1
|
Li X, Liu X, Cai M, Wei S, Wang R, Xu N, Qu J, Wang Y. Investigation of the mechanism of Dan Zhi Qing'e Formula for treating menopausal hot flashes using UHPLC-Q-TOF MS and network pharmacology. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04277-7. [PMID: 40434420 DOI: 10.1007/s00210-025-04277-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
This study aims to investigate the material basis and underlying mechanisms of action of the Dan Zhi Qing'e Formula in the treatment of menopausal hot flashes. The composition analysis of the Dan Zhi Qing'e Formula was conducted using UHPLC-Q-TOF MS, facilitated by MassLynx V4.1 software. Subsequently, Cytoscape 3.10.1 was employed to merge the data with information from Swiss, GeneCards, and OMIM databases to identify the active components and primary targets. GO and KEGG analyses were performed to elucidate the potential mechanisms of action. Finally, the molecular docking technique was applied to validate the results. A total of 118 components, 73 blood-absorbed components, and 89 potential targets were identified. Seven key targets were obtained, which were aldose reductase (AKR1B1, UniProtKB: P15121), carbonic anhydrase 4 (CA4, UniProtKB: P22748), carbonic anhydrase 2 (CA2, UniProtKB: P00918), acetylcholinesterase (ACHE, UniProtKB: P22303), estrogen receptor beta (ESR2, UniProtKB: Q92731), cytochrome P450 19A1 (CYP19A1, UniProtKB: P11511), and matrix metalloproteinase-2 (MMP2, UniProtKB: P08253). Molecular docking studies indicate that these core components exhibit strong affinity for the identified targets. These targets contribute to the tonic function of the liver and kidneys through hormone response. The findings provide a scientific foundation for further in-depth research into the therapeutic mechanisms of Dan Zhi Qing'e Formula for menopausal hot flashes.
Collapse
Affiliation(s)
- Xinyu Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, West Zone Tuanbo New City, Jinghai District, Tianjin, 301617, China
| | - Xinyu Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, West Zone Tuanbo New City, Jinghai District, Tianjin, 301617, China
| | - Minghui Cai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, NO. 88 Changling Road, Xiqing District, Tianjin, 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Shuang Wei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, West Zone Tuanbo New City, Jinghai District, Tianjin, 301617, China
| | - Rui Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, West Zone Tuanbo New City, Jinghai District, Tianjin, 301617, China
| | - Nanjian Xu
- Department of Spine Surgery, No.6 Hospital in Ningbo, 1059# Zhongshan East Road, Ningbo, 315040, Zhejiang Province, People's Republic of China.
| | - Jingtian Qu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, NO. 88 Changling Road, Xiqing District, Tianjin, 300381, China.
| | - Yuming Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, No.10 Poyang Lake Road, West Zone Tuanbo New City, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
2
|
Mbock MA, Kamkumo RG, Iqbal N, Feudjou Fouatio W, Shukla R, Chandra A, Tsouh Fokou PV, Ngueguim Tsofack F, Bibi I, Fekam Boyom F, Essia Ngang JJ, Boyomo O, Nganso Ditchou YO, Nkengfack AE, Lenta Ndjakou B, Sewald N, Siwe Noundou X, Dimo T. Safety assessment of Detarium microcarpum Guil. & Perr. (Leguminosae) root bark extract in rats: Acute, sub-acute toxicity, ADMET, molecular docking, and molecular dynamics simulations approach. Fitoterapia 2025; 184:106635. [PMID: 40398517 DOI: 10.1016/j.fitote.2025.106635] [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: 03/02/2025] [Revised: 04/18/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
Since the last decades, Detarium microcarpum have been used and studied due to the significant properties to threat various ailments. It is therefore crucial to ascertain the safety properties of is phytoconstituents. The present study was taken up to investigate the oral acute toxicity, 28 days repeated oral sub-acute toxicity study of D. microcarpum root bark extract in Albino Wistar rats, molecular docking, and molecular dynamic simulation. Based on acute oral toxicity, the LD50 value of the extract was considered "non-toxic" up to a 5000 mg/kg b.w. The regular administration of the extract post 28-day treatment was also qualified as safe based on the body and organ weight, hematological, biochemical, and histopathological results in the sub-acute toxicity assay. The majority of the previous identified and isolated compounds passed the ADMET analysis, and the molecular docking identified 1,7-dihydroxy-6-methylxanthone (15) as the top one candidate with an effective binding affinity of -8.1, and - 7.5 (kcal/mol), for P450 and UGT2B7 proteins respectively. This compound was then further assessed using MD simulation, which verified the molecule's stability and binding to the targeted protein. Collectively these data build the foundation of support demonstrating that D. microcarpum root bark extract could be considered as a promising medicinal drug and concluded its suitable used for pharmacological purposes.
Collapse
Affiliation(s)
- Michel Arnaud Mbock
- Department of Biochemistry, Laboratory of Biochemistry, Faculty of Science, University of Douala, PO Box 24 157, Douala, Cameroon; Department of Microbiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon; Department of Animal Biology and Physiology, Faculty of Science University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon; Department of Biochemistry, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon.
| | - Raceline Gounoue Kamkumo
- Department of Animal Biology and Physiology, Faculty of Science University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon; Department of Biochemistry, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon.
| | - Naveed Iqbal
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - William Feudjou Fouatio
- Laboratory of Phytochemistry, Centre for Research on Medicinal Plants and Traditional Medicine, Institute of Medical Research and Medicinal Plants Studies, P.O. Box 13033, Yaoundé, Cameroon; Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology Waknaghat, Solan, 173215, H.P., India
| | - Anshuman Chandra
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology Waknaghat, Solan, 173215, H.P., India
| | | | - Florence Ngueguim Tsofack
- Department of Animal Biology and Physiology, Faculty of Science University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon
| | - Ishrat Bibi
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Fabrice Fekam Boyom
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Jean Justin Essia Ngang
- Department of Microbiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon
| | - Onana Boyomo
- Department of Microbiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon
| | | | - Augustin Ephrem Nkengfack
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Bruno Lenta Ndjakou
- Department of Chemistry, Higher Teacher Training College, University of Yaoundé 1, P.O. Box 47, Yaoundé, Cameroon
| | - Norbert Sewald
- Department of Chemistry, Bielefeld University, P.O. Box 100131, 33501 Bielefeld, Germany
| | - Xavier Siwe Noundou
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0204, South Africa.
| | - Théophile Dimo
- Department of Animal Biology and Physiology, Faculty of Science University of Yaounde I, P.O. Box 812, Yaoundé, Cameroon
| |
Collapse
|
3
|
Wu R, Zhao XJ, Du Y, Dong Y, Song X, Zhu Y. Lipid metabolic disorders and their impact on cartilage endplate and nucleus pulposus function in intervertebral disk degeneration. Front Nutr 2025; 12:1533264. [PMID: 40129665 PMCID: PMC11931516 DOI: 10.3389/fnut.2025.1533264] [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: 11/23/2024] [Accepted: 02/06/2025] [Indexed: 03/26/2025] Open
Abstract
Lipid metabolism encompasses the processes of digestion, absorption, synthesis, and degradation of fats within biological systems, playing a crucial role in sustaining normal physiological functions. Disorders of lipid metabolism, characterized by abnormal blood lipid levels and dysregulated fatty acid metabolism, have emerged as significant contributors to intervertebral disk degeneration (IDD). The pathogenesis of IDD is multifaceted, encompassing genetic predispositions, nutritional and metabolic factors, mechanical stressors, trauma, and inflammatory responses, which collectively facilitate the progression of IDD. Although the precise mechanisms underlying IDD remain incompletely elucidated, there is substantial consensus regarding the close association between lipid metabolism disorders and its development. Intervertebral disks are essential for maintaining spinal alignment. Their primary functions encompass shock absorption, preservation of physiological curvature, facilitation of movement, and provision of stability. The elasticity and thickness of these disks effectively absorb daily impacts, safeguard the spine, uphold its natural curvature and flexibility, while also creating space for nerve roots to prevent compression and ensure normal transmission of nerve signals. Research indicates that such metabolic disturbances may compromise the functionality of cartilaginous endplates (CEP) and nucleus pulposus (NP), thereby facilitating IDD's onset and progression. The CEP is integral to internal material exchange and shock absorption while mitigating NP herniation under mechanical load conditions. As the central component of intervertebral disks, NP is essential for maintaining disk height and providing shock-absorbing capabilities; thus, damage to these critical structures accelerates IDD progression. Furthermore, lipid metabolism disorders contribute to IDD through mechanisms including activation of endoplasmic reticulum stress pathways, enhancement of oxidative stress levels, induction of cellular pyroptosis alongside inhibition of autophagy processes-coupled with the promotion of inflammation-induced fibrosis and fibroblast proliferation leading to calcification within intervertebral disks. This review delineates the intricate interplay between lipid metabolism disorders and IDD; it is anticipated that advancing our understanding of this pathogenesis will pave the way for more effective preventive measures and therapeutic strategies against IDD in future research.
Collapse
Affiliation(s)
- Ruixia Wu
- Inner Mongolia Medical University, Hohhot, China
| | - Xiao Juan Zhao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yaxin Du
- Inner Mongolia Medical University, Hohhot, China
| | - Yizhi Dong
- Inner Mongolia Medical University, Hohhot, China
| | - Xinyue Song
- Inner Mongolia Medical University, Hohhot, China
| | - Yong Zhu
- Peking University Cancer Hospital Inner Mongolia Hospital, Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
4
|
Zhang Y, Xing D, Liu Y, Sha S, Xiao Y, Liu Z, Yin Q, Gao Z, Liu W. CREG1 attenuates intervertebral disc degeneration by alleviating nucleus pulposus cell pyroptosis via the PINK1/Parkin-related mitophagy pathway. Int Immunopharmacol 2025; 147:113974. [PMID: 39746276 DOI: 10.1016/j.intimp.2024.113974] [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: 10/28/2024] [Revised: 12/12/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Intervertebral disc degeneration (IVDD) is a chronic degenerative disease with a complex pathophysiological mechanism. Increasing evidence suggests that the NOD-like receptor thermal protein domain associated protein 3 (NLRP3)-mediated pyroptosis of nucleus pulposus cells (NPCs) plays a crucial role in the pathological progression of IVDD. Pyroptosis is a novel form of programmed cell death characterized by the formation of plasma membrane pores by gasdermin family proteins, leading to cell swelling, membrane rupture, and the release of inflammatory cytokines, which trigger an inflammatory response. The close relationship between pyroptosis and mitophagy has been previously described in various diseases, but the crosstalk between pyroptosis and mitophagy in IVDD remains unexplored. Cellular repressor of E1A-stimulated genes 1 (CREG1) is a secreted glycoprotein involved in cell differentiation and homeostasis regulation and has been shown to promote lysosomal biogenesis and function. However, the potential role and underlying mechanisms of CREG1 in the progression of IVDD have not yet been reported. In this study, we first observed that CREG1 is downregulated following IVDD and that pyroptosis occurs. Furthermore, CREG1 knockdown inhibited NPC proliferation and exacerbated apoptosis and degeneration. Moreover, we confirmed that CREG1 knockdown induced NLRP3 activation while also leading to mitophagy inhibition and mitochondrial dysfunction in NPCs. CREG1 overexpression ameliorated LPS-induced mitophagy inhibition and mitochondrial dysfunction by promoting PINK1/Parkin-mediated mitophagy, thereby suppressing NLRP3 inflammasome activation. However, these protective effects were reversed by pretreatment with the mitophagy inhibitor cyclosporin A (CsA). In a rat model of IVDD, imaging and histological assessments revealed that CREG1 overexpression effectively alleviated the progression of IVDD. Additionally, CREG1 overexpression reduced the expression of NLRP3, caspase-1, and IL-1β while increasing the expression of collagen II, PINK1 and LC3, delaying the course of IVDD. Overall, this study highlights the importance of the interplay between CREG1-mediated regulation of mitophagy and pyroptosis in the pathogenesis of IVDD, identifying CREG1 as a promising therapeutic target for IVDD treatment.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Spinal Surgery, Zhongda Hospital, College of Medicine, Southeast University, 87 Dingjiaqiao Street, Nanjing, Jiangsu 210009, People's Republic of China
| | - Deguo Xing
- Department of Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Yi Liu
- Department of Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China.
| | - Shiyu Sha
- Department of Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Yueying Xiao
- Department of Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Zhonghao Liu
- Department of Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Qingfeng Yin
- Department of Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China
| | - Zengxin Gao
- Department of Spinal Surgery, Zhongda Hospital, College of Medicine, Southeast University, 87 Dingjiaqiao Street, Nanjing, Jiangsu 210009, People's Republic of China
| | - Wenguang Liu
- Department of Orthopaedics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, People's Republic of China.
| |
Collapse
|
5
|
Li Q, Guo R, Wu Z, Zhao C, Chen X, Wang H, Shen C. Endplate chondrocyte-derived exosomal miR-128-3p mitigates intervertebral disc degeneration by targeting TRAF6 via the miR-128-3p/TRAF6 axis to suppress pyroptosis. Int Immunopharmacol 2024; 143:113620. [PMID: 39550843 DOI: 10.1016/j.intimp.2024.113620] [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: 10/09/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/19/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a leading cause of chronic back pain and significantly impacts quality of life. The pathogenesis of IVDD is largely driven by inflammation, pyroptosis, and extracellular matrix (ECM) degradation, which current therapies fail to adequately address. In this study, we explore the therapeutic potential of exosomes derived from endplate chondrocytes (EPCs), with a particular focus on the microRNA miR-128-3p. Our findings reveal that exosomes isolated from third-generation EPCs, enriched with miR-128-3p, exhibit potent anti-inflammatory and anti-pyroptotic effects in lipopolysaccharide-treated nucleus pulposus cells, which are key contributors to IVDD pathology. Specifically, we demonstrate that miR-128-3p delivered via EPC-derived exosomes directly targets TRAF6, effectively suppressing activation of the NF-κB signaling pathway, which is known to play a pivotal role in inflammation and ECM breakdown, leading to a marked reduction in pro-inflammatory cytokine release and mitigation of ECM degradation. Importantly, third-generation EPC exosomes, with higher levels of miR-128-3p, showed superior efficacy compared to fifth-generation EPCs, underscoring the critical role of miR-128-3p in mediating these protective effects. Our research highlights the promise of EPC-derived exosomes, particularly those rich in miR-128-3p, as a novel, cell-free therapeutic approach for IVDD. Unlike current treatments that focus primarily on symptom management, our approach targets key molecular pathways underlying IVDD progression, including inflammation, pyroptosis, and ECM degradation. By elucidating the miR-128-3p/TRAF6 axis, this study provides a foundation for the development of targeted, biologically based interventions aimed at halting or even reversing IVDD, thereby offering hope for more effective and lasting therapeutic options.
Collapse
Affiliation(s)
- Qiuwei Li
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
| | - Ruocheng Guo
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
| | - Zuomeng Wu
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
| | - Chenhao Zhao
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China
| | - Xuewu Chen
- Spinal Surgery Division of Yijishan Hospital and Wannan Medical College in Wuhu, Anhui 241000, China
| | - Hong Wang
- Spinal Surgery Division of Yijishan Hospital and Wannan Medical College in Wuhu, Anhui 241000, China
| | - Cailiang Shen
- Department of Orthopedics and Spine Surgery, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China; Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, Anhui 230022, China.
| |
Collapse
|
6
|
Gong W, Zhu Y, Wang L. Qiang Jin Mixture Promotes Osteogenic Differentiation of MC3T3-E1 Cells via BMP2/Smads Pathway and its Network Pharmacology Study. Mol Biotechnol 2024:10.1007/s12033-024-01313-4. [PMID: 39527391 DOI: 10.1007/s12033-024-01313-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
The study aimed to explore the potential of QiangJin mixture (QJM), a Chinese herbal compound prescription, in regulating MC3T3-E1 cell differentiation and to analyze the ingredients and therapeutic targets of QJM against osteoporosis based on network pharmacology. MC3T3-E1 cells were incubated with different concentrations of QJM-contained rat serum (5, 10, or 20%). After 14 days of cell culture, Alizarin Red staining was performed to assess the mineralization ability of osteoblasts. RT-qPCR was used to measure mRNA levels of osteogenesis-related genes. Western blot was conducted to measure protein levels of factors related to the BMP2/Smads pathway. Functional and pathway enrichment of overlapping targets for QJM and osteoporosis were analyzed using gene ontology and KEGG analyses. As shown by experimental results, QJM-contained serum led to calcium deposition, increased expression levels of osteogenesis-related genes, and activated BMP2/Smad/Runx2 signaling in MC3T3-E1 cells. A total of 125 active compounds and 162 disease-related targets were identified. The core targets were MAPK8, TP53, ESR1, STAT3, MAPK3, IL6, NFKB1, JUN, MAPK1 and AKT1. In conclusion, QJM promotes the osteogenic differentiation of MC3T3-E1 cells by activating the BMP2/Smads signaling. Additionally, QJM is an anti-osteoporotic mixture by regulating diverse therapeutic targets and signaling pathways.
Collapse
Affiliation(s)
- Weiyue Gong
- Department of Pharmacy, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, No. 315 South Street, Wuxing District, Huzhou, 313000, China
| | - Yao Zhu
- Department of Pharmacy, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, No. 315 South Street, Wuxing District, Huzhou, 313000, China
| | - Limin Wang
- Department of Pharmacy, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, No. 315 South Street, Wuxing District, Huzhou, 313000, China.
| |
Collapse
|
7
|
Sun Y, Li Z, Duan J, Liu E, Sun F, Yang L, Chen L, Yang S. Unveiling the Gut-Disc Axis: How Microbiome Dysbiosis Accelerates Intervertebral Disc Degeneration. J Inflamm Res 2024; 17:8271-8280. [PMID: 39525319 PMCID: PMC11549883 DOI: 10.2147/jir.s487936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
The gut microbiome (GM), often referred to as the second genome of the human body, plays a crucial role in various metabolic processes and mediates the development of numerous diseases. Intervertebral disc degeneration (IDD) is an age-related degenerative spinal disease characterized by the loss of disc height, hydration, and integrity, leading to pain and reduced mobility. Although the pathogenesis of IDD is not fully understood, recent studies suggest that dysbiosis of the gut microbiome may accelerate the progression of IDD through multiple mechanisms. This article begins by discussing the potential relationship between GM dysbiosis and human diseases, followed by a comprehensive review of the regulatory mechanisms of GM in skeletal diseases within the gut-disc axis framework. Furthermore, it explores three potential pathways through which GM dysbiosis may mediate the development of IDD: immunomodulation, bacterial translocation and colonization, and the decomposition and absorption of intestinal metabolites. These pathways can disrupt disc cell homeostasis and promote degenerative changes. Finally, this paper summarizes for the first time the potential therapeutic approaches for delaying IDD by targeting the gut-disc axis, providing new insights into the pathogenesis and regenerative repair strategies for IDD.
Collapse
Affiliation(s)
- Yu Sun
- Graduate School of Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Zhaoyong Li
- Graduate School of Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Jiahao Duan
- Graduate School of Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Enxu Liu
- Graduate School of Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Fei Sun
- Graduate School of Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China
| | - Lei Yang
- Department of Orthopedics, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Long Chen
- Department of Orthopedics, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Shaofeng Yang
- Department of Orthopedics, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| |
Collapse
|
8
|
Wang N, Chen S, Xie Y, Liu X, Xi Z, Li J, Xue C, Deng R, Min W, Kang R, Xie L. The Sanbi Decoction alleviates intervertebral disc degeneration in rats through intestinal flora and serum metabolic homeostasis modulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 127:155480. [PMID: 38484462 DOI: 10.1016/j.phymed.2024.155480] [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: 11/13/2023] [Revised: 01/29/2024] [Accepted: 02/23/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is an essential cause of low back pain (LBP), the incidence of which has risen in recent years and is progressively younger, but treatment options are limited, placing a serious economic burden on society. Sanbi decoction (SBD) is an important classical formula for the treatment of IVDD, which can significantly improve patients' symptoms and is a promising alternative therapy. PURPOSE The aim of this study is to investigate the safety and efficacy of SBD in the treatment of IVDD and to explore the underlying mechanisms by using an integrated analytical approach of microbiomics and serum metabolomics, as well as by using molecular biology. METHODS A rat IVDD puncture model was established and treated by gavage with different concentrations of SBD, and clean faeces, serum, liver, kidney, and intervertebral disc (IVD) were collected after 4 weeks. We assessed the safety by liver and kidney weighing, functional tests and tissue staining, the expression of tumor necrosis factor-alpha (TNF-ɑ), interleukin 1β (IL-1β) and interleukin 6 (IL-6) inflammatory factors in serum was detected by ELISA kits, and X-ray test, magnetic resonance imaging (MRI) examination, immunohistochemistry (IHC), western blotting (WB), hematoxylin-eosin (HE) staining and safranin O-fast green (SO/FG) staining were used to assess the efficacy. Finally, we performed 16S rRNA sequencing analysis on the faeces of different groups and untargeted metabolomics on serum and analyzed the association between them. RESULTS SBD can effectively reduce the inflammatory response, regulate the metabolic balance of extracellular matrix (ECM), improve symptoms, and restore IVD function. In addition, SBD can significantly improve the diversity of intestinal flora and maintain the balance. At the phylum level, SBD greatly increased the relative abundance of Patescibacteria and Actinobacteriota and decreased the relative abundance of Bacteroidota. At the genus level, SBD significantly increased the relative abundance of Clostridia_UCG-014, Enterorhabdus, and Adlercreutzia, and decreased the relative abundance of Ruminococcaceae_UCG-005 (p < 0.05). Untargeted metabolomics indicated that SBD significantly improved serum metabolites and altered serum expression of 4alpha-phorbol 12,13-didecanoate (4alphaPDD), euscaphic acid (EA), alpha-muricholic acid (α-MCA), 5-hydroxyindoleacetic acid (5-HIAA), and kynurenine (Kyn) (p < 0.05), and the metabolic pathways were mainly lipid metabolism and amino acid metabolism. CONCLUSIONS This study demonstrated that SBD can extensively regulate intestinal flora and serum metabolic homeostasis to reduce inflammatory response, inhibit the degradation of ECM, restore IVD height and water content to achieve apparent therapeutic effect for IVDD.
Collapse
Affiliation(s)
- Nan Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210028, China
| | - Shuang Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210028, China
| | - Yimin Xie
- Department of Orthopedics, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210029, China
| | - Xin Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210028, China
| | - Zhipeng Xi
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210028, China
| | - Jingchi Li
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Congyang Xue
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210028, China
| | - Rongrong Deng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210028, China
| | - Wen Min
- Department of Orthopedics, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210029, China.
| | - Ran Kang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210028, China.
| | - Lin Xie
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210028, China.
| |
Collapse
|
9
|
Yi B, Lv F, Zhang N, Lin J, Xu K, Li C, Li P, Zhao M. Exploring the pharmacological mechanisms of Biyan Qingdu Granula in the treatment after nasopharyngeal carcinoma radiotherapy based on UPLC/Q-TOF MS, network pharmacology and molecular docking. J Pharm Biomed Anal 2024; 239:115830. [PMID: 38096633 DOI: 10.1016/j.jpba.2023.115830] [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: 07/21/2023] [Revised: 09/30/2023] [Accepted: 10/27/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Biyan Qingdu Granula (BYQD) is a traditional Chinese medicine (TCM) formula commonly used for post-radiotherapy treatment of nasopharyngeal carcinoma (NPC). Despite its extensive use, the underlying pharmacological mechanisms have yet to be fully elucidated. METHODS UPLC/Q-TOF MS was used to comprehensively analyze the chemical composition of BYQD. Additionally, an everted gut sac model, coupled with UPLC/Q-TOF MS, was used to screen and identify the active ingredients. Subsequently, we conducted a network pharmacological analysis to delve into the potential mechanisms of these active ingredients. Molecular docking experiments were also performed to assess the interactions between active ingredients and potential core targets. RESULTS The findings revealed the identification of 62 identical ingredients upon comparing the sample solution and intestinal absorbed solution of BYQD. We constructed a protein-protein interaction (PPI) network, which led to the identification of five core targets, namely, TP53, STAT3, MAPK1, SRC and AKT1. Through the construction of a drug-active ingredient-intersection target network, we identified Quercetin, Luteolin, Eupatilin, Magnoflorine, Acacetin and other compound as potential active ingredients. Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis suggested that pathways in cancer, PI3K-Akt signaling pathway, lipid and atherosclerosis, proteoglycans in cancer, and the MAPK signaling pathway might play the key roles in the treatment of NPC after radiotherapy using BYQD. Molecular docking results corroborated strong binding activity between the putative core targets and the corresponding key active ingredients. CONCLUSION This study provides a preliminary revelation of the active ingredients and potential pharmacological mechanisms of BYQD in the post-radiotherapy treatment of NPC. These findings establish a vital theoretical basis and serve as a scientific reference for the future investigating the pharmacological mechanisms and clinical application of BYQD.
Collapse
Affiliation(s)
- Bojiao Yi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China; Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fengyi Lv
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, Shandong, China
| | - Na Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China; Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Juan Lin
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Keyi Xu
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Chuyuan Li
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Peng Li
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China.
| | - Min Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
10
|
Luo Z, Wei Z, Zhang G, Chen H, Li L, Kang X. Achilles' Heel-The Significance of Maintaining Microenvironmental Homeostasis in the Nucleus Pulposus for Intervertebral Discs. Int J Mol Sci 2023; 24:16592. [PMID: 38068915 PMCID: PMC10706299 DOI: 10.3390/ijms242316592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
The dysregulation of intracellular and extracellular environments as well as the aberrant expression of ion channels on the cell membrane are intricately linked to a diverse array of degenerative disorders, including intervertebral disc degeneration. This condition is a significant contributor to low back pain, which poses a substantial burden on both personal quality of life and societal economics. Changes in the number and function of ion channels can disrupt the water and ion balance both inside and outside cells, thereby impacting the physiological functions of tissues and organs. Therefore, maintaining ion homeostasis and stable expression of ion channels within the cellular microenvironment may prove beneficial in the treatment of disc degeneration. Aquaporin (AQP), calcium ion channels, and acid-sensitive ion channels (ASIC) play crucial roles in regulating water, calcium ions, and hydrogen ions levels. These channels have significant effects on physiological and pathological processes such as cellular aging, inflammatory response, stromal decomposition, endoplasmic reticulum stress, and accumulation of cell metabolites. Additionally, Piezo 1, transient receptor potential vanilloid type 4 (TRPV4), tension response enhancer binding protein (TonEBP), potassium ions, zinc ions, and tungsten all play a role in the process of intervertebral disc degeneration. This review endeavors to elucidate alterations in the microenvironment of the nucleus pulposus during intervertebral disc degeneration (IVDD), with a view to offer novel insights and approaches for exploring therapeutic interventions against disc degeneration.
Collapse
Affiliation(s)
- Zhangbin Luo
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Ziyan Wei
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Haiwei Chen
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
| | - Lei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| |
Collapse
|