1
|
Yang R, Deng MY, Yang LK, Wang GZ, Ma J, Wen Q, Gao N, Qiao HL. Identification of cytochrome P450 2E1 as a novel target in neuroinflammation and development of its inhibitor Q11 as a treatment strategy. Free Radic Biol Med 2025; 234:220-232. [PMID: 40122152 DOI: 10.1016/j.freeradbiomed.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
Neuroinflammation is implicated in nearly all pathological processes of central nervous system (CNS) diseases. However, no medications specifically targeting neuroinflammation are clinically available, and conventional anti-inflammatory drugs exhibit limited efficacy. Here, we identified cytochrome P450 2E1 (CYP2E1) as a novel therapeutic target in neuroinflammation. Elevated CYP2E1 levels were observed in hippocampal tissues of mouse and rat neuroinflammation models, as well as in LPS-stimulated primary microglia. Genetic ablation of Cyp2e1 improved spatial learning and memory in neuroinflammatory rats and reduced pro-inflammatory cytokine levels in Cyp2e1-deficient microglia. Furthermore, Q11 (1-(4-methyl-5-thiazolyl) ethanone), a novel CYP2E1 inhibitor developed and synthesized in our laboratory, effectively ameliorated Alzheimer's disease-related spatial learning and memory functions and depression-related anxiety-like behaviors in mice/rats. Mechanistically, Q11 attenuated microglial activation, neuronal damage, oxidative stress, and neuroinflammation by suppressing the PI3K/Akt, STAT1/3, and NF-κB signaling pathways. These findings establish CYP2E1 as a druggable target for neuroinflammation and propose Q11 as a promising candidate for treating neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Rui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Meng-Yan Deng
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Lu-Kui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Guan-Zhe Wang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jun Ma
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Na Gao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Hai-Ling Qiao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
2
|
Cao LH, Wang ZZ, Zhao H, Tian S, He HJ, Miao JX, Huang SN, Wang XY, Song YG, Kang L, Wang HY, Wang C, Bai M, Gao S, Miao MS. The microglial state transition as a novel mechanism by which fresh Baihe Dihuang decoction prevents depression by regulating SIRT1/HMGB1 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156718. [PMID: 40220404 DOI: 10.1016/j.phymed.2025.156718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Fresh Baihe Dihuang decoction (FBD) is a classic Traditional Chinese Medicine (TCM) formula used to treat depression. However, its underlying molecular mechanisms in treating depression still need further exploration. PURPOSE In this study, we investigated whether FBD could prevent depressive-like behaviors by remodeling the microglial state transition via the inhibition of SIRT1/HMGB1 signaling in vivo and in vitro. STUDY DESIGN AND METHODS In vivo, adult male C57BL/6 J mice were subjected to chronic unpredictable mild stress (CUMS) for 6 weeks to investigate whether FBD could produce antidepressant-like behavioral effects. At 9 weeks of age, EX527, a SIRT1 inhibitor, was injected intraperitoneally 30 min before the intragastric administration of FBD, Flx or vehicle daily until 12 weeks of age, and the effects of alterations in SIRT1/HMGB1 signaling on CUMS-mediated depression were investigated. In vitro, the anti-depressive mechanism of FBD was further investigated using BV-2 cells (a microglial cell line) and primary PFC neurons. We examined depression-like behaviors using behavioral tests. Serum and supernatants samples were collected and interleukin-1β (IL-1β), IL-6 and tumor necrosis factor-α (TNF-α) levels were measured using enzyme-linked immunosorbent assays (ELISAs). Prefrontal cortical (PFC) tissues, BV-2 cells and PFC neurons were collected to detect neuronal apoptosis, the microglial state or proteins in the silent information regulator 2 homolog 1 (SIRT1)/high mobility group box 1 (HMGB1) signaling pathway via flow cytometry, immunohistochemical staining, immunofluorescence staining,TUNEL staining or western blot analysis. RESULTS The administration of FBD ameliorated the depressive-like behaviors induced by CUMS. In addition, FBD supplementation promoted the transition from a proinflammatory microglial phenotype to an anti-inflammatory microglial phenotype. The FBD-mediated inhibition of HMGB1 expression and its nucleocytoplasmic translocation were identified as likely due to increased SIRT1 activity, effectively inhibiting the subsequent inflammatory response. Furthermore, our findings revealed that FBD notably attenuated neuronal apoptosis in the PFC. In contrast, the SIRT1 inhibitor EX527 counteracted the antidepressant effect of FBD while also reversing the expression of brain-derived neurotrophic factor (BDNF), NeuN, cleaved caspase-3, bax, and bcl-2 proteins. CONCLUSIONS This study showed that FBD prevents depression by mediating a microglial state transition via the SIRT1/HMGB1 signaling pathway, providing a promising and novel antidepressant therapeutic strategy.
Collapse
Affiliation(s)
- Li-Hua Cao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Zhen-Zhen Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hui Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Shuo Tian
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hong-Juan He
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jin-Xin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Sheng-Nan Huang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Xiao-Yu Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Ya-Gang Song
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Le Kang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hao-Yang Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Can Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Ming Bai
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Song Gao
- Henan Solid Formulation Technology Innovation Center, Zhongjing Wanxi Pharmaceutical Co., Ltd., Xixia 474550, Henan Province, China
| | - Ming-San Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China.
| |
Collapse
|
3
|
Chai C, Jin B, Bi J, Cui Y, Cui X, Shan C, Yu S, Wen H. Exploring of Antidepressant Components and Mechanisms of Zhizichi Decoction: Integration of Serum Pharmacochemistry, Network Pharmacology and Anti-inflammatory Analysis Verification. ANALYTICAL SCIENCE ADVANCES 2025; 6:e70002. [PMID: 39991188 PMCID: PMC11845310 DOI: 10.1002/ansa.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/25/2025]
Abstract
Using LC-MS to screen and analyse the characteristics of components and biological systems is a new approach to study the pharmacological substance basis of traditional Chinese medicine, which has strong novelty in analytical science. This study analyses the antidepressant material basis of Zhizichi decoction through the integration strategy of serum pharmacochemistry, network pharmacology and immunoreactivity verification, which helps in overcoming the limitations of TCM research and provided a new perspective and approach for studying the components of ZZCD. First of all, blood from SD rats was collected before and after Zhizichi decoction administration initially. The migration constituents in the serum were then analysed using ultra-high performance liquid chromatography-Q-TOF-MS/MS. By integrating the TCMSP databases with the serum pharmacochemistry results, we constructed the 'ingredients-targets-pathways' network and the protein-protein interaction network for Zhizichi decoction's depression-relieving. Finally, the inhibitory effects of Zhizichi decoction and active ingredient groups comprised of pharmacodynamic components identified in prior network pharmacology study on IL-1β, IL-6 and TNF-α were measured through an inflammatory cytokines experiments. From the serum pharmacochemistry study, 146 migration constituents in serum and their attribution were hypothesized and characterized. They were identified as 18 prototype components and 128 metabolites, of which 121 were Phase I and 7 were Phase II metabolites. The Zhizichi decoction pharmacology network illustrated the relationships of the 20 definitive ingredients, 85 potential targets and 21 signalling pathways in connection with the depression. The targets predicted by pharmacology and protein-protein interaction network were reported to be associated with neuroinflammation, which suggested that further anti-inflammatory experiment was required. For the anti-inflammatory effect of AIGs 1 composed of 14 pharmacodynamic components was basically equivalent to that of whole ZZCD recipe, AIG 1 was hypothesized to be the critical pharmacodynamic components to inhibit inflammatory factors and defined as the antidepressant components' of Zhizichi decoction, providing a scientific foundation for the pursuit of potential new drugs for depression treatments.
Collapse
Affiliation(s)
- Chuan Chai
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Bo Jin
- China Mobile Group Jiangsu Co. Ltd.NanjingJiangsuChina
| | - Jinghan Bi
- Nanjing Zenkom Pharmaceutical Co. Ltd.NanjingJiangsuChina
| | - Yuhan Cui
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Xiaobing Cui
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Chenxiao Shan
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Sheng Yu
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| | - Hongmei Wen
- School of PharmacyNanjing University of Chinese MedicineNanjingJiangsuChina
| |
Collapse
|
4
|
Luo S, Qi Z, Wu F, Tao D, Huang S, Shi C, Yang C, Shi Y, Luo Y. Flavokawain B ameliorates depressive-like behaviors by inhibiting neddylation in the ventral hippocampus. Neuropharmacology 2025; 276:110508. [PMID: 40368175 DOI: 10.1016/j.neuropharm.2025.110508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/16/2025]
Abstract
Depression is a widespread and serious mental illness. Recent evidence suggests that post-translational modifications, particularly neddylation, play a significant role in the pathophysiology of mood disorders. Neddylation, similar to ubiquitination, involves the attachment of NEDD8 to target proteins, regulating their function. However, its role in depression remains unclear. This study aimed to explore the antidepressant effects of flavokawain B (FKB), a novel neddylation inhibitor, in mouse models of depression induced by lipopolysaccharide (LPS) and chronic unpredictable mild stress (CUMS). We assessed depressive- and anxiety-like behaviors using the sucrose preference test (SPT), open-field test (OFT), forced swimming test (FST), and tail suspension test (TST). Neddylation-related protein expression (NEDD8, NAE1, UBA3, UBC12) was measured in the dorsal and ventral hippocampus (DH, VH), prelimbic cortex (PrL), and nucleus accumbens (NAc). Results showed that FKB reversed the activation of neddylation anxiety- and depressive-like behaviors in both LPS- and CUMS-treated mice. The LPS and CUMS experimental paradigms only impacted the neddylation pathway in the VH and not other brain regions (DH, PrL, IL, NAc). In conclusion, these findings suggest that FKB ameliorates depressive-like behaviors by targeting the NAE1/UBA3/UBC12 pathway in the VH, providing a potential therapeutic approach for depression.
Collapse
Affiliation(s)
- Shuting Luo
- School of Medicine, Hunan Normal University, Changsha, 410013, China; Hunan Province People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, 410081, China
| | - Zihan Qi
- School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Feilong Wu
- School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Dan Tao
- School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China
| | - Cuijie Shi
- College of Forensic Medicine, Hebei Medical University, 050017, Shijiazhuang, China
| | - Chang Yang
- School of Medicine, Hunan Normal University, Changsha, 410013, China; Hunan Province People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, 410081, China
| | - Yan Shi
- School of Medicine, Hunan Normal University, Changsha, 410013, China.
| | - Yixiao Luo
- School of Medicine, Hunan Normal University, Changsha, 410013, China; Hunan Province People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
5
|
Ding Y, Wang Y, Qi M, Zhang X, Wu D. Pioglitazone Modulates Microglia M1/M2 Polarization Through PPAR-γ Pathway and Exerts Neuroprotective Effects in Experimental Subarachnoid Hemorrhage. Mol Neurobiol 2025; 62:5930-5946. [PMID: 39668302 DOI: 10.1007/s12035-024-04664-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Subarachnoid hemorrhage (SAH), a subtype of hemorrhagic stroke primarily resulting from the rupture of intracranial aneurysms, remains a significant contributor to disability and mortality, notwithstanding advancements in treatment. This study investigates the neuroprotective effects of pioglitazone in SAH, focusing on the PPAR-γ pathway and its potential role in mitigating early brain injury (EBI) following SAH. Neuroprotective efficacy was assessed through neurobehavioral assessment, brain water content analysis, TUNEL, immunofluorescence, western blotting, and inflammatory factor assay. Results indicate that pioglitazone treatment effectively mitigated brain edema, reduced neuronal death, and enhanced short-term neurobehavioral function in SAH-afflicted rats. Furthermore, pioglitazone demonstrated sustained improvements in long-term neurobehavioral function and decreased neuronal loss post-SAH. Mechanistically, SAH induced the polarization of microglia towards the M1 phenotype and the release of pro-inflammatory cytokines. Conversely, pioglitazone treatment predominantly shifted microglia polarization towards the M2 phenotype, eliciting a notable release of anti-inflammatory cytokines. Notably, the positive effects of pioglitazone were nullified by the PPAR-γ inhibitor T0070907. In conclusion, our findings suggest that pioglitazone may alleviate neuroinflammation by modulating microglia M1/M2 polarization through the PPAR-γ pathway, thereby conferring neuroprotection against SAH injury and positing itself as a potential therapeutic agent for SAH treatment.
Collapse
Affiliation(s)
- Yingying Ding
- Department of Neurosurgery, The Affiliated Yixing Hospital of Jiangsu University, Jiangsu Province, Yixing, 214200, China
| | - Yue Wang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA) Jiangsu Province, Wuxi, 214044, China
| | - Ming Qi
- Department of Neurosurgery, The Affiliated Yixing Hospital of Jiangsu University, Jiangsu Province, Yixing, 214200, China
| | - Xu Zhang
- Department of Neurosurgery, Wuxi Clinical College of Anhui Medical University (The 904 Hospital of PLA) Jiangsu Province, Wuxi, 214044, China
| | - Da Wu
- Department of Neurosurgery, The Affiliated Yixing Hospital of Jiangsu University, Jiangsu Province, Yixing, 214200, China.
| |
Collapse
|
6
|
Huang M, Chen F, Zhou L, Zhang Q, Wang L, Li L, Yang L, Gao M, Li L, Wang Y, Yang J, Yao G, Li Q, Yang X. The antidepressant effects of kaji-ichigoside F1 via activating PPAR-γ/CX3CR1/Nrf2 signaling and suppressing NF-κB/NLRP3 signaling pathways. Front Pharmacol 2025; 16:1569888. [PMID: 40308754 PMCID: PMC12040888 DOI: 10.3389/fphar.2025.1569888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Depression is a mental illness closely associated with neurological damage and is characterised by high rates of suicide and mood changes. As a traditional medicinal plant, Rosa roxburghii Tratt has been widely used since ancient times in the Miao and Dong regions of Southwest China for the relief of sleep disorders, indigestion, anti-inflammation, neurasthenia and neuroprotection. The total triterpenes of R. roxburghii were previously found to have certain neuroprotective effects, and whether Kaji-ichigoside F1 (KF1), as its main ingredient, plays a relevant pharmacological role needs to be further investigated. Methods Establishment of mouse depression model and BV2 microglia inflammation model using intraperitoneal injection of LPS in mice and LPS stimulated-BV2 microglia, respectively. The antidepressant effects of KF1 were evaluated by forced swim test (FST), sucrose preference test (SPT), tail suspension test (TST) and open field test (OFT). The number of Nissl bodies and apoptotic positive cells in the CA1 region of the hippocampus was observed by Nissl and TUNEL staining. Then, the levels of TNFα, PPAR-γ, TGF-β, and IL-6 cytokines were tested by ELISA kits. Finally, the molecular mechanisms were investigated by Western blotting (WB) and immunofluorescence in vivo and in vitro. Results KF1 dramatically ameliorated LPS-induced depressive like behaviors, neuronal damage, apoptosis, and suppressed the levels of pro-inflammatory cytokines in the serum and hippocampus of mice. Our vitro experiment also showed KF1 significantly reduced cell viability and attenuated apoptosis in LPS-induced BV2 microglia, decreased the mean fluorescence intensity of Caspase-1, TNFα, NF-κB, IL-1β, NLRP3, and Keap1. However, the mean fluorescence intensity of GCLC, GCLM, GST, SOD1, HO-1, and Nrf2 were significantly increased. Finally, Western blot analysis showed that KF1 suppressing the expression of NF-κB/NLRP3 signaling pathway and activating PPARγ/CX3CR1/Nrf2 signaling pathway both in vivo and in vitro. Conclusion In conclusion, these results suggest that KF1 is an effective alleviator of LPS-induced depression-like effects in vivo and in vitro. These effects were associated with activating PPARγ/CX3CR1/Nrf2 signaling, and suppressing NF-κB/NLRP3 signaling pathways.
Collapse
Affiliation(s)
- Maoyang Huang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Faju Chen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Lang Zhou
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Qing Zhang
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Li Wang
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Liangqun Li
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Lishou Yang
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Ming Gao
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Lilang Li
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Yu Wang
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Juan Yang
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Guanping Yao
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Qiji Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| | - Xiaosheng Yang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guiyang, China
| |
Collapse
|
7
|
Zhang H, Zhao Z, Liu P, Wang M, Liu YE, He H, Ge Y, Zhou T, Xiao C, You Z, Zhang J. Gastrodin enhances stress resilience through promoting Wnt/β-Catenin-dependent neurogenesis. J Adv Res 2025:S2090-1232(25)00261-9. [PMID: 40233892 DOI: 10.1016/j.jare.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/24/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Enhancing stress resilience constitutes a pivotal strategy in mitigating the risk of depression, making it a critical component of both prevention and treatment. In the current work, we identified a compound, gastrodin (GAS), as capable of enhancing stress resilience, as demonstrated by its ability to protect against depression following chronic stress exposure. OBJECTIVES To elucidate the potential of GAS to promote neurogenesis under chronic stress, along with the associated cellular and molecular processes involved. METHOD We evaluated the effect of GAS on NSPC proliferation and differentiation using both in vitro and in vivo investigations. Neurogenesis was inhibited using temozolomide to verify GAS's impact on stress resilience. Comprehensive methodologies, including hippocampal transcriptome analysis and western blotting, were utilized to identify the involvement of the Wnt/β-catenin pathway. Immunolocalization was conducted to confirm β-catenin's nuclear translocation in SOX2+ cells within the hippocampal dentate gyrus subgranular zone. RESULTS GAS demonstrated robust stimulation of NSPC proliferation and neuronal differentiation, enhancing adult hippocampal neurogenesis under conditions of chronic stress. Inhibition of neurogenesis negated GAS's protective effects on stress resilience. Integrated analysis pointed to the Wnt/β-catenin signaling pathway within NSPCs as a crucial mechanism facilitating GAS-promoted neurogenesis. Inhibiting Wnt expression or blocking β-catenin's nuclear translocation abolished GAS's neurogenic and stress-resilience enhancing effects. CONCLUSION These results suggested that GAS directly activates the Wnt/β-catenin signaling pathway, which promotes the proliferation and neuronal differentiation of NSPCs, thereby enhancing adult hippocampal neurogenesis and promoting stress resilience to mitigate the risk of depression.
Collapse
Affiliation(s)
- Haili Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Zhihuang Zhao
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Pei Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Meidan Wang
- Faculty of Biology, University of Freiburg, Freiburg 79104, Germany.
| | - Yu-E Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Hui He
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 6100544, China.
| | - Yangyan Ge
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Tao Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Chenghong Xiao
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Zili You
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 6100544, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
8
|
Zhu W, Chang J, Tian L, Yang X, Li W. Depletion of HSP60 in Microglia Leads to Synaptic Dysfunction and Depression-Like Behaviors Through Enhanced Synaptic Pruning in Male Mice. CNS Neurosci Ther 2025; 31:e70394. [PMID: 40237297 PMCID: PMC12001071 DOI: 10.1111/cns.70394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
AIMS Microglia, as resident macrophages in the brain, play an important role in depression. Heat shock protein 60 (HSP60), as a chaperone protein, plays a role in cell stress. However, the role of microglial HSP60 in depression remains unclear. METHODS CX3CR1-CreER was used to generate microglial-specific HSP60 knockout (HSP60 cKO) mice. Behavioral tests, western blotting, Golgi staining, biochemical assays, and proteomics were employed to assess depression-like symptoms, microglial activation, and synaptic changes. RESULTS HSP60 cKO male mice exhibited depressive-like behaviors, without anxiety-like behavior, including increased immobility in the forced swimming and tail suspension tests, reduced sucrose preference, and elevated corticosterone (CORT) levels, indicating HPA axis activation. Microglial activation was confirmed by the increased expression levels of CD68 and CD86, the elevated transcription of the cybb gene, and reduced branch complexity. Enhanced phagocytosis of excitatory synapses, reduced dendritic spine density, and decreased glutamate levels were observed, with downregulation of synaptic proteins (AMPAR2, Synapsin-1, PSD95), indicating dysregulated synaptic pruning. Moreover, GO analysis showed 20 significant differentially expressed proteins (DEPs) from proteomics are associated with the presynaptic endosome, which plays a crucial role in maintaining synaptic function. Treatment with PLX3397, a CSF1R inhibitor, alleviated depressive-like behaviors and restored synaptic density in HSP60 cKO male mice. CONCLUSIONS HSP60 deletion in microglia leads to overactivation of microglia, impaired synaptic function, and depression-like behaviors, highlighting the importance of microglial homeostasis in mood regulation and the potential therapeutic role of microglial modulation.
Collapse
Affiliation(s)
- Wenhui Zhu
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single‐Cell and Extracellular Vesicles, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jinlong Chang
- Department of Physiology and Pathophysiology, School of Basic Medical SciencesDali UniversityDaliChina
| | - Liusuyan Tian
- The Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenChina
| | - Xiuyan Yang
- The Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenChina
| | - Weifen Li
- School of Pharmacy, Shenzhen University Medical SchoolShenzhen UniversityShenzhenChina
- State Key Laboratory of Oncogenomics, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| |
Collapse
|
9
|
Xu X, Tan L, Zhang X. Prenatal Exposure to Valproic Acid may Alter CD200/CD200R Signaling Pathways in a Rat Model of Autism Spectrum Disorder. ALPHA PSYCHIATRY 2025; 26:39444. [PMID: 40352073 PMCID: PMC12059736 DOI: 10.31083/ap39444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/21/2024] [Accepted: 09/26/2024] [Indexed: 05/14/2025]
Abstract
Objective To investigate the potential toxic effects of prenatal exposure to valproic acid (VPA) on microglia-neuron communication in the brain, with a specific focus on the alterations in key molecules involved in this process, namely CX3CL1/CX3CR1 and CD200/CD200R, during the early stages of life in a rat model of autism. Methods Pregnant female rats were administered either sterile saline or VPA on embryonic day 12.5. The brains of the rat offspring were collected on postnatal day 30 for analysis. Immunohistochemical techniques and enzyme-linked immunosorbent assay (ELISA) were employed to assess changes in microglia-neuron crosstalk. Results The study revealed a significant reduction in CD200 levels within the hippocampus of rats on postnatal day 30 following prenatal exposure to VPA, indicating an impairment in CD200/CD200R signaling. Additionally, there was no observed increase in microglial numbers or any pathological alterations in the hippocampus. Additionally, no significant changes in the levels of CX3CL1 and CX3CR1 were noted in the VPA-exposed rats compared with the control group. Conclusion Prenatal exposure to VPA resulted in a decrease in CD200 expression within the hippocampus, potentially disrupting the communication between microglia and neurons. The findings suggest that VPA may modify the interactions between microglia and neurons, which could lead to neuroinflammation due to hyperactivated microglia. These disruptions have the potential to affect synaptic connectivity and contribute to the development of neurodevelopmental disorders, including autism. Further research is necessary to clarify the underlying mechanisms and implications for pathological conditions associated with autism spectrum disorder (ASD).
Collapse
Affiliation(s)
- Xiaoou Xu
- Department for Science and Technology Management and Education, Chongqing Population and Family Planning Science and Technology Research Institute, 401120 Chongqing, China
| | - Li Tan
- Department of Clinical Medicine, Chongqing Medical and Pharmaceutical College, 401331 Chongqing, China
| | - Xiaojuan Zhang
- Department of Clinical Medicine, Chongqing Medical and Pharmaceutical College, 401331 Chongqing, China
| |
Collapse
|
10
|
Yuan NJ, Zhu WJ, Ma QY, Huang MY, Huo RR, She KJ, Pan JP, Wang JG, Chen JX. Luteolin ameliorates chronic stress-induced depressive-like behaviors in mice by promoting the Arginase-1 + microglial phenotype via a PPARγ-dependent mechanism. Acta Pharmacol Sin 2025; 46:575-591. [PMID: 39496862 PMCID: PMC11845711 DOI: 10.1038/s41401-024-01402-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/23/2024] [Indexed: 11/06/2024]
Abstract
Accumulating evidence shows that neuroinflammation substantially contributes to the pathology of depression, a severe psychiatric disease with an increasing prevalence worldwide. Although modulating microglial phenotypes is recognized as a promising therapeutic strategy, effective treatments are still lacking. Previous studies have shown that luteolin (LUT) has anti-inflammatory effects and confers benefits on chronic stress-induced depression. In this study, we investigated the molecular mechanisms by which LUT regulates the functional phenotypes of microglia in mice with depressive-like behaviors. Mice were exposed to chronic restraint stress (CRS) for 7 weeks, and were administered LUT (10, 30, 40 mg· kg-1 ·day-1, i.g.) in the last 4 weeks. We showed that LUT administration significantly ameliorated depressive-like behaviors and decreased hippocampal inflammation. LUT administration induced pro-inflammatory microglia to undergo anti-inflammatory arginase (Arg)-1+ phenotypic polarization, which was associated with its antidepressant effects. Furthermore, we showed that LUT concentration-dependently increased the expression of PPARγ in LPS + ATP-treated microglia and the hippocampus of CRS-exposed mice, promoting the subsequent inhibition of the NLRP3 inflammasome. Molecular dynamics (MD) simulation and microscale thermophoresis (MST) analysis confirmed a direct interaction between LUT and peroxisome proliferator-activated receptor gamma (PPARγ). By using the PPARγ antagonist GW9662, we demonstrated that LUT-driven protection, both in vivo and in vitro, resulted from targeting PPARγ. First, LUT-induced Arg-1+ microglia were no longer detected when PPARγ was blocked. Next, LUT-mediated inhibition of the NLRP3 inflammasome and downregulation of pro-inflammatory cytokine production were reversed by the inhibition of PPARγ. Finally, the protective effects of LUT, which attenuated the microglial engulfment of synapses and prevented apparent synapse loss in the hippocampus of CRS-exposed mice, were eliminated by blocking PPARγ. In conclusion, this study showed that LUT ameliorates CRS-induced depressive-like behaviors by promoting the Arg-1+ microglial phenotype through a PPARγ-dependent mechanism, thereby alleviating microglial pro-inflammatory responses and reversing microglial phagocytosis-mediated synapse loss.
Collapse
Affiliation(s)
- Nai-Jun Yuan
- Department of Critical Care Medicine, Shenzhen Clinical Research Center for Geriatric, and Guangdong Provincial Clinical Research Center for Geriatrics, Integrated Chinese and Western Medicine Postdoctoral Research Station, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Wen-Jun Zhu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Qing-Yu Ma
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Min-Yi Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Rou-Rou Huo
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Kai-Jie She
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Jun-Ping Pan
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478, Lørenskog, Norway
- Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Basic Medicine, Jinan University, Guangzhou, 510632, China
| | - Ji-Gang Wang
- Department of Critical Care Medicine, Shenzhen Clinical Research Center for Geriatric, and Guangdong Provincial Clinical Research Center for Geriatrics, Integrated Chinese and Western Medicine Postdoctoral Research Station, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China.
- State Key Laboratory for Quality Assurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Jia-Xu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China.
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
11
|
Lei X, Zhao D, Chen T, Li Q, Xue A, Hu Z, Jia F, Li X. Exploring the active components and potential mechanisms of Zhimu-Huangbai herb-pair in the treatment of depression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156365. [PMID: 39904199 DOI: 10.1016/j.phymed.2025.156365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/15/2024] [Accepted: 01/01/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND The Zhimu-Huangbai herb-pair (ZB) is commonly used to treat depression. Previous research has verified that ZB is effective as an antidepressant. Nevertheless, its active components and potential mechanism still require further elucidation. PURPOSE This study aims to analyze the compounds of ZB penetrating into the brain using UPLC-MS and investigate the potential mechanisms of ZB in the treatment of depression through in vivo and in vitro experiments. METHODS The compounds of ZB that penetrate into the brain were identified using the UPLC-MS method. Network pharmacology analysis was employed to predict the therapeutic targets and mechanisms of the compounds of ZB in the brain for the treatment of depression. Subsequently, the molecular docking method was used to analyze the binding between active compounds and target proteins. Rat depression models induced by CUMS were used to investigate the impact of ZB on depression. Finally, the mechanism of ZB treatment for depression was investigated using the LPS-induced BV2 cell inflammation model. RESULTS A total of 17 compounds were identified in ZB that crossed the blood-brain barrier (BBB). The network pharmacological analysis showed that the anti-depressant mechanism of ZB is closely related to inflammatory cytokines, including TNF and IL-6. Furthermore, KEGG and PPI analyses demonstrated that ZB regulates the microglia M1/M2 phenotypic polarization by modulating inflammation-related pathways. ZB was found to improve depression-like behavior in vivo. The molecular docking indicated that the compounds in ZB that penetrate into the brain have a strong binding ability to RELA and PPAR-γ. ZB inhibited the expression of p-p65 and increased the expression of PPAR-γ in the mPFC. By rebalancing the ratio of pro-inflammatory/anti-inflammatory cytokines, ZB was able to reduce neuroinflammation in the mPFC and hippocampus regions. The immunofluorescence results showed that ZB-containing serum reduced M1 polarization induced by LPS in BV2 cells. CONCLUSION This study reveals that ZB effectively alleviates depression by regulating the M1/M2 phenotypic polarization of microglial cells. The mechanism may be that the active compounds of ZB reduce M1 phenotypic polarization by inhibiting P65 and increase M2 phenotypic polarization by promoting PPARγ.
Collapse
Affiliation(s)
- Xia Lei
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Deping Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Tongtong Chen
- College of Pharmacy, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Qing Li
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Ao Xue
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Zhuoyi Hu
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Fan Jia
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Xiaoliang Li
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education & International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province & Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, Hainan, 571199, China.
| |
Collapse
|
12
|
Zhang Z, Yuan D, Jin X, Chang W, Zhang Y, Xie W. Asperosaponin VI suppresses ferroptosis in chondrocytes and ameliorates osteoarthritis by modulating the Nrf2/GPX4/HO-1 signaling pathway. Front Pharmacol 2025; 16:1539092. [PMID: 40093317 PMCID: PMC11906723 DOI: 10.3389/fphar.2025.1539092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Background Asperosaponin VI (AVI) is a naturally occurring monosaccharide derived from Dipsacus asperoides renowned for its anti-inflammatory and bone-protective properties. Objective To elucidate the specific mechanism through which AVI affects chondrocytes in osteoarthritis (OA). Methods For the in vitro experiments, primary chondrocytes were to elucidate the molecular mechanisms underlying the action of AVI.For the in vivo experiments, rat OA models were established using a modified Hulth method. The severity of knee osteoarthritis was evaluated 8 weeks post-surgery. Micro-CT imaging, hematoxylin-eosin staining, and Safranin O-fast green staining were used to assess degeneration in rat knee joints. Immunohistochemistry techniques were conducted to measure the levels of collagen II, MMP13, Nrf2, GPX4, ACSL4, and HO-1 within cartilage tissues. ELISA assays were performed to measure those of TNF-α, IL -6, and PGE2 in serum samples. Results AVI alleviated chondrocyte apoptosis and extracellular matrix degradation in rat OA induced by IL-1β. It attenuated the levels of TNF-α, IL-6, and PGE2 while reducing those of Fe2+ and malondialdehyde (MDA). AVI upregulated the expression of Nrf2, HO-1, and GPX4 while downregulating that of ACSL4. Mechanistic studies revealed that ML385-induced inhibition of the Nrf2 signaling pathway reversed the increase in GPX4 and ACSL4 expression and increased Fe2+ and MDA levels; treatment with erastin, a ferroptosis inducer, produced comparable results. In vivo experiments demonstrated that AVI improved the bone volume/tissue volume and trabecular separation values in OA rats; reversed the Osteoarthritis Research Society International score; upregulated Nrf2, HO-1, and GPX4 expression; downregulated ACSL4 and MMP13 expression, and decreased the serum levels of TNF-α, IL-6, and PGE2. Conclusion Our findings suggest that AVI is a promising therapeutic agent for OA. It exerted its protective effect by regulating the Nrf2/GPX4/HO-1 signaling axis to inhibit cartilage cell ferroptosis and improve osteoarthritis.
Collapse
Affiliation(s)
- Zhimeng Zhang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Daotong Yuan
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ximin Jin
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenjie Chang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongkui Zhang
- Department of Orthopedic Surgery, Affliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenpeng Xie
- Department of Orthopedic Surgery, Affliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Zhu Z, Guan Y, Gao S, Guo F, Liu D, Zhang H. Impact of natural compounds on peroxisome proliferator-activated receptor: Molecular effects and its importance as a novel therapeutic target for neurological disorders. Eur J Med Chem 2025; 283:117170. [PMID: 39700874 DOI: 10.1016/j.ejmech.2024.117170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Neurological disorders refer to the pathological changes of the nervous system involving multiple pathological mechanisms characterized by complex pathogenesis and poor prognosis. Peroxisome proliferator-activated receptor (PPAR) is a ligand-activated transcription factor that is a member of the nuclear receptor superfamily. PPAR has attracted considerable attention in the past decades as one of the potential targets for the treatment of neurological disorders. Several in vivo and in vitro studies have confirmed that PPARs play a neuroprotective role by regulating multiple pathological mechanisms. Several selective PPAR ligands, such as thiazolidinediones and fibrates, have been approved as pharmacological agonists. Nevertheless, PPAR agonists cause a variety of adverse effects. Some natural PPAR agonists, including wogonin, bergenin, jujuboside A, asperosaponin VI, monascin, and magnolol, have been introduced as safe agonists, as evidenced by clinical or preclinical experiments. This review summarizes the effects of phytochemicals on PPAR receptors in treating various neurological disorders. Further, it summarizes recent advances in phytochemicals as potential, safe, and promising PPAR agonists to provide insights into understanding the PPAR-dependent and independent cascades mediated by phytochemicals. The phytochemicals exhibited potential for treating neurological disorders by inhibiting neuroinflammation, exerting anti-oxidative stress and anti-apoptotic activities, promoting autophagy, preventing demyelination, and reducing brain edema and neurotoxicity. This review presents data that will help clarify the potential mechanisms by which phytochemicals act as pharmacological agonists of PPARs in the treatment of neurological disorders. It also provides insights into developing new drugs, highlighting phytochemicals as potential, safe, and promising PPAR agonists. Additionally, this review aims to enhance understanding of both PPAR-dependent and independent pathways mediated by phytochemicals.
Collapse
Affiliation(s)
- Zhe Zhu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yadi Guan
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Songlan Gao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Feng Guo
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Dong Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Honglei Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
14
|
Yu S, Gan C, Li W, Zhang Q, Cai Y, Xu J, Huang R, Yao S, Cheng L, Cheng H. Depression decreases immunity and PD-L1 inhibitor efficacy via the hypothalamic-pituitary-adrenal (HPA) axis in triple-negative breast cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167581. [PMID: 39579799 DOI: 10.1016/j.bbadis.2024.167581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Depression weakens antitumor immunity, yet the underlying mechanisms linking depression and tumor growth remain unclear. This study examines the influence of depression on the hypothalamic-pituitary-adrenal (HPA) axis, immunological function, and effectiveness of immunotherapy in triple-negative breast cancer (TNBC) patients. METHODS A mouse model of comorbid TNBC and depression was established via chronic restraint stress (CRS) and 4T1 tumor transplantation. A programmed cell death ligand 1 (PD-L1) inhibitor was used to manage mice with TNBC, and the ability of metyrapone to reverse the immune system changes induced by HPA axis activation in depression was evaluated. Mouse peripheral blood was used to measure HPA axis activity, immune cell numbers and cytokine levels. RESULTS Depression activates the HPA axis, leading to increased levels of glucocorticoids. Depression led to an increase in the B-cell number and a reduction in the CD4+ T-cell and CD8+ T-cell numbers, without a statistically significant difference in the regulatory T (Treg) cell number. Furthermore, depression increased the levels of the cytokines interferon-gamma (IFN-γ), interleukin (IL)-1β, IL-4, IL-6, IL-8, and tumor necrosis factor (TNF)-α while decreasing the levels of IL-2 and IL-10. Similar results were observed in the context of PD-L1 inhibitor therapy. The depressed mice presented an increased tumor burden and a poor response to the PD-L1 inhibitor. The application of metyrapone during PD-L1 inhibitor treatment resulted in partial restoration of these depression-related alterations. CONCLUSIONS Depression reduces the effectiveness of PD-L1 inhibitors by altering the number of immune cells and the levels of cytokines through activation of the HPA axis. TRANSLATIONAL RELEVANCE Depression is common in breast cancer patients and is associated with reduced antitumor immunity. There is limited knowledge regarding the specific mechanisms through which depression impairs antitumor immunity. Immunotherapy, which promotes the restoration of antitumor immunity, represents a promising treatment strategy for TNBC patients. However, the efficacy of immunotherapy can be compromised by depressive symptoms and the administration of glucocorticoids during treatment. It is still uncertain whether increasing glucocorticoid levels can reduce the efficacy of immunotherapy in patients with depression. The potential benefits of combining immunotherapy with glucocorticoid inhibitors compared with immunotherapy alone need to be evaluated for TNBC patients with concurrent depressive symptoms. Therefore, further clarification of the specific mechanisms by which depression impairs antitumor immunity is needed to inform future optimization of immunotherapy strategies.
Collapse
Affiliation(s)
- Sheng Yu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450000, China; Department of Oncology, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Chen Gan
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Wen Li
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Qianqian Zhang
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Yinlian Cai
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Jian Xu
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Runze Huang
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Senbang Yao
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Ling Cheng
- Department of Intensive Care Unit, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China; Department of Oncology, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, 518000, Guangdong, People's Republic of China.
| | - Huaidong Cheng
- Shenzhen Clinical Medical School of Southern Medical University, China; Department of Oncology, Shenzhen Hospital of Southern Medical University, Shenzhen 518000, Guangdong, China.
| |
Collapse
|
15
|
Gao Z, Peng J, Zhang Y, Chen Z, Song R, Song Z, Feng Q, Sun M, Zhu H, Lu X, Yang R, Huang C. Hippocampal SENP3 mediates chronic stress-induced depression-like behaviors by impairing the CREB-BDNF signaling. Neuropharmacology 2025; 262:110203. [PMID: 39486575 DOI: 10.1016/j.neuropharm.2024.110203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Impaired signaling between cyclic adenosine monophosphate response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF) in the hippocampus is generally considered to be the cause of depression. The mechanisms underlying the impairment of CREB-BDNF signaling under stress conditions are largely unclear. Small ubiquitin-like modifier (SUMO) specific peptidase 3 (SENP3) is a molecule that can regulate SUMOylation of target proteins related to synaptic plasticity. Its dynamic changes have been reported to be associated with neuronal damage in various models of central nervous disorders such as cerebral ischemia and traumatic brain injury. However, its role in depression is completely unknown. This problem was addressed in the present study. Our results showed that chronic unpredictable stress (CUS) triggered a specific increase in SENP3 expression in the hippocampus of non-stressed mice. Overexpression of SENP3 in the hippocampus of non-stressed mice elicited depression-like behaviors in the tail suspension test, forced swimming test, and sucrose preference test, accompanied by impairment of the CREB-BDNF signaling cascade in the hippocampus. Conversely, genetic silencing of SENP3 in the hippocampus suppressed the development of depression-like behaviors. Furthermore, infusion of SENP3-shRNA into the hippocampus failed to suppress CUS-induced depression-like behaviors when mice received genetic silencing CREB or BDNF in the hippocampus or inhibition of the BDNF receptor by K252a. Taken together, these results suggest that abnormally elevated SENP3 in the hippocampus leads to the development of depression-like behavior by impairing the CREB-BDNF signaling cascade. SENP3 in the hippocampus could be a promising target for the development of new antidepressants.
Collapse
Affiliation(s)
- Zhiwei Gao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20 Xisi Road, Nantong 226001, Jiangsu Province, China
| | - Jie Peng
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Yi Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu Province, China
| | - Rongrong Song
- Department of Emergency and Critical Care Medicine, Tongzhou People's Hospital, 999 Jianshe Road, Nantong 226300, Jiangsu Province, China
| | - Ze Song
- Department of Neurosurgery, Tongzhou People's Hospital, 999 Jianshe Road, Nantong 226300, Jiangsu Province, China
| | - Qijie Feng
- Department of Orthopedics, Tongzhou People's Hospital, 999 Jianshe Road, Nantong 226300, Jiangsu Province, China
| | - Micona Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20 Xisi Road, Nantong 226001, Jiangsu Province, China.
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
16
|
Guo M, Pei WJ, Liu L, Chen K, Cheng Y, Piao XL. Neuroprotective effects of gypenosides on LPS-induced anxiety and depression-like behaviors. Int Immunopharmacol 2024; 143:113367. [PMID: 39413644 DOI: 10.1016/j.intimp.2024.113367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/25/2024] [Accepted: 10/06/2024] [Indexed: 10/18/2024]
Abstract
AIM Depression, a prevalent mental disorder, significantly impairs the quality of life and social functioning. Targeting neuroinflammation is a promising therapeutic approach, highlighting the need for natural neuroprotective agents. Gypenosides (Gyp) from Gynostemma pentaphyllum exhibit anxiolytic and antidepressant effects, yet the underlying mechanisms remain unclear. We investigated whether Gyp, isolated and purified by our laboratory, can exert neuroprotective effects by modulating neuroinflammation in the hippocampus and prefrontal cortex (PFC) of mice with LPS-induced anxiety and depression, thereby ameliorating behavioral phenotypes. METHODS LPS (1 mg/kg, i.p.) was used to induce anxiety and depression-like behaviors. Gyp was administered at 50, 100, or 200 mg/kg in pretreatment, with fluoxetine hydrochloride (Flu) as a positive control, for 10 consecutive days. RESULTS Gyp, especially at 100 mg/kg, significantly ameliorated LPS-induced anxiety and depression in mice, normalizing cytokine expression in the hippocampus and PFC, with IL-1β showing the most pronounced regulation (Hippocampus: RatioGyp-100/LPS = 30.73 %, PFC: RatioGyp-100/LPS = 55.89 %). Gyp also reversed LPS-induced neuronal loss and necrosis, reduced glial cell activation, and prevented the transition of microglia to the M1 phenotype. Mechanistically, Gyp suppressed the activation of the NLRP3 inflammasome in the PFC, and modulated hippocampal synaptic protein loss, thereby mediating neuroinflammation. CONCLUSIONS Gyp improved anxiety and depression in LPS-induced mice, which may be achieved by balancing systemic inflammatory levels, regulating glial cell activation and phenotypic polarization, regulating hippocampal synaptic plasticity, and suppressing the NLRP3/Caspase-1/ASC signaling pathway in the PFC.
Collapse
Affiliation(s)
- Mei Guo
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing 100081, China; School of Pharmacy, Minzu University of China, Beijing 100081, China; Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Wen-Jing Pei
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing 100081, China; School of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Liming Liu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China; Institute of National Security, Minzu University of China, Beijing 100081, China
| | - Kexuan Chen
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China.
| | - Xiang-Lan Piao
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing 100081, China; School of Pharmacy, Minzu University of China, Beijing 100081, China.
| |
Collapse
|
17
|
Madeshiya AK, Quintanilla B, Whitehead C, Tomlinson S, Pillai A. Systemic Administration of a Site-Targeted Complement Inhibitor Attenuates Chronic Stress-Induced Social Behavior Deficits and Neuroinflammation in Mice. Cells 2024; 13:1988. [PMID: 39682736 PMCID: PMC11640647 DOI: 10.3390/cells13231988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic stress, a risk factor for many neuropsychiatric conditions, causes dysregulation in the immune system in both humans and animal models. Additionally, inflammation and synapse loss have been associated with deficits in social behavior. The complement system, a key player of innate immunity, has been linked to social behavior impairments caused by chronic stress. However, it is not known whether complement inhibition can help prevent neuroinflammation and behavioral deficits caused by chronic stress. In this study, we investigated the potential of a site-targeted complement inhibitor to ameliorate chronic stress-induced changes in social behavior and inflammatory markers in the prefrontal cortex (PFC) and hippocampus. Specifically, we investigated the use of C2-Crry, which comprises a natural antibody-derived single-chain antibody (ScFv) targeting domain-designated C2, linked to Crry, a C3 activation inhibitor. The C2 targeting domain recognizes danger-associated molecular patterns consisting of a subset of phospholipids that become exposed following cell stress or injury. We found that systemic administration of C2-Crry attenuated chronic stress-induced social behavioral impairments in mice. Furthermore, C2-Crry administration significantly decreased microglia/macrophage and astrocyte activation markers in the PFC and hippocampus. These findings suggest that site-targeted complement inhibition could offer a promising, safe, and effective strategy for treating chronic stress induced behavioral and immune function disorders.
Collapse
Affiliation(s)
- Amit Kumar Madeshiya
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77054, USA; (A.K.M.); (C.W.)
| | - Brandi Quintanilla
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77054, USA; (A.K.M.); (C.W.)
| | - Carl Whitehead
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77054, USA; (A.K.M.); (C.W.)
| | - Stephen Tomlinson
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph Johnson VA Medical Center, Charleston, SC 29401, USA
| | - Anilkumar Pillai
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77054, USA; (A.K.M.); (C.W.)
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| |
Collapse
|
18
|
She K, Yuan N, Huang M, Zhu W, Tang M, Ma Q, Chen J. Emerging role of microglia in the developing dopaminergic system: perturbation by early life stress. Neural Regen Res 2024; 21:01300535-990000000-00587. [PMID: 39589170 DOI: 10.4103/nrr.nrr-d-24-00742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/15/2024] [Indexed: 11/27/2024] Open
Abstract
Early life stress correlates with a higher prevalence of neurological disorders, including autism, attention-deficit/hyperactivity disorder, schizophrenia, depression, and Parkinson's disease. These conditions, primarily involving abnormal development and damage of the dopaminergic system, pose significant public health challenges. Microglia, as the primary immune cells in the brain, are crucial in regulating neuronal circuit development and survival. From the embryonic stage to adulthood, microglia exhibit stage-specific gene expression profiles, transcriptome characteristics, and functional phenotypes, enhancing the susceptibility to early life stress. However, the role of microglia in mediating dopaminergic system disorders under early life stress conditions remains poorly understood. This review presents an up-to-date overview of preclinical studies elucidating the impact of early life stress on microglia, leading to dopaminergic system disorders, along with the underlying mechanisms and therapeutic potential for neurodegenerative and neurodevelopmental conditions. Impaired microglial activity damages dopaminergic neurons by diminishing neurotrophic support (e.g., insulin-like growth factor-1) and hinders dopaminergic axon growth through defective phagocytosis and synaptic pruning. Furthermore, blunted microglial immunoreactivity suppresses striatal dopaminergic circuit development and reduces neuronal transmission. Furthermore, inflammation and oxidative stress induced by activated microglia can directly damage dopaminergic neurons, inhibiting dopamine synthesis, reuptake, and receptor activity. Enhanced microglial phagocytosis inhibits dopamine axon extension. These long-lasting effects of microglial perturbations may be driven by early life stress-induced epigenetic reprogramming of microglia. Indirectly, early life stress may influence microglial function through various pathways, such as astrocytic activation, the hypothalamic-pituitary-adrenal axis, the gut-brain axis, and maternal immune signaling. Finally, various therapeutic strategies and molecular mechanisms for targeting microglia to restore the dopaminergic system were summarized and discussed. These strategies include classical antidepressants and antipsychotics, antibiotics and anti-inflammatory agents, and herbal-derived medicine. Further investigations combining pharmacological interventions and genetic strategies are essential to elucidate the causal role of microglial phenotypic and functional perturbations in the dopaminergic system disrupted by early life stress.
Collapse
Affiliation(s)
- Kaijie She
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Naijun Yuan
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- Shenzhen People's Hospital, The 2 Clinical Medical College, Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Shenzhen, Guangdong Province, China
| | - Minyi Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Wenjun Zhu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Manshi Tang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong Province, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
19
|
Wang Q, Zeng F, Fang C, Sun Y, Zhao X, Rong X, Zhang H, Xu Y. Galectin-3 alleviates demyelination by modulating microglial anti-inflammatory polarization through PPARγ-CD36 axis. Brain Res 2024; 1842:149106. [PMID: 38986827 DOI: 10.1016/j.brainres.2024.149106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024]
Abstract
Demyelination is characterized by disruption of myelin sheath and disorders in myelin formation. Currently, there are no effective therapeutic treatments available. Microglia, especially anti-inflammatory phenotype microglia are critical for remyelination. Galectin-3 (Gal-3), which is known to modulate microglia activation, is correlated with myelination. In this study, we aimed to elucidate the roles of Gal-3 during myelin formation and explore the efficiency and mechanism of rGal-3 administration in remyelination. We enrolled Gal-3 knockout (Lgals3 KO) mice and demonstrated Lgals3 KO causes demyelination during spontaneous myelinogenesis. We performed a cuprizone (CPZ) intoxication model and found Lgals3 KO aggravates demyelinated lesions and favors microglial pro-inflammatory phenotype polarization. Recombinant Gal-3 (rGal-3) administration alleviates CPZ intoxication and drives microglial towards anti-inflammatory phenotype. Additionally, RNA sequencing results reveal the correlation between Gal-3 and the PPARγ-CD36 axis. Thus, we performed SSO and GW9662 administration to inhibit the activation of the PPARγ-CD36 axis and found that rGal-3 administration modulates microglial phenotype polarization by regulating the PPARγ-CD36 axis. Together, our findings highlight the importance of Gal-3 in myelination and provide insights into rGal-3 administration for modulating microglial anti-inflammatory phenotype polarization through the PPARγ-CD36 axis.
Collapse
Affiliation(s)
- Qian Wang
- Department of Infectious and Liver Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China; Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China.
| | - Fansen Zeng
- Department of Infectious and Liver Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Chunxiao Fang
- Department of Infectious and Liver Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Yi Sun
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Xiaopeng Zhao
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Xiao Rong
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China
| | - Huayan Zhang
- Department of Neonatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China.
| | - Yi Xu
- Department of Infectious and Liver Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, PR China.
| |
Collapse
|
20
|
Spanoudaki M, Chrysafi M, Papadopoulou SK, Tsourouflis G, Pritsa A, Giaginis C. Naturally Occurring Compounds Targeting Peroxisome Proliferator Receptors: Potential Molecular Mechanisms and Future Perspectives for Promoting Human Health. APPLIED SCIENCES 2024; 14:9994. [DOI: 10.3390/app14219994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background: Peroxisome-proliferator-activated receptors (PPARs) constitute nuclear transcription factors controlling gene expression associated with cell growth and proliferation, diverse proteins, lipids, and glucose metabolism, being related to several other pathophysiological states such as metabolic disorders, atherogenesis, carcinogenesis, etc. The present survey aims to analyze the natural compounds that can act as agonists for the PPAR-α, PPAR-β/δ, and PPAR-γ system targeting, highlighting how the amazing biochemical diversity of natural compounds can yield new insights into this “hotspot” of the scientific field. Methods: A narrative review was performed by searching the recent international literature for the last two decades in the most authoritative scientific databases, like PubMed, Scopus, Web of Science, and Embase, using appropriate keywords. Results: Several natural compounds and/or their synthetic derivatives can act as ligands of PPARs, stimulating their transcriptional activity and enabling their use as preventive and/or therapeutic agents for several disease states, such as inflammation, oxidative stress, metabolic disturbances, atherogenesis, and carcinogenesis. Although synthetic compounds are increasingly used as drugs to manage health problems, serious side effects have been observed, while their natural analogues exhibit only few minor side effects. Conclusions: Further clinical studies on natural compounds such as ligands of PPARs and the evaluation of the related molecular mechanisms are needed to implement an effective strategy concerning the pharmaco-technology, food chemistry, and nutrition to introduce them as part of clinical and dietary practice.
Collapse
Affiliation(s)
- Maria Spanoudaki
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece
- 424 General Military Hospital of Thessaloniki, 54621 Thessaloniki, Greece
| | - Maria Chrysafi
- Department of Food Science and Nutrition, School of Environment, University of Aegean, 81400 Lemnos, Greece
| | - Sousana K. Papadopoulou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| | - Gerasimos Tsourouflis
- Second Department of Propedeutic Surgery, Medical School, University of Athens, 11527 Athens, Greece
| | - Agathi Pritsa
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| | - Constantinos Giaginis
- Department of Food Science and Nutrition, School of Environment, University of Aegean, 81400 Lemnos, Greece
| |
Collapse
|
21
|
Yu W, Lv Y, Xuan R, Han P, Xu H, Ma X. Human placental mesenchymal stem cells transplantation repairs the alveolar epithelial barrier to alleviate lipopolysaccharides-induced acute lung injury. Biochem Pharmacol 2024; 229:116547. [PMID: 39306309 DOI: 10.1016/j.bcp.2024.116547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/18/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are accompanied by high mortality rates and few effective treatments. Transplantation of human placental mesenchymal stem cells (hPMSCs) may attenuate ALI and the mechanism is still unclear. Our study aimed to elucidate the potential protective effect and therapeutic mechanism of hPMSCs against lipopolysaccharide (LPS)-induced ALI, An ALI model was induced by tracheal instillation of LPS into wild-type (WT) and angiotensin-converting enzyme 2 (ACE2) knockout (KO) male mice, followed by injection of hPMSCs by tail vein. Treatment with hPMSCs improved pulmonary histopathological injury, reduced pulmonary injury scores, decreased leukocyte count and protein levels in bronchoalveolar lavage fluid(BALF), protected the damaged alveolar epithelial barrier, and reversed LPS-induced upregulation of pro-inflammatory factors Interleukin-6 (IL-6) and Tumor necrosis factor-α(TNF-α) and downregulation of anti-inflammatory factor Interleukin-6(IL-10) in BALF. Moreover, administration of hPMSCs inhibited Angiotensin (Ang)II activation and promoted the expression levels of ACE2 and Ang (1-7) in ALI mice. Pathological damage, inflammation levels, and disruption of alveolar epithelial barrier in ALI mice were elevated after the deletion of ACE2 gene, and the Renin angiotensin system (RAS) imbalance was exacerbated. The therapeutic effect of hPMSCs was significantly reduced in ACE2 KO mice. Our findings suggest that ACE2 plays a key role in hPMSCs repairing the alveolar epithelial barrier to protect against ALI, laying a new foundation for the clinical treatment of ALI.
Collapse
Affiliation(s)
- Wenqin Yu
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Ningxia Institute of Human Stem Cells, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Yuzhen Lv
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Ningxia Institute of Human Stem Cells, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Ruirui Xuan
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Peipei Han
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Haihuan Xu
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Ningxia Institute of Human Stem Cells, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China
| | - Xiaowei Ma
- Clinical Medical College of Ningxia Medical University, Yinchuan Province 750004, China; Intensive Care Unit, Cardiocerebral Vascular Disease Hospital of General Hospital of Ningxia Medical University, Yinchuan Province 750002, China.
| |
Collapse
|
22
|
Liu YE, Zhao Z, He H, Li L, Xiao C, Zhou T, You Z, Zhang J. Stress-induced obesity in mice causes cognitive decline associated with inhibition of hippocampal neurogenesis and dysfunctional gut microbiota. Front Microbiol 2024; 15:1381423. [PMID: 39539712 PMCID: PMC11557545 DOI: 10.3389/fmicb.2024.1381423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Effects of stress on obesity have been thoroughly studied in high-fat diet fed mice, but not in normal diet fed mice, which is important to clarify because even on a normal diet, some individuals will become obese under stress conditions. Here we compared mice that showed substantial weight gain or loss under chronic mild stress while on a normal diet; we compared the two groups in terms of cognitive function, hypothalamic-pituitary-adrenal signaling, neurogenesis and activation of microglia in hippocampus, gene expression and composition of the gut microbiome. Chronic mild stress induced diet-independent obesity in approximately 20% of animals, and it involved inflammatory responses in peripheral and central nervous system as well as hyperactivation of the hypothalamic-pituitary-adrenal signaling and of microglia in the hippocampus, which were associated with cognitive deficits and impaired hippocampal neurogenesis. It significantly increased in relative abundance at the phylum level (Firmicutes), at the family level (Prevotellaceae ucg - 001 and Lachnospiraceae NK4a136), at the genus level (Dubosiella and Turicibacter) for some enteric flora, while reducing the relative abundance at the family level (Lactobacillaceae and Erysipelotrichaceae), at the genus level (Bacteroidota, Alistipes, Alloprevotella, Bifidobacterium and Desulfovibrio) for some enteric flora. These results suggest that stress, independently of diet, can induce obesity and cognitive decline that involve dysfunctional gut microbiota. These insights imply that mitigation of hypothalamic-pituitary-adrenal signaling and microglial activation as well as remodeling of gut microbiota may reverse stress-induced obesity and associated cognitive decline.
Collapse
Affiliation(s)
- Yu-e Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhihuang Zhao
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Haili He
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Liangyuan Li
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Chenghong Xiao
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Tao Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zili You
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
23
|
Qiao J, Zhong C, Zhang Q, Yang G, Li S, Jin J. ASA VI controls osteoarthritis in mice by maintaining mitochondrial homeostasis through Sirtuin 3. Int Immunopharmacol 2024; 140:112858. [PMID: 39111145 DOI: 10.1016/j.intimp.2024.112858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
OBJECTIVE The aim of this study was to investigate whether ASA VI controls osteoarthritis (OA) by regulating mitochondrial function. METHODS Primary chondrocytes were isolated and cultured from rat knee joints. The chondrocytes were treated with ASA VI and interleukin-1β (IL-1β) to simulate the inflammatory environment of OA. Cell viability, apoptosis, inflammatory cytokine levels, and extracellular matrix (ECM) component levels were assessed. Mitochondrial function, including ATP levels, mitochondrial membrane potential, reactive oxygen species (ROS) levels, and mitochondrial DNA content, was evaluated. The expression of Sirtuin 3 (Sirt3), a key regulator of mitochondrial homeostasis, was examined. Additionally, a rat OA model was established by destabilizing the medial meniscus, and the effects of ASA VI on cartilage degeneration were assessed. RESULTS ASA VI treatment improved cell viability, reduced apoptosis, and decreased IL-6 and TNF-α levels in IL-1β-induced chondrocytes. ASA VI also upregulated Collagen II and Aggrecan expression, while downregulating ADAMTS5 and MMP-13 expression. Furthermore, ASA VI mitigated IL-1β-induced mitochondrial dysfunction by increasing ATP levels, restoring mitochondrial membrane potential, reducing ROS production, and preserving mitochondrial DNA content. These effects were accompanied by the activation of Sirt3. In the rat OA model, ASA VI treatment increased Sirt3 expression and alleviated cartilage degeneration. CONCLUSION ASA VI exerts chondroprotective and anti-inflammatory effects on IL-1β-induced chondrocytes by improving mitochondrial function through Sirt3 activation. ASA VI also attenuates cartilage degeneration in a rat OA model. These findings suggest that ASA VI may be a potential therapeutic agent for the treatment of osteoarthritis by targeting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jie Qiao
- Department of Orthopedics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China; Hubei Sizhen Laboratory, Wuhan, Hubei, China; Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Chuanqi Zhong
- Department of Orthopedics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China; Hubei Sizhen Laboratory, Wuhan, Hubei, China; Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Qing Zhang
- Department of Orthopedics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China; Hubei Sizhen Laboratory, Wuhan, Hubei, China; Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Gongxu Yang
- Department of Orthopedics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China; Hubei Sizhen Laboratory, Wuhan, Hubei, China; Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Shuying Li
- School of Acupuncture and Orthopedics, Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Jun Jin
- Department of Orthopedics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China; Hubei Sizhen Laboratory, Wuhan, Hubei, China; Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China.
| |
Collapse
|
24
|
Zhao H, Liang K, Yu Z, Wen Y, Shi J, Zhang T, Yu X, Zu X, Fang Y. Hyperbaric oxygen preconditioning rescues prolonged underwater exercise-induced hippocampal dysfunction by regulating microglia activation and polarization. Neurosci Res 2024; 207:26-36. [PMID: 38848903 DOI: 10.1016/j.neures.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
Underwater exercise is becoming increasingly prevalent, during which brain function is necessary but is also at risk. However, no study has explored how prolonged exercise affect the brain in underwater environment. Previous studies have indicated that excessive exercise in common environment causes brain dysfunction but have failed to provide appropriate interventions. Numerous evidence has indicated the neuroprotective effect of hyperbaric oxygen preconditioning (HBO-PC). The objective of this study was to investigate the cognitive effect of prolonged underwater exercise (PUE) and to explore the potential neuroprotective effect of HBO-PC in underwater environment. Rats swimming for 3 h in a simulated hyperbaric chamber (2.0 ATA) was used to establish the PUE animal model and HBO-PC (2.5 ATA for 1, 3,5 times respectively) was administrated before PUE. The results demonstrated that PUE triggers anxiety-like behaviors, cognitive impairment accompanied by hippocampal dysfunction, microglia activation and neuroinflammation. Conversely, 3 HBO-PC rescued anxiety-like behaviors and cognitive impairment. Mechanistically, 3 HBO-PC reduced microglia activation and switched the activated microglia from a pro-inflammatory to neuroprotective phenotype. These findings illustrated that PUE induces anxiety-like behaviors and cognitive impairment and HBO-PC of proper frequency may provide an appropriate and less invasive intervention for protecting the brain in underwater exercise.
Collapse
Affiliation(s)
- Houyu Zhao
- Department of Diving and Hyperbaric Medicine, Navy Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Kun Liang
- Department of Diving and Hyperbaric Medicine, Navy Special Medical Center, Naval Medical University, Shanghai 200433, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200433, China
| | - Zeyuan Yu
- Department of Diving and Hyperbaric Medicine, Navy Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Yukun Wen
- Department of Diving and Hyperbaric Medicine, Navy Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Jin Shi
- Department of Diving and Hyperbaric Medicine, Navy Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Tingting Zhang
- Department of Diving and Hyperbaric Medicine, Navy Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Xuhua Yu
- Department of Diving and Hyperbaric Medicine, Navy Special Medical Center, Naval Medical University, Shanghai 200433, China
| | - Xianpeng Zu
- School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Yiqun Fang
- Department of Diving and Hyperbaric Medicine, Navy Special Medical Center, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
25
|
He H, Zhang X, He H, Xu G, Li L, Yang C, Liu Y, You Z, Zhang J. Microglial priming by IFN-γ involves STAT1-mediated activation of the NLRP3 inflammasome. CNS Neurosci Ther 2024; 30:e70061. [PMID: 39392762 PMCID: PMC11468839 DOI: 10.1111/cns.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Inflammatory and immune responses in the brain that contribute to various neuropsychiatric disorders may begin as microglial "priming". Interferon (IFN)-γ is known to cause microglial priming, but the mechanism is unclear. METHODS We examined the effects of IFN-γ on gene expression, microglial activation, inflammatory and immune responses and activity of the NLRP3 inflammasome in primary microglia and in the brains of mice. RESULTS Our results showed that treating microglial cultures with IFN-γ induced a hedgehog-like morphology and upregulated markers of microglial activation (CD86, CD11b) and pro-inflammatory molecules (IL-1β, IL-6, TNF-α, iNOS), while downregulating markers of microglial homeostasis (CX3CR1, CD200R1), anti-inflammatory molecules (MCR1, Arg-1) and neurotrophic factors (IGF-1, BDNF). IFN-γ also upregulated markers of NLRP3 inflammasome activation (NLRP3, caspase-1, gasdermin D, IL-18). This particular transcriptional profiling makes IFN-γ-primed microglia with exaggerated responses upon lipopolysaccharide (LPS) stimulation. The level of NLRP3, caspase-1, gasdermin D, IL-1β, IL-18, TNF-α and iNOS in microglia cultures treated with both IFN-γ and LPS were highest than with either one alone. Injecting IFN-γ into the lateral ventricle of mice induced similar morphological and functional changes in hippocampal microglia as in primary microglial cultures. The effects of IFN-γ on NLRP3 inflammasome and microglia from cultures or hippocampus were abolished when STAT1 was inhibited using fludarabin. Injecting mice with IFN-γ alone or together with LPS induced anxiety- and depression-like behaviors and impaired hippocampus-dependent spatial memory; these effects were mitigated by fludarabin. CONCLUSIONS IFN-γ primes microglia by activating STAT1, which upregulates genes that activate the NLRP3 inflammasome. Inhibiting the IFN-γ/STAT1 axis may be a way to treat neurodegenerative diseases and psychiatric disorders that involve microglial priming.
Collapse
Affiliation(s)
- Haili He
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese MedicineGuiyangChina
| | - Xiaomei Zhang
- School of Life Science and Technology, Center for Informational BiologyUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Hui He
- School of Life Science and Technology, Center for Informational BiologyUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Gaojie Xu
- School of Life Science and Technology, Center for Informational BiologyUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Liangyuan Li
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese MedicineGuiyangChina
| | - Chengyan Yang
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese MedicineGuiyangChina
| | - Yu‐e Liu
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese MedicineGuiyangChina
| | - Zili You
- School of Life Science and Technology, Center for Informational BiologyUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Jinqiang Zhang
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese MedicineGuiyangChina
| |
Collapse
|
26
|
Yang C, Jiang W, Su D, Yang C, Yuan Q, Kang C, Xiao C, Wang L, Peng C, Zhou T, Zhang J. Contamination of the traditional medicine Radix Dipsaci with aflatoxin B1 impairs hippocampal neurogenesis and cognitive function in a mouse model of osteoporosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116831. [PMID: 39151374 DOI: 10.1016/j.ecoenv.2024.116831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/16/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Aflatoxin B1, which can penetrate the blood-brain barrier and kill neural cells, can contaminate traditional herbal medicines, posing a significant risk to human health. The present study examined cellular, cognitive and behavioral consequences of aflatoxin B1 contamination of the anti-osteoporotic medicine Radix Dipsaci. METHODS A mouse model of osteoporosis was created by treating the animals with all-trans-retinoic acid. Then the animals were treated intragastically with water decoctions of Radix Dipsaci that contained detectable aflatoxin B1 or not. The animals were compared in terms of mineral density and mineral salt content of bone, production of pro-inflammatory factors, neurogenesis and microglial activation in hippocampus, as well as behavior and cognitive function. RESULTS Contamination of Radix Dipsaci with aflatoxin B1 significantly reduced the medicine's content of bioactive saponins. It destroyed the ability of the herbal decoction to improve mineral density and mineral salt content in the bones of diseased mice, and it induced the production of the oxidative stress marker malondialdehyde as well as the pro-inflammatory cytokines interleukin-1β and tumor necrosis factor-α. Aflatoxin B1 contamination inhibited formation of new neurons and increased the proportion of activated microglia in the hippocampus. These neurological changes were associated with anhedonia, behavioral despair, and deficits in short-term memory and social memory. CONCLUSION Contamination of Radix Dipsaci with aflatoxin B1 not only eliminates the herbal decoction's anti-osteoporotic effects, but it also induces neurotoxicity that can lead to cognitive decline and behavioral abnormalities. Such contamination should be avoided through tightly regulated production and quality control of medicinal herbs.
Collapse
Affiliation(s)
- Chengyan Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Weike Jiang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Dapeng Su
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Changgui Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Qingsong Yuan
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Chuanzhi Kang
- Resource Institute for Chinese and Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Chenghong Xiao
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Lulu Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Cheng Peng
- Chengdu University of Traditional Chinese Medicine.
| | - Tao Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
27
|
Li Z, Liang S, Cui X, Shen C, Xu Z, Chen W, Wu M, Liang C, Liu J, Huang J, Li W. Network pharmacology- and molecular docking-based investigation on the mechanism of action of Si-ni San in the treatment of depression combined with anxiety and experimental verification in adolescent rats. Front Psychiatry 2024; 15:1414242. [PMID: 39247617 PMCID: PMC11378754 DOI: 10.3389/fpsyt.2024.1414242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/02/2024] [Indexed: 09/10/2024] Open
Abstract
Background The incidence rate of adolescent depression and anxiety has been increasing since the outbreak of COVID-19, which there are no effective therapeutic drugs available. Si-ni San is commonly used in traditional Chinese medicine for the treatment of depression-like as well as anxiety-like behavior, but its mechanism for treating depression combined with anxiety during adolescence is not yet clear. Methods Network pharmacology was used to explore potential drug molecules and related targets, molecular docking and molecular dynamics (MD) simulation were used to evaluate the interaction between the potential drug molecules and related targets, and a model of anxiety combined with depression in adolescent rats as well as the following behavioral tests and molecular biology tests were used to verify the results from network pharmacology and molecular docking. Results As a result, 256 active ingredients of Si-ni San and 1128 potential targets were screened out. Among them, quercetin, Luteolin, kaempferol, 7-Methoxy-2-methyl isoflavone, formononetin showed to be the most potential ingredients; while STAT3, IL6, TNF, AKT1, AKT1, TP53, IL1B, MAPK3, VEGFA, CASP3, MMP9 showed to be the most potential targets. AGE-RAGE signaling pathway in diabetic complications, IL-17 signaling pathway, HIF-1 signaling pathway, PI3K-Akt signaling pathway and TNF signaling pathway, which are involved in anti-inflammation processes, showed to be the most probable pathways regulated by Si-ni San. Molecular docking and MD simulation between the compounds to inflammation-associated targets revealed good binding abilities of quercetin, Luteolin, kaempferol, nobiletin and formononetin to PTGS2 and PPARγ. In the experiment with adolescent rats, Si-ni San markedly suppressed early maternal separation (MS) combined with adolescent chronic unpredictable mild stress (CUMS)-induced depression combined with anxiety. The qPCR results further indicated that Si-ni San regulated the oxidative stress and inflammatory response. Conclusion This study demonstrates that adolescent anxiety- and depression-like behavior induced by MS combined CUMS can be ameliorated by Si-ni San by improved inflammation in hippocampus via targeting TNF pathway and Nrf2 pathway, helping to reveal the mechanism of Si-ni San in treating adolescent depression combined with anxiety.
Collapse
Affiliation(s)
- Zhiping Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shimin Liang
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xulan Cui
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chongkun Shen
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zaibin Xu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Chen
- Rehabilitation Center Massage Clinic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mingan Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chao Liang
- Deparment of Acupuncture, Haikou Hospital of Traditional Chinese Medicine, Haikou, China
| | - Jinman Liu
- Department of Encephalopathy, Affiliated Jiangmen Traditional Chinese Medicine (TCM) Hospital of Ji'nan University, Jiangmen, China
| | - Jiawen Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
28
|
He H, Zhang X, He H, Xiao C, Xu G, Li L, Liu YE, Yang C, Zhou T, You Z, Zhang J. Priming of hippocampal microglia by IFN-γ/STAT1 pathway impairs social memory in mice. Int Immunopharmacol 2024; 134:112191. [PMID: 38759369 DOI: 10.1016/j.intimp.2024.112191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/29/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024]
Abstract
Social behavior is inextricably linked to the immune system. Although IFN-γ is known to be involved in social behavior, yet whether and how it encodes social memory remains unclear. In the current study, we injected with IFN-γ into the lateral ventricle of male C57BL/6J mice, and three-chamber social test was used to examine the effects of IFN-γ on their social preference and social memory. The morphology of microglia in the hippocampus, prelimbic cortex and amygdala was examined using immunohistochemistry, and the phenotype of microglia were examined using immunohistochemistry and enzyme-linked immunosorbent assays. The IFN-γ-injected mice were treated with lipopolysaccharide, and effects of IFN-γ on behavior and microglial responses were evaluated. STAT1 pathway and microglia-neuron interactions were examined in vivo or in vitro using western blotting and immunohistochemistry. Finally, we use STAT1 inhibitor or minocycline to evaluated the role of STAT1 in mediating the microglial priming and effects of primed microglia in IFN-γ-induced social dysfunction. We demonstrated that 500 ng of IFN-γ injection results in significant decrease in social index and social novelty recognition index, and induces microglial priming in hippocampus, characterized by enlarged cell bodies, shortened branches, increased expression of CD68, CD86, CD74, CD11b, CD11c, CD47, IL-33, IL-1β, IL-6 and iNOS, and decreased expression of MCR1, Arg-1, IGF-1 and BDNF. This microglia subpopulation is more sensitive to LPS challenge, which characterized by more significant morphological changes and inflammatory responses, as well as induced increased sickness behaviors in mice. IFN-γ upregulated pSTAT1 and STAT1 and promoted the nuclear translocation of STAT1 in the hippocampal microglia and in the primary microglia. Giving minocycline or STAT1 inhibitor fludarabin blocked the priming of hippocampal microglia induced by IFN-γ, ameliorated the dysfunction in hippocampal microglia-neuron interactions and synapse pruning by microglia, thereby improving social memory deficits in IFN-γ injected mice. IFN-γ initiates STAT1 pathway to induce priming of hippocampal microglia, thereby disrupts hippocampal microglia-neuron interactions and neural circuit link to social memory. Blocking STAT1 pathway or inhibiting microglial priming may be strategies to reduce the effects of IFN-γ on social behavior.
Collapse
Affiliation(s)
- Haili He
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xiaomei Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hui He
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Chenghong Xiao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Gaojie Xu
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Liangyuan Li
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Yu-E Liu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chengyan Yang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Tao Zhou
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Zili You
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Jinqiang Zhang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
29
|
Ma Y, Zhao Y, Luo M, Jiang Q, Liu S, Jia Q, Bai Z, Wu F, Xie J. Advancements and challenges in pharmacokinetic and pharmacodynamic research on the traditional Chinese medicine saponins: a comprehensive review. Front Pharmacol 2024; 15:1393409. [PMID: 38774213 PMCID: PMC11106373 DOI: 10.3389/fphar.2024.1393409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/12/2024] [Indexed: 05/24/2024] Open
Abstract
Recent research on traditional Chinese medicine (TCM) saponin pharmacokinetics has revealed transformative breakthroughs and challenges. The multicomponent nature of TCM makes it difficult to select representative indicators for pharmacokinetic studies. The clinical application of saponins is limited by their low bioavailability and short half-life, resulting in fluctuating plasma concentrations. Future directions should focus on novel saponin compounds utilizing colon-specific delivery and osmotic pump systems to enhance oral bioavailability. Optimizing drug combinations, such as ginsenosides with aspirin, shows therapeutic potential. Rigorous clinical validation is essential for practical applications. This review emphasizes a transformative era in saponin research, highlighting the need for clinical validation. TCM saponin pharmacokinetics, guided by traditional principles, are in development, utilizing multidisciplinary approaches for a comprehensive understanding. This research provides a theoretical basis for new clinical drugs and supports rational clinical medication.
Collapse
Affiliation(s)
- Yuhan Ma
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yongxia Zhao
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Mingxia Luo
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qin Jiang
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Sha Liu
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qi Jia
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Zhixun Bai
- Organ Transplant Center, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Faming Wu
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jian Xie
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| |
Collapse
|
30
|
Liu GX, Li ZL, Lin SY, Luo ZY, Yin YN, Zhou YL, Ning YP. NEFA can serve as good biological markers for the diagnosis of depression in adolescents. J Affect Disord 2024; 352:342-348. [PMID: 38364978 DOI: 10.1016/j.jad.2024.01.274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 01/13/2024] [Accepted: 01/31/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND The incidence of adolescent depression has markedly risen in recent years, with a high recurrence rate into adulthood. Diagnosis in adolescents is challenging due to subjective factors, highlighting the crucial need for objective diagnostic markers. METHODS Our study enrolled 204 participants, including healthy controls (n = 88) and first-episode adolescent depression patients (n = 116). Serum samples underwent gas chromatography-mass spectrometry (GC-MS) analysis to assess non-esterified fatty acids (NEFA) expression. Machine learning and ROC analysis were employed to identify potential biomarkers, followed by bioinformatics analysis to explore underlying mechanisms. RESULTS Nearly all differentially expressed NEFA exhibited significant downregulation. Notably, nonanoic acid, cis-10-pentadecenoic acid, cis-10-carboenoic acid, and cis-11-eicosenoic acid demonstrated excellent performance in distinguishing adolescent depression patients. Metabolite-gene interaction analysis revealed these NEFAs interacted with multiple genes. KEGG pathway analysis on these genes suggested that differentially expressed NEFA may impact PPAR and cAMP signaling pathways. LIMITATIONS Inclusion of diverse populations for evaluation is warranted. Biomarkers identified in this study require samples that are more in line with the experimental design for external validation, and further basic research is necessary to validate the potential depressive mechanisms of NEFA. CONCLUSIONS The overall reduction in NEFA expression in first-episode adolescent depression patients suggests a potential mediation of depression symptoms through cAMP and PPAR signaling pathways. NEFA levels show promise as a diagnostic tool for identifying first-episode adolescent depression patients.
Collapse
Affiliation(s)
- Guan-Xi Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Ze-Lin Li
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Su-Yan Lin
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zheng-Yi Luo
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Ya-Nan Yin
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Yan-Ling Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| | - Yu-Ping Ning
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
31
|
Bu Y, Yang S, Wang D, Hu S, Zhang Q, Wu Z, Yang C. Role of soluble epoxide hydrolase in pain and depression comorbidity. Neurobiol Dis 2024; 193:106443. [PMID: 38395315 DOI: 10.1016/j.nbd.2024.106443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024] Open
Abstract
The coexistence of chronic pain and depression in clinical practice places a substantial social burden and profoundly impacts in patients. Although a clear correlation exists, the underlying mechanism of comorbidity between chronic pain and depression remains elusive. Research conducted in recent decades has uncovered that soluble epoxide hydrolase, a pivotal enzyme in the metabolism of polyunsaturated fatty acids, plays a crucial role in inflammation. Interestingly, this enzyme is intricately linked to the development of both pain and depression. With this understanding, this review aims to summarize the roles of soluble epoxide hydrolase in pain, depression, and their comorbidity. Simultaneously, we will also explore the underlying mechanisms, providing guidance for future research and drug development.
Collapse
Affiliation(s)
- Yuchen Bu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qi Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
32
|
Gan H, Ma Q, Hao W, Yang N, Chen ZS, Deng L, Chen J. Targeting autophagy to counteract neuroinflammation: A novel antidepressant strategy. Pharmacol Res 2024; 202:107112. [PMID: 38403256 DOI: 10.1016/j.phrs.2024.107112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Depression is a common disease that affects physical and mental health and imposes a considerable burden on afflicted individuals and their families worldwide. Depression is associated with a high rate of disability and suicide. It causes a severe decline in productivity and quality of life. Unfortunately, the pathophysiological mechanisms underlying depression have not been fully elucidated, and the risk of its treatment is still presented. Studies have shown that the expression of autophagic markers in the brain and peripheral inflammatory mediators are dysregulated in depression. Autophagy-related genes regulate the level of autophagy and change the inflammatory response in depression. Depression is related to several aspects of immunity. The regulation of the immune system and inflammation by autophagy may lead to the development or deterioration of mental disorders. This review highlights the role of autophagy and neuroinflammation in the pathophysiology of depression, sumaries the autophagy-targeting small moleculars, and discusses a novel therapeutic strategy based on anti-inflammatory mechanisms that target autophagy to treat the disease.
Collapse
Affiliation(s)
- Hua Gan
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Qingyu Ma
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Wenzhi Hao
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Nating Yang
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Lijuan Deng
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Jiaxu Chen
- Guangzhou Key Laboratory of Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
33
|
He H, He H, Mo L, Yuan Q, Xiao C, Ma Q, Yi S, Zhou T, You Z, Zhang J. Gut microbiota regulate stress resistance by influencing microglia-neuron interactions in the hippocampus. Brain Behav Immun Health 2024; 36:100729. [PMID: 38317780 PMCID: PMC10840119 DOI: 10.1016/j.bbih.2024.100729] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Communication among the brain, gut and microbiota in the gut is known to affect the susceptibility to stress, but the mechanisms involved are unclear. Here we demonstrated that stress resistance in mice was associated with more abundant Lactobacillus and Akkermansia in the gut, but less abundant Bacteroides, Alloprevotella, Helicobacter, Lachnoclostridium, Blautia, Roseburia, Colidextibacter and Lachnospiraceae NK4A136. Stress-sensitive animals showed higher permeability and stronger immune responses in their colon, as well as higher levels of pro-inflammatory cytokines in serum. Their hippocampus also showed more extensive microglial activation, abnormal interactions between microglia and neurons, and lower synaptic plasticity. Transplanting fecal microbiota from stress-sensitive mice into naïve ones perturbed microglia-neuron interactions and impaired synaptic plasticity in the hippocampus, translating to more depression-like behavior after stress exposure. Conversely, transplanting fecal microbiota from stress-resistant mice into naïve ones protected microglia from activation and preserved synaptic plasticity in the hippocampus, leading to less depression-like behavior after stress exposure. These results suggested that gut microbiota may influence resilience to chronic psychological stress by regulating microglia-neuron interactions in the hippocampus.
Collapse
Affiliation(s)
- Haili He
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Hui He
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Li Mo
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Qingsong Yuan
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Chenghong Xiao
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Qiman Ma
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Saini Yi
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Tao Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Zili You
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| |
Collapse
|
34
|
Zhang H, Xie X, Xu S, Wang C, Sun S, Song X, Li R, Li N, Feng Y, Duan H, Li D, Liu Z. Oligodendrocyte-derived exosomes-containing SIRT2 ameliorates depressive-like behaviors and restores hippocampal neurogenesis and synaptic plasticity via the AKT/GSK-3β pathway in depressed mice. CNS Neurosci Ther 2024; 30:e14661. [PMID: 38439616 PMCID: PMC10912796 DOI: 10.1111/cns.14661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
AIMS To investigate the antidepressant role of oligodendrocyte-derived exosomes (ODEXs)-containing sirtuin 2 (SIRT2) and the underlying mechanism both in vivo and in vitro. METHODS Oligodendrocyte-derived exosomes isolated from mouse serum were administered to mice with chronic unpredictable mild stress (CUMS)-induced depression via the tail vein. The antidepressant effects of ODEXs were assessed through behavioral tests and quantification of alterations in hippocampal neuroplasticity. The role of SIRT2 was confirmed using the selective inhibitor AK-7. Neural stem/progenitor cells (NSPCs) were used to further validate the impact of overexpressed SIRT2 and ODEXs on neurogenesis and synapse formation in vitro. RESULTS Oligodendrocyte-derived exosome treatment alleviated depressive-like behaviors and restored neurogenesis and synaptic plasticity in CUMS mice. SIRT2 was enriched in ODEXs, and blocking SIRT2 with AK-7 reversed the antidepressant effects of ODEXs. SIRT2 overexpression was sufficient to enhance neurogenesis and synaptic protein expression. Mechanistically, ODEXs mediated transcellular delivery of SIRT2, targeting AKT deacetylation and AKT/GSK-3β signaling to regulate neuroplasticity. CONCLUSION This study establishes how ODEXs improve depressive-like behaviors and hippocampal neuroplasticity and might provide a promising therapeutic approach for depression.
Collapse
Affiliation(s)
- Honghan Zhang
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Xin‐hui Xie
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Shu‐xian Xu
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Chao Wang
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Siqi Sun
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Xinhua Song
- Clinical College of Traditional Chinese MedicineHubei University of Chinese MedicineWuhanChina
| | - Ruiling Li
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Ningyuan Li
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Yuqi Feng
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Hao Duan
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Di Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhongchun Liu
- Department of PsychiatryRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Taikang Center for Life and Medical SciencesWuhan UniversityWuhanChina
| |
Collapse
|
35
|
Zhou S, Liu Y, Xue B, Yuan P. Low-dose Esketamine suppresses NLRP3-mediated apoptotic and pyroptotic cell death in microglial cells to ameliorate LPS-induced depression via ablating GSK-3β. Behav Brain Res 2024; 459:114782. [PMID: 38029844 DOI: 10.1016/j.bbr.2023.114782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/19/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
Esketamine is verified as a potential therapeutic drug for the treatment of depression, but it is still unclear the detailed underlying mechanisms by which Esketamine ameliorates depression-related symptoms, which seriously limits the utilization of this drug in clinical practices. In this study, the C57BL6/J mice and mouse primary microglial cells were subjected to lipopolysaccharide (LPS)-induced depressive models in vivo and in vitro, and our results confirmed that LPS-induced neuroinflammation, pyroptotic and apoptotic death contributed to the development of LPS-induced depressive symptoms. Then, the following experiments verified that low-dose Esketamine treatment decreased the expression levels of IL-6, TNF-α and IL-18 to restrain cellular inflammation, downregulated NLRP3, cleaved Caspase-1, IL-1β and GSDMD-N to hamper pyroptotic cell death, and inhibited cleaved caspase-3 and Bax, but upregulated Bcl-2 to restrict apoptotic cell death in the LPS-treated mice hippocampus tissues and mouse microglial cells, leading to the suppression of depression development. However, high-dose Esketamine did not have those effects. Next, by conducting mechanical experiments, we verified that low-dose Esketamine downregulated GSK-3β to inactivate NLRP3 inflammasome, and the effects of low-dose Esketamine on cell pyroptosis, neuroinflammation and apoptosis in the LPS-treated microglial cells were all abrogated by overexpressing GSK-3β and NLRP3. Taken together, low-dose Esketamine ameliorated LPS-induced depressive symptoms in mice through regulating the GSK-3β/NLRP3 pathway, and our work suggested that appropriate doses of Esketamine were essential for the treatment of depression in clinic.
Collapse
Affiliation(s)
- Sen Zhou
- Department of Anesthesiology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Jinxiu Road No. 75, Wenzhou City 325000, Zhejiang, China
| | - Yang Liu
- Department of Physical Examination Center, Panhealth Medical Center, Ouyue Street No. 1929, Wenzhou City 325000, Zhejiang, China
| | - Binbin Xue
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Fuxue Road No. 2, Wenzhou City 325000, Zhejiang, China
| | - Peigen Yuan
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Fuxue Road No. 2, Wenzhou City 325000, Zhejiang, China.
| |
Collapse
|
36
|
Cui L, Li S, Wang S, Wu X, Liu Y, Yu W, Wang Y, Tang Y, Xia M, Li B. Major depressive disorder: hypothesis, mechanism, prevention and treatment. Signal Transduct Target Ther 2024; 9:30. [PMID: 38331979 PMCID: PMC10853571 DOI: 10.1038/s41392-024-01738-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
Worldwide, the incidence of major depressive disorder (MDD) is increasing annually, resulting in greater economic and social burdens. Moreover, the pathological mechanisms of MDD and the mechanisms underlying the effects of pharmacological treatments for MDD are complex and unclear, and additional diagnostic and therapeutic strategies for MDD still are needed. The currently widely accepted theories of MDD pathogenesis include the neurotransmitter and receptor hypothesis, hypothalamic-pituitary-adrenal (HPA) axis hypothesis, cytokine hypothesis, neuroplasticity hypothesis and systemic influence hypothesis, but these hypothesis cannot completely explain the pathological mechanism of MDD. Even it is still hard to adopt only one hypothesis to completely reveal the pathogenesis of MDD, thus in recent years, great progress has been made in elucidating the roles of multiple organ interactions in the pathogenesis MDD and identifying novel therapeutic approaches and multitarget modulatory strategies, further revealing the disease features of MDD. Furthermore, some newly discovered potential pharmacological targets and newly studied antidepressants have attracted widespread attention, some reagents have even been approved for clinical treatment and some novel therapeutic methods such as phototherapy and acupuncture have been discovered to have effective improvement for the depressive symptoms. In this work, we comprehensively summarize the latest research on the pathogenesis and diagnosis of MDD, preventive approaches and therapeutic medicines, as well as the related clinical trials.
Collapse
Affiliation(s)
- Lulu Cui
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Shu Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Siman Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Xiafang Wu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yingyu Liu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Weiyang Yu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yijun Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling/Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine/Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang, China.
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Centre of Forensic Investigation, Shenyang, China.
| |
Collapse
|
37
|
Yang X, Liang J, Shu Y, Wei L, Wen C, Luo H, Ma L, Qin T, Wang B, Zeng S, Liu Y, Zhou C. Asperosaponin VI facilitates the regeneration of skeletal muscle injury by suppressing GSK-3β-mediated cell apoptosis. J Cell Biochem 2024; 125:115-126. [PMID: 38079224 DOI: 10.1002/jcb.30510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/26/2023] [Accepted: 11/26/2023] [Indexed: 01/16/2024]
Abstract
Asperosaponin VI (ASA VI) is a bioactive triterpenoid saponin extracted from Diptychus roots, of Diptyl, and has previously shown protective functions in rheumatoid arthritis and sepsis. This study investigates the effects and molecular mechanisms of ASA VI on skeletal muscle regeneration in a cardiotoxin (CTX)-induced skeletal muscle injury mouse model. Mice were subjected to CTX-induced injury in the tibialis anterior and C2C12 myotubes were treated with CTX. Muscle fiber histology was analyzed at 7 and 14 days postinjury. Apoptosis and autophagy-related protein expression were evaluated t s by Western blot, and muscle regeneration markers were quantified by quantitative polymerase chain reaction. Docking studies, cell viability assessments, and glycogen synthase kinase-3β (GSK-3β) activation analyses were performed to elucidate the mechanism. ASA VI was observed to improve muscle interstitial fibrosis, remodeling, and performance in CTX-treated mice, thereby increased skeletal muscle size, weight, and locomotion. Furthermore, ASA VI modulated the expression of apoptosis and autophagy-related proteins through GSK-3β inhibition and activated the transcription of regeneration genes. Our results suggest that ASA VI mitigates skeletal muscle injury by modulating apoptosis and autophagy via GSK-3β signaling and promotes regeneration, thus presenting a probable therapeutic agent for skeletal muscle injury.
Collapse
Affiliation(s)
- Xinru Yang
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jian Liang
- Department of Pediatrics, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yue Shu
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Linlin Wei
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Cailing Wen
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Luo
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Liqing Ma
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Tian Qin
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Bin Wang
- Department of Cardiovascular Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Siyu Zeng
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ying Liu
- Department of Pharmacology, School of Pharmacy, Macau University of Science and Technology, Taipa, Macao, China
- Department of Pharmacology, School of Pharmacy, Guangzhou Xinhua University, Guangzhou, China
| | - Chun Zhou
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Liu M, Fan G, Liu H. Integrated bioinformatics and network pharmacology identifying the mechanisms and molecular targets of Guipi Decoction for treatment of comorbidity with depression and gastrointestinal disorders. Metab Brain Dis 2024; 39:183-197. [PMID: 37847347 DOI: 10.1007/s11011-023-01308-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Guipi decoction (GPD) not only improves gastrointestinal (GI) function, but also depressive mood. The bioinformatics study aimed to reveal potential crosstalk genes and related pathways between depression and GI disorders. A network pharmacology approach was used to explore the molecular mechanisms and potential targets of GPD for the simultaneous treatment of depression comorbid GI disorders. METHODS Differentially expressed genes (DEGs) of major depressive disorder (MDD) were identified based on GSE98793 and GSE19738, and GI disorders-related genes were screened from the GeneCards database. Overlapping genes between MDD and GI disorders were obtained to identify potential crosstalk genes. Protein-protein interaction (PPI) network was constructed to screen for hub genes, signature genes were identified by LASSO regression analysis, and single sample gene set enrichment analysis (ssGSEA) was performed to analyze immune cell infiltration. In addition, based on the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, we screened the active ingredients and targets of GPD and identified the intersection targets of GPD with MDD and GI disorder-related genes, respectively. A "component-target" network was constructed using Cytoscape, the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. RESULTS The MDD-corrected dataset contained 2619 DEGs, and a total of 109 crosstalk genes were obtained. 14 hub genes were screened, namely SOX2, CRP, ACE, LEP, SHH, CDH2, CD34, TNF, EGF, BDNF, FN1, IL10, PPARG, and KIT. These genes were identified by LASSO regression analysis for 3 signature genes, including TNF, EGF, and IL10. Gamma.delta.T.cell was significantly positively correlated with all three signature genes, while Central.memory.CD4.T.cell and Central.memory.CD8.T.cell were significantly negatively correlated with EGF and TNF. GPD contained 134 active ingredients and 248 targets, with 41 and 87 relevant targets for the treatment of depression and GI disorders, respectively. EGF, PPARG, IL10 and CRP overlap with the hub genes of the disease. CONCLUSION We found that GPD may regulate inflammatory and oxidative stress responses through EGF, PPARG, IL10 and CRP targets, and then be involved in the treatment of both depression and GI disorders.
Collapse
Affiliation(s)
- Menglin Liu
- Tianjin University of Chinese Medicine, Tianjin, China
| | - Genhao Fan
- Tianjin University of Chinese Medicine, Tianjin, China
| | - Huayi Liu
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China.
| |
Collapse
|
39
|
He H, He H, Mo L, You Z, Zhang J. Priming of microglia with dysfunctional gut microbiota impairs hippocampal neurogenesis and fosters stress vulnerability of mice. Brain Behav Immun 2024; 115:280-294. [PMID: 37914097 DOI: 10.1016/j.bbi.2023.10.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Mental disorders may be involved in neuroinflammatory processes that are triggered by gut microbiota. How gut microbiota influence microglia-mediated sensitivity to stress remains unclear. Here we explored in an animal model of depression whether disruption of the gut microbiome primes hippocampal microglia, thereby impairing neurogenesis and sensitizing to stress. METHODS Male C57BL/6J mice were exposed to chronic unpredictable mild stress (CUMS) for 4 weeks, and effects on gut microbiota were assessed using 16S rRNA sequencing. Fecal microbiota was transplanted from control or CUMS mice into naïve animals. The depression-like behaviors of recipients were evaluated in a forced swimming test and sucrose preference test. The morphology and phenotype of microglia in the hippocampus of recipients were examined using immunohistochemistry, quantitative PCR, and enzyme-linked immunosorbent assays. The recipients were treated with lipopolysaccharide or chronic stress exposure, and effects were evaluated on behavior, microglial responses and hippocampal neurogenesis. Finally, we explored the ability of minocycline to reverse the effects of CUMS on hippocampal neurogenesis and stress sensitivity in recipients. RESULTS CUMS altered the gut microbiome, leading to higher relative abundance of some bacteria (Helicobacter, Bacteroides, and Desulfovibrio) and lower relative abundance of some bacteria (Lactobacillus, Bifidobacterium, and Akkermansia). Fecal microbiota transplantation from CUMS mice to naïve animals induced microglial priming in the dentate gyrus of recipients. This microglia showed hyper-ramified morphology, and became more sensitive to LPS challenge or chronic stress, which characterized by more significant morphological changes and inflammatory responses, as well as impaired hippocampal neurogenesis and increased depressive-like behaviors. Giving minocycline to recipients reversed these effects of fecal transplantation. CONCLUSIONS These findings suggest that gut microbiota from stressed animals can induce microglial priming in the dentate gyrus, which is associated with a hyper-immune response to stress and impaired hippocampal neurogenesis. Remodeling the gut microbiome or inhibiting microglial priming may be strategies to reduce sensitivity to stress.
Collapse
Affiliation(s)
- Hui He
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Haili He
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Li Mo
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Zili You
- Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
40
|
Liu Y, Zhou X, Xue K, Sun R, Tang Y, Tang C. Reviving: restoring depression-like behaviour through glial cell-derived neurotrophic factor treatment in the medial prefrontal cortex. J Psychiatry Neurosci 2024; 49:E23-E34. [PMID: 38302136 PMCID: PMC10843345 DOI: 10.1503/jpn.230079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/04/2023] [Accepted: 10/17/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Depression is a prevalent nonmotor symptom in Parkinson disease and can greatly reduce the quality of life for patients; the dopamine receptors found in glutamatergic pyramidal cells in the medial prefrontal cortex (mPFC) play a role in regulating local field activity, which in turn affects behavioural and mood disorders. Given research showing that glial cell-derived neurotrophic factor (GDNF) may have an antidepressant effect, we sought to evaluate the impact of exogenous GDNF on depression-like behaviour in mouse models of Parkinson disease. METHODS We used an established subacute model of Parkinson disease in mice involving intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), followed by brain stereotaxic injection of GDNF into the mPFC region. Subsequently, we assessed depression-like behaviour using the sucrose preference test, forced swimming test and tail suspension test, while also evaluating protein expression in the mPFC. RESULTS We included 60 mice, divided into 3 groups, including a control group (saline injection), an MPTP plus saline injection group and an MPTP plus GDNF injection group. We found that exogenous GDNF injection into the mPFC led to an increase in dopamine receptor D1 (DRD1) protein levels. We also observed that activating the protein kinase A pathway through DRD1 produced a prolonged antidepressant response. Under GDNF stimulation, the expression of dopamine receptor D2 (DRD2) remained constant, suggesting that the DRD2 signal was ineffective in alleviating depression-like symptoms. Moreover, our investigation involved Golgi staining and Western blot techniques, which found enhanced synaptic plasticity, including increased dendritic branches, dendritic spines and retrograde protection after GDNF treatment in Parkinson disease models. LIMITATIONS A subtle motor phenotype became evident only toward the conclusion of the behavioural testing period. The study exclusively involved male mice, and no separate control group receiving only GDNF treatment was included in the experimental design. CONCLUSION Our findings support a positive effect of exogenous GDNF on synaptic plasticity, mediated by DRD1 signalling in the mPFC, which could facilitate depression remission in Parkinson disease.
Collapse
Affiliation(s)
- Yehao Liu
- From the Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (Liu, Xue, Sun, C. Tang); the Department of Neurology, the Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu, China (Zhou); the Department of Neurology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Jiangsu, China (Y. Tang)
| | - Xiaoyu Zhou
- From the Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (Liu, Xue, Sun, C. Tang); the Department of Neurology, the Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu, China (Zhou); the Department of Neurology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Jiangsu, China (Y. Tang)
| | - Ke Xue
- From the Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (Liu, Xue, Sun, C. Tang); the Department of Neurology, the Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu, China (Zhou); the Department of Neurology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Jiangsu, China (Y. Tang)
| | - Ruiao Sun
- From the Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (Liu, Xue, Sun, C. Tang); the Department of Neurology, the Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu, China (Zhou); the Department of Neurology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Jiangsu, China (Y. Tang)
| | - Yan Tang
- From the Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (Liu, Xue, Sun, C. Tang); the Department of Neurology, the Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu, China (Zhou); the Department of Neurology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Jiangsu, China (Y. Tang)
| | - Chuanxi Tang
- From the Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China (Liu, Xue, Sun, C. Tang); the Department of Neurology, the Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu, China (Zhou); the Department of Neurology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Jiangsu, China (Y. Tang)
| |
Collapse
|
41
|
Quan H, Zhang R. Microglia dynamic response and phenotype heterogeneity in neural regeneration following hypoxic-ischemic brain injury. Front Immunol 2023; 14:1320271. [PMID: 38094292 PMCID: PMC10716326 DOI: 10.3389/fimmu.2023.1320271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Hypoxic-ischemic brain injury poses a significant threat to the neural niche within the central nervous system. In response to this pathological process, microglia, as innate immune cells in the central nervous system, undergo rapid morphological, molecular and functional changes. Here, we comprehensively review these dynamic changes in microglial response to hypoxic-ischemic brain injury under pathological conditions, including stroke, chronic intermittent hypoxia and neonatal hypoxic-ischemic brain injury. We focus on the regulation of signaling pathways under hypoxic-ischemic brain injury and further describe the process of microenvironment remodeling and neural tissue regeneration mediated by microglia after hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Hongxin Quan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Runrui Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| |
Collapse
|
42
|
Wang Y, Lai H, Zhang T, Wu J, Tang H, Liang X, Ren D, Huang J, Li W. Mitochondria of intestinal epithelial cells in depression: Are they at a crossroads of gut-brain communication? Neurosci Biobehav Rev 2023; 153:105403. [PMID: 37742989 DOI: 10.1016/j.neubiorev.2023.105403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
The role of gut dysbiosis in depression is well established. However, recent studies have shown that gut microbiota is regulated by intestinal epithelial cell (IEC) mitochondria, which has yet to receive much attention. This review summarizes the recent developments about the critical role of IEC mitochondria in actively maintaining gut microbiota, intestinal metabolism, and immune homeostasis. We propose that IEC mitochondrial dysfunction alters gut microbiota composition, participates in cell fate, mediates oxidative stress, activates the peripheral immune system, causes peripheral inflammation, and transmits peripheral signals through the vagus and enteric nervous systems. These pathological alterations lead to brain inflammation, disruption of the blood-brain barrier, activation of the hypothalamic-pituitary-adrenal axis, activation of microglia and astrocytes, induction of neuronal loss, and ultimately depression. Furthermore, we highlight the prospect of treating depression through the mitochondria of IECs. These new findings suggest that the mitochondria of IECs may be a newly found important factor in the pathogenesis of depression and represent a potential new strategy for treating depression.
Collapse
Affiliation(s)
- Yi Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China
| | - Han Lai
- School of Foreign Languages, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China
| | - Tian Zhang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China
| | - Jing Wu
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China
| | - Huiling Tang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China
| | - Xuanwei Liang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China
| | - Dandan Ren
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China
| | - Jinzhu Huang
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Weihong Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| |
Collapse
|
43
|
Li CC, Munalisa R, Lee HY, Lien TS, Chan H, Hung SC, Sun DS, Cheng CF, Chang HH. Restraint Stress-Induced Immunosuppression Is Associated with Concurrent Macrophage Pyroptosis Cell Death in Mice. Int J Mol Sci 2023; 24:12877. [PMID: 37629059 PMCID: PMC10454201 DOI: 10.3390/ijms241612877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Psychological stress is widely acknowledged as a major contributor to immunosuppression, rendering individuals more susceptible to various diseases. The complex interplay between the nervous, endocrine, and immune systems underlies stress-induced immunosuppression. However, the underlying mechanisms of psychological-stress-induced immunosuppression remain unclear. In this study, we utilized a restraint stress mouse model known for its suitability in investigating physiological regulations during psychological stress. Comparing it with cold exposure, we observed markedly elevated levels of stress hormones corticosterone and cortisol in the plasma of mice subjected to restraint stress. Furthermore, restraint-stress-induced immunosuppression differed from the intravenous immunoglobulin-like immunosuppression observed in cold exposure, with restraint stress leading to increased macrophage cell death in the spleen. Suppression of pyroptosis through treatments of inflammasome inhibitors markedly ameliorated restraint-stress-induced spleen infiltration and pyroptosis cell death of macrophages in mice. These findings suggest that the macrophage pyroptosis associated with restraint stress may contribute to its immunosuppressive effects. These insights have implications for the development of treatments targeting stress-induced immunosuppression, emphasizing the need for further investigation into the underlying mechanisms.
Collapse
Affiliation(s)
- Chi-Cheng Li
- Department of Hematology and Oncology, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan;
- Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Hualien 970, Taiwan
| | - Rina Munalisa
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (H.-Y.L.); (T.-S.L.); (H.C.); (S.-C.H.); (D.-S.S.)
| | - Hsuan-Yun Lee
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (H.-Y.L.); (T.-S.L.); (H.C.); (S.-C.H.); (D.-S.S.)
| | - Te-Sheng Lien
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (H.-Y.L.); (T.-S.L.); (H.C.); (S.-C.H.); (D.-S.S.)
| | - Hao Chan
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (H.-Y.L.); (T.-S.L.); (H.C.); (S.-C.H.); (D.-S.S.)
| | - Shih-Che Hung
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (H.-Y.L.); (T.-S.L.); (H.C.); (S.-C.H.); (D.-S.S.)
| | - Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (H.-Y.L.); (T.-S.L.); (H.C.); (S.-C.H.); (D.-S.S.)
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei 231, Taiwan;
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan; (R.M.); (H.-Y.L.); (T.-S.L.); (H.C.); (S.-C.H.); (D.-S.S.)
| |
Collapse
|
44
|
Chen X, Cui QQ, Hu XH, Ye J, Liu ZC, Mei YX, Wang F, Hu ZL, Chen JG. CD200 in dentate gyrus improves depressive-like behaviors of mice through enhancing hippocampal neurogenesis via alleviation of microglia hyperactivation. J Neuroinflammation 2023; 20:157. [PMID: 37391731 DOI: 10.1186/s12974-023-02836-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Neuroinflammation and microglia play critical roles in the development of depression. Cluster of differentiation 200 (CD200) is an anti-inflammatory glycoprotein that is mainly expressed in neurons, and its receptor CD200R1 is primarily in microglia. Although the CD200-CD200R1 pathway is necessary for microglial activation, its role in the pathophysiology of depression remains unknown. METHODS The chronic social defeat stress (CSDS) with behavioral tests were performed to investigate the effect of CD200 on the depressive-like behaviors. Viral vectors were used to overexpress or knockdown of CD200. The levels of CD200 and inflammatory cytokines were tested with molecular biological techniques. The status of microglia, the expression of BDNF and neurogenesis were detected with immunofluorescence imaging. RESULTS We found that the expression of CD200 was decreased in the dentate gyrus (DG) region of mice experienced CSDS. Overexpression of CD200 alleviated the depressive-like behaviors of stressed mice and inhibition of CD200 facilitated the susceptibility to stress. When CD200R1 receptors on microglia were knocked down, CD200 was unable to exert its role in alleviating depressive-like behavior. Microglia in the DG brain region were morphologically activated after exposure to CSDS. In contrast, exogenous administration of CD200 inhibited microglia hyperactivation, alleviated neuroinflammatory response in hippocampus, and increased the expression of BDNF, which in turn ameliorated adult hippocampal neurogenesis impairment in the DG induced by CSDS. CONCLUSIONS Taken together, these results suggest that CD200-mediated alleviation of microglia hyperactivation contributes to the antidepressant effect of neurogenesis in dentate gyrus in mice.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian-Qian Cui
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Hai Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Ye
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Cun Liu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Xi Mei
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.
| |
Collapse
|
45
|
Afridi R, Suk K. Microglial Responses to Stress-Induced Depression: Causes and Consequences. Cells 2023; 12:1521. [PMID: 37296642 PMCID: PMC10252665 DOI: 10.3390/cells12111521] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Chronic stress is a major risk factor for various psychiatric diseases, including depression; it triggers various cellular and structural changes, resulting in the alteration of neurocircuitry and subsequent development of depression. Accumulating evidence suggests that microglial cells orchestrate stress-induced depression. Preclinical studies of stress-induced depression revealed microglial inflammatory activation in regions of the brain that regulate mood. Although studies have identified several molecules that trigger inflammatory responses in microglia, the pathways that regulate stress-induced microglial activation remain unclear. Understanding the exact triggers that induce microglial inflammatory activation can help find therapeutic targets in order to treat depression. In the current review, we summarize the recent literature on possible sources of microglial inflammatory activation in animal models of chronic stress-induced depression. In addition, we describe how microglial inflammatory signaling affects neuronal health and causes depressive-like behavior in animal models. Finally, we propose ways to target the microglial inflammatory cascade to treat depressive disorders.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu 41940, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Kyungpook National University, Daegu 41940, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
46
|
Su D, Jiang W, Yuan Q, Guo L, Liu Q, Zhang M, Kang C, Xiao C, Yang C, Li L, Xu C, Zhou T, Zhang J. Chronic exposure to aflatoxin B1 increases hippocampal microglial pyroptosis and vulnerability to stress in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 258:114991. [PMID: 37172405 DOI: 10.1016/j.ecoenv.2023.114991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Chronic aflatoxin B1 (AFB1) exposure may increase the risk of multiple neuropsychiatric disorders. Stress is considered one of the main contributors to major depressive disorder. Whether and how chronic AFB1 exposure affects vulnerability to stress is unclear. METHODS Mice were exposed for three weeks to AFB1 (100 µg/kg/d) and/or chronic mild stress (CMS). The vulnerability behaviors in response to stress were assessed in the forced swimming test (FST), sucrose preference test (SPT), and tail suspension test (TST). Microglial pyroptosis was investigated using immunofluorescence, enzyme-linked immunosorbent assays, and western blot assay in the hippocampus of mice. Hippocampal neurogenesis and the effects of AFB1-treated microglia on proliferation and differentiation of neural stem/precursor cells (NSPCs) were assessed via immunofluorescence in the hippocampus of mice. RESULTS Mice exposed to CMS in the presence of AFB1 exhibited markedly greater vulnerability to stress than mice treated with CMS or AFB1 alone, as indicated by reduced sucrose preference and longer immobility time in the forced swimming test. Chronic aflatoxin B1 exposure resulted in changes in the microglial morphology and increase in TUNEL+ microglia and GSDMD+ microglia in the hippocampal dentate gyrus. When mice were exposed to both CMS and AFB1, pyroptosis-related molecules (such as NLRP3, caspase-1, GSDMD-N, and interleukin-1β) were significantly upregulated in the hippocampus. These molecules were also significantly enhanced by AFB1 in primary microglial cultures. AFB1-treated mice showed decrease in the numbers of BrdU+, BrdU-DCX+, and BrdU-NeuN+ cells in the hippocampal dentate gyrus, as well as the percentages of BrdU+ cells that were NeuN+ in the presence or absence of CMS when compared with vehicle-treated mice. The combination of AFB1 and CMS exacerbated these effects to an even greater extent. The number of DCX+ cells correlated negatively with the percentage of ameboid microglia, TUNEL+ microglia and GSDMD+ microglia in the hippocampal dentate gyrus. AFB1-treated microglia suppressed the proliferation and neuronal differentiation of NSPCs in vitro. CONCLUSION Chronic AFB1 exposure induces microglial pyroptosis, promoting an adverse neurogenic microenvironment that impairs hippocampal neurogenesis, which may render mice more vulnerable to stress.
Collapse
Affiliation(s)
- Dapeng Su
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Weike Jiang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Qingsong Yuan
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qin Liu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Mengmeng Zhang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chuangzhi Kang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chenghong Xiao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Changgui Yang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Liangyuan Li
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chunyun Xu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Tao Zhou
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Jinqiang Zhang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
47
|
Zhang J, Li L, Liu Q, Zhao Z, Su D, Xiao C, Jin T, Chen L, Xu C, You Z, Zhou T. Gastrodin programs an Arg-1 + microglial phenotype in hippocampus to ameliorate depression- and anxiety-like behaviors via the Nrf2 pathway in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 113:154725. [PMID: 36867963 DOI: 10.1016/j.phymed.2023.154725] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/05/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Regulating the microglial phenotype is an attractive strategy for treating diseases of the central nervous system such as depression and anxiety. Gastrodin can quickly cross the blood-brain barrier and mitigate microglia-mediated inflammation, which widely used to treat a variety of central nervous system diseases associated with microglial dysfunction. However, the molecular mechanism by which gastrodin regulates the functional phenotype of microglia remains unclear. PURPOSE Since the transcription factor "nuclear factor erythroid 2-related factor 2″ (Nrf2) is associated with the anti-inflammatory effects of gastrodin, we hypothesized that gastrodin induces Nrf2 expression in microglia and thereby programs an anti-inflammatory phenotype. STUDY DESIGN Male C57BL/6 mice, treated or not with gastrodin, were given lipopolysaccharide (LPS) at 0.25 mg/kg/d for 10 days to induce chronic neuroinflammation. The effects of gastrodin on microglial phenotypes, neuroinflammation and depression- and anxiety-like behaviors were evaluated. In another experiment, animals were treated with Nrf2 inhibitor ML385 throughout the 13-day gastrodin intervention period. METHODS The effects of gastrodin on depression- and anxiety-like behaviors were evaluated through the sucrose preference test, forced swimming test, open field test and elevated plus-maze test; as well as its effects on morphology and molecular and functional phenotypes of hippocampal microglia through immunohistochemistry, real-time PCR and enzyme-linked immunosorbent assays. RESULTS Chronic exposure to LPS caused hippocampal microglia to secrete inflammatory cytokines, their somata to enlarge, and their dendrites to lose branches. These changes were associated with depression- and anxiety-like behaviors. Gastrodin blocked these LPS-induced alterations and promoted an Arg-1+ microglial phenotype that protected neurons from injury. The effects of gastrodin were associated with Nrf2 activation, whereas blockade of Nrf2 antagonized gastrodin. CONCLUSION These results suggest that gastrodin acts via Nrf2 to promote an Arg-1+ microglial phenotype, which buffers the harmful effects of LPS-induced neuroinflammation. Gastrodin may be a promising drug against central nervous system diseases that involve microglial dysfunction.
Collapse
Affiliation(s)
- Jinqiang Zhang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Liangyuan Li
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Qin Liu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Zhihuang Zhao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Dapeng Su
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chenghong Xiao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Ting Jin
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Li Chen
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chunyun Xu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Zili You
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Tao Zhou
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
48
|
Skała E, Szopa A. Dipsacus and Scabiosa Species-The Source of Specialized Metabolites with High Biological Relevance: A Review. Molecules 2023; 28:molecules28093754. [PMID: 37175164 PMCID: PMC10180103 DOI: 10.3390/molecules28093754] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
The genera Dipsacus L. and Scabiosa L. of the Caprifoliaceae family are widely distributed in Europe, Asia, and Africa. This work reviews the available literature on the phytochemical profiles, ethnomedicinal uses, and biological activities of the most popular species. These plants are rich sources of many valuable specialized metabolites with beneficial medicinal properties, such as triterpenoid derivatives, iridoids, phenolic acids, and flavonoids. They are also sources of essential oils. The genus Dipsacus has been used for centuries in Chinese and Korean folk medicines to treat bone (osteoporosis) and joint problems (rheumatic arthritis). The Korean Herbal Pharmacopoeia and Chinese Pharmacopoeia include Dipsaci radix, the dried roots of D. asperoides C.Y.Cheng & T.M.Ai. In addition, S. comosa Fisch. ex Roem & Schult. and S. tschiliiensis Grunning are used in traditional Mongolian medicine to treat liver diseases. The current scientific literature data indicate that these plants and their constituents have various biological properties, including inter alia antiarthritic, anti-neurodegenerative, anti-inflammatory, antioxidant, anticancer, and antimicrobial activities; they have also been found to strengthen tendon and bone tissue and protect the liver, heart, and kidney. The essential oils possess antibacterial, antifungal, and insecticidal properties. This paper reviews the key biological values of Dipsacus and Scabiosa species, as identified by in vitro and in vivo studies, and presents their potential pharmacological applications.
Collapse
Affiliation(s)
- Ewa Skała
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Agnieszka Szopa
- Chair and Department of Pharmaceutical Botany, Medical College, Jagiellonian University, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
49
|
Zhou Y, Xu Y, Yang J, Yu Z, Wang W, Yuan M, Wang Y, Bai Q, Li Z. Spinal cannabinoid receptor 2 activation alleviates neuropathic pain by regulating microglia and suppressing P2X7 receptor. Front Mol Neurosci 2023; 16:1061220. [PMID: 36969555 PMCID: PMC10030493 DOI: 10.3389/fnmol.2023.1061220] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/13/2023] [Indexed: 03/29/2023] Open
Abstract
Neuropathic pain (NP) is the chronic pain in patients resulting from injuries or diseases in the somatosensory nervous system. However, effective treatment remains limited to opioids. Currently, there is an urgent need to develop new specific pharmaceuticals with low abuse potentiality. Cannabinoid receptor 2 (CB2R) is one of the significant receptors in the endocannabinoid system. It is widely expressed in the central nervous system, especially enriched in glial cells, and plays an important role in the occurrence and development of inflammation in the nervous system. CB2R activation has a neuroprotective effect on nerve injury. In this study, we report increased and more reactive microglia (with larger cell body, shorter processes, and fewer endpoints) observed in the spinal dorsal horn of spared nerve injury (SNI) rats. Continuous intrathecal administration of CB2R agonist PM226 attenuated mechanical and cold hyperalgesia in rats and prevented the transition of microglia to the proinflammatory stage. Thus, microglia transitioned into the neuroprotective stage. Meanwhile, the proinflammatory factors TNF-α and iNOS decreased, and the levels of anti-inflammatory factors Arg-1 and IL-10 increased. The content of P2X7 receptors in the spinal dorsal horn of rats increases with time after SNI. After continuous intrathecal administration of PM226, the content of P2X7 protein decreases significantly. The administration of P2X7 inhibitor A-438079 alleviated the mechanical hyperalgesia of rats, reduced the number of microglia, and decreased the content of P2X7. These results indicate that P2X7 is involved in the neuroprotective effect caused by CB2R activation. In conclusion, this study provides new insights into the neuroprotective mechanism of CB2R activation.
Collapse
Affiliation(s)
- Yifan Zhou
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaowei Xu
- Institute of Neuroscience, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jingjie Yang
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhixiang Yu
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenting Wang
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Yuan
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Wang
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qian Bai
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Qian Bai,
| | - Zhisong Li
- Department of Anesthesiology and Perioperative Medicine, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Zhisong Li,
| |
Collapse
|
50
|
Han W, Wang N, Han M, Ban M, Sun T, Xu J. Reviewing the role of gut microbiota in the pathogenesis of depression and exploring new therapeutic options. Front Neurosci 2022; 16:1029495. [PMID: 36570854 PMCID: PMC9772619 DOI: 10.3389/fnins.2022.1029495] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
The relationship between gut microbiota (GM) and mental health is one of the focuses of psychobiology research. In recent years, the microbial-gut-brain axis (MGBA) concept has gradually formed about this bidirectional communication between gut and brain. But how the GM is involved in regulating brain function and how they affect emotional disorders these mechanisms are tenuous and limited to animal research, and often controversial. Therefore, in this review, we attempt to summarize and categorize the latest advances in current research on the mechanisms of GM and depression to provide valid information for future diagnoses and therapy of mental disorders. Finally, we introduced some antidepressant regimens that can help restore gut dysbiosis, including classic antidepressants, Chinese materia medica (CMM), diet, and exogenous strains. These studies provide further insight into GM's role and potential pathways in emotion-related diseases, which holds essential possible clinical outcomes for people with depression or related psychiatric disorders. Future research should focus on clarifying the causal role of GM in disease and developing microbial targets, applying these findings to the prevention and treatment of depression.
Collapse
Affiliation(s)
- Wenjie Han
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Pharmacology, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China
| | - Na Wang
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Pharmacology, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China
| | - Mengzhen Han
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Pharmacology, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China
| | - Meng Ban
- Liaoning Microhealth Biotechnology Co., Ltd., Shenyang, China
| | - Tao Sun
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| | - Junnan Xu
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Pharmacology, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China,*Correspondence: Junnan Xu,
| |
Collapse
|