenges in preclinical stroke recovery research. Brain Circ. 2021, 7, 230–240. [CrossRef]
81. Chen, R.; Ovbiagele, B.; Feng, W. Diabetes and Stroke: Epidemiology, Pathophysiology, Pharmaceuticals and Outcomes. Am. J.
Med. Sci. 2016, 351, 380–386. [CrossRef]
82. Johansson, B.B. Hypertension mechanisms causing stroke. Clin. Exp. Pharmacol. Physiol. 1999, 26, 563–565. [CrossRef] [PubMed]
83. Gajurel, B.P.; Gurung, A.; Ojha, R.; Rajbhandari, R.; Karn, R. Dyslipidemia and Obesity in Ischemic Stroke. Cureus 2023, 15, e45409.
[CrossRef] [PubMed]
Life 2024, 14, 1110 24 of 31
84. Popa-Wagner, A.; Petcu, E.B.; Capitanescu, B.; Hermann, D.M.; Radu, E.; Gresita, A. Ageing as a risk factor for cerebral ischemia:
Underlying mechanisms and therapy in animal models and in the clinic. Mech. Ageing Dev. 2020, 190, 111312. [CrossRef]
[PubMed]
85. Popa-Wagner, A.; Udristoiu, I.; Gresita, A.; Lledós, M.; Cadenas, I. Post-Stroke Depression: Genetics, Mechanisms, and Treatment;
Springer International Publishing: Cham, Switzerland, 2022; pp. 4467–4478.
86. Kohen, R.; Cain, K.C.; Mitchell, P.H.; Becker, K.; Buzaitis, A.; Millard, S.P.; Navaja, G.P.; Teri, L.; Tirschwell, D.; Veith, R. Association
of serotonin transporter gene polymorphisms with poststroke depression. Arch. Gen. Psychiatry 2008, 65, 1296–1302. [CrossRef]
87. Zhang, E.; Liao, P. Brain-derived neurotrophic factor and post-stroke depression. J. Neurosci. Res. 2020, 98, 537–548. [CrossRef]
88. Qayyum, A.; Zai, C.C.; Hirata, Y.; Tiwari, A.K.; Cheema, S.; Nowrouzi, B.; Beitchman, J.H.; Kennedy, J.L. The Role of the Catechol-
o-Methyltransferase (COMT) GeneVal158Met in Aggressive Behavior, a Review of Genetic Studies. Curr. Neuropharmacol. 2015,
13, 802–814. [CrossRef]
89. Kwon, S.; Hartzema, A.G.; Duncan, P.W.; Min-Lai, S. Disability measures in stroke: Relationship among the Barthel Index, the
Functional Independence Measure, and the Modified Rankin Scale. Stroke 2004, 35, 918–923. [CrossRef]
90. Notsu, Y.; Nabika, T.; Park, H.Y.; Masuda, J.; Kobayashi, S. Evaluation of genetic risk factors for silent brain infarction. Stroke 1999,
30, 1881–1886. [CrossRef]
91. Devereux, N.; Berns, A.M. Evaluation & Treatment of Psychological Effects of Stroke. Dela J. Public. Health 2023, 9, 62–69.
[CrossRef]
92. Ladwig, S.; Werheid, K.; Südmeyer, M.; Volz, M. Predictors of post-stroke depression: Validation of established risk factors and
introduction of a dynamic perspective in two longitudinal studies. Front. Psychiatry 2023, 14, 1093918. [CrossRef] [PubMed]
93. Taylor-Rowan, M.; Momoh, O.; Ayerbe, L.; Evans, J.J.; Stott, D.J.; Quinn, T.J. Prevalence of pre-stroke depression and its association
with post-stroke depression: A systematic review and meta-analysis. Psychol. Med. 2019, 49, 685–696. [CrossRef] [PubMed]
94. Wist, S.; Clivaz, J.; Sattelmayer, M. Muscle strengthening for hemiparesis after stroke: A meta-analysis. Ann. Phys. Rehabil. Med.
2016, 59, 114–124. [CrossRef]
95. Cuccurullo, S.J.; Fleming, T.K.; Zinonos, S.; Cosgrove, N.M.; Cabrera, J.; Kostis, J.B.; Greiss, C.; Ray, A.R.; Eckert, A.; Scarpati,
R.; et al. Stroke Recovery Program with Modified Cardiac Rehabilitation Improves Mortality, Functional & Cardiovascular
Performance. J. Stroke Cerebrovasc. Dis. 2022, 31, 106322. [CrossRef] [PubMed]
96. Moore, N.; Reeder, S.; O’Keefe, S.; Alves-Stein, S.; Schneider, E.; Moloney, K.; Radford, K.; Lannin, N.A. “I’ve still got a job to go
back to”: The importance of early vocational rehabilitation after stroke. Disabil. Rehabil. 2023, 46, 2769–2776. [CrossRef]
97. Deepradit, S.; Powwattana, A.; Lagampan, S.; Thiangtham, W. Effectiveness of a family-based program for post-stroke patients
and families: A cluster randomized controlled trial. Int. J. Nurs. Sci. 2023, 10, 446–455. [CrossRef]
98. Pérez-de la Cruz, S. Influence of an Aquatic Therapy Program on Perceived Pain, Stress, and Quality of Life in Chronic Stroke
Patients: A Randomized Trial. Int. J. Environ. Res. Public Health 2020, 17, 4796. [CrossRef]
99. Dayuan, Z.; Lan, L.; Hui, C.; Huanjie, L.; Deliang, L.; Yihui, D. The effect of music as an intervention for post-stroke depression:
A systematic review and meta-analysis. Complement. Ther. Med. 2022, 71, 102901. [CrossRef]
100. Wang, S.B.; Wang, Y.Y.; Zhang, Q.E.; Wu, S.L.; Ng, C.H.; Ungvari, G.S.; Chen, L.; Wang, C.X.; Jia, F.J.; Xiang, Y.T. Cognitive
behavioral therapy for post-stroke depression: A meta-analysis. J. Affect. Disord. 2018, 235, 589–596. [CrossRef]
101. Gao, W.; Xue, F.; Yu, B.; Yu, S.; Zhang, W.; Huang, H. Repetitive transcranial magnetic stimulation for post-stroke depression:
An overview of systematic reviews. Front. Neurol. 2023, 14, 930558. [CrossRef]
102. Machová, K.; Procházková, R.; Říha, M.; Svobodová, I. The Effect of Animal-Assisted Therapy on the State of Patients’ Health
After a Stroke: A Pilot Study. Int. J. Environ. Res. Public Health 2019, 16, 3272. [CrossRef] [PubMed]
103. Zulkifli, W.; Shamsuddin, S.; Lim, T.H. Animal Robot Assisted-therapy for Rehabilitation of Patient with Post-Stroke Depression.
IOP Conf. Ser. Mater. Sci. Eng. 2017, 210, 012005. [CrossRef]
104. Remes, O.; Mendes, J.F.; Templeton, P. Biological, Psychological, and Social Determinants of Depression: A Review of Recent
Literature. Brain Sci. 2021, 11, 1633. [CrossRef] [PubMed]
105. Liu, F.; McCullough, L.D. Middle cerebral artery occlusion model in rodents: Methods and potential pitfalls. J. Biomed. Biotechnol.
2011, 2011, 464701. [CrossRef] [PubMed]
106. Fluri, F.; Schuhmann, M.K.; Kleinschnitz, C. Animal models of ischemic stroke and their application in clinical research. Drug Des.
Devel Ther. 2015, 9, 3445–3454. [CrossRef]
107. Kuts, R.; Melamed, I.; Shiyntum, H.N.; Frank, D.; Grinshpun, J.; Zlotnik, A.; Brotfain, E.; Dubilet, M.; Natanel, D.; Boyko, M.
A Middle Cerebral Artery Occlusion Technique for Inducing Post-stroke Depression in Rats. J. Vis. Exp. 2019, 147, e58875.
[CrossRef]
108. Liu, S.; Zhen, G.; Meloni, B.P.; Campbell, K.; Winn, H.R. RODENT STROKE MODEL GUIDELINES FOR PRECLINICAL STROKE
TRIALS (1ST EDITION). J. Exp. Stroke Transl. Med. 2009, 2, 2–27. [CrossRef]
109. Gresita, A.; Mihai, R.; Hermann, D.M.; Amandei, F.S.; Capitanescu, B.; Popa-Wagner, A. Effect of environmental enrichment and
isolation on behavioral and histological indices following focal ischemia in old rats. GeroScience 2022, 44, 211–228. [CrossRef]
110. Boboc, I.K.S.; Chirea, A.C.; Gheorman, V.; Gresita, A.; Balseanu, T.A.; Catalin, B.; Calina, D. Investigating the Neuroprotective and
Neuroregenerative Effect of Trazodone Regarding Behavioral Recovery in a BL6C57 Mice Stroke Model. Curr. Health Sci. J. 2023,
49, 210–219. [CrossRef]
Life 2024, 14, 1110 25 of 31
111. Pinosanu, L.R.; Boboc, I.K.S.; Balseanu, T.A.; Gresita, A.; Hermann, D.M.; Popa-Wagner, A.; Catalin, B. Beam narrowing test:
A motor index of post-stroke motor evaluation in an aged rat model of cerebral ischemia. J. Neural Transm. 2024, 131, 763–771.
[CrossRef]
112. Boboc, I.K.S.; Rotaru-Zavaleanu, A.D.; Calina, D.; Albu, C.V.; Catalin, B.; Turcu-Stiolica, A. A Preclinical Systematic Review and
Meta-Analysis of Behavior Testing in Mice Models of Ischemic Stroke. Life 2023, 13, 567. [CrossRef] [PubMed]
113. Zhang, G.; Chen, L.; Yang, L.; Hua, X.; Zhou, B.; Miao, Z.; Li, J.; Hu, H.; Namaka, M.; Kong, J.; et al. Combined use of spatial
restraint stress and middle cerebral artery occlusion is a novel model of post-stroke depression in mice. Sci. Rep. 2015, 5, 16751.
[CrossRef]
114. Cunningham, C.J.; Wong, R.; Barrington, J.; Tamburrano, S.; Pinteaux, E.; Allan, S.M. Systemic conditioned medium treatment
from interleukin-1 primed mesenchymal stem cells promotes recovery after stroke. Stem Cell Res. Ther. 2020, 11, 32. [CrossRef]
115. Kawai, H.; Yamashita, T.; Ohta, Y.; Deguchi, K.; Nagotani, S.; Zhang, X.; Ikeda, Y.; Matsuura, T.; Abe, K. Tridermal tumorigenesis
of induced pluripotent stem cells transplanted in ischemic brain. J. Cereb. Blood Flow. Metab. 2010, 30, 1487–1493. [CrossRef]
116. Patkar, S.; Tate, R.; Modo, M.; Plevin, R.; Carswell, H.V. Conditionally immortalised neural stem cells promote functional recovery
and brain plasticity after transient focal cerebral ischaemia in mice. Stem Cell Res. 2012, 8, 14–25. [CrossRef] [PubMed]
117. Sun, R.; Peng, M.; Xu, P.; Huang, F.; Xie, Y.; Li, J.; Hong, Y.; Guo, H.; Liu, Q.; Zhu, W. Low-density lipoprotein receptor (LDLR)
regulates NLRP3-mediated neuronal pyroptosis following cerebral ischemia/reperfusion injury. J. Neuroinflamm. 2020, 17, 330.
[CrossRef]
118. Cao, Z.; Balasubramanian, A.; Pedersen, S.E.; Romero, J.; Pautler, R.G.; Marrelli, S.P. TRPV1-mediated Pharmacological Hypother-
mia Promotes Improved Functional Recovery Following Ischemic Stroke. Sci. Rep. 2017, 7, 17685. [CrossRef] [PubMed]
119. Suenaga, J.; Hu, X.; Pu, H.; Shi, Y.; Hassan, S.H.; Xu, M.; Leak, R.K.; Stetler, R.A.; Gao, Y.; Chen, J. White matter injury and
microglia/macrophage polarization are strongly linked with age-related long-term deficits in neurological function after stroke.
Exp. Neurol. 2015, 272, 109–119. [CrossRef] [PubMed]
120. Ding, Z.; Gao, J.; Feng, Y.; Wang, M.; Zhao, H.; Wu, R.; Zheng, X.; Feng, X.; Lai, M. Electroacupuncture Ameliorates
Depression-Like Behaviors in Post-Stroke Rats via Activating AMPK-Mediated Mitochondrial Function. Neuropsychiatr. Dis. Treat.
2023, 19, 2657–2671. [CrossRef]
121. Wang, S.; Sun, H.; Liu, S.; Wang, T.; Guan, J.; Jia, J. Role of hypothalamic cannabinoid receptors in post-stroke depression in rats.
Brain Res. Bull. 2016, 121, 91–97. [CrossRef]
122. Qian, L.; Huang, S.; Liu, X.; Jiang, Y.; Jiang, Y.; Hu, Y.; Yang, Z. Morroniside improves the symptoms of post-stroke depression in
mice through the BDNF signaling pathway mediated by MiR-409-3p. Phytomedicine 2024, 123, 155224. [CrossRef] [PubMed]
123. Xu, Y.; Liang, L. Vitamin D3/vitamin D receptor signaling mitigates symptoms of post-stroke depression in mice by upregulating
hippocampal BDNF expression. Neurosci. Res. 2021, 170, 306–313. [CrossRef] [PubMed]
124. Gaignier, F.; Legrand-Frossi, C.; Stragier, E.; Mathiot, J.; Merlin, J.L.; Cohen-Salmon, C.; Lanfumey, L.; Frippiat, J.P. A Model of
Chronic Exposure to Unpredictable Mild Socio-Environmental Stressors Replicates Some Spaceflight-Induced Immunological
Changes. Front. Physiol. 2018, 9, 514. [CrossRef] [PubMed]
125. Zhang, M.; Li, A.; Yang, Q.; Li, J.; Zheng, L.; Wang, G.; Sun, Y.; Huang, Y.; Zhang, M.; Song, Z.; et al. Matrine alleviates
depressive-like behaviors via modulating microbiota–gut–brain axis in CUMS-induced mice. J. Transl. Med. 2023, 21, 145.
[CrossRef]
126. Yan, W.; Dong, Z.; Zhao, D.; Li, J.; Zeng, T.; Mo, C.; Gao, L.; Lv, Z. Xiaoyaosan Exerts Antidepressant Effect by Downregulating
RAGE Expression in Cingulate Gyrus of Depressive-Like Mice. Front. Pharmacol. 2021, 12, 703965. [CrossRef] [PubMed]
127. Mus, at, M.I.; Mitran, S.I.; Udris, toiu, I.; Albu, C.V.; Cătălin, B. The impact of stress on the behavior of C57BL/6 mice with liver
injury: A comparative study. Front. Behav. Neurosci. 2024, 18, 1358964. [CrossRef]
128. Wu, J.; Li, J.; Gaurav, C.; Muhammad, U.; Chen, Y.; Li, X.; Chen, J.; Wang, Z. CUMS and dexamethasone induce depression-like
phenotypes in mice by differentially altering gut microbiota and triggering macroglia activation. Gen. Psychiatr. 2021, 34, e100529.
[CrossRef]
129. Wang, Y.-l.; Wu, H.-r.; Zhang, S.-s.; Xiao, H.-l.; Yu, J.; Ma, Y.-y.; Zhang, Y.-d.; Liu, Q. Catalpol ameliorates depressive-like behaviors
in CUMS mice via oxidative stress-mediated NLRP3 inflammasome and neuroinflammation. Transl. Psychiatry 2021, 11, 353.
[CrossRef]
130. Wang, G.; Lei, C.; Tian, Y.; Wang, Y.; Zhang, L.; Zhang, R. Rb1, the Primary Active Ingredient in Panax ginseng C.A. Meyer, Exerts
Antidepressant-Like Effects via the BDNF-Trkb-CREB Pathway. Front. Pharmacol. 2019, 10, 1034. [CrossRef]
131. Wen, G.; Yao, H.; Li, Y.; Ding, R.; Ren, X.; Tan, Y.; Ren, W.; Yu, H.; Zhan, X.; Wang, X.; et al. Regulation of Tau Protein on the
Antidepressant Effects of Ketamine in the Chronic Unpredictable Mild Stress Model. Front. Psychiatry 2019, 10, 287. [CrossRef]
132. Li, M.; Fu, Q.; Li, Y.; Li, S.; Xue, J.; Ma, S. Emodin opposes chronic unpredictable mild stress induced depressive-like behavior in
mice by upregulating the levels of hippocampal glucocorticoid receptor and brain-derived neurotrophic factor. Fitoterapia 2014,
98, 1–10. [CrossRef] [PubMed]
133. Xie, M.; Wang, H.; Peng, J.; Qing, D.; Zhang, X.; Guo, D.; Meng, P.; Luo, Z.; Wang, X.; Peng, Q. Acacetin protects against
depression-associated dry eye disease by regulating ubiquitination of NLRP3 through gp78 signal. Front. Pharmacol. 2022,
13, 984475. [CrossRef]
134. Wassouf, Z.; Hentrich, T.; Casadei, N.; Jaumann, M.; Knipper, M.; Riess, O.; Schulze-Hentrich, J.M. Distinct Stress Response and
Altered Striatal Transcriptome in Alpha-Synuclein Overexpressing Mice. Front. Neurosci. 2018, 12, 1033. [CrossRef] [PubMed]
Life 2024, 14, 1110 26 of 31
135. Wang, Y.; Gu, J.H.; Liu, L.; Liu, Y.; Tang, W.Q.; Ji, C.H.; Guan, W.; Zhao, X.Y.; Sun, Y.F.; Xu, D.W.; et al. Hippocampal PPARα
Plays a Role in the Pharmacological Mechanism of Vortioxetine, a Multimodal-Acting Antidepressant. Front. Pharmacol. 2021,
12, 673221. [CrossRef]
136. Kalliokoski, O.; Teilmann, A.C.; Jacobsen, K.R.; Abelson, K.S.; Hau, J. The lonely mouse—Single housing affects serotonergic
signaling integrity measured by 8-OH-DPAT-induced hypothermia in male mice. PLoS ONE 2014, 9, e111065. [CrossRef]
137. Zhang, Y.; Yu, P.; Liu, H.; Yao, H.; Yao, S.; Yuan, S.Y.; Zhang, J.C. Hyperforin improves post-stroke social isolation-induced
exaggeration of PSD and PSA via TGF-β. Int. J. Mol. Med. 2019, 43, 413–425. [CrossRef] [PubMed]
138. Chen, H.H.; Zhang, N.; Li, W.Y.; Fang, M.R.; Zhang, H.; Fang, Y.S.; Ding, M.X.; Fu, X.Y. Overexpression of brain-derived
neurotrophic factor in the hippocampus protects against post-stroke depression. Neural Regen. Res. 2015, 10, 1427–1432.
[CrossRef]
139. Niu, L.; Jin, X.; Zhang, Y.; Liu, B.; Li, C. Feasibility of focal cerebral ischemia and reperfusion surgery combined with chronic
unpredictable mild stress to simulate the post-stroke depressive state in rats. Behav. Brain Funct. 2015, 11, 39. [CrossRef]
140. Wang, A.R.; Mi, L.F.; Zhang, Z.L.; Hu, M.Z.; Zhao, Z.Y.; Liu, B.; Li, Y.B.; Zheng, S. Saikosaponin A improved depression-like
behavior and inhibited hippocampal neuronal apoptosis after cerebral ischemia through p-CREB/BDNF pathway. Behav. Brain
Res. 2021, 403, 113138. [CrossRef]
141. Aarsland, T.I.; Landaas, E.T.; Hegvik, T.A.; Ulvik, A.; Halmøy, A.; Ueland, P.M.; Haavik, J. Serum concentrations of kynurenines
in adult patients with attention-deficit hyperactivity disorder (ADHD): A case-control study. Behav. Brain Funct. 2015, 11, 36.
[CrossRef]
142. Pałucha-Poniewiera, A.; Podkowa, K.; Rafało-Ulińska, A. The group II mGlu receptor antagonist LY341495 induces a rapid
antidepressant-like effect and enhances the effect of ketamine in the chronic unpredictable mild stress model of depression in
C57BL/6J mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 109, 110239. [CrossRef]
143. Zain, M.A.; Pandy, V.; Majeed, A.B.A.; Wong, W.F.; Mohamed, Z. Chronic restraint stress impairs sociability but not social
recognition and spatial memoryin C57BL/6J mice. Exp. Anim. 2019, 68, 113–124. [CrossRef] [PubMed]
144. Zhi, L.; Zhang, F.; Liu, H.; Jiang, X.; Zhang, Y.; Yang, Q.; Zhang, X.; Liu, M.; Zhang, Z.; Song, J. CRS induces depression-like
behavior after MCAO in rats possibly by activating p38 MAPK. Behav. Brain Res. 2023, 437, 114104. [CrossRef] [PubMed]
145. Tekam, C.S.; Shinde, S.; Ranjana, P.; Mahto, S. Bilateral Common Carotid Artery Occlusion: Stroke Model; Springer International
Publishing: Cham, Switzerland, 2021; pp. 41–56.
146. Di Lorenzo, A.; Nabavi, S.F.; Sureda, A.; Moghaddam, A.H.; Khanjani, S.; Arcidiaco, P.; Nabavi, S.M.; Daglia, M. Antidepressive-like
effects and antioxidant activity of green tea and GABA green tea in a mouse model of post-stroke depression. Mol. Nutr. Food Res.
2016, 60, 566–579. [CrossRef] [PubMed]
147. Daglia, M.; Di Lorenzo, A.; Nabavi, S.F.; Sureda, A.; Khanjani, S.; Moghaddam, A.H.; Braidy, N.; Nabavi, S.M. Improvement of
Antioxidant Defences and Mood Status by Oral GABA Tea Administration in a Mouse Model of Post-Stroke Depression. Nutrients
2017, 9, 446. [CrossRef]
148. Nabavi, S.M.; Nabavi, S.F.; Sureda, A.; Caprioli, G.; Iannarelli, R.; Sokeng, A.J.T.; Braidy, N.; Khanjani, S.; Moghaddam, A.H.;
Atanasov, A.G.; et al. The water extract of tutsan (Hypericum androsaemum L.) red berries exerts antidepressive-like effects and
in vivo antioxidant activity in a mouse model of post-stroke depression. Biomed. Pharmacother. 2018, 99, 290–298. [CrossRef]
149. Aggarwal, A.; Gaur, V.; Kumar, A. Nitric oxide mechanism in the protective effect of naringin against post-stroke depression
(PSD) in mice. Life Sci. 2010, 86, 928–935. [CrossRef] [PubMed]
150. Lee, S.R.; Choi, B.; Paul, S.; Seo, J.H.; Back, D.B.; Han, J.S.; Choi, D.H.; Kwon, K.J.; Shin, C.Y.; Lee, J.; et al. Depressive-like
behaviors in a rat model of chronic cerebral hypoperfusion. Transl. Stroke Res. 2015, 6, 207–214. [CrossRef]
151. Hu, G.; Zhou, C.; Wang, J.; Ma, X.; Ma, H.; Yu, H.; Peng, Z.; Huang, J.; Cai, M. Electroacupuncture treatment ameliorates
depressive-like behavior and cognitive dysfunction via CB1R dependent mitochondria biogenesis after experimental global
cerebral ischemic stroke. Front. Cell Neurosci. 2023, 17, 1135227. [CrossRef]
152. Molcho, L.; Ben-Zur, T.; Barhum, Y.; Offen, D. DJ-1 based peptide, ND-13, promote functional recovery in mouse model of focal
ischemic injury. PLoS ONE 2018, 13, e0192954. [CrossRef]
153. Vahid-Ansari, F.; Lagace, D.C.; Albert, P.R. Persistent post-stroke depression in mice following unilateral medial prefrontal
cortical stroke. Transl. Psychiatry 2016, 6, e863. [CrossRef] [PubMed]
154. Happ, D.; Tasker, R.A.; Wegener, G.P. 2.038—Endothelin-1 injection into the left medial prefrontal cortex induces anxiety-like
symptoms—A possible model for post-stroke anxiety? Eur. Neuropsychopharmacol. 2018, 28, S48–S49. [CrossRef]
155. Sommer, C.J. Ischemic stroke: Experimental models and reality. Acta Neuropathol. 2017, 133, 245–261. [CrossRef]
156. Kim, G.W.; Sugawara, T.; Chan, P.H. Involvement of oxidative stress and caspase-3 in cortical infarction after photothrombotic
ischemia in mice. J. Cereb. Blood Flow. Metab. 2000, 20, 1690–1701. [CrossRef] [PubMed]
157. Talley Watts, L.; Zheng, W.; Garling, R.J.; Frohlich, V.C.; Lechleiter, J.D. Rose Bengal Photothrombosis by Confocal Optical Imaging
In Vivo: A Model of Single Vessel Stroke. J. Vis. Exp. 2015, 100, e52794. [CrossRef]
158. Wester, P.; Watson, B.D.; Prado, R.; Dietrich, W.D. A photothrombotic ‘ring’ model of rat stroke-in-evolution displaying putative
penumbral inversion. Stroke 1995, 26, 444–450. [CrossRef]
159. Lunardi Baccetto, S.; Lehmann, C. Microcirculatory Changes in Experimental Models of Stroke and CNS-Injury Induced
Immunodepression. Int. J. Mol. Sci. 2019, 20, 5184. [CrossRef]
Life 2024, 14, 1110 27 of 31
160. Lindholm, J.S.; Castrén, E. Mice with altered BDNF signaling as models for mood disorders and antidepressant effects. Front.
Behav. Neurosci. 2014, 8, 143. [CrossRef]
161. Holmes, A.; Murphy, D.L.; Crawley, J.N. Abnormal behavioral phenotypes of serotonin transporter knockout mice: Parallels with
human anxiety and depression. Biol. Psychiatry 2003, 54, 953–959. [CrossRef]
162. Craske, M.G.; Meuret, A.E.; Ritz, T.; Treanor, M.; Dour, H.J. Treatment for Anhedonia: A Neuroscience Driven Approach. Depress.
Anxiety 2016, 33, 927–938. [CrossRef]
163. Sah, A.; Schmuckermair, C.; Sartori, S.B.; Gaburro, S.; Kandasamy, M.; Irschick, R.; Klimaschewski, L.; Landgraf, R.;
Aigner, L.; Singewald, N. Anxiety- rather than depression-like behavior is associated with adult neurogenesis in a female mouse
model of higher trait anxiety- and comorbid depression-like behavior. Transl. Psychiatry 2012, 2, e171. [CrossRef] [PubMed]
164. Labenz, C.; Huber, Y.; Michel, M.; Nagel, M.; Galle, P.R.; Kostev, K.; Schattenberg, J.M. Nonalcoholic Fatty Liver Disease Increases
the Risk of Anxiety and Depression. Hepatol. Commun. 2020, 4, 1293–1301. [CrossRef] [PubMed]
165. Wu, H.H.; Wang, S. Strain differences in the chronic mild stress animal model of depression. Behav. Brain Res. 2010, 213, 94–102.
[CrossRef]
166. Primo, M.J.; Fonseca-Rodrigues, D.; Almeida, A.; Teixeira, P.M.; Pinto-Ribeiro, F. Sucrose preference test: A systematic review of
protocols for the assessment of anhedonia in rodents. Eur. Neuropsychopharmacol. 2023, 77, 80–92. [CrossRef]
167. Wang, L.; Cui, Q.; Liu, J.; Zou, H. Emotion Reactivity and Suicide Risk in Patients With Depression: The Mediating Role of
Non-Suicidal Self-Injury and Moderating Role of Childhood Neglect. Front. Psychiatry 2021, 12, 707181. [CrossRef] [PubMed]
168. Busquet, P.; Nguyen, N.K.; Schmid, E.; Tanimoto, N.; Seeliger, M.W.; Ben-Yosef, T.; Mizuno, F.; Akopian, A.; Striessnig, J.; Singewald, N.
CaV1.3 L-type Ca2+ channels modulate depression-like behaviour in mice independent of deaf phenotype.
Int. J. Neuropsychopharmacol. 2010, 13, 499–513. [CrossRef]
169. Seo, J.S.; Wei, J.; Qin, L.; Kim, Y.; Yan, Z.; Greengard, P. Cellular and molecular basis for stress-induced depression. Mol. Psychiatry
2017, 22, 1440–1447. [CrossRef]
170. Sasaki, K.; Halder, S.K.; Matsunaga, H.; Ueda, H. Beneficial actions of prothymosin alpha-mimetic hexapeptide on central
post-stroke pain, reduced social activity, learning-deficit and depression following cerebral ischemia in mice. Peptides 2020,
126, 170265. [CrossRef]
171. Partoazar, A.; Seyyedian, Z.; Zamanian, G.; Saffari, P.M.; Muhammadnejad, A.; Dehpour, A.R.; Goudarzi, R. Neuroprotective
phosphatidylserine liposomes alleviate depressive-like behavior related to stroke through neuroinflammation attenuation in the
mouse hippocampus. Psychopharmacology 2021, 238, 1531–1539. [CrossRef]
172. Wu, D.; Zhang, G.; Zhao, C.; Yang, Y.; Miao, Z.; Xu, X. Interleukin-18 from neurons and microglia mediates depressive behaviors
in mice with post-stroke depression. Brain Behav. Immun. 2020, 88, 411–420. [CrossRef]
173. Weiss, S.M.; Wadsworth, G.; Fletcher, A.; Dourish, C.T. Utility of ethological analysis to overcome locomotor confounds in
elevated maze models of anxiety. Neurosci. Biobehav. Rev. 1998, 23, 265–271. [CrossRef] [PubMed]
174. Li, X.; Zhang, J.; Niu, R.; Manthari, R.K.; Yang, K.; Wang, J. Effect of fluoride exposure on anxiety- and depression-like behavior in
mouse. Chemosphere 2019, 215, 454–460. [CrossRef] [PubMed]
175. Sweeney, P.; O’Hara, K.; Xu, Z.; Yang, Y. HFD-induced energy states-dependent bidirectional control of anxiety levels in mice.
Int. J. Obes. 2017, 41, 1237–1245. [CrossRef] [PubMed]
176. Pitzer, C.; La Porta, C.; Treede, R.D.; Tappe-Theodor, A. Inflammatory and neuropathic pain conditions do not primarily evoke
anxiety-like behaviours in C57BL/6 mice. Eur. J. Pain. 2019, 23, 285–306. [CrossRef] [PubMed]
177. Blasco-Serra, A.; González-Soler, E.M.; Cervera-Ferri, A.; Teruel-Martí, V.; Valverde-Navarro, A.A. A standardization of the
Novelty-Suppressed Feeding Test protocol in rats. Neurosci. Lett. 2017, 658, 73–78. [CrossRef]
178. Pietri, M.; Djillani, A.; Mazella, J.; Borsotto, M.; Heurteaux, C. First evidence of protective effects on stroke recovery and
post-stroke depression induced by sortilin-derived peptides. Neuropharmacology 2019, 158, 107715. [CrossRef]
179. Bourin, M.; Hascoët, M. The mouse light/dark box test. Eur. J. Pharmacol. 2003, 463, 55–65. [CrossRef]
180. Takao, K.; Miyakawa, T. Light/dark transition test for mice. J. Vis. Exp. 2006, 1, e104. [CrossRef]
181. Njung’e, K.u.; Handley, S.L. Evaluation of marble-burying behavior as a model of anxiety. Pharmacol. Biochem. Behav. 1991,
38, 63–67. [CrossRef]
182. Bahi, A.; Dreyer, J.L. Hippocampus-specific deletion of tissue plasminogen activator “tPA” in adult mice impairs depression- and
anxiety-like behaviors. Eur. Neuropsychopharmacol. 2012, 22, 672–682. [CrossRef]
183. Dorninger, F.; Zeitler, G.; Berger, J. Nestlet Shredding and Nest Building Tests to Assess Features of Psychiatric Disorders in Mice.
Bio Protoc. 2020, 10, e3863. [CrossRef] [PubMed]
184. Teo, A.R.; Nelson, S.; Strange, W.; Kubo, H.; Katsuki, R.; Kurahara, K.; Kanba, S.; Kato, T.A. Social withdrawal in major depressive
disorder: A case-control study of hikikomori in japan. J. Affect. Disord. 2020, 274, 1142–1146. [CrossRef] [PubMed]
185. Katayama, Y.; Nishiyama, M.; Shoji, H.; Ohkawa, Y.; Kawamura, A.; Sato, T.; Suyama, M.; Takumi, T.; Miyakawa, T.; Nakayama,
K.I. CHD8 haploinsufficiency results in autistic-like phenotypes in mice. Nature 2016, 537, 675–679. [CrossRef]
186. Peça, J.; Feliciano, C.; Ting, J.T.; Wang, W.; Wells, M.F.; Venkatraman, T.N.; Lascola, C.D.; Fu, Z.; Feng, G. Shank3 mutant mice
display autistic-like behaviours and striatal dysfunction. Nature 2011, 472, 437–442. [CrossRef]
187. Wilson, C.A.; Koenig, J.I. Social interaction and social withdrawal in rodents as readouts for investigating the negative symptoms
of schizophrenia. Eur. Neuropsychopharmacol. 2014, 24, 759–773. [CrossRef] [PubMed]
Life 2024, 14, 1110 28 of 31
188. Kaidanovich-Beilin, O.; Lipina, T.; Vukobradovic, I.; Roder, J.; Woodgett, J.R. Assessment of social interaction behaviors. J. Vis.
Exp. 2011, 48, e2473. [CrossRef]
189. Ni, R.J.; Tian, Y.; Dai, X.Y.; Zhao, L.S.; Wei, J.X.; Zhou, J.N.; Ma, X.H.; Li, T. Social avoidance behavior in male tree shrews and
prosocial behavior in male mice toward unfamiliar conspecifics in the laboratory. Zool. Res. 2020, 41, 258–272. [CrossRef]
190. Liu, Y.; Deng, S.L.; Li, L.X.; Zhou, Z.X.; Lv, Q.; Wang, Z.Y.; Wang, F.; Chen, J.G. A circuit from dorsal hippocampal CA3 to
parvafox nucleus mediates chronic social defeat stress-induced deficits in preference for social novelty. Sci. Adv. 2022, 8, eabe8828.
[CrossRef]
191. Piccin, A.; Contarino, A. Long-lasting pseudo-social aggressive behavior in opiate-withdrawn mice. Prog. Neuropsychopharmacol.
Biol. Psychiatry 2020, 97, 109780. [CrossRef]
192. Alsaeed, I.; Al-Somali, F.; Sakhnini, L.; Aljarallah, O.S.; Hamdan, R.M.; Bubishate, S.A.; Sarfaraz, Z.K.; Kamal, A. Autism-relevant
social abnormalities in mice exposed perinatally to extremely low frequency electromagnetic fields. Int. J. Dev. Neurosci. 2014,
37, 58–64. [CrossRef]
193. Bevins, R.A.; Besheer, J. Object recognition in rats and mice: A one-trial non-matching-to-sample learning task to study ‘recognition
memory’. Nat. Protoc. 2006, 1, 1306–1311. [CrossRef] [PubMed]
194. Takahashi, H.; Tsuboi, A. Olfactory Habituation-dishabituation Test (Mouse). Bio Protoc. 2017, 7, e2154. [CrossRef] [PubMed]
195. Zou, J.; Wang, W.; Pan, Y.W.; Lu, S.; Xia, Z. Methods to measure olfactory behavior in mice. Curr. Protoc. Toxicol. 2015,
63, 11.18.11–11.18.21. [CrossRef]
196. Choi, K.W.; Kim, Y.K.; Jeon, H.J. Comorbid Anxiety and Depression: Clinical and Conceptual Consideration and Transdiagnostic
Treatment. Adv. Exp. Med. Biol. 2020, 1191, 219–235. [CrossRef]
197. Koolhaas, J.M.; Coppens, C.M.; de Boer, S.F.; Buwalda, B.; Meerlo, P.; Timmermans, P.J. The resident-intruder paradigm:
A standardized test for aggression, violence and social stress. J. Vis. Exp. 2013, 77, e4367. [CrossRef]
198. Fan, Z.; Zhu, H.; Zhou, T.; Wang, S.; Wu, Y.; Hu, H. Using the tube test to measure social hierarchy in mice. Nat. Protoc. 2019, 14,
819–831. [CrossRef]
199. Yang, C.R.; Bai, Y.Y.; Ruan, C.S.; Zhou, H.F.; Liu, D.; Wang, X.F.; Shen, L.J.; Zheng, H.Y.; Zhou, X.F. Enhanced aggressive behaviour
in a mouse model of depression. Neurotox. Res. 2015, 27, 129–142. [CrossRef]
200. Douglas, K.M.; Gallagher, P.; Robinson, L.J.; Carter, J.D.; McIntosh, V.V.; Frampton, C.M.; Watson, S.; Young, A.H.;
Ferrier, I.N.; Porter, R.J. Prevalence of cognitive impairment in major depression and bipolar disorder. Bipolar Disord. 2018,
20, 260–274. [CrossRef] [PubMed]
201. Liu, X.; Zhang, M.; Liu, H.; Zhu, R.; He, H.; Zhou, Y.; Zhang, Y.; Li, C.; Liang, D.; Zeng, Q.; et al. Bone marrow mesenchymal stem
cell-derived exosomes attenuate cerebral ischemia-reperfusion injury-induced neuroinflammation and pyroptosis by modulating
microglia M1/M2 phenotypes. Exp. Neurol. 2021, 341, 113700. [CrossRef]
202. Balseanu, A.T.; Buga, A.M.; Catalin, B.; Wagner, D.C.; Boltze, J.; Zagrean, A.M.; Reymann, K.; Schaebitz, W.; Popa-Wagner, A.
Multimodal Approaches for Regenerative Stroke Therapies: Combination of Granulocyte Colony-Stimulating Factor with Bone
Marrow Mesenchymal Stem Cells is Not Superior to G-CSF Alone. Front. Aging Neurosci. 2014, 6, 130. [CrossRef]
203. Yang, K.; Tan, Y.; Wang, F.; Zhang, Q.; Sun, P.; Zhang, Y.; Yao, N.; Zhao, Y.; Wang, X.; Fan, A.; et al. The improvement of spatial
memory deficits in APP/V717I transgenic mice by chronic anti-stroke herb treatment. Exp. Biol. Med. 2014, 239, 1007–1017.
[CrossRef]
204. Pitts, M.W. Barnes Maze Procedure for Spatial Learning and Memory in Mice. Bio Protoc. 2018, 8, e2744. [CrossRef]
205. Kraeuter, A.K.; Guest, P.C.; Sarnyai, Z. The Y-Maze for Assessment of Spatial Working and Reference Memory in Mice. Methods
Mol. Biol. 2019, 1916, 105–111. [CrossRef] [PubMed]
206. Huo, K.; Wei, M.; Zhang, M.; Wang, Z.; Pan, P.; Shaligram, S.S.; Huang, J.; Prado, L.B.D.; Wong, J.; Su, H. Reduction of
neuroinflammation alleviated mouse post bone fracture and stroke memory dysfunction. J. Cereb. Blood Flow. Metab. 2021,
41, 2162–2173. [CrossRef]
207. Ahnstedt, H.; Patrizz, A.; Chauhan, A.; Roy-O’Reilly, M.; Furr, J.W.; Spychala, M.S.; D’Aigle, J.; Blixt, F.W.; Zhu, L.; Bravo Alegria,
J.; et al. Sex differences in T cell immune responses, gut permeability and outcome after ischemic stroke in aged mice. Brain Behav.
Immun. 2020, 87, 556–567. [CrossRef] [PubMed]
208. Toshkezi, G.; Kyle, M.; Longo, S.L.; Chin, L.S.; Zhao, L.R. Brain repair by hematopoietic growth factors in the subacute phase of
traumatic brain injury. J. Neurosurg. 2018, 129, 1286–1294. [CrossRef] [PubMed]
209. Olton, D.S.; Collison, C.; Werz, M.A. Spatial memory and radial arm maze performance of rats. Learn. Motiv. 1977, 8, 289–314.
[CrossRef]
210. Kohler, J.; Mei, J.; Banneke, S.; Winter, Y.; Endres, M.; Emmrich, J.V. Assessing spatial learning and memory in mice: Classic radial
maze versus a new animal-friendly automated radial maze allowing free access and not requiring food deprivation. Front. Behav.
Neurosci. 2022, 16, 1013624. [CrossRef]
211. Borlongan, C.V.; Cahill, D.W.; Sanberg, P.R. Locomotor and passive avoidance deficits following occlusion of the middle cerebral
artery. Physiol. Behav. 1995, 58, 909–917. [CrossRef]
212. Senechal, Y.; Kelly, P.H.; Dev, K.K. Amyloid precursor protein knockout mice show age-dependent deficits in passive avoidance
learning. Behav. Brain Res. 2008, 186, 126–132. [CrossRef]
213. Singh, N.; Ma, B.; Leonardo, C.C.; Ahmad, A.S.; Narumiya, S.; Doré, S. Role of PGE2 EP1 receptor in intracerebral hemorrhage-
induced brain injury. Neurotox. Res. 2013, 24, 549–559. [CrossRef] [PubMed]
Life 2024, 14, 1110 29 of 31
214. Deacon, R.M. Measuring motor coordination in mice. J. Vis. Exp. 2013, 75, e2609. [CrossRef]
215. Jacobs, J.R.; Carey, M.R. Move Over Rotarod, Here Comes RotaWheel. Neuroscience 2021, 466, 258–259. [CrossRef]
216. Nguyen, K.P.; Sharma, A.; Gil-Silva, M.; Gittis, A.H.; Chase, S.M. Distinct Kinematic Adjustments over Multiple Timescales
Accompany Locomotor Skill Development in Mice. Neuroscience 2021, 466, 260–272. [CrossRef]
217. Syeara, N.; Bagchi, S.; Al Shoyaib, A.; Karamyan, S.T.; Alamri, F.F.; Karamyan, V.T. The Finer Aspects of Grid-Walking and
Cylinder Tests for Experimental Stroke Recovery Studies in Mice. Methods Mol. Biol. 2023, 2616, 345–353. [CrossRef]
218. Zalewska, K.; Pietrogrande, G.; Ong, L.K.; Abdolhoseini, M.; Kluge, M.; Johnson, S.J.; Walker, F.R.; Nilsson, M. Sustained
administration of corticosterone at stress-like levels after stroke suppressed glial reactivity at sites of thalamic secondary
neurodegeneration. Brain Behav. Immun. 2018, 69, 210–222. [CrossRef]
219. Modo, M.; Stroemer, R.P.; Tang, E.; Veizovic, T.; Sowniski, P.; Hodges, H. Neurological sequelae and long-term behavioural
assessment of rats with transient middle cerebral artery occlusion. J. Neurosci. Methods 2000, 104, 99–109. [CrossRef]
220. Hayashi, K.; Hasegawa, Y.; Takemoto, Y.; Cao, C.; Mukasa, A.; Kim-Mitsuyama, S. Enhanced oxidative stress contributes to worse
prognosis and delayed neurofunctional recovery after striatal intracerebral hemorrhage in 5XFAD mice. Eur. J. Neurosci. 2020,
51, 1806–1814. [CrossRef] [PubMed]
221. Carter, R.J.; Morton, J.; Dunnett, S.B. Motor coordination and balance in rodents. Curr. Protoc. Neurosci. 2001, 15, 8–12. [CrossRef]
222. Hu, M.Z.; Wang, A.R.; Zhao, Z.Y.; Chen, X.Y.; Li, Y.B.; Liu, B. Antidepressant-like effects of paeoniflorin on post-stroke depression
in a rat model. Neurol. Res. 2019, 41, 446–455. [CrossRef]
223. Du, Y.; Liang, H.; Zhang, L.; Fu, F. Administration of Huperzine A exerts antidepressant-like activity in a rat model of post-stroke
depression. Pharmacol. Biochem. Behav. 2017, 158, 32–38. [CrossRef]
224. Ji, S.; Kronenberg, G.; Balkaya, M.; Färber, K.; Gertz, K.; Kettenmann, H.; Endres, M. Acute neuroprotection by pioglitazone after
mild brain ischemia without effect on long-term outcome. Exp. Neurol. 2009, 216, 321–328. [CrossRef]
225. Feng, L.; Han, C.-X.; Cao, S.-Y.; Zhang, H.-M.; Wu, G.-Y. Deficits in motor and cognitive functions in an adult mouse model of
hypoxia-ischemia induced stroke. Sci. Rep. 2020, 10, 20646. [CrossRef] [PubMed]
226. Shi, X.; Bai, H.; Wang, J.; Wang, J.; Huang, L.; He, M.; Zheng, X.; Duan, Z.; Chen, D.; Zhang, J.; et al. Behavioral Assessment
of Sensory, Motor, Emotion, and Cognition in Rodent Models of Intracerebral Hemorrhage. Front. Neurol. 2021, 12, 667511.
[CrossRef] [PubMed]
227. Metz, G.A.; Whishaw, I.Q. The ladder rung walking task: A scoring system and its practical application. J. Vis. Exp. 2009,
28, e1204. [CrossRef]
228. Tamakoshi, K.; Ishida, A.; Takamatsu, Y.; Hamakawa, M.; Nakashima, H.; Shimada, H.; Ishida, K. Motor skills training promotes
motor functional recovery and induces synaptogenesis in the motor cortex and striatum after intracerebral hemorrhage in rats.
Behav. Brain Res. 2014, 260, 34–43. [CrossRef]
229. Sun, J.; Wei, Z.Z.; Gu, X.; Zhang, J.Y.; Zhang, Y.; Li, J.; Wei, L. Intranasal delivery of hypoxia-preconditioned bone marrow-
derived mesenchymal stem cells enhanced regenerative effects after intracerebral hemorrhagic stroke in mice. Exp. Neurol. 2015,
272, 78–87. [CrossRef]
230. Beray-Berthat, V.; Delifer, C.; Besson, V.C.; Girgis, H.; Coqueran, B.; Plotkine, M.; Marchand-Leroux, C.; Margaill, I. Long-term
histological and behavioural characterisation of a collagenase-induced model of intracerebral haemorrhage in rats. J. Neurosci.
Methods 2010, 191, 180–190. [CrossRef] [PubMed]
231. Schaar, K.L.; Brenneman, M.M.; Savitz, S.I. Functional assessments in the rodent stroke model. Exp. Transl. Stroke Med. 2010, 2, 13.
[CrossRef]
232. de Oliveira, J.L.; Ávila, M.; Martins, T.C.; Alvarez-Silva, M.; Winkelmann-Duarte, E.C.; Salgado, A.S.I.; Cidral-Filho, F.J.; Reed,
W.R.; Martins, D.F. Medium- and long-term functional behavior evaluations in an experimental focal ischemic stroke mouse
model. Cogn. Neurodyn 2020, 14, 473–481. [CrossRef]
233. Zarruk, J.; Garcia-Yebenes, I.; Romera, V.G.; Ballesteros, I.; Moraga, A.; Cuartero, M.; Hurtado, O.; Sobrado, M.; Pradillo, J.;
Fernandez-Lopez, D.; et al. Neurological tests for functional outcome assessment in rodent models of ischaemic stroke.
Rev. Neurol. 2011, 53, 607–618. [PubMed]
234. Hao, J.; Mdzinarishvili, A.; Abbruscato, T.J.; Klein, J.; Geldenhuys, W.J.; Van der Schyf, C.J.; Bickel, U. Neuroprotection in mice by
NGP1-01 after transient focal brain ischemia. Brain Res. 2008, 1196, 113–120. [CrossRef]
235. Narayan, S.K.; Grace Cherian, S.; Babu Phaniti, P.; Babu Chidambaram, S.; Rachel Vasanthi, A.H.; Arumugam, M. Preclinical
animal studies in ischemic stroke: Challenges and some solutions. Anim. Model. Exp. Med. 2021, 4, 104–115. [CrossRef] [PubMed]
236. Allred, R.P.; Adkins, D.L.; Woodlee, M.T.; Husbands, L.C.; Maldonado, M.A.; Kane, J.R.; Schallert, T.; Jones, T.A. The vermicelli
handling test: A simple quantitative measure of dexterous forepaw function in rats. J. Neurosci. Methods 2008, 170, 229–244.
[CrossRef] [PubMed]
237. Yoshida, H.M.; Lima, F.O.; Barreira, J.; Appenzeller, S.; Fernandes, P.T. Is there a correlation between depressive symptoms and
motor skills in post-stroke patients? Arq. Neuropsiquiatr. 2019, 77, 155–160. [CrossRef]
238. Ritzel, R.M.; Lai, Y.J.; Crapser, J.D.; Patel, A.R.; Schrecengost, A.; Grenier, J.M.; Mancini, N.S.; Patrizz, A.; Jellison, E.R.;
Morales-Scheihing, D.; et al. Aging alters the immunological response to ischemic stroke. Acta Neuropathol. 2018, 136, 89–110.
[CrossRef]
239. Gu, S.; He, Z.; Xu, Q.; Dong, J.; Xiao, T.; Liang, F.; Ma, X.; Wang, F.; Huang, J.H. The Relationship Between 5-Hydroxytryptamine
and Its Metabolite Changes With Post-stroke Depression. Front. Psychiatry 2022, 13, 871754. [CrossRef]
Life 2024, 14, 1110 30 of 31
240. Sullivan, G.M.; Ogden, R.T.; Huang, Y.Y.; Oquendo, M.A.; Mann, J.J.; Parsey, R.V. Higher in vivo serotonin-1a binding in
posttraumatic stress disorder: A PET study with [11C]WAY-100635. Depress. Anxiety 2013, 30, 197–206. [CrossRef]
241. Spasojevic, N.; Jovanovic, P.; Dronjak, S. Chronic fluoxetine treatment affects gene expression of catecholamine enzymes in the
heart of depression model rats. Indian. J. Exp. Biol. 2012, 50, 771–775.
242. Starr, L.R.; Hammen, C.; Brennan, P.A.; Najman, J.M. Serotonin transporter gene as a predictor of stress generation in depression.
J. Abnorm. Psychol. 2012, 121, 810–818. [CrossRef]
243. Kim, J.M.; Stewart, R.; Kim, S.W.; Shin, I.S.; Kim, J.T.; Park, M.S.; Park, S.W.; Kim, Y.H.; Cho, K.H.; Yoon, J.S. Associations of
cytokine gene polymorphisms with post-stroke depression. World J. Biol. Psychiatry 2012, 13, 579–587. [CrossRef] [PubMed]
244. Ogłodek, E. Changes in the Serum Levels of Cytokines: IL-1β, IL-4, IL-8 and IL-10 in Depression with and without Posttraumatic
Stress Disorder. Brain Sci. 2022, 12, 387. [CrossRef] [PubMed]
245. Wang, L.; Wang, R.; Liu, L.; Qiao, D.; Baldwin, D.S.; Hou, R. Effects of SSRIs on peripheral inflammatory markers in patients with
major depressive disorder: A systematic review and meta-analysis. Brain Behav. Immun. 2019, 79, 24–38. [CrossRef] [PubMed]
246. Zhang, S.; Zong, Y.; Ren, Z.; Hu, J.; Wu, X.; Xiao, H.; Qin, S.; Zhou, G.; Ma, Y.; Zhang, Y.; et al. Regulation of indoleamine 2,
3-dioxygenase in hippocampal microglia by NLRP3 inflammasome in lipopolysaccharide-induced depressive-like behaviors. Eur.
J. Neurosci. 2020, 52, 4586–4601. [CrossRef]
247. Holsboer, F. The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 2000, 23, 477–501. [CrossRef]
248. Pace, T.W.; Hu, F.; Miller, A.H. Activation of cAMP-protein kinase A abrogates STAT5-mediated inhibition of glucocorticoid
receptor signaling by interferon-alpha. Brain Behav. Immun. 2011, 25, 1716–1724. [CrossRef]
249. El Husseini, N.; Laskowitz, D.T. The role of neuroendocrine pathways in prognosis after stroke. Expert. Rev. Neurother. 2014,
14, 217–232. [CrossRef]
250. Kim, J.M.; Stewart, R.; Kang, H.J.; Kim, S.Y.; Kim, S.W.; Shin, I.S.; Park, M.S.; Kim, H.R.; Shin, M.G.; Cho, K.H.; et al. A longitudinal
study of BDNF promoter methylation and genotype with poststroke depression. J. Affect. Disord. 2013, 149, 93–99. [CrossRef]
251. Zhang, Y.; Jiang, H.; Yue, Y.; Yin, Y.; Zhang, Y.; Liang, J.; Li, S.; Wang, J.; Lu, J.; Geng, D.; et al. The protein and mRNA expression
levels of glial cell line-derived neurotrophic factor in post stroke depression and major depressive disorder. Sci. Rep. 2017, 7, 8674.
[CrossRef]
252. Kunze, A.; Zierath, D.; Drogomiretskiy, O.; Becker, K. Strain differences in fatigue and depression after experimental stroke.
Transl. Stroke Res. 2014, 5, 604–611. [CrossRef]
253. Lavu, V.K.; Mohamed, R.A.; Huang, R.; Potla, S.; Bhalla, S.; Al Qabandi, Y.; Nandula, S.A.; Boddepalli, C.S.; Gutlapalli, S.D.; Mohammed, L.
Evaluation and Treatment of Depression in Stroke Patients: A Systematic Review. Cureus 2022, 14, e28137. [CrossRef]
254. Starkstein, S.E.; Hayhow, B.D. Treatment of Post-Stroke Depression. Curr. Treat. Options Neurol. 2019, 21, 31. [CrossRef]
255. Medeiros, G.C.; Roy, D.; Kontos, N.; Beach, S.R. Post-stroke depression: A 2020 updated review. Gen. Hosp. Psychiatry 2020,
66, 70–80. [CrossRef] [PubMed]
256. Zhao, Z.; Zhang, W.; Zhang, Y.; Zhao, Y.; Zheng, C.; Tian, H.; Lei, J.; Liu, Y.; Zhao, R.; Tang, Q. Multimodal Magnetic Resonance
Imaging and Therapeutic Intervention With Yi-nao-jie-yu Decoction in a Rat Model of Post-stroke Depression. Front. Psychiatry
2020, 11, 557423. [CrossRef]
257. Chen, C.; Dong, Y.; Liu, F.; Gao, C.; Ji, C.; Dang, Y.; Ma, X.; Liu, Y. A Study of Antidepressant Effect and Mechanism on Intranasal
Delivery of BDNF-HA2TAT/AAV to Rats with Post-Stroke Depression. Neuropsychiatr. Dis. Treat. 2020, 16, 637–649. [CrossRef]
258. Ngwa, C.; Al Mamun, A.; Qi, S.; Sharmeen, R.; Xu, Y.; Liu, F. Regulation of microglial activation in stroke in aged mice:
A translational study. Aging 2022, 14, 6047–6065. [CrossRef] [PubMed]
259. Morioka, T.; Kalehua, A.N.; Streit, W.J. Characterization of microglial reaction after middle cerebral artery occlusion in rat brain.
J. Comp. Neurol. 1993, 327, 123–132. [CrossRef] [PubMed]
260. Lambertsen, K.L.; Biber, K.; Finsen, B. Inflammatory cytokines in experimental and human stroke. J. Cereb. Blood Flow. Metab.
2012, 32, 1677–1698. [CrossRef]
261. Kang, J.B.; Son, H.K.; Shah, M.A.; Koh, P.O. Retinoic acid attenuates ischemic injury-induced activation of glial cells and
inflammatory factors in a rat stroke model. PLoS ONE 2024, 19, e0300072. [CrossRef] [PubMed]
262. Cojocaru, A.; Burada, E.; Băls, eanu, A.T.; Deftu, A.F.; Cătălin, B.; Popa-Wagner, A.; Osiac, E. Roles of Microglial Ion Channel in
Neurodegenerative Diseases. J. Clin. Med. 2021, 10, 1239. [CrossRef]
263. Miao, Z.; Wang, Y.; Sun, Z. The Relationships Between Stress, Mental Disorders, and Epigenetic Regulation of BDNF. Int. J. Mol.
Sci. 2020, 21, 1375. [CrossRef]
264. Wu, X.; Gu, J.; Zou, Z.; Yu, M.; Zhang, C.; Xiao, Q.; Chen, X.; Li, C. Suppressive Effects of Isofraxidin on Depressive-like Behaviors
Induced by Chronic Unpredictable Mild Stress in Mice. Brain Sci. 2022, 12, 1376. [CrossRef] [PubMed]
265. Kim, J.S.; Choi-Kwon, S. Poststroke depression and emotional incontinence: Correlation with lesion location. Neurology 2000,
54, 1805–1810. [CrossRef]
266. Harciarek, M.; Mańkowska, A. Hemispheric stroke: Mood disorders. Handb. Clin. Neurol. 2021, 183, 155–167. [CrossRef]
267. Carson, A.J.; MacHale, S.; Allen, K.; Lawrie, S.M.; Dennis, M.; House, A.; Sharpe, M. Depression after stroke and lesion location:
A systematic review. Lancet 2000, 356, 122–126. [CrossRef] [PubMed]
268. Singh, A.; Herrmann, N.; Black, S.E. The importance of lesion location in poststroke depression: A critical review. Can. J. Psychiatry
1998, 43, 921–927. [CrossRef] [PubMed]
Life 2024, 14, 1110 31 of 31
269. Dockman, R.L.; Carpenter, J.M.; Diaz, A.N.; Benbow, R.A.; Filipov, N.M. Sex differences in behavior, response to LPS, and glucose
homeostasis in middle-aged mice. Behav. Brain Res. 2022, 418, 113628. [CrossRef]
270. Furman, O.; Tsoory, M.; Chen, A. Differential chronic social stress models in male and female mice. Eur. J. Neurosci. 2022,
55, 2777–2793. [CrossRef]
271. Sohrabji, F.; Okoreeh, A.; Panta, A. Sex hormones and stroke: Beyond estrogens. Horm. Behav. 2019, 111, 87–95. [CrossRef]
272. Jiang, H.; Xiao, L.; Jin, K.; Shao, B. Estrogen administration attenuates post-stroke depression by enhancing CREB/BDNF/TrkB
signaling in the rat hippocampus. Exp. Ther. Med. 2021, 21, 433. [CrossRef]
273. Zeng, P.Y.; Tsai, Y.H.; Lee, C.L.; Ma, Y.K.; Kuo, T.H. Minimal influence of estrous cycle on studies of female mouse behaviors.
Front. Mol. Neurosci. 2023, 16, 1146109. [CrossRef] [PubMed]
274. Tsao, C.-H.; Wu, K.-Y.; Su, N.C.; Edwards, A.; Huang, G.-J. The influence of sex difference on behavior and adult hippocampal
neurogenesis in C57BL/6 mice. Sci. Rep. 2023, 13, 17297. [CrossRef] [PubMed]
275. Tariq, M.B.; Lee, J.; McCullough, L.D. Sex differences in the inflammatory response to stroke. Semin. Immunopathol. 2023,
45, 295–313. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.