1.Mechanism of Chaishao Kaiyu Decoction in ameliorating hippocampal neuroinflammation in depressed rats based on complement component C3/C3aR pathway.
Ying-Juan TANG ; Hai-Peng GUO ; Man-Shu ZOU ; Yuan-Shan HAN ; Jun-Cheng LIU ; Yu-Hong WANG
China Journal of Chinese Materia Medica 2025;50(1):1-9
This study investigated the mechanism of Chaishao Kaiyu Decoction in improving hippocampal neuroinflammation in depressed rats based on complement component 3(C3)/C3 receptor(C3aR). A total of 60 SD rats were randomly divided into a blank group, a model group, high, medium, and low dose groups of Chaishao Kaiyu Decoction, and a positive drug group, with 10 rats in each group. Except for the blank group, chronic unpredictable mild stress(CUMS) was used to construct depression models in other groups. Sucrose preference, open-field experiment, forced swimming, and water maze were used to detect the changes in depression-like behavior in each group. Enzyme-linked immunosorbent assay(ELISA) was used to detect the serum inflammatory factor level in rats, and hematoxylin-eosin(HE) staining and Nissl staining were employed to observe the pathological damage of hippocampal neurons. Golgi-Cox staining was used to observe the dendritic spine damage of hippocampal neurons, and immunofluorescence and Western blot were utilized to detect the expression of microglial marker Iba-1 and C3/C3aR protein in the hippocampus of rats. The behavioral results showed that compared with the model group, Chaishao Kaiyu Decoction could significantly strengthen the sugar water preference, increase the distance and number of voluntary activities, shorten the immobility time in forced swimming and the successful incubation period of positioning navigation, and prolong the stay time of space exploration in the target quadrant. ELISA results showed that the content of inflammatory factors in the hippocampus of depressed rats was significantly higher than that of the blank group, and the content of inflammatory factors decreased significantly after the intervention of Chaishao Kaiyu Decoction. In addition, Chaishao Kaiyu Decoction could relieve pathological damage such as cell swelling and loose arrangement of hippocampus tissue. In the Western blot experiment, the expression levels of C3 and C3aR proteins in the model group were higher than those in the blank group, while the expression of C3 and C3aR in Chaishao Kaiyu Decoction could be down-regulated. Immunofluorescence results showed that compared with the model group, the fluorescence intensity of microglia marker Iba-1 decreased significantly after the intervention of Chaishao Kaiyu Decoction and positive drugs. The antidepressant effect of Chaishao Kaiyu Decoction may be related to the down-regulation of C3/C3aR signaling pathway-related proteins, thus alleviating hippocampal inflammation.
Animals
;
Hippocampus/metabolism*
;
Rats, Sprague-Dawley
;
Drugs, Chinese Herbal/administration & dosage*
;
Rats
;
Male
;
Depression/metabolism*
;
Complement C3/metabolism*
;
Receptors, Complement/metabolism*
;
Humans
;
Neuroinflammatory Diseases/genetics*
2.Congrong San ameliorates cognitive impairment and neuroinflammation in rat model of Alzheimer's disease by alleviating endoplasmic reticulum stress to inhibit NLRP3 inflammasome activation.
Yuan-Qin CAI ; Yang XIANG ; Qing-Hua LONG ; Xi WANG ; Chu-Hua ZENG
China Journal of Chinese Materia Medica 2025;50(7):1881-1888
This study aims to investigate the effect of Congrong San(CRS) on endoplasmic reticulum stress-induced neuroinflammation in the rat model of Aβ_(1-42)-induced Alzheimer's disease(AD). Sixty male Sprague-Dawley rats(2 months old) were randomized into blank(CON), model(MOD), low-dose Congrong San(L-CRS), medium-dose Congrong San(M-CRS), high-dose Congrong San(H-CRS), and memantine hydrochloride(MJG) groups. The Morris water maze test was carried out to examine the learning and memory abilities of rats in each group. Hematoxylin-eosin staining and Nissl staining were employed to observe the morphology and number of CA1 neurons in the hippocampus of rats in each group. The morphology and structure of the endoplasmic reticulum in the hippocampus were observed by transmission electron microscopy. The immunofluorescence assay was employed to detect the expression of 78 kDa glucose-regulated protein(GRP78) in the hippocampus. Western blot was employed to determine the expression of apoptosis-associated speck-like protein containing a CARD(ASC), cysteinyl aspartate-specific proteinase(caspase-1), interleukin-18(IL-18), interleukin-1β(IL-1β), GRP78, and pathway proteins including protein kinase RNA-like endoplasmic reticulum kinase(PERK), phosphorylated PERK(p-PERK), C/EBP homologous protein(CHOP), and NOD-like receptor pyrin domain-containing protein 3(NLRP3) in the rat hippocampus. Compared with the MOD group, the M-CRS and H-CRS groups showed improved learning and memory abilities, reduced neuron losses in the hippocampus, alleviated endoplasmic reticulum stress, inhibited PERK-CHOP-NLRP3 pathway, and lowered levels of IL-1β, IL-6, and tumor necrosis factor-alpha(TNF-α). The results suggest that CRS can alleviate cognitive impairment and hippocampal neuron damage and reduce neuroinflammation in AD rats by alleviating endoplasmic reticulum stress to inhibit the activation of NLRP3 inflammasomes.
Animals
;
Endoplasmic Reticulum Stress/drug effects*
;
Male
;
Alzheimer Disease/psychology*
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Rats, Sprague-Dawley
;
Rats
;
Inflammasomes/genetics*
;
Drugs, Chinese Herbal/administration & dosage*
;
Cognitive Dysfunction/metabolism*
;
Disease Models, Animal
;
Hippocampus/drug effects*
;
Humans
;
Neuroinflammatory Diseases/drug therapy*
3.4'-O-methylbavachalcone improves vascular cognitive impairment by inhibiting neuroinflammation via EPO/Nrf2/HO-1 pathway.
Xin-Yuan ZHANG ; Chen WANG ; Hong-Qing CHEN ; Xiang-Bing ZENG ; Jun-Jie WANG ; Qing-Guang ZHANG ; Jin-Wen XU ; Shuang LING
China Journal of Chinese Materia Medica 2025;50(14):3990-4002
This study aims to explore the effects and mechanisms of 4'-O-methylbavachalcone(MeBavaC), an active compound from Psoraleae Fructus, in regulating white matter neuroinflammation to improve vascular cognitive impairment. Male Sprague-Dawley(SD) rats were randomly divided into four groups: sham group, model group, high-dose MeBavaC group(14 mg·kg~(-1)), and low-dose MeBavaC group(7 mg·kg~(-1)). The rat model of chronic cerebral hypoperfusion(CCH) was established using bilateral common carotid artery occlusion. The Morris water maze test was performed to evaluate the learning and memory abilities of the rats. Luxol fast blue staining, Nissl staining, immunofluorescence, immunohistochemistry, and transmission electron microscopy were utilized to observe the morphology and ultrastructure of the white matter myelin sheaths, axon integrity, the morphology and number of hippocampal neurons, and the loss and activation of glial cells in the white matter. Transcriptome analysis was performed to explore the potential mechanisms of white matter injury induced by CCH. Western blot and quantitative real-time polymerase chain reaction(qRT-PCR) assays were conducted to measure the expression levels of NOD-like receptor protein 3(NLRP3), absent in melanoma 2(AIM2), gasdermin D(GSDMD), cysteinyl aspartate-specific proteinase-1(caspase-1), interleukin-18(IL-18), interleukin-1β(IL-1β), erythropoietin(EPO), nuclear factor erythroid 2-related factor 2(Nrf2), and heme oxygenase-1(HO-1) in the white matter of rats. The results showed that compared with the model group, MeBavaC significantly improved the learning and memory abilities of rats with CCH, improved the damage of white matter myelin sheath, maintained axonal integrity, reduced the loss of hippocampal neurons and oligodendrocytes in the white matter, inhibited the activation of microglia and the proliferation of astrocytes in the white matter, and suppressed the NLRP3/AIM2/caspase-1/GSDMD pathway. The expression levels of inflammatory cytokines IL-1β and IL-18 were significantly reduced, while EPO expression and the expression of Nrf2/HO-1 antioxidant pathway were notably elevated. In conclusion, MeBavaC can alleviate cognitive impairment in rats with CCH and suppress neuroinflammation in cerebral white matter. The mechanism of action may involve activation of EPO activity, promotion of endogenous antioxidant pathways, and inhibition of neuroinflammation in the white matter. This study suggests that MeBavaC exhibits antioxidant and anti-neuroinflammatory effects, showing potential application in improving cognitive dysfunction.
Animals
;
Male
;
Rats, Sprague-Dawley
;
NF-E2-Related Factor 2/immunology*
;
Rats
;
Chalcones/administration & dosage*
;
Cognitive Dysfunction/metabolism*
;
Signal Transduction/drug effects*
;
Neuroinflammatory Diseases/drug therapy*
;
Heme Oxygenase-1/metabolism*
;
Humans
;
Heme Oxygenase (Decyclizing)/genetics*
4.WNK1 Alleviates Chloride Efflux-Induced NLRP3 Inflammasome Activation and Subsequent Neuroinflammation in Early Brain Injury Following Subarachnoid Hemorrhage.
Panpan ZHAO ; Huimiao FENG ; Xinyu ZHOU ; Jingyuan ZHOU ; Fangbo HU ; Taotao HU ; Yong SUN
Neuroscience Bulletin 2025;41(9):1570-1588
The nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome plays a crucial role in the prognosis of subarachnoid hemorrhage (SAH). WNK1 kinase negatively regulates NLRP3 in various inflammatory conditions, but its role in early brain injury (EBI) after SAH remains unclear. In this study, we used an in vivo SAH model in rats/mice and AAV-WNK1 intraventricular injection to investigate its neuroprotective mechanisms. WNK1 expression was significantly reduced in SAH patient blood and SAH model brain tissue, correlating negatively with microglial activation. AAV-WNK1 alleviated brain edema, neuronal necrosis, behavioral deficits, and inflammation by inhibiting NLRP3 inflammasome activation. In hemin-stimulated BV-2 cells, WNK1 overexpression reduced NLRP3 activation and inflammatory cytokines. Chloride counteracted WNK1's inhibitory effects, and WNK1 suppressed P2X7R-induced NLRP3 activation. Mechanistically, WNK1 functioned via the OXSR1/STK39 pathway. These findings highlight WNK1 as a key regulator of intracellular chloride balance and neuroinflammation, presenting a potential therapeutic target for SAH treatment.
Animals
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Subarachnoid Hemorrhage/complications*
;
Inflammasomes/metabolism*
;
Rats
;
Mice
;
Neuroinflammatory Diseases/metabolism*
;
WNK Lysine-Deficient Protein Kinase 1/genetics*
;
Male
;
Humans
;
Chlorides/metabolism*
;
Mice, Inbred C57BL
;
Rats, Sprague-Dawley
;
Brain Injuries/metabolism*
;
Microglia/metabolism*
;
Protein Serine-Threonine Kinases
5.SOX11-mediated CBLN2 Upregulation Contributes to Neuropathic Pain through NF-κB-Driven Neuroinflammation in Dorsal Root Ganglia of Mice.
Ling-Jie MA ; Tian WANG ; Ting XIE ; Lin-Peng ZHU ; Zuo-Hao YAO ; Meng-Na LI ; Bao-Tong YUAN ; Xiao-Bo WU ; Yong-Jing GAO ; Yi-Bin QIN
Neuroscience Bulletin 2025;41(12):2201-2217
Neuropathic pain, a debilitating condition caused by dysfunction of the somatosensory nervous system, remains difficult to treat due to limited understanding of its molecular mechanisms. Bioinformatics analysis identified cerebellin 2 (CBLN2) as highly enriched in human and murine proprioceptive and nociceptive neurons. We found that CBLN2 expression is persistently upregulated in dorsal root ganglia (DRG) following spinal nerve ligation (SNL) in mice. In addition, transcription factor SOX11 binds to 12 cis-regulatory elements within the Cbln2 promoter to enhance its transcription. SNL also induced SOX11 upregulation, with SOX11 and CBLN2 co-localized in nociceptive neurons. The siRNA-mediated knockdown of Sox11 or Cbln2 attenuated SNL-induced mechanical allodynia and thermal hyperalgesia. High-throughput sequencing of DRG following intrathecal injection of CBLN2 revealed widespread gene expression changes, including upregulation of numerous NF-κB downstream targets. Consistently, CBLN2 activated NF-κB signaling, and inhibition with pyrrolidine dithiocarbamate reduced CBLN2-induced pain hypersensitivity, proinflammatory cytokines and chemokines production, and neuronal hyperexcitability. Together, these findings identified the SOX11/CBLN2/NF-κB axis as a critical mediator of neuropathic pain and a promising target for therapeutic intervention.
Animals
;
Neuralgia/metabolism*
;
Ganglia, Spinal/metabolism*
;
Up-Regulation
;
Mice
;
NF-kappa B/metabolism*
;
SOXC Transcription Factors/genetics*
;
Male
;
Neuroinflammatory Diseases/metabolism*
;
Mice, Inbred C57BL
;
Nerve Tissue Proteins/genetics*
;
Hyperalgesia/metabolism*
;
Signal Transduction
;
Spinal Nerves
6.Stem-leaf saponins of Panax notoginseng attenuate experimental Parkinson's disease progression in mice by inhibiting microglia-mediated neuroinflammation via P2Y2R/PI3K/AKT/NFκB signaling pathway.
Hui WU ; Chenyang NI ; Yu ZHANG ; Yingying SONG ; Longchan LIU ; Fei HUANG ; Hailian SHI ; Zhengtao WANG ; Xiaojun WU
Chinese Journal of Natural Medicines (English Ed.) 2025;23(1):43-53
Stem-leaf saponins from Panax notoginseng (SLSP) comprise numerous PPD-type saponins with diverse pharmacological properties; however, their role in Parkinson's disease (PD), characterized by microglia-mediated neuroinflammation, remains unclear. This study evaluated the effects of SLSP on suppressing microglia-driven neuroinflammation in experimental PD models, including the 1-methyl-4-phenylpyridinium (MPTP)-induced mouse model and lipopolysaccharide (LPS)-stimulated BV-2 microglia. Our findings revealed that SLSP mitigated behavioral impairments and excessive microglial activation in models of PD, including MPTP-treated mice. Additionally, SLSP inhibited the upregulation of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) and attenuated the phosphorylation of PI3K, protein kinase B (AKT), nuclear factor-κB (NFκB), and inhibitor of NFκB protein α (IκBα) both in vivo and in vitro. Moreover, SLSP suppressed the production of inflammatory markers such as interleukin (IL)-1β, IL-6, and tumor necrosis factor alpha (TNF-α) in LPS-stimulated BV-2 cells. Notably, the P2Y2R agonist partially reversed the inhibitory effects of SLSP in LPS-treated BV-2 cells. These results suggest that SLSP inhibit microglia-mediated neuroinflammation in experimental PD models, likely through the P2Y2R/PI3K/AKT/NFκB signaling pathway. These novel findings indicate that SLSP may offer therapeutic potential for PD by attenuating microglia-mediated neuroinflammation.
Animals
;
Panax notoginseng/chemistry*
;
Saponins/pharmacology*
;
Microglia/immunology*
;
Mice
;
NF-kappa B/immunology*
;
Signal Transduction/drug effects*
;
Proto-Oncogene Proteins c-akt/immunology*
;
Phosphatidylinositol 3-Kinases/genetics*
;
Male
;
Parkinson Disease/immunology*
;
Mice, Inbred C57BL
;
Disease Models, Animal
;
Plant Leaves/chemistry*
;
Neuroinflammatory Diseases/drug therapy*
;
Humans
7.Shionone protects cerebral ischemic injury through alleviating microglia-mediated neuroinflammation.
Lushan XU ; Chenggang LI ; ChenChen ZHAO ; Zibu WANG ; Zhi ZHANG ; Xin SHU ; Xiang CAO ; Shengnan XIA ; Xinyu BAO ; Pengfei SHAO ; Yun XU
Chinese Journal of Natural Medicines (English Ed.) 2025;23(4):471-479
Microglia, the resident immune cells in the central nervous system (CNS), rapidly transition from a resting to an active state in the acute phase of ischemic brain injury. This active state mediates a pro-inflammatory response that can exacerbate the injury. Targeting the pro-inflammatory response of microglia in the semi-dark band during this acute phase may effectively reduce brain injury. Shionone (SH), an active ingredient extracted from the dried roots and rhizomes of the genus Aster (Asteraceae), has been reported to regulate the inflammatory response of macrophages in sepsis-induced acute lung injury. However, its function in post-stroke neuroinflammation, particularly microglia-mediated neuroinflammation, remains uninvestigated. This study found that SH significantly inhibited lipopolysaccharide (LPS)-induced elevation of inflammatory cytokines, including interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and inducible nitric oxide synthase (iNOS), in microglia in vitro. Furthermore, the results demonstrated that SH alleviated infarct volume and improved behavioral performance in middle cerebral artery occlusion (MCAO) mice, which may be attributed to the inhibition of the microglial inflammatory response induced by SH treatment. Mechanistically, SH potently inhibited the phosphorylation of serine-threonine protein kinase B (AKT), mammalian target of rapamycin (mTOR), and signal transducer and activator of transcription 3 (STAT3). These findings suggest that SH may be a potential therapeutic agent for relieving ischemic stroke (IS) by alleviating microglia-associated neuroinflammation.
Animals
;
Microglia/immunology*
;
Mice
;
Male
;
Mice, Inbred C57BL
;
Brain Ischemia/immunology*
;
Neuroinflammatory Diseases/drug therapy*
;
Neuroprotective Agents/administration & dosage*
;
Interleukin-1beta/genetics*
;
STAT3 Transcription Factor/genetics*
;
TOR Serine-Threonine Kinases/genetics*
;
Tumor Necrosis Factor-alpha/genetics*
;
Proto-Oncogene Proteins c-akt/immunology*
;
Nitric Oxide Synthase Type II/genetics*
;
Lipopolysaccharides
8.Parkin deletion affects PINK1/Parkin-mediated mitochondrial autophagy to exacerbate neuroinflammation and accelerate progression of Parkinson's disease in mice.
Chengcheng JIANG ; Yangyang LI ; Kexin DUAN ; Tingting ZHAN ; Zilong CHEN ; Yongxue WANG ; Rui ZHAO ; Caiyun MA ; Yu GUO ; Changqing LIU
Journal of Southern Medical University 2024;44(12):2359-2366
OBJECTIVES:
To investigate the role of mitochondrial autophagy disorder caused by deletion of E3 ubiquitin ligase Parkin in neuroinflammation in a mouse model of MPTP-induced Parkinson's disease (PD).
METHODS:
Wild-type (WT) male C57BL/6 mice and Parkin-/- mice were given intraperitoneal injections with MPTP or PBS for 5 consecutive days, and the changes in motor behaviors of the mice were observed using open field test. The effects of Parkin deletion on PD development and neuroinflammation were evaluated using immunofluorescence and Western blotting. The changes of the PINK 1/Parkin signaling pathway in the midbrain substantia nigra of the mice were examined to explore the molecular mechanism of Parkin-mediated regulation of mitochondrial autophagy and its effect on neuroinflammation in PD mice.
RESULTS:
Compared with their WT counterparts, the Parkin-/- mice with MPTP injections exhibited significant impairment of motor function with decreased TH+ neurons, increased α-synuclein (α-syn) accumulation, and increased numbers of GFAP+ and I-ba1+ cells in the midbrain substantia nigra. Parkin deletion obviously affected PINK1/Parkin-mediated mitochondrial autophagy to result in significantly increased mtDNA and upregulated expressions of STING and NLRP3 inflammatosomes in the midbrain substantia nigra of MPTP-treated transgenic mice.
CONCLUSIONS
Parkin deletion causes mitochondrial autophagy disorder to accelerate PD progression and exacerbates neuroinflammation in mice by affecting the PINK1/Parkin signaling pathway, suggesting the important role of Parkin in early pathogenesis of PD.
Animals
;
Ubiquitin-Protein Ligases/genetics*
;
Mice
;
Mice, Inbred C57BL
;
Male
;
Parkinson Disease/genetics*
;
Protein Kinases/genetics*
;
Mitochondria/metabolism*
;
Disease Models, Animal
;
Autophagy
;
Signal Transduction
;
Neuroinflammatory Diseases/metabolism*
;
Mice, Knockout
;
alpha-Synuclein/metabolism*
;
Substantia Nigra/metabolism*
;
Mitophagy
;
Disease Progression
9.Xixin Decoction improves learning and memory ability of SAMP8 by enhancing neuroprotective effect and inhibiting neuroinflammation.
En-Long ZHAO ; Yong-Chang DIWU ; Hu ZHANG ; Li-Qi DUAN ; Xin-Yue HAN ; Ya-Li WANG ; Yuan ZHOU
China Journal of Chinese Materia Medica 2023;48(18):5032-5040
This study aimed to explore the possible effect of Xixin Decoction(XXD) on the learning and memory ability of Alzheimer's disease(AD) model senescence-accelerated mouse-prone 8(SAMP8) and the related mechanism in enhancing neuroprotective effect and reducing neuroinflammation. Forty SAMP8 were randomly divided into a model group(10 mL·kg~(-1)·d~(-1)), a probiotics group(0.39 g·kg~(-1)·d~(-1)), a high-dose group of XXD granules(H-XXD, 5.07 g·kg~(-1)·d~(-1)), a medium-dose group of XXD granules(M-XXD, 2.535 g·kg~(-1)·d~(-1)), and a low-dose group of XXD granules(L-XXD, 1.267 5 g·kg~(-1)·d~(-1)). Eight senescence-accelerated mouse-resistant 1(SAMR1) of the same age and strain were assigned to the control group(10 mL·kg~(-1)·d~(-1)). After ten weeks of intragastric administration, the Morris water maze was used to test the changes in spatial learning and memory ability of mice after treatment. Meanwhile, immunofluorescence staining was used to detect the positive expression of receptor for advanced glycation end products(AGER), Toll-like receptor 1(TLR1), and Toll-like receptor 2(TLR2) in the hippocampal CA1 region of mice. Western blot was employed to test the protein expression levels of silencing information regulator 2 related enzyme 1(SIRT1), AGER, TLR1, and TLR2 in the hippocampus of mice. Enzyme linked immunosorbent assay(ELISA) was applied to assess the levels of Aβ_(1-42) in the hippocampus of mice and the levels of nuclear factor κB p65(NF-κB p65), NOD-like receptor protein 3(NLRP3), tumor necrosis factor-α(TNF-α), and interleukin-1β(IL-1β) in the serum and hippocampus of mice. Compared with the model group, XXD significantly improved the spatial learning and memory ability of SAMP8, increased the expression of neuroprotective factors in the hippocampus, decreased the levels of neuroinflammatory factors, and inhibited the expression of Aβ_(1-42). In particular, H-XXD significantly increased the expression of SIRT1 in the hippocampus of mice, reduced the expression levels of NF-κB p65, NLRP3, TNF-α, and IL-1β in the serum and hippocampus of mice, and decreased the expression of AGER, TLR1, and TLR2 in the hippocampus of mice(P<0.05 or P<0.01). XXD may improve the spatial learning and memory ability of AD model SAMP8 by enhancing the neuroprotective effect and inhibiting neuroinflammation.
Humans
;
Neuroprotective Agents/therapeutic use*
;
Sirtuin 1/metabolism*
;
Toll-Like Receptor 2/metabolism*
;
NF-kappa B/metabolism*
;
Tumor Necrosis Factor-alpha/metabolism*
;
Neuroinflammatory Diseases
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Toll-Like Receptor 1/metabolism*
;
Alzheimer Disease/genetics*
;
Hippocampus
10.Neuroprotective effect and mechanism of Zuogui Jiangtang Jieyu Formula on diabetes mellitus complicated with depression model rats based on CX3CL1-CX3CR1 axis.
Ping LI ; Yang LIU ; Man-Shu ZOU ; Ting-Ting WANG ; Hai-Peng GUO ; Ting-Ting REN ; Ying HE ; Hua WANG ; Yu-Hong WANG
China Journal of Chinese Materia Medica 2023;48(21):5822-5829
Based on the CX3C chemokine ligand 1(CX3CL1)-CX3C chemokine receptor 1(CX3CR1) axis, this study explored the potential mechanism by which Zuogui Jiangtang Jieyu Formula(ZGJTJY) improved neuroinflammation and enhanced neuroprotective effect in a rat model of diabetes mellitus complicated with depression(DD). The DD rat model was established by feeding a high-fat diet combined with streptozotocin(STZ) intraperitoneal injection for four weeks and chronic unpredictable mild stress(CUMS) combined with isolated cage rearing for five weeks. The rats were divided into a control group, a model group, a positive control group, an inhibitor group, and a ZGJTJY group. The open field test and forced swimming test were used to assess the depression-like behaviors of the rats. Enzyme-linked immunosorbent assay(ELISA) was performed to measure the expression levels of the pro-inflammatory cytokines interleukin-1β(IL-1β) and tumor necrosis factor-α(TNF-α) in plasma. Immunofluorescence staining was used to detect the expression of ionized calcium-binding adapter molecule 1(Iba1), postsynaptic density protein-95(PSD95), and synapsin-1(SYN1) in the hippocampus. Hematoxylin-eosin(HE) staining, Nissl staining, and TdT-mediated dUTP nick end labeling(TUNEL) fluorescence staining were performed to assess hippocampal neuronal damage. Western blot was used to measure the expression levels of CX3CL1, CX3CR1, A2A adenosine receptor(A2AR), glutamate receptor 2A(NR2A), glutamate receptor 2B(NR2B), and brain-derived neurotrophic factor(BDNF) in the hippocampus. Compared with the model group, the ZGJTJY group showed improved depression-like behaviors in DD rats, enhanced neuroprotective effect, increased expression of PSD95, SYN1, and BDNF(P<0.01), and decreased expression of Iba1, IL-1β, and TNF-α(P<0.01), as well as the expression of CX3CL1, CX3CR1, A2AR, NR2A, and NR2B(P<0.01). These results suggest that ZGJTJY may exert its neuroprotective effect by inhibiting the CX3CL1-CX3CR1 axis and activation of hippocampal microglia, thereby improving neuroinflammation and abnormal activation of N-methyl-D-aspartate receptor(NMDAR) subunits, and ultimately enhancing the expression of synaptic-related proteins PSD95, SYN1, and BDNF in the hippocampus.
Rats
;
Animals
;
Depression/drug therapy*
;
Brain-Derived Neurotrophic Factor
;
Neuroprotective Agents
;
Tumor Necrosis Factor-alpha/metabolism*
;
Neuroinflammatory Diseases
;
Diabetes Mellitus
;
Receptors, Glutamate
;
CX3C Chemokine Receptor 1/genetics*

Result Analysis
Print
Save
E-mail