1.Therapeutic effect of Ziziphi Spinosae Semen extracts on chronic unpredictable mild stress-induced depression and insomnia-like behavior in mice.
Hong-Bo CHENG ; Xian LIU ; Hui-Ying SHANG ; Rong GAO ; Wan-Yun DANG ; Ye-Hui GAO ; Cheng-Rong XIAO ; Yue GAO ; Zeng-Chun MA
China Journal of Chinese Materia Medica 2025;50(7):1817-1829
This paper aims to study the effect of Ziziphi Spinosae Semen extracts on chronic unpredictable mild stress(CUMS)-induced depression-like and insomnia behavior models of mice. The CUMS-induced depression-like and insomnia behavior model of mice was established by CUMS treatment for three weeks. The mice were randomly divided into control group, model group, positive drug diazepam group(2 mg·kg~(-1)), as well as low-dose group(1.95 g·kg~(-1)), medium-dose group(3.9 g·kg~(-1)), and high-dose group(7.8 g·kg~(-1)) of Ziziphi Spinosae Semen extracts, with 18 mice in each group. On the 15th day of modeling, the drug was administered intragastrically once a day for one week. Then, the pentobarbital sodium cooperative righting experiment, open field experiment, and elevated plus maze experiment were carried out, respectively. The contents of neurotransmitters 5-hydroxytryptamine(5-HT) and 5-hydroxyindoleacetic acid(5-HIAA) in serum and thalamus of mice, as well as the levels of corticotropin releasing hormone(CRH), adrenocorticotropic hormone(ACTH), and corticosterone(CORT) in serum, were determined by enzyme-linked immunosorbent assay(ELISA). The neuron damage in the hippocampus of mice was observed by hematoxylin-eosin(HE) staining and Nissl staining. Western blot was used to detect the expressions of tryptophan hydroxylase 2(TPH2), serotonin transporter(SERT), monoamine oxidase A(MAOA), five prime repressors under dual repression binding protein 1(Freud1), synaptic plasticity-related proteins [cellular gene FOS(C-FOS), postsynaptic density protein 95(PSD95), synapsin 1(SYN1), and activity-regulated cytoskeleton-associated gene(ARC)], blood-brain barrier(BBB) permeability-related proteins [zonula occludens 1(ZO-1), occludin, and claudin 1], inflammatory factors [NOD-, LRR-and pyrin domain-containing protein 3(NLRP3), apoptosis-associated speck-like protein(ASC), gasdermin D(GSDMD), caspase-3, and caspase-8], and antioxidant factors [nuclear factor erythroid 2-related factor 2(NRF2) and heme oxygenase 1(HO1)] in thalamic tissue of mice. The results indicated that compared with that in the model group, the sleep latency was significantly shortened, and the sleep duration was significantly prolonged in each dose group of Ziziphi Spinosae Semen extracts. The number of visits to the central area of the open field and the distance and time of visits were significantly increased in each dose group of Ziziphi Spinosae Semen extracts. In addition, the proportion of distance and time of entering the open arm area of the elevated plus maze was significantly increased in each dose group of Ziziphi Spinosae Semen extracts. The contents of 5-HT and 5-HIAA in serum and thalamus of mice increased to varying degrees in each dose group of Ziziphi Spinosae Semen extracts; the contents of CRH, ACTH, and CORT in serum of mice were significantly decreased. The protein expression of TPH2 was significantly increased. The protein expression of MAOA, SERT, and Freud1 was significantly decreased. Ziziphi Spinosae Semen extracts could also significantly reduce the protein expression of C-FOS but significantly increase the protein expression of PSD95, ARC, and SYN1. They could reduce the pathological damage of the hippocampus in mice and significantly increase the protein expression of ZO-1, occluding, and claudin 1. The protein expression of NLRP3, GSDMD, ASC, caspase-3, and caspase-8 in the thalamic tissue of mice was significantly decreased, and the protein expression of HO1 and NRF2 was significantly increased. In conclusion, Ziziphi Spinosae Semen extracts could effectively improve sleep disorders and depression-like behaviors in CUMS-induced model mice, which may be related to regulating the 5-HT anabolism process and hypothalamic-pituitary-adrenal(HPA) axis-related hormone levels, reducing pathological damage in the hippocampus, improving synaptic plasticity, repairing BBB integrity, and alleviating inflammatory response and oxidative stress damage.
Animals
;
Ziziphus/chemistry*
;
Mice
;
Male
;
Depression/psychology*
;
Drugs, Chinese Herbal/administration & dosage*
;
Sleep Initiation and Maintenance Disorders/psychology*
;
Stress, Psychological/complications*
;
Behavior, Animal/drug effects*
;
Humans
;
Disease Models, Animal
2.Identification of blood-entering components of Anshen Dropping Pills based on UPLC-Q-TOF-MS/MS combined with network pharmacology and evaluation of their anti-insomnia effects and mechanisms.
Xia-Xia REN ; Jin-Na YANG ; Xue-Jun LUO ; Hui-Ping LI ; Miao QIAO ; Wen-Jia WANG ; Yi HE ; Shui-Ping ZHOU ; Yun-Hui HU ; Rui-Ming LI
China Journal of Chinese Materia Medica 2025;50(7):1928-1937
This study identified blood-entering components of Anshen Dropping Pills and explored their anti-insomnia effects and mechanisms. The main blood-entering components of Anshen Dropping Pills were detected and identified by UPLC-Q-TOF-MS/MS. The rationality of the formula was assessed by using enrichment analysis based on the relationship between drugs and symptoms, and core targets of its active components were selected as the the potential anti-insomnia targets of Anshen Dropping Pills through network pharmacology analysis. Furthermore, protein-protein interaction(PPI) network, Gene Ontology(GO) enrichment analysis, and Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway analysis were performed on the core targets. An active component-core target network for Anshen Dropping Pills was constructed. Finally, the effects of low-, medium-, and high-dose groups of Anshen Dropping Pills on sleep episodes, sleep duration, and sleep latency in mice were measured by supraliminal and subliminal pentobarbital sodium experiments. Moreover, total scores of the Pittsburgh sleep quality index(PSQI) scale was used to evaluate the changes before and after the treatment with Anshen Dropping Pills in a clinical study. The enrichment analysis based on the relationship between drugs and symptoms verified the rationality of the Anshen Dropping Pills formula, and nine blood-entering components of Anshen Dropping Pills were identified by UPLC-Q-TOF-MS/MS. The network proximity revealed a significant correlation between eight components and insomnia, including magnoflorine, liquiritin, spinosin, quercitrin, jujuboside A, ginsenoside Rb_3, glycyrrhizic acid, and glycyrrhetinic acid. Network pharmacology analysis indicated that the major anti-insomnia pathways of Anshen Dropping Pills involved substance and energy metabolism, neuroprotection, immune system regulation, and endocrine regulation. Seven core genes related to insomnia were identified: APOE, ALB, BDNF, PPARG, INS, TP53, and TNF. In summary, Anshen Dropping Pills could increase sleep episodes, prolong sleep duration, and reduce sleep latency in mice. Clinical study results demonstrated that Anshen Dropping Pills could decrease total scores of PSQI scale. This study reveals the pharmacodynamic basis and potential multi-component, multi-target, and multi-pathway effects of Anshen Dropping Pills, suggesting that its anti-insomnia mechanisms may be associated with the regulation of insomnia-related signaling pathways. These findings offer a theoretical foundation for the clinical application of Anshen Dropping Pills.
Animals
;
Drugs, Chinese Herbal/administration & dosage*
;
Tandem Mass Spectrometry/methods*
;
Sleep Initiation and Maintenance Disorders/metabolism*
;
Mice
;
Network Pharmacology
;
Male
;
Chromatography, High Pressure Liquid
;
Humans
;
Protein Interaction Maps/drug effects*
;
Sleep/drug effects*
;
Female
;
Adult
3.Effect of Fushen Decoction on 5-HT system and GABA expression in mouse model of PCPA-induced insomnia.
Jun-Hang HU ; Fei XU ; Tong-Sheng WANG ; Hua-Sheng PENG ; Li LI
China Journal of Chinese Materia Medica 2025;50(6):1581-1591
This study aims to observe the mind-tranquilizing effect of Fushen Decoction on mice and investigate its effects on the 5-hydroxytryptamine(5-HT) system and γ-aminobutyric acid(GABA) in the brain of the mouse model of 4-chloro-DL-phenylalanine(PCPA)-induced insomnia. ICR mice were administrated with coffee(1 g·kg~(-1)) for 3 days, and the effects of Fushen Decoction(10, 20, and 40 g·kg~(-1)) on the autonomic activities of normal mice and coffee-treated mice were observed. Furthermore, the effects of Fushen Decoction on the autonomic activity and sleep induced by a suprathreshold dose of pentobarbital sodium in the mouse model of PCPA(350 mg·kg~(-1) for 3 consecutive days)-induced insomnia were observed. The levels of tryptophan hydroxylase(TPH), 5-hydroxytryptophan(5-HTP), and 5-HT in the serum, as well as those of 5-HTP and 5-HT in the brain stem, hippocampus, and cortex, were measured by enzyme-linked immunosorbent assay(ELISA). The fluorescence intensity of 5-HT in the raphe nucleus, hippocampus, and cortex was measured by the immunofluorescence method. The protein levels of tryptophan hydroxylase-2(TPH2) and 5-HT_(1A) receptor(5-HT_(1A)R) in the brain stem, hippocampus, and cortex were measured by Western blot. The levels of GABA in the hypothalamus, hippocampus, and cortex were measured by ELISA and immunohistochemistry methods. The results showed that Fushen Decoction(20, 40 g·kg~(-1)) reduced the number of autonomous activities in normal mice, coffee-treated mice, and the mouse model of PCPA-induced insomnia, and prolonged the duration of sleep induced by a suprathreshold dose of pentobarbital sodium in the mouse model. Fushen Decoction(20, 40 g·kg~(-1)) elevated the levels of TPH, 5-HTP, and 5-HT in the serum, and TPH2, 5-HTP, 5-HT, and 5-HT_(1A)R in the brain stem, hippocampus, and cortex, and up-regulated GABA expression in the hypothalamus, cortex, and hippocampus of the mouse model of PCPA-induced insomnia. In conclusion, Fushen Decoction(20, 40 g·kg~(-1)) exerted a mind-tranquilizing effect on mice by up-regulating the expression of TPH2, enhancing the 5-HT system, and elevating the GABA level in the brain.
Animals
;
Serotonin/genetics*
;
Sleep Initiation and Maintenance Disorders/genetics*
;
Mice
;
Drugs, Chinese Herbal/administration & dosage*
;
Male
;
Mice, Inbred ICR
;
gamma-Aminobutyric Acid/genetics*
;
Disease Models, Animal
;
Fenclonine/adverse effects*
;
Tryptophan Hydroxylase/genetics*
;
Brain/metabolism*
;
Sleep/drug effects*
;
Humans
;
5-Hydroxytryptophan/metabolism*
4.Antidepressant effects of Ziziphi Spinosae Semen extract on depressive-like behaviors in sleep deprivation rats based on integrated serum metabolomics and gut microbiota.
Liang-Lei SONG ; Ya-Yu SUN ; Ze-Jia NIU ; Jia-Ying LIU ; Xiang-Ping PEI ; Yan YAN ; Chen-Hui DU
China Journal of Chinese Materia Medica 2025;50(16):4510-4524
Based on serum metabolomics and gut microbiota technology, this study explores the effects and mechanisms of the water extract of Ziziphi Spinosae Semen(SZRW) and the petroleum ether extract of Ziziphi Spinosae Semen(SZRO) in improving depressive-like behaviors induced by sleep deprivation. A modified multi-platform water environment method was employed to establish a rat model of sleep deprivation. Depressive-like behaviors in rats were assessed through the sucrose preference test and forced swim test. The expression of barrier proteins, such as Occludin, in the colon was determined by immunofluorescence. UPLC-Q-Orbitrap MS was utilized to analyze the serum metabolic profiles of sleep-deprived rats, screen for differential metabolites, and analyze metabolic pathways. The diversity of the gut microbiota was detected using 16S rRNA gene sequencing. Spearman correlation coefficient analysis was conducted to assess the correlation between differential metabolites and gut microbiota. The results indicated that SZRO significantly increased the sucrose preference index and decreased the immobility time in the forced swim test in rats. A total of 34 differential metabolites were identified through serum metabolomics. SZRW and SZRO shared five metabolic pathways, including phenylalanine metabolism. SZRW uniquely featured taurine and hypotaurine metabolism, while SZRO uniquely featured linoleic acid metabolism and tyrosine metabolism. Correlation analysis revealed that SZRW could upregulate the abundance of Bilophila, promoting the production of indole-3-propionic acid and subsequently upregulating the expression levels of intestinal tight junction proteins such as ZO-1, Occludin, and Claudin-1. SZRO could indirectly influence metabolic pathways such as arginine metabolism and linoleic acid metabolism by upregulating the abundance of gut microbiota such as Coprococcus and Eubacterium species. Both SZRW and SZRO can regulate endogenous metabolism, including amino acids, energy, and lipids, alter the gut microbiota microecology, and improve depressive-like behaviors. SZRO demonstrated superior effects in regulating metabolic pathways and gut microbiota structure compared to SZRW. The findings of this study provide a scientific basis for elucidating the pharmacodynamic material basis of Ziziphi Spinosae Semen.
Animals
;
Rats
;
Gastrointestinal Microbiome/drug effects*
;
Male
;
Metabolomics
;
Drugs, Chinese Herbal/administration & dosage*
;
Depression/blood*
;
Rats, Sprague-Dawley
;
Sleep Deprivation/complications*
;
Ziziphus/chemistry*
;
Antidepressive Agents/administration & dosage*
;
Behavior, Animal/drug effects*
;
Humans
5.Mechanism of Chaijin Jieyu Anshen Formula in regulating synaptic damage in nucleus accumbens neurons of rats with insomnia complicated with depression through TREM2/C1q axis.
Ying-Juan TANG ; Jia-Cheng DAI ; Song YANG ; Xiao-Shi YU ; Yao ZHANG ; Hai-Long SU ; Zhi-Yuan LIU ; Zi-Xuan XIANG ; Jun-Cheng LIU ; Hai-Xia HE ; Jian LIU ; Yuan-Shan HAN ; Yu-Hong WANG ; Man-Shu ZOU
China Journal of Chinese Materia Medica 2025;50(16):4538-4545
This study aims to investigate the effect of Chaijin Jieyu Anshen Formula on the neuroinflammation of rats with insomnia complicated with depression through the regulation of triggering receptor expressed on myeloid cells 2(TREM2)/complement protein C1q signaling pathway. Rats were randomly divided into a normal group, a model group, a positive drug group, as well as a high, medium, and low-dose groups of Chaijin Jieyu Anshen Formula, with 10 rats in each group. Except for the normal group, the other groups were injected with p-chlorophenylalanine and exposed to chronic unpredictable mild stress to establish the rat model of insomnia complicated with depression. The sucrose preference experiment, open field experiment, and water maze test were performed to evaluate the depression in rats. Enzyme-linked immunosorbent assay was employed to detect serum 5-hydroxytryptamine(5-HT), dopamine(DA), and norepinephrine(NE) levels. Hematoxylin and eosin staining and Nissl staining were used to observe the damage in nucleus accumbens neurons. Western blot and immunofluorescence were performed to detect TREM2, C1q, postsynaptic density 95(PSD-95), and synaptophysin 1(SYN1) expressions in rat nucleus accumbens, respectively. Golgi-Cox staining was utilized to observe the synaptic spine density of nucleus accumbens neurons. The results show that, compared with the model group, Chaijin Jieyu Anshen Formula can significantly increase the sucrose preference as well as the distance and number of voluntary activities, shorten the immobility time in forced swimming test and the successful incubation period of positioning navigation, and prolong the stay time of space exploration in the target quadrant test. The serum 5-HT, DA, and NE contents in the model group are significantly lower than those in the normal group, with the above contents significantly increased after the intervention of Chaijin Jieyu Anshen Formula. In addition, Chaijin Jieyu Anshen Formula can alleviate pathological damages such as swelling and loose arrangement of tissue cells in the nucleus accumbens, while increasing the Nissl body numbers. Chaijin Jieyu Anshen Formula can improve synaptic damage in the nucleus accumbens and increase the synaptic spine density. Compared to the normal group, the expression of C1q protein was significantly higher in the model group, while the expression of TREM2 protein was significantly lower. Compared to the model group, the intervention with Chaijin Jieyu Anshen Formula significantly downregulated the expression of C1q protein and significantly upregulated the expression of TREM2. Compared with the model group, the PSD-95 and SYN1 fluorescence intensity is significantly increased in the groups receiving different doses of Chaijin Jieyu Anshen Formula. In summary, Chaijin Jieyu Anshen Formula can reduce the C1q protein expression, relieve the TREM2 inhibition, and promote the synapse-related proteins PSD-95 and SNY1 expression. Chaijin Jieyu Anshen Formula improves synaptic injury of the nucleus accumbens neurons, thereby treating insomnia complicated with depression.
Animals
;
Male
;
Rats
;
Nucleus Accumbens/metabolism*
;
Drugs, Chinese Herbal/administration & dosage*
;
Depression/complications*
;
Membrane Glycoproteins/genetics*
;
Rats, Sprague-Dawley
;
Sleep Initiation and Maintenance Disorders/complications*
;
Neurons/metabolism*
;
Receptors, Immunologic/genetics*
;
Signal Transduction/drug effects*
;
Synapses/metabolism*
6.Beneficial Effects of Dendrobium officinale Extract on Insomnia Rats Induced by Strong Light and Noise via Regulating GABA and GABAA Receptors.
Heng-Pu ZHOU ; Jie SU ; Ke-Jian WEI ; Su-Xiang WU ; Jing-Jing YU ; Yi-Kang YU ; Zhuang-Wei NIU ; Xiao-Hu JIN ; Mei-Qiu YAN ; Su-Hong CHEN ; Gui-Yuan LYU
Chinese journal of integrative medicine 2025;31(6):490-498
OBJECTIVE:
To explore the therapeutic effects and underlying mechanisms of Dendrobium officinale (Tiepi Shihu) extract (DOE) on insomnia.
METHODS:
Forty-two male Sprague-Dawley rats were randomly divided into 6 groups (n=7 per group): normal control, model control, melatonin (MT, 40 mg/kg), and 3-dose DOE (0.25, 0.50, and 1.00 g/kg) groups. Rats were raised in a strong-light (10,000 LUX) and -noise (>80 db) environment (12 h/d) for 16 weeks to induce insomnia, and from week 10 to week 16, MT and DOE were correspondingly administered to rats. The behavior tests including sodium pentobarbital-induced sleep experiment, sucrose preference test, and autonomous activity test were used to evaluate changes in sleep and emotions of rats. The metabolic-related indicators such as blood pressure, blood viscosity, blood glucose, and uric acid in rats were measured. The pathological changes in the cornu ammonis 1 (CA1) region of rat brain were evaluated using hematoxylin and eosin staining and Nissl staining. Additionally, the sleep-related factors gamma-aminobutyric acid (GABA), glutamate (GA), 5-hydroxytryptamine (5-HT), and interleukin-6 (IL-6) were measured using enzyme linked immunosorbent assay. Finally, we screened potential sleep-improving receptors of DOE using polymerase chain reaction (PCR) array and validated the results with quantitative PCR and immunohistochemistry.
RESULTS:
DOE significantly improved rats' sleep and mood, increased the sodium pentobarbital-induced sleep time and sucrose preference index, and reduced autonomic activity times (P<0.05 or P<0.01). DOE also had a good effect on metabolic abnormalities, significantly reducing triglyceride, blood glucose, blood pressure, and blood viscosity indicators (P<0.05 or P<0.01). DOE significantly increased the GABA content in hippocampus and reduced the GA/GABA ratio and IL-6 level (P<0.05 or P<0.01). In addition, DOE improved the pathological changes such as the disorder of cell arrangement in the hippocampus and the decrease of Nissel bodies. Seven differential genes were screened by PCR array, and the GABAA receptors (Gabra5, Gabra6, Gabrq) were selected for verification. The results showed that DOE could up-regulate their expressions (P<0.05 or P<0.01).
CONCLUSION
DOE demonstrated remarkable potential for improving insomnia, which may be through regulating GABAA receptors expressions and GA/GABA ratio.
Animals
;
Dendrobium/chemistry*
;
Rats, Sprague-Dawley
;
Male
;
Sleep Initiation and Maintenance Disorders/blood*
;
Plant Extracts/therapeutic use*
;
Receptors, GABA-A/metabolism*
;
Noise/adverse effects*
;
Light/adverse effects*
;
gamma-Aminobutyric Acid/metabolism*
;
Sleep/drug effects*
;
Rats
;
Receptors, GABA/metabolism*
7.Competitive roles of slow/delta oscillation-nesting-mediated sleep disruption under acute methamphetamine exposure in monkeys.
Xin LV ; Jie LIU ; Shuo MA ; Yuhan WANG ; Yixin PAN ; Xian QIU ; Yu CAO ; Bomin SUN ; Shikun ZHAN
Journal of Zhejiang University. Science. B 2025;26(7):694-707
Abuse of amphetamine-based stimulants is a primary public health concern. Recent studies have underscored a troubling escalation in the inappropriate use of prescription amphetamine-based stimulants. However, the neurophysiological mechanisms underlying the impact of acute methamphetamine exposure (AME) on sleep homeostasis remain to be explored. This study employed non-human primates and electroencephalogram (EEG) sleep staging to evaluate the influence of AME on neural oscillations. The primary focus was on alterations in spindles, delta oscillations, and slow oscillations (SOs) and their interactions as conduits through which AME influences sleep stability. AME predominantly diminishes sleep-spindle waves in the non-rapid eye movement 2 (NREM2) stage, and impacts SOs and delta waves differentially. Furthermore, the competitive relationships between SO/delta waves nesting with sleep spindles were selectively strengthened by methamphetamine. Complexity analysis also revealed that the SO-nested spindles had lost their ability to maintain sleep depth and stability. In summary, this finding could be one of the intrinsic electrophysiological mechanisms by which AME disrupted sleep homeostasis.
Animals
;
Methamphetamine
;
Electroencephalography
;
Male
;
Sleep/drug effects*
;
Central Nervous System Stimulants
;
Delta Rhythm/drug effects*
;
Sleep Stages/drug effects*
8.Cannabidiol regulates circadian rhythm to improve sleep disorders following general anesthesia in rats.
Xinshun WU ; Jingcao LI ; Ying LIU ; Renhong QIU ; Henglin WANG ; Rui XYE ; Yang ZHANG ; Shuo LI ; Qiongyin FAN ; Huajin DONG ; Youzhi ZHANG ; Jiangbei CAO
Journal of Southern Medical University 2025;45(4):744-750
OBJECTIVES:
To assess the regulatory effect of cannabidiol (CBD) on circadian rhythm sleep disorders following general anesthesia and explore its potential mechanism in a rat model of propofol-induced rhythm sleep disorder.
METHODS:
An electrode was embedded in the skull for cortical EEG recording in 24 male SD rats, which were randomized into control, propofol, CBD treatment, and diazepam treatment groups (n=6). Eight days later, a single dose of propofol (10 mg/kg) was injected via the tail vein with anesthesia maintenance for 3 h in the latter 3 groups, and daily treatment with saline, CBD or diazepam was administered via gavage; the control rats received only saline injection. A wireless system was used for collecting EEG, EMG, and body temperature data within 72 h after propofol injection. After data collection, blood samples and hypothalamic tissue samples were collected for determining serum levels of oxidative stress markers and hypothalamic expressions of the key clock proteins.
RESULTS:
Compared with the control rats, the rats with CBD treatment showed significantly increased sleep time at night (20:00-6:00), especially during the time period of 4:00-6:00 am. Compared with the rats in propofol group, which had prolonged SWS time and increased sleep episodes during 18:00-24:00 and sleep-wake transitions, the CBD-treated rats exhibited a significant reduction of SWS time and fewer SWS-to-active-awake transitions with increased SWS aspects and sleep-wake transitions at night (24:00-08:00). Diazepam treatment produced similar effect to CBD but with a weaker effect on sleep-wake transitions. Propofol caused significant changes in protein expressions and redox state, which were effectively reversed by CBD treatment.
CONCLUSIONS
CBD can improve sleep structure and circadian rhythm in rats with propofol-induced sleep disorder possibly by regulating hypothalamic expressions of the key circadian clock proteins, suggesting a new treatment option for perioperative sleep disorders.
Animals
;
Rats, Sprague-Dawley
;
Male
;
Cannabidiol/therapeutic use*
;
Rats
;
Circadian Rhythm/drug effects*
;
Propofol/adverse effects*
;
Anesthesia, General/adverse effects*
;
Sleep Wake Disorders/chemically induced*
;
Hypothalamus/metabolism*
;
Electroencephalography
9.Melatonin Ameliorates Abnormal Sleep-Wake Behavior via Facilitating Lipid Metabolism in a Zebrafish Model of Parkinson's Disease.
Meng-Zhu PANG ; Han-Xing LI ; Xue-Qin DAI ; Xiao-Bo WANG ; Jun-Yi LIU ; Yun SHEN ; Xing XU ; Zhao-Min ZHONG ; Han WANG ; Chun-Feng LIU ; Fen WANG
Neuroscience Bulletin 2024;40(12):1901-1914
Sleep-wake disorder is one of the most common nonmotor symptoms of Parkinson's disease (PD). Melatonin has the potential to improve sleep-wake disorder, but its mechanism of action is still unclear. Our data showed that melatonin only improved the motor and sleep-wake behavior of a zebrafish PD model when melatonin receptor 1 was present. Thus, we explored the underlying mechanisms by applying a rotenone model. After the PD zebrafish model was induced by 10 nmol/L rotenone, the motor and sleep-wake behavior were assessed. In situ hybridization and real-time quantitative PCR were used to detect the expression of melatonin receptors and lipid-metabolism-related genes. In the PD model, we found abnormal lipid metabolism, which was reversed by melatonin. This may be one of the main pathways for improving PD sleep-wake disorder.
Animals
;
Zebrafish
;
Melatonin/pharmacology*
;
Lipid Metabolism/drug effects*
;
Disease Models, Animal
;
Rotenone/pharmacology*
;
Sleep Wake Disorders/metabolism*
;
Parkinson Disease/metabolism*
;
Motor Activity/drug effects*
;
Sleep/drug effects*
10.Ginsenoside Rg1 promotes non-rapid eye movement sleep via inhibition of orexin neurons of the lateral hypothalamus and corticotropin-releasing hormone neurons of the paraventricular hypothalamic nucleus.
Yi-Yuan WANG ; Yi WU ; Ke-Wei YU ; Hong-Yu XIE ; Yi GUI ; Chang-Rui CHEN ; Nian-Hong WANG
Journal of Integrative Medicine 2024;22(6):719-728
OBJECTIVE:
This study investigates the sleep-modulating effects of ginsenoside Rg1 (Rg1, C42H72O14), a key bioactive component of ginseng, and elucidates its underlying mechanisms.
METHODS:
C57BL/6J mice were intraperitoneally administered doses of Rg1 ranging from 12.5 to 100 mg/kg. Sleep parameters were assessed to determine the average duration of each sleep stage by monitoring the electrical activity of the brain and muscles. Further, orexin neurons in the lateral hypothalamus (LH) and corticotropin-releasing hormone (CRH) neurons in the paraventricular hypothalamic nucleus (PVH) were ablated using viral vector surgery and electrode embedding. The excitability of LHorexin and PVHCRH neurons was evaluated through the measurement of cellular Finkel-Biskis-Jinkins murine osteosarcoma viral oncogene homolog (c-Fos) expression.
RESULTS:
Rg1 (12.5-100 mg/kg) augmented the duration of non-rapid eye movement (NREM) sleep phases, while reducing the duration of wakefulness, in a dose dependent manner. The reduced latency from wakefulness to NREM sleep indicates an accelerated sleep initiation time. We found that these sleep-promoting effects were weakened in the LHorexin and PVHCRH neuron ablation groups, and disappeared in the orexin and CRH double-ablation group. Decreased c-Fos protein expression in the LH and PVH confirmed that Rg1 promoted NREM sleep by inhibiting orexin and CRH neurons.
CONCLUSION
Rg1 increases the duration of NREM sleep, underscoring the essential roles of LHorexin and PVHCRH neurons in facilitating the sleep-promoting effects of Rg1. Please cite this article as: Wang YY, Wu Y, Yu KW, Xie HY, Gui Y, Chen CR, Wang NH. Ginsenoside Rg1 promotes non-rapid eye movement sleep via inhibition of orexin neurons of the lateral hypothalamus and corticotropin-releasing hormone neurons of the paraventricular hypothalamic nucleus. J Integr Med. 2024; 22(6): 721-730.
Animals
;
Ginsenosides/pharmacology*
;
Orexins/metabolism*
;
Mice, Inbred C57BL
;
Neurons/metabolism*
;
Paraventricular Hypothalamic Nucleus/metabolism*
;
Male
;
Hypothalamic Area, Lateral/metabolism*
;
Corticotropin-Releasing Hormone/metabolism*
;
Mice
;
Sleep/drug effects*

Result Analysis
Print
Save
E-mail