Abstract
Post‐stroke depression (PSD) is a common psychiatric complication affecting nearly one‐third of stroke survivors, leading to increased disability, mortality, and cognitive decline. Traditional Chinese Medicine (TCM) has proven effective in treating PSD through syndrome differentiation, yet existing animal models primarily reflect Western medical concepts and fail to incorporate the TCM principle of “同病异治” (treating the same disease with different methods). This paper provides a review of the current methods for constructing animal models of post‐stroke depression (PSD) from the perspective of Traditional Chinese Medicine (TCM) syndrome differentiation and proposes multi‐dimensional assessment indicators. By integrating TCM theories with modern biomedical techniques, this study offers a comprehensive framework for deepening the understanding of the pathogenesis and therapeutic evaluation of PSD. This approach not only contributes to advancing PSD research but also paves the way for innovative treatment strategies that combine traditional and modern medicine.
Keywords: animal models, integrative medicine, post‐stroke depression, traditional Chinese medicine
Post‐stroke depression (PSD) animal model of traditional Chinese medicine (TCM) combined with disease and syndrome often uses western medicine surgery (including: middle cerebral artery embolization to construct cerebral ischemia, or brain injection of collagenase to construct cerebral hemorrhage) combined with other methods (including: isolation, restraint, sleep deprivation, high‐fat diet, Chinese medicine gavage, etc.) to construct. The characteristics of the model are different due to the constructed syndromes. It is mainly manifested in external symptoms (e.g. mental state; color changes of eyes, ears, tongue; hair state; diet, water, urine, and feces, etc.) and behavioral performance (e.g. irritability, fear of cold, slow response, reduced activity, etc.).

1. INTRODUCTION
Post‐stroke depression (PSD) is a common and debilitating psychiatric condition affecting approximately one‐third of stroke survivors. Its impact on rehabilitation outcomes, quality of life, and mortality rates underscores the urgent need for effective interventions. 1 Despite advances in Western medicine, current PSD therapies are limited in their ability to address the condition's multifaceted pathophysiology, which includes inflammation, oxidative stress, neuroinflammation, neurotrophic deficiencies, and impaired neuroplasticity. 2 , 3 Additionally, the roles of the immune system, autonomic nervous system, and hypothalamic–pituitary–adrenal (HPA) axis dysfunction have been identified as critical contributors to PSD. 4 , 5
From the perspective of Traditional Chinese Medicine (TCM), PSD is understood as a combination of “郁证” (depression syndrome) (depression) and “中风” (stroke), rooted in “气血失衡”(imbalances of qi and blood) and dysfunction of the “脏腑” (zang‐fu organs). This conceptualization provides a holistic framework for diagnosis and treatment, emphasizing individualized approaches. However, most current PSD animal models are based on Western medical definitions, overlooking TCM principles such as “同病异治” (treating the same disease with different methods). 6 This disconnect presents a significant gap in research, as models that fail to incorporate TCM syndrome differentiation may not fully capture PSD's complex pathogenesis.
Recent studies integrating TCM with modern biomedical approaches have explored novel animal models for PSD, offering insights into disease mechanisms and therapeutic strategies. However, existing models often lack standardization, and their evaluation criteria remain inconsistent, which limits their applicability in the context of TCM treatment. 7 To address these challenges, this review proposes a comprehensive framework for PSD research, focusing on the construction and evaluation of animal models that align with TCM syndrome differentiation and modern pathological mechanisms. By constructing animal models from the perspective of TCM, this approach not only helps deepen the understanding of the pathogenesis of PSD but also promotes the practical application of integrative treatment strategies, providing effective reference and support for the TCM treatment of PSD.
2. ETIOLOGICAL ANALYSIS OF POST‐STROKE DEPRESSION BASED ON TCM THEORY
Although the term PSD is not explicitly mentioned in classical texts, modern TCM categorizes PSD within the scope of “郁证” (depression syndrome) (depression), reflecting the dual pathology of “中风” (stroke) (cerebrovascular accident) and “郁证” (depression syndrome) (emotional depression). For example, the “《素问·六元正纪大论》” (Su Wen Liuyuan Zheng Ji Da Lun) introduces the concept of “五行郁证” (depression syndromes of the five elements), which describes that “木郁达之,火郁发之,土郁夺之,金郁泄之,水郁折之” (wood depression is resolved by dispersing, fire depression by releasing, earth depression by draining, metal depression by alleviating, and water depression by overcoming).
Furthermore, the text emphasizes that “百病皆生于郁” (all diseases arise from depression) 8 and “凡病无不起于郁” (no disease exists without originating from depression). 9 These statements demonstrate that TCM has long recognized the pivotal role of emotional depression in the onset of various diseases. Similarly, the term “中风” (stroke) first appears in the “黄帝内经” (The Yellow Emperor’s Canon of Internal Medicine), 10 which attributes its etiology to external pathogenic factors, emotional disturbances such as anger, individual constitution, and dietary habits. This reflects the TCM view that stroke and depression are interrelated conditions with reciprocal causative interactions, rooted in internal imbalances of “气” (qi), “血” (blood), and “脏腑功能” (organ function).
In modern medicine, PSD is classified as a neuropsychiatric complication of cerebrovascular disorders. However, in the theoretical framework of TCM's theory of “络” (collaterals) or collateral disorders, cerebral vessels and brain collaterals are believed to share both structural and functional homology. This offers a distinctive lens for understanding post‐stroke pathology within TCM.
For instance, Academician Wang 11 highlighted the critical interplay of “血瘀” (blood stasis), “热毒” (heat toxin), and “痰毒” (phlegm toxin) after a stroke as key pathogenic factors. He proposed that therapeutic strategies aimed at eliminating these “toxins” could significantly enhance recovery from cerebrovascular diseases.
Furthermore, Yang et al. 12 identified PSD as a prototypical model of “因病致郁” (depression induced by illness). From this perspective, stroke‐related dysfunction of the “脏腑” (zang‐fu organs) leads to impaired circulation of “气” (qi), “血” (blood), and “津液” (body fluids). These disruptions gradually accumulate, giving rise to “郁证” (depression syndrome). Over time, the persistent functional impairments—particularly involving the “心” (heart zang), “肝” (liver zang), “脾” (spleen zang), and “肾” (kidneys zang)‐converge into a complex and interwoven pathological mechanism underlying the development of PSD.
Additionally, modern research indicates that patients with PSD present with diverse constitutional types, most commonly including “气虚” (qi deficiency), “气滞” (stagnation of qi), and “阴虚” (yin deficiency). 13 These constitutional types not only shape the underlying pathogenesis of PSD but also influence the disease's location, severity, and prognosis. Consequently, identifying and differentiating these constitutional types holds significant clinical value, providing a theoretical basis for individualized diagnosis and treatment strategies in TCM.
3. INVESTIGATION OF THE MODELING METHODS AND MOLECULAR MECHANISMS OF ISCHEMIC AND HEMORRHAGIC STROKE COMBINED WITH TCM SYNDROMES
Under the guidance of TCM syndrome differentiation, PSD animal models are typically constructed based on observable behavioral and physiological characteristics of the animals, such as mental state, water intake, locomotor activity, fur condition, and patterns of urination and defecation. 14 Commonly used approaches involve combining stroke modeling techniques (e.g. middle cerebral artery occlusion (MCAO) 15 or intracerebral collagenase injection) with multiple interventions to simulate disease‐syndrome models that correspond to TCM syndromes. Representative models include “肾虚肝郁” (kidney deficiency with liver depression), “气虚血瘀” (qi deficiency with blood stasis), “痰瘀互结” (phlegm and blood stasis interlocking), “火毒证” (fire toxin syndrome), and “肾阳虚证” (kidney yang deficiency syndrome).
There is a certain degree of correspondence between modern modeling methods and TCM pathophysiological concepts. For instance, focal cerebral ischemia induced by MCAO reflects the TCM notion of “血瘀” (blood stasis), 16 while hemorrhagic injury induced by collagenase injection corresponds to the theory of “离经之血为瘀” (extravasated blood becoming stasis). 17 Additional interventions such as high‐fat diets, forced swimming, and fasting are employed to mimic the TCM mechanisms of “劳则气耗” (over‐strain consuming essence), “饮食自倍,肠胃乃伤” (spleen injury from improper diet), and “饮食劳倦即伤脾” (phlegm formation due to dampness), resulting in “气虚” (qi deficiency) and “血瘀” (blood stasis) or phlegm‐blood stasis interlocking syndromes. 18 , 19 Emotional disturbances induced via the chronic unpredictable mild stress (CUMS) model simulate the “气滞” (qi stagnation) mechanism described in “《灵枢 本神》” (Lingshu·Ben Shen): “愁忧者,气闭塞而不行” (Worry and sorrow cause the qi to stagnate). 20 , 21 “肾阳虚” (kidney yang deficiency) is modeled through cold exposure, cold diets, or hydrocortisone injection, based on “《素问 至真要大论》” (Suwen·Zhi Zhen Yao Da Lun): “诸寒收引,皆属于肾” (all cold‐induced contractions pertain to the kidney). 22 , 23 Administration of hot herbs like Aconitum carmichaelii or hypertonic saline replicates “阳盛则热” (excessive yang transforming into fire), thereby modeling syndromes related to yang excess. 24 , 25 , 26
3.1. Ischemic stroke
The ischemic stroke model integrated with TCM syndrome differentiation is commonly established using MCAO, combined with various interventions such as high‐fat diet feeding, intraperitoneal injection of carrageenan solution, forced swimming, or fasting. MCAO induces transient focal cerebral ischemia, leading to increased blood–brain barrier (BBB) permeability, 27 activation of inflammatory and oxidative stress pathways, and ultimately causing neuronal injury.
This model reliably reproduces core pathological features of human ischemic stroke, including the formation of an ischemic penumbra and the development of substantial infarct volumes. 28 , 29 Moreover, it offers high reproducibility and controllable reperfusion durations, making it a widely accepted tool for investigating neuroprotective mechanisms.
When integrated with TCM principles, these interventions are selected to simulate specific syndromes, such as “气虚” (qi deficiency), “血瘀” (blood stasis), or “痰瘀互结” (phlegm‐damp obstruction), based on classical theories like “劳则气耗” (over‐strain consuming essence) and “饮食劳倦即伤脾” (dietary and emotional strain impair the spleen). 18 , 19 Such combinations aim to enhance the models relevance to both biomedical pathology and TCM syndrome differentiation.
3.1.1. Modeling method for PSD with “肾虚肝郁证” (kidney deficiency and liver qi stagnation syndrome)
In TCM, PSD is classified as a combination of “郁证” (depression syndrome) and “中风” (stroke), with its occurrence closely linked to liver and kidney dysfunction. Clinically, this syndrome corresponds to symptoms such as emotional instability, cognitive decline, and, in severe cases, dementia‐like presentations. To replicate this TCM syndrome in animal models, Chen et al. 30 established a rat model of PSD characterized by kidney deficiency and liver qi stagnation, utilizing the middle cerebral artery occlusion (MCAO) method combined with chronic restraint stress (CRS) and social isolation. Similarly, Zhi et al. 31 adopted a comparable modeling protocol and observed typical signs in rats, including dull fur, hypoactivity, weight loss, anhedonia, and general lethargy. Behavioral assessments, such as the sucrose preference test (SPT), forced swimming test (FST), open field test (OFT), and Y‐maze test revealed significant anxiety‐ and depression‐like phenotypes, supporting the models validity for simulating PSD with this specific TCM syndrome type.
Mechanistically, this modeling strategy is believed to trigger aberrant activation of multiple molecular pathways implicated in neuropsychiatric and neuroinflammatory processes, including JAK2/STAT3, 30 RhoA‐ROCK, 32 and p38MAPK. 31 These signaling cascades are known to mediate key aspects of stroke pathophysiology, such as inflammation, oxidative stress, apoptosis, neuronal injury, and impaired synaptic plasticity. Consequently, their dysregulation contributes to the neurological dysfunctions observed in PSD, including emotional disturbances and behavioral deficits such as anxiety 31 and depressive‐like behaviors. 33
3.1.2. Modeling method for PSD with “气虚血瘀证” (qi deficiency and blood stasis syndrome)
The ancient medical text “《医家四要》” (Four Key Points of Doctor) states, 33 “气为血之帅,血为气之母,气即病矣,则血不得独行,故亦从而病焉” (qi is the commander of blood, and blood is the mother of qi. When qi becomes deficient, blood cannot flow freely and thus also becomes pathological). In stroke patients, the depletion of qi impairs cerebral circulation, leading to stagnation of blood in the brain and contributing to stroke onset and progression. 34 To replicate this TCM syndrome, Li et al. 35 established a model by inducing “气虚” (qi deficiency) exhaustive forced swimming and fasting, while implementing MCAO to simulate “血瘀” (blood stasis), Similarly, Yang et al. 36 applied a multifactorial strategy‐combining exhaustive swimming, sleep deprivation, fasting, high‐fat diet, and MCAO —to establish a more comprehensive “气虚血瘀证” (qi deficiency and blood stasis syndrome) model.
Typical symptoms observed in these models include signs of both “气虚血瘀” (qi deficiency and blood stasis), such as fatigue, lethargy, low responsiveness, hypoactivity, 37 dry and shedding fur, darkened tongue, red eyes, and purplish discoloration of the tail and footpads. 38 Biochemical assessments revealed significant increases in total cholesterol, triglycerides, and low‐density lipoprotein cholesterol levels, indicating dysregulated lipid metabolism and systemic blood stasis.
Mechanistically, the combination of physical stressors and ischemia activates the NF‐κB/iNOS‐COX‐2 signaling pathway, 38 promoting inflammation and oxidative stress. 39 Concurrently, suppression of the brain‐derived neurotrophic factor (BDNF)‐TrkB pathway contributes to impaired synaptic plasticity, 40 neuronal injury, delayed cellular repair, and increased apoptosis. 41 These pathological alterations result in anxiety‐like behaviors and cognitive impairments in the model animals, as evidenced by reduced time in the target quadrant during the open field test, prolonged escape latency in the Morris water maze, and decreased accuracy in the Y‐maze test. 39
3.1.3. Modeling method for PSD with “痰瘀互结证” (phlegm and blood stasis syndrome)
In TCM, the combination of phlegm and blood stasis is regarded as a core pathological mechanism in stroke, reflecting the critical role of vascular obstruction and impaired circulation in disease progression. To replicate this pathological state in animals, researchers commonly induce hyperlipidemia by administering a high‐fat diet to simulate phlegm syndrome, while employing MCAO to model blood stasis.
For instance, Zhang et al. 42 and Li et al. 43 successfully established PSD models with phlegm and blood stasis features using this combined approach. The animals exhibited classic signs including reduced activity, dull and greasy fur, limb paralysis, hyperlipidemia, and impaired coagulation—closely mirroring clinical manifestations of PSD with this TCM syndrome.
At the molecular level, the PPARγ/LXRα/ABCG1 signaling axis is essential for cholesterol efflux and reverse transport. 39 , 40 Disruption of this pathway accelerates atherosclerosis and promotes lipid accumulation. Furthermore, dysregulated cholesterol metabolism activates the NF‐κB/MAPK pathway, which initiates a cascade of inflammatory responses. 44 This leads to elevated expression of pro‐inflammatory cytokines such as TNF‐α, IL‐6, and IL‐1β, resulting in chronic inflammation, vascular endothelial damage, and increased stroke susceptibility.
In addition, activation of the mTOR/HIF‐1α/VEGF signaling pathway 42 , 43 exacerbates oxidative stress and abnormal cell proliferation, contributing to lipid peroxidation, reactive oxygen species (ROS) accumulation, mitochondrial dysfunction, and DNA damage, ultimately inducing apoptosis. 45
In summary, these findings offer valuable insights into the pathological basis of stroke in the context of phlegm and blood stasis syndrome. They also highlight potential molecular targets for intervention. 46 For example, activating PPARγ or inhibiting the mTOR pathway may help mitigate lipid metabolism disorders and systemic inflammation, providing novel therapeutic strategies for PSD associated with “痰瘀互结” (phlegm and blood stasis).
3.1.4. Modeling method for PSD with “火毒证” (fire toxin syndrome)
During the Qing Dynasty, Chen described in “ 《洞天奥旨》” (Insight into the Profound Themes of Truth) 47 that “火未必退而气先失,毒未必化而血先涸” (the fire may not dissipate, but the qi is already lost; the toxin may not be cleared, but the blood has already dried up).
This statement encapsulates the pathogenesis of “火毒证” (fire toxin syndrome) in TCM, wherein retained pathogenic heat disrupts the equilibrium of qi and blood, ultimately resulting in impaired consciousness and systemic dysfunction. In modern biomedical terms, this syndrome aligns with severe neuroinflammatory and metabolic dysregulation, both of which contribute to cerebral damage and vascular disorders implicated in stroke.
To simulate this syndrome in animal models, researchers often employ intraperitoneal injection of carrageenan, a known inflammatory inducer, in conjunction with MCAO followed by reperfusion. Liu Bo et al. 48 successfully established a fire toxin model using this method, observing hallmark signs such as decreased activity, reduced food intake, dull fur, piloerection, dark purplish coloration of ear vasculature, and swollen limbs. Ma et al. 49 further expanded on this model to investigate the co‐occurrence of fire toxin and blood stasis syndromes, highlighting their potential pathological synergy.
Dong et al. 50 introduced an enhanced protocol by inducing blood stasis through MCAO, while administering subcutaneous yeast suspension to trigger “火毒证” (fire toxin syndrome). This approach produced a compound stroke model characterized by severe neuroinflammation, vascular congestion, and thrombotic events.
Mechanistically, carrageenan and yeast extract activated proinflammatory signaling cascades such as NF‐κB, ERK1/2, and p38, 51 , 52 resulting in elevated levels of TNF‐α and IL‐6 and widespread inflammatory cell infiltration. These responses exacerbated vascular stasis and thrombosis, 53 confirmed by reductions in tail blood flow and pathological changes consistent with Fire Toxin, including lethargy, piloerection, and purplish discoloration of the ears. 54 , 55
This model highlights the strong link between “火毒证” (fire toxin syndrome) and ischemic stroke pathology, particularly the central roles of inflammation and oxidative stress. Fire toxin not only intensifies localized cerebral injury but also induces systemic inflammation via pathways. multiple signaling pathways, these findings closely mirror inflammation‐driven mechanisms in modern stroke pathology and suggest actionable molecular targets for intervention. For instance, inhibition of NF‐κB 47 or ERK1/2 53 signaling has been shown to attenuate inflammatory responses and prevent stroke exacerbation.
The integration of this syndrome‐based model with targeted pathway analysis provides a deeper understanding of fire toxin syndrome from a TCM perspective, while simultaneously offering translational value for contemporary biomedical research and therapeutic development.
3.1.5. Modeling method for PSD with “肾阳虚证” (kidney yang deficiency syndrome)
In TCM, the kidney is regarded as the root of the body's yang energy. Stroke patients, who are typically elderly, often experience age‐related decline in kidney function, resulting in yang deficiency. 56 This deficiency disrupts the circulation of qi and blood, leading to insufficient nourishment of the brain and constituting a key pathological basis of PSD. 57
Zhang et al. 58 developed a rat model of ischemic stroke with Kidney Yang Deficiency Syndrome by combining MCAO with hydrocortisone injection. Successful modeling was confirmed by classic yang deficiency symptoms, including hypo‐responsiveness, reduced locomotor activity, aversion to cold, and preference for warmth. Hydrocortisone not only induced autophagy in microvascular endothelial cells and promoted apoptosis but also impaired angiogenesis, leading to hippocampal neuronal damage and affecting cognitive and memory functions. 59 Moreover, by inhibiting the cAMP/PKA signaling pathway, hydrocortisone reduced the expression of neurotransmitters such as 5‐hydroxytryptamine (5‐HT), ACh, dopamine (DA), and norepinephrine (NE), resulting in pronounced cognitive deficits and anxiety‐like behaviors. 60
When hydrocortisone treatment was combined with CUMS, depressive‐ and anxiety‐like behaviors were further exacerbated. Rats exhibited characteristic signs of “肾阳虚” (kidney yang deficiency), such as dull,lusterless fur and increased urine output, Behavioral assessments including the sucrose preference test (SPT), open field test (OFT), and forced swimming test (FST) revealed significant depression‐like phenotypes. Biochemical analyses showed marked reductions in serum 5‐HT and NE levels, 61 confirming the stability and reproducibility of the kidney yang deficiency PSD model. 62
In addition, environmental exposure to cold temperature combined with cold diet has also been used to induce “肾阳虚” (kidney yang deficiency) in animal models. 23 Cold stress initially activate the PKA pathway to maintain temperature, 63 but prolonged exposure disrupts the HPA axis, elevating angiotensin 1 (ET‐1) levels, 64 which in turn impair coagulation homeostasis. These environmental factors further aggravate Kidney Yang Deficiency and deepen systemic metabolic and circulatory disorders, as evidenced by alterations in WBC, RBC, and PLT levels. 65
At the molecular level, these studies elucidate the multifaceted role of “肾阳虚” (kidney yang deficiency) in PSD pathology Both hydrocortisone and cold stress impair cognitive function through direct neuronal injury and autophagy activation, while also inducing systemic immune metabolic dysregulation via signaling pathway modulation. Targeting pathways such as cAMP/PKA, or restoring neurotransmitter balance, may represent promising strategies for treating PSD associated with kidney yang deficiency, addressing both cognitive impairment and emotional disturbances.
3.2. Hemorrhagic stroke
Similar to ischemic stroke, hemorrhagic stroke models are often established using a combination of collagenase injection and other interventions. Collagenase injection into the brain is one of the commonly used methods for inducing cerebral hemorrhage. Studies have shown that it mainly forms hematomas by damaging microvasculature at the injection site, 66 reducing cell viability around the hematoma, and increasing blood–brain barrier disruption. 67 This process generates ROS, triggers inflammatory responses, and results in neurological deficits. 68
3.2.1. Modeling method for PSD with “肝阳化风证” (hyperactive liver‐yang causing syndrome of liver‐wind)
Hemorrhagic stroke is fundamentally related to “肝肾阴虚” (liver and kidney yin deficiency), with “肝阳上亢” (hyperactivity of liver‐yang) and “肝阳化风” (hyperactive liver‐yang causing syndrome of liver‐wind) as secondary features. Yuan et al. 69 established a model of “肝阳化风” (hyperactive liver‐yang causing syndrome of liver‐wind) Syndrome by combining oral administration of Fuzi Decoction, hypertonic saline intake, and collagenase injection into the brain. The model was considered successful when rats exhibited neurological deficits, thirst, reduced food intake, and dull fur. 70 Oral administration of Fuzi Decoction further aggravated irritability, increased water intake, and increased susceptibility to startle, inducing renal arteriole sclerosis, which is consistent with the manifestations of “肝阳上亢” (hyperactivity of liver‐yang syndrome). 70 , 71 However, hypertonic saline may inhibit VEGF expression, thereby preventing activation of the Notch signaling pathway and reducing BBB permeability. 72 , 73
Li Z et al. 71 compared hypertensive rats with “肝阳上亢证” (hyperactivity of liver‐yang syndrome) to general hypertensive rats, finding elevated ROS and Akt levels, as well as renal vascular proliferation, in the former. This suggests that ROS may induce oxidative stress via the PI3K/Akt pathway. Additionally, this model showed increased levels of malondialdehyde (MDA), heme oxygenase‐1, and oxidized glutathione, along with decreased glutathione, which may be associated with the Nrf2 signaling pathway. 74 , 75
Other studies using intraperitoneal dopamine injections established similar models, showing significantly elevated levels of renin and ET‐1, with gradual normalization of heart rate and respiration after treatment cessation. 76
3.2.2. Modeling method for PSD with “痰热腑实证” (phlegm‐heat internal excess syndrome)
Sun et al. 77 successfully constructed a hemorrhagic stroke model associated with Phlegm‐Heat Internal Excess Syndrome by combining intracerebral collagenase injection with oral administration of rat fecal slurry. Ping 78 further refined the model by applying MCAO in conjunction with a high‐fat diet and fecal slurry gavage, which significantly elevated serum total cholesterol (TC), triglycerides (TG), and low‐density lipoprotein cholesterol (LDL‐C) in rats.
Successful modeling was indicated by hallmark symptoms of the syndrome, including neurological deficits, polydipsia, irritability, and pharyngeal congestion with sputum. 78 Compared to conventional ischemic stroke models, rats with Phlegm‐Heat Internal Excess Syndrome displayed more severe dyslipidemia, vascular pathologies such as atherosclerotic changes, and decreased motilin levels, indicating exacerbated metabolic dysfunction. 79 , 80
According to Ping, 78 fecal slurry more accurately represents the “internal excess” component of the syndrome, while a high‐fat diet simulates the “phlegm‐heat” aspect. Their combination offers a more comprehensive and pathophysiologically relevant model for studying this specific TCM syndrome in the context of stroke.
Mechanistically, gut microbiota play a key role in regulating host immune and metabolic responses. 81 Fecal slurry gavage can activate the AMPK‐ULK1‐P62 signaling pathway via butyrate, leading to mitophagy induction, Nrf2‐mediated antioxidant activity, inhibition of ferroptosis, and attenuation of hepatic injury. 82 This highlights the interconnected roles of metabolic disorders, inflammation, and neural damage in post‐stroke pathology.
Overall, this model reveals the complex pathophysiology of Phlegm‐Heat Internal Excess Syndrome, suggesting that targeting cholesterol metabolism and gut microecological balance 83 may offer novel therapeutic strategies for hemorrhagic stroke and its related TCM syndromes.
4. EVALUATION OF PSD MODELS
Behavioral evaluations play a critical role in validating PSD animal models and assessing the therapeutic efficacy of interventions. Given that PSD encompasses both neurological impairment and depressive‐like behaviors, comprehensive assessments should include sensory, motor, cognitive, and emotional domains. 84 Integrating multiple behavioral paradigms while minimizing environmental interference enhances the scientific rigor and reliability of PSD model evaluation.
4.1. Behavioral assessment
Behavioral assessment is essential for verifying model validity and investigating the mechanisms underlying PSD. As PSD represents a combination of stroke and depression syndrome in TCM theory, behavioral tests should concurrently evaluate neurological deficits and affective symptoms. 84 , 85
Stroke‐induced neurological dysfunction is typically characterized by impairments in motor coordination, sensory processing, learning, and memory, whereas depressive‐like behaviors are manifested as anhedonia, reduced locomotor activity, and diminished exploratory behavior.
Commonly used neurological evaluation tools include:
Longa Score.
Bederson Score.
Modified Neurological Severity Score (mNSS).
Foot Fault Test (TFFT).
Modified Grip Traction Test (MGTT).
Standard depression‐ and anxiety‐related behavioral tests include:
Sucrose Preference Test (SPT).
Open Field Test (OFT).
Morris Water Maze (MWM).
Forced Swim Test (FST).
Tail Suspension Test (TST).
These methods enable comprehensive assessment of motor‐sensory deficits, cognitive impairments, and emotional states, providing multidimensional insights into PSD pathology. However, due to the co‐occurrence of motor impairments in PSD animals, performance in certain cognitive and emotional tests (e.g. MGTT, FST, MWM) may be confounded. 86 Therefore, careful selection and combination of behavioral paradigms is essential, especially when evaluating PSD induced by different pathological mechanisms or syndromes.
For instance, Li et al. 87 assessed the impact of gut microbiota modulation on post‐stroke neurological and emotional outcomes using neurological scoring, SPT, and OFT. Lai et al. 74 , 75 investigated the antidepressant effects of bitter almond extract through SPT and neurological scores. Jiang 88 explored the protective role of oxytocin receptor activation in PSD mice using SPT, OFT, and TST.
Despite the availability of diverse behavioral assays lack of standardization, variability in experimental conditions (e.g. temperature, noise, odor), and observer bias contribute to inconsistent results. Future research should aim to (1) minimize environmental interference, (2) incorporate automated video tracking systems, and (3) apply computer vision‐based image recognition technologies to objectively quantify behavioral outcomes and reduce human error, thereby improving reproducibility, accuracy, and inter‐laboratory consistency.
4.2. Physiological and biochemical indicators
In addition to behavioral assessments, physiological and biochemical markers are essential for evaluating the pathological mechanisms and therapeutic responses in PSD models. Key indicators include inflammatory cytokines, oxidative stress markers, neurotrophic factors, and neurotransmitters.
4.2.1. Inflammatory cytokines (IL‐1, IL‐6, IL‐18, TNF‐α)
Pro‐inflammatory cytokines such as IL‐1, IL‐6, IL‐18, and tumor necrosis factor‐alpha (TNF‐α) are critically involved in the development and progression of PSD. 89 Elevated levels of these cytokines contribute to neuroinflammation, which disrupts the hypothalamic–pituitary–adrenal (HPA) axis, alters neurotransmitter homeostasis, impairs neurotrophic signaling, and disturbs gut microbiota balance—all of which play integral roles in PSD pathophysiology. 90 , 91
Research has shown that vagus nerve stimulation (VNS) can reduce infarct size and improve neurological outcomes by downregulating TNF‐α and other pro‐inflammatory cytokines, thereby suppressing post‐stroke inflammatory responses. 92
Moreover, compounds such as resveratrol have demonstrated therapeutic potential by lowering TNF‐α, IL‐1β, and IL‐6 levels, reducing oxidative stress, and modulating key neurotransmitters such as 5‐hydroxytryptamine (5‐HT) and brain‐derived neurotrophic factor (BDNF). These effects collectively alleviate depressive‐like behaviors in PSD animal models. 92
4.2.2. Oxidative stress indicators (SOD, MDA)
Superoxide dismutase (SOD) and malondialdehyde (MDA) are widely recognized markers of oxidative stress. While SOD reflects the organism's antioxidant defense capacity, MDA indicates the severity of lipid peroxidation and cellular oxidative damage. Imbalances in these markers are frequently observed in PSD and correlate with neuronal degeneration and apoptosis. 93 , 94
Pharmacological agents such as scutellarin have been shown to enhance SOD activity and reduce MDA levels, thereby mitigating neuronal injury and improving oxidative resilience in stroke models. 95 Likewise, the synthetic triterpenoid CDDO‐Im has been reported to decrease both MDA and inflammatory cytokines (e.g. IL‐6, IL‐1β), resulting in reduced neuronal loss and alleviation of depression‐like symptoms in PSD rats. 96
These findings underscore the importance of modulating oxidative stress pathways as a therapeutic strategy for PSD and its neurological complications.
4.2.3. Neurotrophic factors (BDNF)
BDNF is a key molecular indicator in PSD, playing a vital role in neuronal survival, synaptic plasticity, and central nervous system recovery. 97 Decreased BDNF levels, especially under chronic stress, are associated with impaired neuroplasticity and are closely linked to the onset and severity of depressive symptoms. 98 , 99
As a result, BDNF is widely regarded as a predictive biomarker for identifying individuals at risk of PSD and as a therapeutic indicator for evaluating treatment efficacy. 100 Pharmacological agents such as antidepressants have been shown to enhance BDNF expression, thereby promoting neural regeneration and mitigating depression‐like behaviors. 101
In addition, TCM formulations such as “柴胡疏肝散” (Chaihu Shugan San) have demonstrated the ability to upregulate BDNF levels in PSD animal models, often accompanied by improvements in emotional regulation and behavioral performance. 102
Moreover, studies have shown that inhibiting the JAK2/STAT3 signaling pathway can further enhance BDNF expression, 103 offering neuroprotective effects by preventing hippocampal neuronal damage and supporting cognitive recovery. 104 , 105 , 106 , 107
Overall, BDNF serves as a central biomarker that reflects both pathological changes and therapeutic responses in PSD, providing a valuable target for future interventions.
4.2.4. Neurotransmitters (5‐HT, NE, DA)
5‐HT, NE, and DA are central to the regulation of emotion, motivation, and cognition. 108 , 109 Decreased levels of these monoamines are strongly associated with the development of PSD. Research has shown that DA and NE pathways TCM formulations (e.g. “解郁汤” (Jieyu Decoction) 110 ) and acupuncture therapies (e.g. “调神运枢” (Tiaoshen Yunshu acupuncture) 111 ) have demonstrated efficacy in upregulating 5‐HT and NE levels, thereby improving depressive symptoms in both PSD models and patients. Stroke‐induced neural damage compromises 5‐HT synthesis, storage, and uptake, while pharmacological enhancement of DA signaling has been shown to reverse motor dysfunction and depressive behaviors. 112 , 113 As such, neurotransmitter levels in brain tissue serve as important biochemical markers for evaluating PSD pathology and treatment efficacy.
4.3. Histopathological evaluation (infarct size, microglial polarization, synaptic plasticity)
Following stroke, insufficient cerebral perfusion, mitochondrial dysfunction, and neuroinflammation result in extensive neuronal injury or death, leading to the formation of ischemic cores and penumbra zones. These pathological changes activate the brain's innate immune system, particularly microglia, which play dual roles in neuroinflammation and repair.
Among them, M2‐polarized microglia are associated with tissue repair, angiogenesis, and synaptic remodeling. Promoting M2 polarization has been shown to reduce infarct volume, attenuate axonal injury, and enhance nerve fiber regeneration. 114
Synapses, the primary sites for neuronal communication and plasticity, are fundamental to information processing, learning, and memory storage. 115 Microglial polarization profoundly influences synaptic plasticity. Facilitating synaptogenesis and dendritic spine formation is critical for restoring neural circuit integrity and improving cognitive and emotional outcomes in PSD. 116 , 117
Therefore, synaptic plasticity, infarct size, and microglial activation states are important histopathological indicators for evaluating PSD progression and therapeutic efficacy. The mechanism of stroke is shown in Figure 1 and for the cellular mechanisms of PSD, refer to Figure 2. For a summary of stroke disease model modeling methods and other related information, please see Table 1.
FIGURE 1.

This diagram illustrates the key biological pathways involved in the development of PSD following ischemic stroke, highlighting the roles of inflammation, oxidative stress, and dysregulation of the HPA axis and sympathetic nervous system in neuronal injury and neuroplasticity reduction.
FIGURE 2.

Integrated Model of Post‐Stroke Depression (PSD) and Its Mechanism Pathways. This diagram outlines the key aspects of PSD modeling, including ischemic and hemorrhagic stroke models, classification of Traditional Chinese Medicine (TCM) syndromes, and the related mechanistic pathways. It illustrates the integration of TCM principles in stroke modeling, including various interventions and syndrome typing based on external symptoms, behavioral manifestations, and biomarkers. The figure also emphasizes the involvement of inflammation, oxidative stress, and neurological damage pathways, as well as the impact on the hypothalamic–pituitary–adrenal (HPA) axis and gut‐brain axis.
TABLE 1.
Disease combination model and related pathway analysis of depression.
| Disease type | Syndrome type | Research object | Behavioral performance | Biochemical indicators | Pathological histomorphology | Signaling pathway(s) | Refs. | ||
|---|---|---|---|---|---|---|---|---|---|
| Animal | Modeling Method | Auxiliary means | |||||||
| Ischemic stroke | Kidney Deficiency and Liver Qi Stagnation Syndrome | Male SD rats, body weight (300 ± 20) g | Suture embolism (ischemia for 2 h followed by reperfusion, with suture removal after 2 h) | Chronic Restraint Stress (CRS) + Social Isolation | Dull coat, lethargy, and reduced activity | Hippocampal dentate gyrus neural stem cells; hippocampal and cortical neurons | JAK2/STAT3, RhoA‐ROCK, p38MAPK | [30, 31, 32] | |
| Qi Deficiency and Blood Stasis Syndrome | Male SD rats, body weight (200 ± 20) g | Suture embolism (permanent occlusion) | Forced Swimming + Fasting | External symptoms: Dry, shedding furPurple tongue, dark red eyesPurplish tail, dark footpads Behavioral manifestations:Fatigue, lethargy, reduced activity Slow response to stimuli |
1. PTGS2, SOD, MDA, GSH‐Px ; 2. TLR4, IL‐1β, IL‐6, IL‐17, TNF Circulatory and related markers: F2R, cerebral blood flow, CHO, TG, HDL‐C, LDL‐C, FDP, D‐D, VEGF, ATP |
Cerebral ischemic area; ATP content | NF‐κB/iNOS‐COX‐2; PI3K/Akt/Nrf2;BDNF–TrkB | [35, 36, 37, 39] | |
| Compound Factors (Exhaustive Swimming, Sleep Deprivation, Fasting, High‐Fat Diet) | |||||||||
| Phlegm and Blood Stasis Syndrome | Male SD rats, body weight (200 ± 20) g | Suture embolism (ischemia for 2 h followed by reperfusion) | High‐fat diet feeding | Dull coat, limb paralysis, drooping eyelids, purple tongue, slow reactions, and sluggish movement |
(1) MPO, SOD, MDA; (2) IL‐1α, IL‐1β, IL‐6, TNF‐α, serum NO, VEGF, GSDMD; (3) TC, TG, LDL‐C, TBA, AST, ALT (4) Lipid levels, APTT, PT |
Infarct area; brain cells; liver cells; abdominal aorta and vascular cell morphology | PPARγ/LXRα/ABCG1; NF‐kB/MAPK; mTOR/HIF‐1α/VEGF; NLRP3, Caspase‐1 | [42, 43, 44, 118, 119, 120, 121] | |
| Fire Toxin Syndrome | Male SD rats, body weight (200 ± 20) g | Suture embolism (ischemia for 1.5 h followed by reperfusion) | Intraperitoneal injection of carrageenan solution | Fluffy fur, dull coloration, reddened ear edges, swollen and red‐purple claws, blackened tail | (1) IL‐1β, IL‐6, TNF‐α, MCP‐1, IFN‐γ (2) PT, FIB, APTT, TT | Brain neurons and ultrastructure, mitochondria; infarct area; brain, spleen, and tail tissues; spleen weight; tail blood perfusion | NF‐kB, ERK1/2, p38MAPK | [48, 49, 50, 53, 55, 122] | |
| Kidney Yang Deficiency Syndrome | Male SD rats, body weight (200 ± 20) g | Suture embolism (permanent occlusion) | Intramuscular injection of hydrocortisone injection | Hair loss, polyuria, lethargy, easily startled, hunched posture, cold intolerance with a preference for warmth, reduced appetite, slow reactions, and decreased activity | (1) 5‐HT, DA, NE (2) ACh, WBC, RBC, PLT, plasma corticosterone, angiotensin | Nissl bodies in cortical neurons; organ indices of liver, spleen, and kidney | cAMP/PKA | [58, 60, 61, 64, 65] | |
| Hemorrhagic stroke | Hyperactive Liver‐Yang causing Syndrome of Liver‐Wind | Wistar rats, half male and half female, 8–10 weeks old, body weight (200 ± 20) g | Collagenase Injection into the Brain: 0.4 U of Type VII collagenase dissolved in 2 μL of sterile saline (injection site: 1.4 mm posterior to the bregma, 3.2 mm lateral to the right, and 5.6 mm vertically into the brain) | Fuzi Decoction gavage + high‐salt water drinking | Increased irritability, fluffy and dull fur, thirst, and reduced food intake | ROS, MDA, HO‐1, NQO1, GSSG | Brain tissue water content; peri‐hematoma tissue (e.g. inflammatory cells, red blood cells, neurons) | PI3K/Akt;Notch;Nrf2 | [69, 74] |
| Phlegm‐Heat Internal Excess Syndrome | Wistar rats, half male and half female, 8–10 weeks old, body weight (200 ± 20) g | Injection of Collagenase VII‐Heparin Mixture: 1.25 μL (containing 0.5 U of Collagenase VII and 7 U of Heparin), (injection site: 0.2 mm anterior to the bregma, 4.0 mm lateral to the right of the midline, and 5.5 mm vertically into the brain) | Autologous fecal gavage | Restlessness, increased water intake, increased nasal secretions, phlegm sounds in the throat, dry stools, and increased stool quantity and weight |
(1) Oxidative DNA damage, lipid peroxidation, reactive aldehydes (2) Serum TC, TG, LDL‐C;Plasma AST, ALT levels;Serum motilin, somatostatin levels |
Brain index; brain tissue water content | AMPK‐ULK1‐P62 | [77, 78, 82] | |
5. CONCLUSION
PSD is a multifactorial disorder involving complex biological and psychosocial mechanisms. Western medicine primarily focuses on neuroinflammation, oxidative stress, mitochondrial dysfunction, and neuronal apoptosis, aiming to elucidate the biological basis of PSD‐related neuronal injury. In contrast, TCM emphasizes holistic regulation of organ function and the dynamic balance of qi, blood, and body fluids. Despite differing theoretical frameworks, there is considerable overlap in the underlying mechanisms recognized by both systems.
Current TCM‐based PSD models are typically developed around classic syndromes such as kidney deficiency with liver depression, qi deficiency with blood stasis, and phlegm‐heat internal excess. These models highlight the diversity of pathological processes observed in PSD and reflect TCM's syndrome differentiation approach. However, most models still rely heavily on biomedical techniques, such as MCAO and high‐fat diets, and may fail to fully capture the dynamic, individualized, and holistic nature of TCM syndromes. The gap between animal models and clinical syndromic presentations remains a key limitation. observation, listening/smelling, questioning, and palpation.
While modern medical methods can align with TCM mechanisms to some extent, several limitations persist. From the perspective of Traditional Chinese Medicine (TCM), the understanding of PSD involves a holistic approach that includes the regulation of “气” (qi), “血” (blood), and the balance of the “脏腑” (zang‐fu organs). However, current animal models primarily focus on Western medical definitions and methods, which fail to fully integrate TCM's diagnostic framework, such as its four diagnostic methods “望, 闻, 问, 切” (observation, listening/smelling, questioning, and palpation). This gap significantly limits the ability of these models to accurately capture the dynamic nature of TCM syndromes. Furthermore, the differences in disease progression in animal models versus clinical symptoms in humans present additional challenges in replicating the full complexity of TCM syndrome differentiation. Thus, more precise model selection is needed to ensure better alignment with both modern pathophysiology and TCM's holistic understanding of disease mechanisms.
This study summarizes the key TCM syndrome models for PSD and their corresponding mechanisms, offering a framework for future integration of TCM theory with biomedical evidence. A multidimensional, integrative modeling approach will not only deepen our understanding of PSD pathogenesis but also advance the clinical application of TCM in global contexts.
AUTHOR CONTRIBUTIONS
Jielin Wang: Conceptualization; writing – original draft. Wenlu Ma: Investigation; project administration; supervision. Wei Wu: Software; supervision. Yujuan Fu: Funding acquisition; supervision. Hui Li: Conceptualization; methodology; supervision; validation; writing – review and editing.
FUNDING INFORMATION
This work was supported by Jilin Scientific and Technological Development Program (YDZJ202401092ZYTS), Jilin Provincial Department of Education (JJKH20230958KJ).
CONFLICT OF INTEREST STATEMENT
The authors declare no financial or non‐financial conflicts of interest in this study.
ETHICS APPROVAL AND CONSENT TO PARTICIPATE
As this is a review article, ethics approval and consent to participate are not applicable.
CONSENT FOR PUBLICATION
All authors have given their consent for the publication of this manuscript.
ACKNOWLEDGMENTS
We would also like to acknowledge Figdraw software for providing the tools used to create Figures 1 and 2 in this manuscript.
Wang J, Ma W, Wu W, Fu Y, Li H. Research progress on traditional Chinese medicine animal models of post‐stroke depression and pathological insights. Anim Models Exp Med. 2025;8:1387‐1399. doi: 10.1002/ame2.70068
Contributor Information
Yujuan Fu, Email: fuyj@ccucm.edu.cn.
Hui Li, Email: lihuiterrisa@163.com.
DATA AVAILABILITY STATEMENT
As this is a review article, no new datasets were generated. All data analyzed in this article are available from the cited sources.
REFERENCES
- 1. Ming L. Investigation of post‐stroke depression and anxiety. Chin J Misdiagn. 2008;19:4783‐4784. [Google Scholar]
- 2. Villa RF, Ferrari F, Moretti A. Post‐stroke depression: mechanisms and pharmacological treatment. Pharmacol Ther. 2018;184:131‐144. [DOI] [PubMed] [Google Scholar]
- 3. Jing D, Hou X, Guo X, et al. Astrocytes in post‐stroke depression: roles in inflammation, neurotransmission, and neurotrophin signaling. Cell Mol Neurobiol. 2023;43(7):3301‐3313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Dorrance AM, Fink G. Effects of stroke on the autonomic nervous system. Compr Physiol. 2015;5(3):1241‐1263. [DOI] [PubMed] [Google Scholar]
- 5. Radak D, Resanovic I, Isenovic ER. Changes in hypothalamus‐pituitary–adrenal axis following transient ischemic attack. Angiology. 2014;65(8):723‐732. [DOI] [PubMed] [Google Scholar]
- 6. Juanjuan Y, Haidong W, Jinghai W, et al. Animal model of ankylosing spondylitis based on its clinical symptoms of traditional Chinese and Western medicine. Modernization of Traditional Chinese Medicine and Materia Medica‐World Science and Technology. 2024;26(4):944‐950. [Google Scholar]
- 7. Danhong P, Yanping W, Xiaoqi L, et al. Analysis on present situation of evaluation system of disease and TcM syndrome combinated animal model. Chin Arch Tradit Chin Med. 2017;35(8):2027‐2030. [Google Scholar]
- 8. Mengyao H. Yi Bian [M]. People's Medical Publishing House; 1994. [Google Scholar]
- 9. Peiqin L. Lei Zheng Zhi Cai: Treatment of Different Kinds of Diseases [M]. China Traditional Chinese Medicine Press; 1997. [Google Scholar]
- 10. Wenzhang F. Huang Di Nei Jing: The Yellow Emperor's Canon of Internal Medicine [M]. Sichuan People's Publishing House; 2018. [Google Scholar]
- 11. Fan Z, Jialin L, Junchen J, et al. Discussing the new implications of “toxins damaging brain collaterals” from the pathogenesis of acquired cerebral amyloid angiopathy. J Beijing Univ Tradit Chin Med. 2024;47(5):598‐602. [Google Scholar]
- 12. Baoping Z, Xiaoqing Z, Ping F. Yang Qunyu's experience in treating post‐stroke emotional disorders. J Guangzhou Univ Tradit Chin Med. 2021;38(5):1042‐1046. [Google Scholar]
- 13. Yifan L, Yongchen Z, Dongli Z, et al. Progress in research on post‐stroke depression from the perspective of TCM body constitution. Chin J Tradit Chin Med. 2022;37(12):7249‐7251. [Google Scholar]
- 14. Xuwei Z, Zeting Y, Zuqing S, et al. Construction of a spleen deficiency and damp obstruction type psoriasis animal model and preliminary exploration of its gut microbiota. Liaoning J Tradit Chin Med. 1‐18. [Online ahead of print]. Accessed July 22, 2025. https://link.cnki.net/urlid/21.1128.r.20250226.1644.040 [Google Scholar]
- 15. Messmer SJ, Salmeron KE, Frank JA, et al. Extended middle cerebral artery occlusion (MCAO) model to Mirror stroke patients undergoing thrombectomy. Transl Stroke Res. 2022;13(4):604‐615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Zhiheng L, Yuli C, Yu L, et al. Establishment and evaluation of a combination of disease and syndrome model of post‐stroke depression based on the theory of “disease‐syndrome‐drug”. Acta Lab Anim Sci Sin. 2025;33(3):388‐398. [Google Scholar]
- 17. Huizhen Y, Guang H, Lan M, et al. Study on plasma metabolomics of rat model of chronic blood stasis syndrome induced by adrenaline. J Basic Chin Med. 2022;28(2):205‐209. [Google Scholar]
- 18. Xiangning H, Weiyi L, Yifan S, et al. Analysis of animal model of type 2 diabetes mellitus based on clinical characteristics of traditional Chinese and western medicine. Chin J Exp Tradit Med Formulae. 1‐14 [Online ahead of print]. Accessed July 22, 2025. https://link.cnki.net/doi/10.13422/j.cnki.syfjx.20241714 [Google Scholar]
- 19. Chunyan X, Lu L, Ruichen S, et al. Analysis of medicinal usage patterns by Ll Dongyuan in treating dyspepsia. New Chin Med. 2025;57(8):12‐17. [Google Scholar]
- 20. Z Q, Lu L, C J, et al. Study on theory of treating depression syndrome from spleen: origin and development. J Liaoning Univ Tradit Chin Med. 2025;27(2):50‐53. [Google Scholar]
- 21. Xiaojuan L, Qingyu M, Yan Z, et al. To explore the relationship between stress and TCM syndrome. China J Tradit Chin Med Pharm. 2016;31(11):4424‐4427. [Google Scholar]
- 22. Xiaowei H, Chao M, Guangfu L, et al. Research progress in the preparation of experimental animal models of TCM syndromes of depression. Global Tradit Chin Med. 2023;16(2):354‐359. [Google Scholar]
- 23. Xuanwei L, Han L, Jing W, et al. Effects of Jin Gui Shen qi Wan on monoamine neurotransmitter levels in rats with kidney yang deficiency. J Basic Chin Med. 2024;30(11):1872‐1876. [Google Scholar]
- 24. Yongkang S, Xinzhi W, Xiangzhe L, et al. Review of animal models and modern biological studies of liver yang hyperactivity syndrome. J Basic Chin Med. 2023;29(5):851‐855. [Google Scholar]
- 25. Wenxiu Q, Jianqiang D, Junfeng X, et al. Research progress in the construction of experimental animal model of lschemic stroke with combination of disease and syndrome. Chin J Inf Tradit Chin Med. 2023;30(10):191‐196. [Google Scholar]
- 26. Yong T, Xinru L, Cheng L, et al. Metabolic profiling reveals therapeutic biomarkers of processed Aconitum carmichaeli Debx in treating hydrocortisone induced kidney‐yang deficiency syndrome rats. J Ethnopharmacol. 2014;152(3):585‐593. [DOI] [PubMed] [Google Scholar]
- 27. Zhang W, Ren C, Yang Y, Xu J, Tong F, Wu X. Ginseng aconitum decoction (Shenfu Tang) provides neuroprotection by ameliorating impairment of blood–brain barrier in cerebral ischemia–reperfusion injury. Brain Res. 2024;1842:149098. [DOI] [PubMed] [Google Scholar]
- 28. Qin L, Peng L, Sun Y, et al. Shengjiang powder ameliorates cell pyroptosis and inflammation induced by MCAO in rats through the NLRP3/Caspase‐1 pathway. J Stroke Cerebrovasc Dis. 2025;34(7):108314. [DOI] [PubMed] [Google Scholar]
- 29. Fluri F, Schuhmann MK, Kleinschnitz C. Animal models of ischemic stroke and their application in clinical research. Drug Des Devel Ther. 2015;9:3445‐3454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Lin C, Qisheng T, Haipeng L, et al. Effects of Yinao Jieyu formula on the JAK/STAT signaling pathway in the hippocampus of rats with kidney deficiency and liver depression syndrome of post‐stroke depression. J Beijing Univ Tradit Chin Med. 2020;43(12):1003‐1010. [Google Scholar]
- 31. Zhi L, Zhang F, Liu H, et al. CRS induces depression‐like behavior after MCAO in rats possibly by activating p38 MAPK. Behav Brain Res. 2023;437:114104. [DOI] [PubMed] [Google Scholar]
- 32. Zhao S, Zhao M, Xiao T, Jolkkonen J, Zhao C. Constraint‐induced movement therapy overcomes the intrinsic axonal growth‐inhibitory signals in stroke rats. Stroke. 2013;44(6):1698‐1705. [DOI] [PubMed] [Google Scholar]
- 33. Daizhen L, Xi C. Yi Jia Si Yao: Four Key Points of Doctor [M]. Shanxi Science and Technology Press; 2012. [Google Scholar]
- 34. Xuefeng F, Manzhong L, Yu Z, et al. Buyang Huanwu decoction promotes remodeling of brain tissue microstructure in ischemic stroke rats. J Beijing Univ Tradit Chin Med. 2021;44(6):500‐509. [Google Scholar]
- 35. Zhuohang L, Mingzhu Q, Helan H, et al. Analysis of the intervention effects and compatibility efficacy of the Qiqiong drug pair in ischemic stroke based on network regulation. China J Chin Mater Med. 2024;49(9):2326‐2335. [DOI] [PubMed] [Google Scholar]
- 36. Luping Y. Mechanism of Buyang Huanwu decoction and its Components in the Treatment of Ischemic Stroke Based on Mitochondrial Energy Metabolism. Beijing University of Traditional Chinese Medicine; 2022. [Google Scholar]
- 37. Yu L, Wei L, Peng W. Effects of berberine on cognitive function through the NF‐κB/iNOS‐COX‐2 signaling pathway in sleep‐deprived rats. Chin J Gerontol. 2024;44(2):459‐462. [Google Scholar]
- 38. Nirmal S, Olatunde OO, Medhe S, Vitti S, Khemtong C, Nirmal NP. Betalains alleviate exercise‐induced oxidative stress, inflammation, and fatigue and improve sports performance: an update on recent advancement. Curr Nutr Rep. 2023;12(4):778‐787. [DOI] [PubMed] [Google Scholar]
- 39. Hongli C, Jingjing G, Zhongdi J, et al. The effects of phototherapy on learning and memory, as well as the BDNF–TrkB signaling pathway in sleep‐deprived mice. Chin J Lasers. 2022;49(5):230‐240. [Google Scholar]
- 40. Xiaowen S, Wei S, Xiaofeng S. Ultrastructural study of autophagy and apoptosis in oocytes of starved mice. Zool Res. 2020;55(4):485‐490. [Google Scholar]
- 41. Ping L, Mingzhu L, Rong F, et al. Experimental study on the improvement of cognitive function in sleep‐deprived rats by hydrogen‐rich water. Acta Nutrimenta Sin. 2021;43(3):289‐293. [Google Scholar]
- 42. Linglin Z, Ge F, Xinfu L, et al. Experimental study of phlegm and blood stasis syndrome in local cerebral ischemia–reperfusion animal models. Chin J Integr Tradit West Med. 2018;38(12):1479‐1483. [Google Scholar]
- 43. Shujie L, Hualing L, Xiaoling C, et al. The effect of Danlou tablets on the NLRP3/GSDMD pyroptosis pathway in rats with phlegm‐blood stasis type ischemic stroke. Chin J Tradit Chin Med. 2022;37(8):4708‐4714. [Google Scholar]
- 44. Muñoz A, Costa M. Nutritionally mediated oxidative stress and inflammation. Oxidative Med Cell Longev. 2013;2013:610950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Hurtubise J, Mclellan K, Durr K, et al. The different facets of dyslipidemia and hypertension in atherosclerosis. Curr Atheroscler Rep. 2016;18(12):82. [DOI] [PubMed] [Google Scholar]
- 46. Yanjin T, Shaojun Q, Zhengduo Z, et al. Effects of supplementing multiple trace elements on antioxidative, anti‐inflammatory, and immune function in rats on a high‐fat diet. China Food Nutr. 1‐6. [Online ahead of print]. Accessed July 22, 2025. https://link.cnki.net/doi/10.19870/j.cnki.11‐3716/ts.20230505.001 [Google Scholar]
- 47. Shiduo C. Dong Tian Ao Zhi: Insight into the Profound Themes of Truth [M]. Shanxi Science and Technology Press; 2011. [Google Scholar]
- 48. Bo L. Effects of Kudiezi on the Notch Signaling Pathway in the Ischemic Penumbra of Rats with Fire Toxin Syndrome of Cerebral Ischemia–Reperfusion Injury. Beijing University of Traditional Chinese Medicine; 2019. [Google Scholar]
- 49. Ruixue M, Qihui Z. Effects of Kudiezi injection pretreatment on neurological function and hippocampal neurons in a model of blood stasis and poison syndrome after acute ischemic stroke. J Pract Cardiovasc Pulm Vasc Dis. 2021;29(12):67‐72, +85. [Google Scholar]
- 50. Yilei D, Yue L, Junyuan L, et al. Exploration of the “brain‐spleen inflammation coupling” mechanism in a rat model of acute ischemic stroke with blood stasis and poison syndrome. Chin Comp Med J. 2024;34(2):45‐54. [Google Scholar]
- 51. Lim SM, Jang HM, Jang SE, Han MJ, Kim DH. Lactobacillus fermentum IM12 attenuates inflammation in mice by inhibiting NF‐κB‐STAT3 signalling pathway. Benefic Microbes. 2017;8(3):407‐419. [DOI] [PubMed] [Google Scholar]
- 52. Ziyi H, Shiyi S, Jinxin Z, et al. Establishment of a chronic thrombosis mouse model induced by carrageenan. Chin J Lab Anim Sci. 2021;29(1):78‐84. [Google Scholar]
- 53. Jidong Z, Honghua L, Xiangzi L, et al. Exploration of the effects of Jingfang granules on thrombosis in carrageenan‐induced mice based on the ERK/p38 MAPK signaling pathway. Zhongguo Zhong Yao Za Zhi. 2022;47(8):2195‐2199. [DOI] [PubMed] [Google Scholar]
- 54. Lei L, Shilan D, Ying C, et al. Thrombolytic effect and impact on hemodynamics of Xueshuantong for injection. Zhongguo Zhong Yao Za Zhi. 2020;45(10):2446‐2453. [DOI] [PubMed] [Google Scholar]
- 55. Yue L, Mingjiang Y, Guojing F, et al. Reevaluation of rat model of blood stasis syndrome induced by carrageenan. J Basic Chin Med. 2023;29(12):2005‐2009. [Google Scholar]
- 56. Tu WJ, Zhao Z, Yin P, et al. Estimated burden of stroke in China in 2020. JAMA Netw Open. 2023;6(3):e231455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Siyuan C, Siyu W, Mingyuan Q, et al. Effect of Cordyceps militarisextract on CYP19/CaM/CaMKII signaling in testes of kidney yang deficiency model rats. Chin J Comp Med. 2023;33(1):1‐8. [Google Scholar]
- 58. Yuwei Z, Yanjie W, Bo Z, et al. Effects of Yougui pills on behavior and its mechanism in a rat model of ischemic stroke with kidney deficiency syndrome. Chin Med Equip J. 2019;16(7):166‐170. [Google Scholar]
- 59. Feifan L, Weiguo W, Wanshou G, et al. Glucocorticoids induce apoptosis of femoral head microvascular endothelial cells via the PI3K‐Akt–mTOR signaling pathway. Chin J Osteoporos. 2022;28(5):631‐636, +669. [Google Scholar]
- 60. Rui Z, Songrui D, Shuhui Y, et al. Mechanism study of Noni's improvement of memory impairment in mice with hydrocortisone‐induced kidney yang deficiency syndrome. J Tradit Chin Med. 2022;50(3):17‐22. [Google Scholar]
- 61. Mengmeng L, Jing L, Weijia L, et al. Metabolomics comparison of three depression rat models. Chin J Exp Tradit Med Formulae. 2024;30(16):149‐159. [Google Scholar]
- 62. Xin L, Bing Z, Xiaoqing L, et al. Dynamic observation of hydrocortisone‐induced yang deficiency state in rats. Chin J Tradit Chin Med. 2011;26(1):126‐128. [Google Scholar]
- 63. Hong AE, Ryu MS, Lim IK. Proper regulation of β‐adrenergic signal requires Btg2 gene for lipolysis and thermogenesis in response to starvation or cold acclimation in female mice. J Nutr Biochem. 2023;111:109160. [DOI] [PubMed] [Google Scholar]
- 64. Yang G, Xi ZX, Wan Y, et al. Changes of angiotensin II contents in rat plasma, brain, cardiovascular system and adrenal during stress. Sheng Li Xue Bao: [Acta Physiol Sin]. 1993;45(5):505‐509. [PubMed] [Google Scholar]
- 65. Xuhui Y, Pin J, Chengjun Y, et al. Effects of hypothermia on blood components in rats. Occup Health. 2008;15:1518‐1519. [Google Scholar]
- 66. Nakamura T, Xi G, Hua Y, Schallert T, Hoff JT, Keep RF. Intracerebral hemorrhage in mice: model characterization and application for genetically modified mice. J Cereb Blood Flow Metab. 2004;24(5):487‐494. [DOI] [PubMed] [Google Scholar]
- 67. Lu X, Chen‐Roetling J, Regan RF. Systemic hemin therapy attenuates blood–brain barrier disruption after intracerebral hemorrhage. Neurobiol Dis. 2014;70:70‐251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Kaneko S, Wang J, Kaneko M, et al. Protecting axonal degeneration by increasing nicotinamide adenine dinucleotide levels in experimental autoimmune encephalomyelitis models. J Neurosci. 2006;26(38):9794‐9804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Chunyun Y, Sisi Y, Dahua W, et al. Tianma Gouteng decoction alleviates neural injury in hypertensive intracerebral hemorrhage rats with liver yang rising syndrome by inhibiting NF‐κB expression and nuclear translocation. J Tradit Chin Med. 2019;37(12):2829‐2832, +3089–3090. [Google Scholar]
- 70. Xiaojun D, Xing Z, Xian Z, et al. Effects of Tianma Gouteng decoction on ang II, ALD, and liver protein expression in rats with spontaneous hypertension and liver yang rising syndrome. China J Exp Tradit Med Formulae. 2010;16(16):160‐162, +165. [Google Scholar]
- 71. Li Z, Wu M, Liu D, et al. Pingyang Jiangya fang pretreatment reduces the blood pressure of spontaneously hypertensive rats with liver‐Yang hyperactivity syndrome via ROS/Akt oxidative stress pathway. Ann Palliat Med. 2021;10(2):1904‐1919. [DOI] [PubMed] [Google Scholar]
- 72. Wei C, Yiyu D, Linqiang H, et al. Effects of hypertonic saline on blood–brain barrier permeability in rats with cerebral ischemia–reperfusion injury. J Pract Med. 2013;29(9):1412‐1415. [Google Scholar]
- 73. Wenxin Z, Yongli H, Gaofeng Z, et al. Hypertonic saline attenuates expression of Notch signaling and proinflammatory mediators in activated microglia in experimentally induced cerebral ischemia and hypoxic BV‐2 microglia. BMC Neurosci. 2017;18(1):32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Xiaoming L, Hongyu Z, Yanqiu L, et al. Effects of Zhen Gan Xi Feng tang on oxidative stress in brain tissues of rats with Parkinson's disease and liver‐Yang hyperactivity syndrome. Shiz Trad Chin Med Mater Med Res. 2016;27(11):2588‐2590. [Google Scholar]
- 75. Xiaoli W, Lanqin Z, Yanqiu Q, et al. Effects of Zhen Gan Xi Feng tang on the glutathione antioxidant system in brain tissues of rats with Parkinson's disease and liver‐Yang hyperactivity syndrome. Acta Chin Med. 2018;33(7):1289‐1293. [Google Scholar]
- 76. Wenquan H, Lingui Y, Hong X. Preliminary study on the experimental animal model of liver‐Yang hyperactivity syndrome. Chin J Emerg Med. 1996;1:36‐37. [Google Scholar]
- 77. Jingbo S, Maocai L, Rong H, et al. Therapeutic effects of Tongfu Xingshen capsules on phlegm‐heat‐fu syndrome in rats with stroke. J Emerg Trad Chin Med. 2001;6:341‐343. [Google Scholar]
- 78. Xingfeng P, Junying L, Kai L, et al. Establishment and evaluation of an animal model of ischemic stroke with phlegm‐heat‐fu syndrome. Chin J Tissue Eng Res. 2025;29(11):2301‐2309. [Google Scholar]
- 79. Chenfei H, Haoge C, Chulong R, et al. Research Progress on biomarkers of phlegm‐heat and bowel‐excess syndrome in stroke based on multiomics. Modernization of Traditional Chinese Medicine and Materia Medica‐World Science and Technology. 2024;26(10):2600‐2606. [Google Scholar]
- 80. Peng Z, Xue W, Jia X, et al. Research on the role of test indexes on TCM syndrome differentiation of acute cerebral infarction. Beijing J Trad Chin Med. 2020;39(12):1307‐1310. [Google Scholar]
- 81. Y J, C B, D Y, et al. Mechanism of Yiqi Jiedu formula in treating ischemic stroke based on the microbiota‐gut‐brain axis. China J Exp Tradit Med Formulae. 2024;30(6):91‐100. [Google Scholar]
- 82. Yang CJ, Chang HC, Sung PC, et al. Oral fecal transplantation enriches Lachnospiraceae and butyrate to mitigate acute liver injury. Cell Rep. 2024;43(1):113591. [DOI] [PubMed] [Google Scholar]
- 83. Mengjun L, Xiangzhe L, Chunlong R, et al. Effect of Zhongfeng Xinglou Tongfu (中风星萎通腑) capsules on intestinal Flora in patients with AcuteIschemic stroke of Tanrefushi (痰热腑实) syndrome and correlation between intestinal Flora and TanrefushiSyndrome. Pharmacol Clin Chin Mater Med. 2023;39(9):70‐75. [Google Scholar]
- 84. Mușat MI, Cătălin B, Hadjiargyrou M, Popa‐Wagner A, Greșiță A. Advancing post‐stroke depression research: insights from murine models and behavioral analyses. Life. 2024;14(9):1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Deacon RMJ. Measuring motor coordination in mice. J Vis Exp. 2013;75:e2609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Xinyi L, Yujie L, Kechong D, et al. Regulation of gut microbiota improves neurological function and depression symptoms in post‐stroke rats. South Med Univ J. 2024;44(2):405‐410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. St J, Sy L, Yh S, et al. The oxytocin receptor is essential for the protective effect of pair housing on post‐stroke depression in mice. Exp Gerontol. 2024;190:112432. [DOI] [PubMed] [Google Scholar]
- 89. Li CG, Xu LS, Sun L, et al. Xiangshao granules ameliorate post‐stroke depression by inhibiting activation of microglia and IDO1 expression in hippocampus and prefrontal cortex. Chin J Integr Med. 2025;31(1):28‐38. [DOI] [PubMed] [Google Scholar]
- 90. Zhang Y, Yang Y, Li H, Feng Q, Ge W, Xu X. Investigating the potential mechanisms and therapeutic targets of inflammatory cytokines in post‐stroke depression. Mol Neurobiol. 2024;61(1):132‐147. [DOI] [PubMed] [Google Scholar]
- 91. Zhang J, Ling L, Xiang L, Li W, Bao P, Yue W. Role of the gut microbiota in complications after ischemic stroke. Front Cell Infect Microbiol. 2024;14:1334581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Du L, He X, Xiong X, Zhang X, Jian Z, Yang Z. Vagus nerve stimulation in cerebral stroke: biological mechanisms, therapeutic modalities, clinical applications, and future directions. Neural Regen Res. 2024;19(8):1707‐1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Kim SH, Chung DK, Lee YJ, Song CH, Ku SK. Neuroprotective effects of Danggui‐Jakyak‐San on rat stroke model through antioxidant/antiapoptotic pathway. J Ethnopharmacol. 2016;188:123‐133. [DOI] [PubMed] [Google Scholar]
- 94. Kaur T, Huang ACW, Shyu BC. Modulation of melatonin in pain behaviors associated with oxidative stress and neuroinflammation responses in an animal model of central post‐stroke pain. Int J Mol Sci. 2023;24(6):5413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Ma XJ, Wang L, Yin XJ, et al. Protective effect and mechanism of scutellarin ethyl ester on focal cerebral ischemia induced by ligation of middle cerebral artery in rats. Zhongguo Zhong Yao Za Zhi. 2017;42(7):1350‐1357. [DOI] [PubMed] [Google Scholar]
- 96. Yong L, Li L. Protective effects of tanshinone on cerebral ischemia/reperfusion injury in rats through the PPARδ pathway. Chin J Immunol. 2024;40(9):1827‐1832. [Google Scholar]
- 97. Chaulagain B, Singh J. Penetratin and mannose‐functionalized cannabidiol lipid nanoparticles encapsulating the BDNF gene reduce amyloid‐induced inflammation. Mol Pharm. 2025;22(1):154‐167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Tan L, Li J, Sun D, Tian X, Zhong X, Shan Y. TLR4 as a therapeutic target: antidepressant mechanism of saikosaponin a in regulating the NF‐κB/BDNF axis and mitigating oxidative stress and inflammation in vivo and in vitro. Front Pharmacol. 2025;16:1585290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Veltri A, Nicolì V, Marino R, et al. Plasma brain‐derived neurotrophic factor (BDNF) levels and BDNF promoters ‘DNA methylation in workers exposed to occupational stress and suffering from psychiatric disorders. Brain Sci. 2024;14(11):1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Chang X, He Y, Liu Y, et al. Serum brain derived neurotrophic factor levels and post‐stroke depression in ischemic stroke patients. J Affect Disord. 2024;361:341‐347. [DOI] [PubMed] [Google Scholar]
- 101. Jin Y, Yu L, Li Y. Paroxetine effect on nerve growth factor, human neurotrophin‐4, brain‐derived neurotrophic factor levels in post‐stroke depression. Mol Neurobiol. 2024;61(10):7890‐7897. [DOI] [PubMed] [Google Scholar]
- 102. Wu Q, Xie Z, Cao X, et al. Chaihu‐Shugan‐san alleviates post‐stroke depression in mice: mechanistic insights into exosome‐mediated neuroprotection. J Ethnopharmacol. 2025;347:119700. [DOI] [PubMed] [Google Scholar]
- 103. Liu Y, Jin F, Chen Q, et al. PDGFR‐α mediated the neuroinflammation and autophagy via the JAK2/STAT3 signaling pathway contributing to depression‐like behaviors in myofascial pain syndrome rats. Mol Neurobiol. 2025;62(5):5650‐5663. [DOI] [PubMed] [Google Scholar]
- 104. Chunhua L, Weimin H, Dandan Y, et al. Amygdalin improves post‐stroke depression by inhibiting the JNK signaling pathway. J Shenyang Pharm Univ. 2024;41(1):112‐120. [Google Scholar]
- 105. Fioranelli M, Roccia MG, Przybylek B, Garo ML. The Role of Brain‐Derived Neurotrophic Factor (BDNF) in Depression and Cardiovascular Disease: A Systematic Review. Life (Basel, Switzerland). 2023;13(10):1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Duman RS, Li N. A neurotrophic hypothesis of depression: role of synaptogenesis in the actions of NMDA receptor antagonists. Philos Trans R Soc Lond Ser B Biol Sci. 2012;367(1601):2475‐2484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Kui J, Long F, Chenjie H, et al. Effects of Chaihu Shugan san on behavior and expression of 5‐HT2A and BDNF in the brain of rats with post‐stroke depression. Chinese Patent Medicine. 2023;45(12):3943‐3949. [Google Scholar]
- 108. Bhatt S, Devadoss T, Manjula SN, et al. 5‐HT3 receptor antagonism: a potential therapeutic approach for the treatment of depression and other disorders. Curr Neuropharmacol. 2021;19(9):1545‐1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Hamon M, Blier P. Monoamine neurocircuitry in depression and strategies for new treatments. Prog Neuro‐Psychopharmacol Biol Psychiatry. 2013;45:54‐63. [DOI] [PubMed] [Google Scholar]
- 110. Xia Z, Yanling Z, Minghao C, et al. Effect of Jieyu decoction (解郁汤) on behavior improvement and expression of 5‐HT and NE in rats with post‐stroke depression. J Pract Tradit Chin Intern Med. 2021;35(6):70‐73. [Google Scholar]
- 111. Yan S, Yang G, Runze W, et al. Effect of Tiaoshen Yunshu acupuncture on lntestinal Flora and lts metabolites and RelatedNeurotransmitters in treatment of post‐StrokeDepression: a randomized control trial. J Clin Acup Moxibust. 2024;40(1):33‐38. [Google Scholar]
- 112. Villa M, Martínez‐Vega M, Del Pozo A, et al. The role of the dopamine system in post‐stroke mood disorders in newborn rats. Int J Mol Sci. 2023;24(4):3229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Thorstensen JR, Taylor JL, Kavanagh JJ. 5‐HT2 receptor antagonism reduces human motoneuron output to antidromic activation but not to stimulation of corticospinal axons. Eur J Neurosci. 2022;56(1):3674‐3686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Mc L, Jt J, Yx W, et al. Astragaloside IV promotes cerebral tissue restoration through activating AMPK‐ mediated microglia polarization in ischemic stroke rats. J Ethnopharmacol. 2024;334:118532. [DOI] [PubMed] [Google Scholar]
- 115. Gipson CD, Olive MF. Structural and functional plasticity of dendritic spines ‐ root or result of behavior? Genes Brain Behav. 2017;16(1):101‐117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Pan R, Cai J, Zhan L, et al. Buyang Huanwu decoction facilitates neurorehabilitation through an improvement of synaptic plasticity in cerebral ischemic rats. BMC Complement Altern Med. 2017;17(1):173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Wang H, He Y, Sun Z, et al. Microglia in depression: an overview of microglia in the pathogenesis and treatment of depression. J Neuroinflammation. 2022;19(1):132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Fang C, Shen Y, Ma Z, et al. L‐theanine prevents colonic damage via NF‐κB/MAPK signaling pathways induced by a high‐fat diet in rats. Mol Nutr Food Res. 2024;68(8):e2300797. [DOI] [PubMed] [Google Scholar]
- 119. Yufang Z, Jian C, Yixin Z, et al. Mechanism of astragaloside total saponins and lotus leaf total alkaloids in preventing and treating hyperlipidemia based on the PPARγ/LXRα/ABCG1 signaling pathway. China J Exp Tradit Med Formulae. 2024;30(13):37‐44. [Google Scholar]
- 120. Qian X, Liyuan L, Rongxian Z, et al. Effects of moxibustion at “Zusanli” on vascular damage and oxidative stress in rats on a high‐fat diet based on the mTOR/HIF‐1α/VEGF signaling pathway. Chin Acupunct Moxibust. 2024;44(4):433‐440. [DOI] [PubMed] [Google Scholar]
- 121. Wencheng C, Shuang L, Jiali L, et al. Tea saponin combined with aerobic exercise improves fat metabolism and oxidative stress levels in obese mice induced by a high‐fat diet. Food Sci. 2023;44(11):106‐114. [Google Scholar]
- 122. Annamalai P, Thangam EB. Vitex trifolia L. modulates inflammatory mediators via down‐regulation of the NF‐κB signaling pathway in carrageenan‐induced acute inflammation in experimental rats. J Ethnopharmacol. 2022;298:115583. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
As this is a review article, no new datasets were generated. All data analyzed in this article are available from the cited sources.
