1
|
Hu H, Zhang J, Xin X, Jin Y, Zhu Y, Zhang H, Fan R, Ye Y, Jiang Y, Li D. Bushen Jianpi Tiaoxue Decoction (BJTD) inhibits the LIF-mTOR signaling axis to regulate mitochondrial function and alleviate cyclophosphamide-induced diminished ovarian reserve. Apoptosis 2025; 30:1331-1350. [PMID: 40042747 DOI: 10.1007/s10495-025-02093-1] [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] [Accepted: 02/08/2025] [Indexed: 06/16/2025]
Abstract
Diminished ovarian reserve (DOR) is a challenging obstacle impacting women' fertility globally with limited treatment option. Bushen Jianpi Tiaoxue Decoction (BJTD) has shown significant efficacy and safety in treating DOR patients, yet the molecular mechanisms behind its effect remain uncertain. Our study aimed to uncover the pharmacology and signaling pathway of BJTD in cyclophosphamide (Cy)-provoked DOR mice and 4-hydroperoxy cyclophosphamide (4-HC)-irritated KGN cells (human granulosa-like cell line) damage models. Granulosa cells from DOR patients and Cy-induced models were reanalyzed utilizing transcriptomics to predict differentially expressed genes and crucial signaling pathways. Validation experiments were conducted in vitro using KGN cells treated with 4-HC, a Cy metabolite, to establish a DOR model. Cell viability, apoptosis, oxidative stress, mitochondrial function, and ferroptosis markers were assessed using the cck8 assay, flow cytometry, fluorescence staining, RT-qPCR, and western blotting analyses. BJTD-serum was evaluated for its protective effects on 4-HC-triggered KGN damages. In vivo, a Cy-induced DOR mouse model was treated with BJTD to evaluate ovarian morphology, estrous cycle, follicle counts, hormone markers, mitochondrial apoptosis and ferroptosis levels, respectively via the vaginal smear, histological analysis, immunostaining, gene and protein expression experiments. The UPLC-MS analysis and network pharmacology were applied to identify BJTD's active ingredients, followed by molecular dockings to assess interactions with the target protein. To confirm the BJTD's mechanism of action, mTOR signaling modulation was analyzed using a specific inhibitor or activator in vitro. Transcriptomic reanalysis revealed significant gene expression differences, with LIF identified as a key target associated with apoptosis pathway. In vitro, 4-HC exposure induced apoptosis, mitochondrial dysfunction, and ferroptosis in KGN cells, accompanied by upregulation of LIF, mTOR, and FoxO3a signalings. BJTD-serum treatment significantly improved cell viability, reduced apoptosis, and alleviated oxidative stress by modulating mitochondrial function and ferroptosis markers, such as Nrf2, HO-1, and GPX4. In vivo, BJTD alleviated Cy-induced ovarian damage, improving ovarian index, estrous cycle, follicle development, and hormone levels, while reducing follicular atresia and granulosa cells apoptosis. Mechanically, BJTD suppressed Cy-induced activation of the LIF-mTOR axis and downstream mitochondrial apoptosis markers, including Cleaved Caspase 9/3, BAX, and γH2AX, while enhancing OPA1 and Bcl-2 expressions. The UPLC-MS outcome combining with network pharmacology identified mainly 20 active compounds in BJTD, with astragaloside IV exhibiting the strongest binding to the mTOR protein. The mTOR pathway modulation experiments confirmed that BJTD's protective effects are mediated through inhibition of hyperactivated mTOR phosphorylation and mitochondrial apoptosis cascades. BJTD demonstrates efficacy in alleviating Cy- and 4-HC-induced DOR models through targeting the LIF-mTOR signaling axis to suppress granulosa cells mitochondrial apoptosis and ferroptosis. These results might highlight promising therapeutic potential of BJTD for ovarian reserve preservation.
Collapse
Affiliation(s)
- Hangqi Hu
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Jiacheng Zhang
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Xiyan Xin
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yuxin Jin
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yutian Zhu
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Haolin Zhang
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Ruiwen Fan
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yang Ye
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China.
| | - Yong Jiang
- School of Pharmaceutical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China.
| | - Dong Li
- State Key Laboratory of Female Fertility Promotion, Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
2
|
Zhang X, Zhang N, Dong Z, Sun H, Diao Z, Li Y, Du D, Ma Y. The role of Chinese herbal medicine in diminished ovarian reserve management. J Ovarian Res 2025; 18:90. [PMID: 40307895 PMCID: PMC12042416 DOI: 10.1186/s13048-025-01669-4] [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: 02/22/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
Diminished Ovarian Reserve (DOR) is characterized by a reduction in the number of available follicles in the ovaries, leading to hormonal imbalances, decreased ovarian reserve, and reduced fertility. Clinically, it presents with elevated follicle-stimulating hormone (FSH) levels, decreased anti-Müllerian hormone (AMH) levels, and a lower antral follicle count (AFC). In recent years, Traditional Chinese Medicine (TCM) has gained recognition for its multi-target, holistic regulation in treating DOR, offering broad therapeutic effects with minimal side effects. This review aims to summarize the mechanisms and clinical efficacy of Chinese herbal medicine (CHM) formulas and active compounds in the treatment of DOR, providing theoretical support for their clinical application and future research. A systematic literature search was conducted from June 2019 to June 2024, and 12 clinical studies along with 38 basic research papers were selected. The findings suggest that CHM formulas primarily act by counteracting oxidative stress, regulating immune defense, modulating sex hormone secretion via the hypothalamic-pituitary-ovarian axis, and inhibiting excessive apoptosis of ovarian granulosa cells. This review highlights the therapeutic potential of TCM for improving ovarian function, regulating endocrine balance, and alleviating DOR symptoms, offering valuable insights for clinical practice and research.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Na Zhang
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhibin Dong
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hao Sun
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhihao Diao
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yujie Li
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Dongqing Du
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yuxia Ma
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
3
|
Niu Y, Wu Z, Hu Q, Wu Y, Jiang Q, Yang X. Discovery of acetohydroxyacid synthase inhibitors as anti-tuberculosis lead compounds from natural products. Bioorg Med Chem 2025; 118:118041. [PMID: 39708691 DOI: 10.1016/j.bmc.2024.118041] [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: 09/04/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024]
Abstract
Acetohydroxy acid synthase (AHAS) is a key enzyme that catalyzes the synthesis of branched-chain amino acids, which is indispensable for the survival and growth of Mycobacterium tuberculosis (Mtb). Aim to discover new AHAS inhibitors from natural products, here we performed computer assistant target-based screening for Mtb-AHAS inhibitors using Discovery Studio on TCMSP and SELLECK libraries. Mtb-AHAS structure was first simulated and verified for docking, and 80 compounds with top LIBDOCK and CDDOCK scores were obtained. By experimental verification, four compounds namely Salvianolic acid A, Embelin, Celastrol and Wushanicaritin showed inhibition potency against Mtb-AHAS with IC50 ranging from 805.5 nM-32.36 μM. The most potential inhibitor Celastrol exhibited bacteriostatic activity for both Mycobacterium smegmatis and Mycobacterium tuberculosis with MIC of 62.5 μM and 80 μM, respectively. This study revealed that Celastrol is the potential Mtb-AHAS inhibitor as an anti-tuberculosis lead compound.
Collapse
Affiliation(s)
- Yanhong Niu
- Key Laboratory of Medical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong Dist, Chongqing 400016, China
| | - Zhili Wu
- Key Laboratory of Medical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong Dist, Chongqing 400016, China
| | - Qianfang Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuchen Wu
- Key Laboratory of Medical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong Dist, Chongqing 400016, China
| | - Qihua Jiang
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Xiaolan Yang
- Key Laboratory of Medical Laboratory Diagnostics of the Education Ministry, College of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong Dist, Chongqing 400016, China.
| |
Collapse
|
4
|
Xu X, Wang J, Jin X, Ma Q, Li H, Zhou Q, Chen W. Bu-Shen-Ning-Xin decoction ameliorates premature ovarian insufficiency by suppressing oxidative stress through rno_circRNA_012284/rno_miR-760-3p/HBEGF pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155920. [PMID: 39126922 DOI: 10.1016/j.phymed.2024.155920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND POI (premature ovarian insufficiency) refers to premature and rapid decline of ovarian reserve function in women before the age of 40, which can be manifested as menstrual disorders, endocrine abnormalities and low fertility. Bu-Shen-Ning-Xin decoction (BSNXD) has been found to have therapeutic effects on POI. Nevertheless, how it exerts therapeutic effects remains elusive. PURPOSE This research aims to clarify the pharmacological mechanisms of BSNXD. METHODS We applied Ultra Performance Liquid Chromatography (UPLC) to identify the main components of BSNXD.4-vinylcyclohexene diepoxide(VCD)was used to induce POI models. ELISA detected the serum level of hormones. H&E staining evaluated the morphology of ovarian tissues.CircRNA and mRNA expression profiles in the ovaries of both POI rats and those treated with BSNXD were detected. Then, dysregulated circRNAs and mRNAs that were potentially altered by BSNXD were screened. Network pharmacology analysis was performed to identify drug targets of BSNXD active ingredients. A circRNA-miRNA-mRNA network and an oxidative stress(OS)-related subnetwork were constructed. Expression of rno_circRNA_012284, rno_miR-760-3p, and HBEGF(Heparin-binding epidermal growth factor-like growth factor) was measured by RT-PCR and their binding were verified by dual-luciferase reporter assays. ROS was measured through DCFH-DA fluorescence probes. The HBEGF target was selected for molecular docking with key active ingredients.Surface plasmon resonance(SPR) was applied to verify the binding ability and affinity between components and HBEGF. RESULTS UPLC analysis indicated that 6 chemical compounds including berberine, paeoniflorin, morroniside,gallic acid, loganin, baicalin were identified.Elevated FSH and LH levels, suppressed E2 and AMH levels in the serum, and inhibited follicles and corpus luteums in the ovarian tissues of VCD-induced rats were notably reversed by BSNXD.In total, 992 up- and 1135 down-regulated circRNAs, and 205 up- and 243 down-regulated mRNAs were found in POI rat ovaries following BSNXD administration. Furthermore, 198 drug targets of BSNXD were identified. An OS-related and BSNXD-targeted ceRNA subnetwork composed of rno_circRNA_012284/rno_miR-760-3p/HBEGF was established. rno_circRNA_012284 and HBEGF were up-regulated and rno_miR-760-3p was down-regulated in POI ovarian granulosa cells (OGCs) after BSNXD administration. rno_circRNA_012284 was a sponge of rno_miR-760-3p to elevate HBEGF expression. Moreover, rno_circRNA_012284 overexpression alleviated POI-induced excessive ROS generation in ovarian granulosa cells, while rno_circRNA_012284 inhibition exerted the opposite effect. Finally,molecular docking speculated active ingredients of each herb acted on HBEGF to reduce the OS. SPR tests showed that Berberine,Baicalein,Quercetin,Pachymic acid,Paeoniflorin exhibited satisfying affinity with HBEGF protein. CONCLUSION This study demonstrates that BSNXD ameliorates POI partly by attenuating OS in ovarian granulosa cells via rno_circRNA_012284/rno_miR-760-3p/HBEGF axis, uncovering the pharmacological mechanisms of BSNXD in alleviating POI.
Collapse
Affiliation(s)
- Xianli Xu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, 310006 Hangzhou, Zhejiang, China
| | - Jiajing Wang
- School of Pharmacy, Zhejiang Chinese Medical University,311402 Hangzhou, Zhejiang, China
| | - Xin Jin
- Department of Tuina, The First Affiliated Hospital of Zhejiang Chinese Medical University, 310006 Hangzhou, Zhejiang, China
| | - Qianwen Ma
- Department of Traditional Chinese Medicine, Hangzhou Ninth People's Hospital,311225, Hangzhou, Zhejiang, China
| | - Huifang Li
- Department of TCM Gynecology, Tongxiang Maternal and Child Health-Care Center, 314500, Jiaxing, Zhejiang, China
| | - Qun Zhou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, 310006 Hangzhou, Zhejiang, China
| | - Wenjun Chen
- School of Nursing, Hangzhou Medical College, 311399 Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Yan Z, Chen C, Zhai S, Tang H, Zhu M, Yu Y, Zheng H. Mechanism of Qingchang compound against coccidiosis based on network pharmacology-molecular docking. Front Vet Sci 2024; 11:1361552. [PMID: 38496310 PMCID: PMC10940363 DOI: 10.3389/fvets.2024.1361552] [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: 12/26/2023] [Accepted: 02/22/2024] [Indexed: 03/19/2024] Open
Abstract
The aim of this study was to investigate the anti-Eimeria tenella mechanism of Qingchang Compound (QCC) and provide a basis for its clinical application. The active ingredients, active ingredient-disease intersection targets, and possible pathways of QCC for the treatment of chicken coccidiosis were analyzed, the binding ability of pharmacodynamic components and target proteins was determined by network pharmacology and the molecular docking, and a model of infection with coccidiosis was constructed to verify and analyze the mechanism of action of QCC against coccidiosis. Among the 57 components that met the screening conditions, the main bioactive components were quercetin, dichroine, and artemisinin, with IL-1β, IL-6, IL-10, IFN-γ, and IL-8 as the core targets. Simultaneously, the KEGG signaling pathway of QCC anti-coccidiosis in chickens was enriched, including cytokine-cytokine receptor interactions. The results showed that the main pharmacodynamic components of QCC and the core targets could bind well; artemisinin and alpine possessed the largest negative binding energies and presented the most stable binding states. In addition, in vivo studies showed that QCC reduced blood stool in chickens with coccidiosis, restored cecal injury, and significantly reduced the mRNA and protein expression levels of IL-1β, IL-10, and IFN-γ in ceca (p < 0.01). Our results suggest that the main active ingredients of QCC are artemisinin and alpine and its mechanism of action against coccidiosis may be related to the reduction of the inflammatory response by acting on specific cytokines.
Collapse
Affiliation(s)
- Zhiqiang Yan
- Chongqing Academy of Animal Sciences, Rongchang, China
| | - Chunlin Chen
- Chongqing Academy of Animal Sciences, Rongchang, China
- Chongqing Research Center of Veterinary Biological Products Engineering Technology, Chongqing, China
| | - Shaoqin Zhai
- Chongqing Academy of Animal Sciences, Rongchang, China
| | - Hongmei Tang
- Chongqing Academy of Animal Sciences, Rongchang, China
| | - Maixun Zhu
- Chongqing Academy of Animal Sciences, Rongchang, China
| | - Yuandi Yu
- Chongqing Academy of Animal Sciences, Rongchang, China
| | - Hua Zheng
- Chongqing Research Center of Veterinary Biological Products Engineering Technology, Chongqing, China
| |
Collapse
|