1
|
Yang W, Cui M, Yang P, Liu C, Han X, Yao W, Li Z. Gut microbiota and blood biomarkers in IBD-Related arthritis: insights from mendelian randomization. Sci Rep 2025; 15:514. [PMID: 39747467 PMCID: PMC11696716 DOI: 10.1038/s41598-024-84116-6] [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: 09/12/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
With the ongoing rise in the incidence of inflammatory bowel disease (IBD), its extraintestinal manifestations have garnered significant attention. IBD-related arthritis is notable for its insidious onset and unpredictability, presenting considerable challenges for clinical diagnosis and management. Factors such as gut microbiota, plasma proteins, inflammatory proteins, and biomarkers found in blood and urine may be closely associated with IBD-related arthritis. However, the mechanisms by which these factors influence this condition remain poorly understood and require urgent investigation. We employed the method of linkage disequilibrium and the two-sample Mendelian randomization (MR) approach, utilizing single nucleotide polymorphisms (SNPs) identified from large-scale genome-wide association studies as instrumental variables. In this scientifically rigorous manner, we explored the potential causal relationship between gut microbiota, plasma proteins, inflammatory proteins, and blood and urine biomarkers in relation to arthritis resulting from inflammatory bowel disease (IBD). This method aids in elucidating the potential roles of these biomarkers in the development of arthritis following IBD, while minimizing the confounding factors and reverse causality commonly encountered in observational studies. To further verify and strengthen our findings, we conducted subsequent sensitivity analyses. These analyses will evaluate the strength of the association between SNPs and the studied biomarkers, as well as post-IBD arthritis, while accounting for variations in SNP distribution among populations and other potential genetic influencing factors. Through these rigorous analytical steps, our objective is to enhance the robustness and credibility of the research findings and provide more reliable scientific evidence regarding the pathogenesis of post-IBD arthritis. MR analysis provides evidence for the association between genetically predicted gut microbiota, plasma proteins, inflammatory proteins, and blood and urine biomarkers with the risk of IBD-related arthritis. This analysis investigates the characteristics of the associations between specific gut microbiota, plasma proteins, inflammatory proteins, and blood and urine biomarkers in relation to IBD-related arthritis. Among the plasma proteins, pterin-4-alpha-carbinolamine dehydratase, aldo-keto reductase family 1 member C4, cathepsin L2, angiostatin, hepatocyte growth factor-like protein, hepatitis A virus cellular receptor 2, protein O-linked mannose beta-1,4-N-acetylglucosaminyltransferase 2, epididymal-specific alpha-mannosidase, and platelet-derived growth factor receptor-like protein are associated with Crohn's disease-related arthritis. In contrast, agrin, methylenetetrahydrofolate synthetase domain-containing protein, neurotrophin-3 (NT-3) growth factor receptor, and neuropilin-1 are associated with ulcerative colitis-related arthritis. Furthermore, regarding gut bacterial pathway abundance, adenosylcobalamin, N-acetylglucosamine, N-acetylmannosamine, and N-acetylneuraminic acid degradation, as well as glycolysis metabolism and degradation pathways, are associated with Crohn's disease-related arthritis. Meanwhile, gut bacterial pathway abundance (pentose phosphate pathway) and gut microbiota abundance (Bacteroidetes, Bacteroidia, Bacteroidales, Porphyromonadaceae, Faecalibacterium, Eubacterium eligens) are linked to ulcerative colitis-related arthritis. Notably, we did not identify any connections between inflammatory protein factors, blood and urine biomarkers, and IBD-related arthritis. Lastly, in the reverse MR study, the insufficient number of SNPs available for analysis precluded the detection of a reverse causal relationship. This study employs the MR method to elucidate the potential causal relationships among gut microbiota, plasma proteins, inflammatory proteins, and blood and urine biomarkers in relation to the occurrence and progression of IBD-related arthritis. This research offers a novel perspective for a deeper understanding of the pathogenesis of IBD-related arthritis and highlights future directions for the diagnosis and treatment strategies of this condition.
Collapse
Affiliation(s)
- Wei Yang
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Jilin Province, Changchun, 130117, People's Republic of China
| | - Miao Cui
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Peng Yang
- South China Normal University, Guangdong, Guangzhou, 510006, China
| | - Chenlin Liu
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Jilin Province, Changchun, 130117, People's Republic of China
| | - Xiuzhen Han
- Jiangsu Province Hospital of Traditional Chinese Medicine, Jiangsu, Nanjing, 210004, China
| | - Wenyi Yao
- The Seventh Affiliated Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, Chongqing, 200137, China
| | - Zhenhua Li
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Jilin Province, Changchun, 130117, People's Republic of China.
| |
Collapse
|
2
|
Saxena J, Agarwal G, Das S, Kumar A, Thakkar K, Kaushik S, Srivatsava VK, Siddiqui AJ, Jyoti A. Immunopharmacological Insights into Cordyceps spp.: Harnessing Therapeutic Potential for Sepsis. Curr Pharm Des 2025; 31:823-842. [PMID: 39694962 DOI: 10.2174/0113816128326301240920040036] [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: 04/26/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 12/20/2024]
Abstract
Cordyceps spp. (CS), a well-known medicinal mushroom that belongs to Tibetan medicine and is predominantly found in the high altitudes in the Himalayas. CS is a rich reservoir of various bioactive substances including nucleosides, sterols flavonoids, peptides, and phenolic compounds. The bioactive compounds and CS extract have antibacterial, antioxidant, immunomodulatory, and inflammatory properties in addition to organ protection properties across a range of disease states. The study aimed to review the potential of CS, a medicinal mushroom, as a treatment for sepsis. While current sepsis drugs have side effects, CS shows promise due to its anti-inflammatory, antioxidant, and antibacterial properties. We have performed an extensive literature search based on published original and review articles in Scopus and PubMed. The keywords used were Cordyceps, sepsis, and inflammation. Studies indicate that CS extract and bioactive compounds target free radicals including oxidative as well as nitrosative stress, lower inflammation, and modulate the immune system, all of which are critical components in sepsis. The brain, liver, kidneys, lungs, and heart are among the organs that CS extracts may be able to shield against harm during sepsis. Traditional remedies with anti-inflammatory and protective qualities, such as Cordyceps mushrooms, are promising in sepsis. However, more research including clinical trials is required to validate the usefulness of CS metabolites in terms of organ protection and fight infections in sepsis.
Collapse
Affiliation(s)
- Juhi Saxena
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara, Gujarat, India
| | - Gaurang Agarwal
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Sarvjeet Das
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Anshu Kumar
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| | - Krish Thakkar
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara, Gujarat, India
| | - Sanket Kaushik
- Amity Institute of Biotechnology, Amity University, Jaipur, Rajasthan, India
| | | | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Anupam Jyoti
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
3
|
Xie Q, Gong S, Cao J, Li A, Kulyar MF, Wang B, Li J. Mesenchymal stem cells: a novel therapeutic approach for feline inflammatory bowel disease. Stem Cell Res Ther 2024; 15:409. [PMID: 39522034 PMCID: PMC11550560 DOI: 10.1186/s13287-024-04038-y] [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: 09/20/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) poses a significant and growing global health challenge, affecting both humans and domestic cats. Research on feline IBD has not kept pace with its widespread prevalence in human populations. This study aimed to develop a model of feline IBD by incorporating dextran sulfate sodium (DSS) to evaluate the therapeutic potential of MSCs and to elucidate the mechanisms that enhance their action. METHODS We conducted a comprehensive clinical assessment, including magnetic resonance imaging (MRI), endoscopy, and histopathological examination. Additionally, alterations in intestinal microbiota were characterized by 16 S rDNA sequencing, and the influence of MSCs on IBD-related gene expression was investigated through transcriptome analysis. RESULTS According to our findings, MSC treatment significantly mitigated DSS-induced clinical manifestations, reduced inflammatory cell infiltration, decreased the production of inflammatory mediators, and promoted mucosal repair. Regarding the intestinal microbiota, MSC intervention effectively corrected the DSS-induced dysbiosis, increasing the presence of beneficial bacteria and suppressing the proliferation of harmful bacteria. Transcriptome analysis revealed the ability of MSCs to modulate various inflammatory and immune-related signaling pathways, including cytokine-cytokine receptor interactions, TLR signaling pathways, and NF-κB pathways. CONCLUSION The collective findings indicate that MSCs exert multifaceted therapeutic effects on IBD, including the regulation of intestinal microbiota balance, suppression of inflammatory responses, enhancement of intestinal barrier repair, and modulation of immune responses. These insights provide a solid scientific foundation for employing MSCs as an innovative therapeutic strategy for IBD and pave the way for future clinical explorations.
Collapse
Affiliation(s)
- Qiyun Xie
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Saisai Gong
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Jintao Cao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Aoyun Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, P.R. China
| | - Md F Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Bingyun Wang
- School of Life Science and Engineering, Foshan University, Foshan, P.R. China.
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, P.R. China.
| |
Collapse
|
4
|
Wang S, Liu W, Wei B, Wang A, Wang Y, Wang W, Gao J, Jin Y, Lu H, Ka Y, Yue Q. Traditional herbal medicine: Therapeutic potential in acute gouty arthritis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118182. [PMID: 38621464 DOI: 10.1016/j.jep.2024.118182] [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: 01/15/2024] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute gouty arthritis (AGA) is characterized by a rapid inflammatory reaction caused by the build-up of monosodium urate (MSU) crystals in the tissues surrounding the joints. This condition often associated with hyperuricemia (HUA), is distinguished by its symptoms of intense pain, active inflammation, and swelling of the joints. Traditional approaches in AGA management often fall short of desired outcomes in clinical settings. However, recent ethnopharmacological investigations have been focusing on the potential of Traditional Herbal Medicine (THM) in various forms, exploring their therapeutic impact and targets in AGA treatment. AIM OF THE REVIEW This review briefly summarizes the current potential pharmacological mechanisms of THMs - including active ingredients, extracts, and prescriptions -in the treatment of AGA, and discusses the relevant potential mechanisms and molecular targets in depth. The objective of this study is to offer extensive information and a reference point for the exploration of targeted AGA treatment using THMs. MATERIALS AND METHODS This review obtained scientific publications focused on in vitro and in vivo studies of anti-AGA THMs conducted between 2013 and 2023. The literature was collected from various journals and electronic databases, including PubMed, Elsevier, ScienceDirect, Web of Science, and Google Scholar. The retrieval and analysis of relevant articles were guided by keywords such as "acute gouty arthritis and Chinese herbal medicine," "acute gouty arthritis herbal prescription," "acute gouty arthritis and immune cells," "acute gouty arthritis and inflammation," "acute gouty arthritis and NOD-like receptor thermoprotein domain associated protein 3 (NLRP3)," "acute gouty arthritis and miRNA," and "acute gouty arthritis and oxidative stress." RESULTS We found that AGA has a large number of therapeutic targets, highlighting the effectiveness the potential of THMs in AGA treatment through in vitro and in vivo studies. THMs and their active ingredients can mitigate AGA symptoms through a variety of therapeutic targets, such as influencing macrophage polarization, neutrophils, T cells, natural killer (NK) cells, and addressing factors like inflammation, NLRP3 inflammasome, signaling pathways, oxidative stress, and miRNA multi-target interactions. The anti-AGA properties of THMs, including their active components and prescriptions, were systematically summarized and categorized based on their respective therapeutic targets. CONCLUSION phenolic, flavonoid, terpenoid and alkaloid compounds in THMs are considered the key ingredients to improve AGA. THMs and their active ingredients achieve enhanced efficacy through interactions with multiple targets, of which NLRP3 is a main therapeutic target. Nonetheless, given the intricate composition of traditional Chinese medicine (TCM), additional research is required to unravel the underlying mechanisms and molecular targets through which THMs alleviate AGA.
Collapse
Affiliation(s)
- Siwei Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China.
| | - Bowen Wei
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Aihua Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yiwen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Wen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Jingyue Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yue Jin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Hang Lu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Yuxiu Ka
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Qingyun Yue
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| |
Collapse
|
5
|
Fan QQ, Zhai BT, Zhang D, Zhang XF, Cheng JX, Guo DY, Tian H. Study on the Underlying Mechanism of Yinhua Gout Granules in the Treatment of Gouty Arthritis by Integrating Transcriptomics and Network Pharmacology. Drug Des Devel Ther 2024; 18:3089-3112. [PMID: 39050804 PMCID: PMC11268870 DOI: 10.2147/dddt.s475442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Purpose Yinhua Gout Granules (YGG) is a traditional Chinese medicine preparation with a variety of pharmacological effects, and its clinical efficacy in the treatment of gouty arthritis (GA) has been fully confirmed. However, the pharmacodynamic basis of YGG and its anti-inflammatory mechanism of action in GA are unknown. The objective of this study was to identify the active components and molecular mechanisms of YGG in the treatment of GA. Methods Ultra-performance liquid chromatography-electrospray ionization tandem mass spectrometry (UPLC-ESI-MS/MS) and network pharmacology were used to identify and predict the potential active ingredients and related signaling pathways. Then, we revealed the anti-GA effects of YGG based on pharmacodynamic experiments in GA rats. Finally, we integrated transcriptomics and network pharmacology to elucidate the potential mechanism of action and verified the putative mechanism by molecular docking, immunohistochemical (IHC) and Western blot. Results We have identified 10 major active components of YGG that may have anti-GA effects, such as ferulic acid, rutin, luteolin, etc. Using molecular docking, we found that 10 major compounds could bind well to TNF, PTGS2, IL-6, IL1β, NOS2 and PTGS1, and the binding energies were all less than -5 kcal/mol. Animal studies have shown that YGG can improve joint inflammation and inflammatory cell infiltration, reduce serum UA, BUN and Cr levels (p<0.01), and decrease IL-1β, IL-6, TNF-α, COX-2 and PGE2 levels in synovial tissue (p<0.01), which are associated with the pathogenesis of GA. IHC and Western blot results showed that YGG could regulate TLR4/MYD88/NF-κB pathway to inhibit the inflammatory response induced by GA. Conclusion This study found that YGG could not only improve the disease of GA by inhibiting the production of UA in the body, but also target the regulation of TLR4/MYD88/NF-κB signaling pathway through a variety of active components to achieve effective therapeutic effects on GA.
Collapse
Affiliation(s)
- Qiang-qiang Fan
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Bing-tao Zhai
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Dan Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Xiao-fei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jiang-xue Cheng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Dong-yan Guo
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Huan Tian
- Xi’an Hospital of Traditional Chinese Medicine, Xi’an, 712046, People’s Republic of China
| |
Collapse
|
6
|
Liu W, Dong P, Li C, Guo W, Zhao K, Man S, Zhang L, Wu H, Song H. Clinical implications of persistent joint pain after gout flare. REUMATOLOGIA CLINICA 2024; 20:291-296. [PMID: 38991822 DOI: 10.1016/j.reumae.2024.03.003] [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: 12/13/2023] [Revised: 02/05/2024] [Accepted: 03/10/2024] [Indexed: 07/13/2024]
Abstract
OBJECTIVES A subset of gout patients developed persistent joint pain after flares. Analysis of this clinical phenomenon may shed further light on the factors related to worsening gout and even provide clues to its pathogenesis. METHODS We analyzed the clinical, laboratory, and ultrasound data of gout patients to explore the associations of these data with persistent joint pain after gout flares. RESULTS A total of 1029 gout patients were included: 182 (17.7%) patients with persistent joint pain and 847 (82.3%) patients with nonpersistent joint pain. Patients with persistent joint pain had more total involved joints, more gout flares in the past year, and more joints with simultaneous gout flares (P<0.01). Among the ultrasound-detected lesions, patients with persistent joint pain had a higher incidence of tophus (36.4% vs. 21.1%) and bone erosion (18.6% vs. 8.6%) (P<0.05). Higher UA and lower TBil were found in patients with persistent joint pain (P<0.001). Hypertension (54.9% vs. 38.7%) and metabolic syndrome (58.8% vs. 46.4%) were both more frequent in patients with persistent joint pain (P<0.05). TBil was negatively correlated with the incidence of persistent joint pain (P<0.001, r=-0.190), UA values (P<0.001, r=-0.125), and metabolic syndrome scores (P<0.001, r=-0.192). A correlation curve was fitted using LOESS (locally weighted region). CONCLUSION Persistent joint pain after gout flares is a marker of increased disease burden in gout. The significance of the level of total bilirubin for the exacerbation of gout deserves further study.
Collapse
Affiliation(s)
- Wei Liu
- Department of Rheumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Peng Dong
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, China
| | - Chunyan Li
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Wen Guo
- Department of Ultrasound, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Kaiping Zhao
- Department of Medical Record Management and Statistics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Siliang Man
- Department of Rheumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Liang Zhang
- Department of Orthopedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Husheng Wu
- Department of Rheumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Hui Song
- Department of Rheumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Lu Y, Wu Y, Sun L, Yang S, Kuang H, Li R, Meng Y, Wu Y. Identifying the Anti-inflammatory Effects of Astragalus Polysaccharides in Anti-N-Methyl-D-Aspartate Receptor Encephalitis: Network Pharmacology and Experimental Validation. Comb Chem High Throughput Screen 2024; 27:1022-1032. [PMID: 37587811 DOI: 10.2174/1386207326666230816162113] [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: 04/21/2023] [Revised: 05/17/2023] [Accepted: 06/09/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Astragalus polysaccharides (APS), a group of bioactive compounds obtained from the natural source Astragalus membranaceus (AM), exhibits numerous pharmacological actions in the central nervous system, such as anti-inflammatory, antioxidant, and immunomodulatory properties. Despite the remarkable benefits, the effectiveness of APS in treating anti- N-methyl-D-aspartate receptor (NMDAR) encephalitis and the corresponding mechanism have yet to be fully understood. As such, this study aims to investigate the impact of APS on anti-NMDAR encephalitis and explore the potential molecular network mechanism. METHODS The impact of APS intervention on mice with anti-NMDAR encephalitis was assessed, and the possible molecular network mechanism was investigated utilizing network pharmacology and bioinformatics techniques such as Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG),protein-protein interaction (PPI) network, and molecular docking. Enzymelinked immunosorbent assay (ELISA) was applied to detect the expression of core target proteins. RESULTS APS significantly ameliorated cognitive impairment and reduced susceptibility to PTZinduced seizures in mice with anti-NMDAR encephalitis, confirming the beneficial effect of APS on anti-NMDAR encephalitis. Seventeen intersecting genes were identified between APS and anti- NMDAR encephalitis. GO and KEGG analyses revealed the characteristics of the intersecting gene networks. STRING interaction in the PPI network was applied to find crucial molecules. The results of molecular docking suggested that APS may regulate interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) as potential targets in anti-NMDAR encephalitis. Furthermore, the levels of IL-1β, IL-6, and TNF-α detected by ELISA in anti-NMDAR encephalitis mice were significantly downregulated in response to the administration of APS. CONCLUSION The findings of this study demonstrate the significant role of APS in the treatment of anti-NMDAR encephalitis, as it effectively suppresses inflammatory cytokines. These results suggest that APS has the potential to be considered as a viable herbal medication for the treatment of anti-NMDAR encephalitis.
Collapse
Affiliation(s)
- Yuling Lu
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Ying Wu
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Lanfeng Sun
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Shengyu Yang
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Huimin Kuang
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Rida Li
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Youshi Meng
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Yuan Wu
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
8
|
Sharma H, Sharma N, An SSA. Unique Bioactives from Zombie Fungus ( Cordyceps) as Promising Multitargeted Neuroprotective Agents. Nutrients 2023; 16:102. [PMID: 38201932 PMCID: PMC10780653 DOI: 10.3390/nu16010102] [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: 11/17/2023] [Revised: 12/08/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Cordyceps, also known as "zombie fungus", is a non-poisonous mushroom that parasitizes insects for growth and development by manipulating the host system in a way that makes the victim behave like a "zombie". These species produce promising bioactive metabolites, like adenosine, β-glucans, cordycepin, and ergosterol. Cordyceps has been used in traditional medicine due to its immense health benefits, as it boosts stamina, appetite, immunity, longevity, libido, memory, and sleep. Neuronal loss is the typical feature of neurodegenerative diseases (NDs) (Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS)) and neurotrauma. Both these conditions share common pathophysiological features, like oxidative stress, neuroinflammation, and glutamatergic excitotoxicity. Cordyceps bioactives (adenosine, N6-(2-hydroxyethyl)-adenosine, ergosta-7, 9 (11), 22-trien-3β-ol, active peptides, and polysaccharides) exert potential antioxidant, anti-inflammatory, and anti-apoptotic activities and display beneficial effects in the management and/or treatment of neurodegenerative disorders in vitro and in vivo. Although a considerable list of compounds is available from Cordyceps, only a few have been evaluated for their neuroprotective potential and still lack information for clinical trials. In this review, the neuroprotective mechanisms and safety profile of Cordyceps extracts/bioactives have been discussed, which might be helpful in the identification of novel potential therapeutic entities in the future.
Collapse
Affiliation(s)
| | - Niti Sharma
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea;
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea;
| |
Collapse
|
9
|
Lee SH, Seo D, Lee KH, Park SJ, Park S, Kim H, Kim T, Joo IH, Park JM, Kang YH, Lim GH, Kim DH, Yang JY. Biometabolites of Citrus unshiu Peel Enhance Intestinal Permeability and Alter Gut Commensal Bacteria. Nutrients 2023; 15:nu15020319. [PMID: 36678190 PMCID: PMC9862503 DOI: 10.3390/nu15020319] [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: 12/13/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Flavanones in Citrus unshiu peel (CUP) have been used as therapeutic agents to reduce intestinal inflammation; however, the anti-inflammatory effects of their biometabolites remain ambiguous. Here, we identified aglycone-type flavanones, such as hesperetin and naringenin, which were more abundant in the bioconversion of the CUP than in the ethanol extracts of the CUP. We found that the bioconversion of the CUP induced the canonical nuclear factor-κB pathway via degradation of IκB in Caco-2 cells. To check the immune suppressive capacity of the aglycones of the CUP in vivo, we orally administered the bioconversion of the CUP (500 mg/kg) to mice for two weeks prior to the 3% dextran sulfate sodium treatment. The CUP-pretreated group showed improved body weight loss, colon length shortage, and intestinal inflammation than the control mice. We also found a significant decrease in the population of lamina propria Th17 cells in the CUP-pretreated group following dextran sodium sulfate (DSS) treatment and an increase in mRNA levels of occludin in CUP-treated Caco-2 cells. Pyrosequencing analysis revealed a decreased abundance of Alistipes putredinis and an increased abundance of Muribaculum intestinale in the feces of the CUP-pretreated mice compared to those of the control mice. Overall, these findings suggest that the pre-administration of CUP biometabolites may inhibit the development of murine colitis by modulating intestinal permeability and the gut microbiome.
Collapse
Affiliation(s)
- Se-Hui Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Dongju Seo
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Kang-Hee Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - So-Jung Park
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Sun Park
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Hyeyun Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Taekyung Kim
- Department of Biology Education, Pusan National University, Busan 46241, Republic of Korea
| | - In Hwan Joo
- Department of Pathology, College of Korean Medicine, Daejeon University, Daejeon 34520, Republic of Korea
| | - Jong-Min Park
- Department of Pathology, College of Korean Medicine, Daejeon University, Daejeon 34520, Republic of Korea
| | - Yun-Hwan Kang
- Department of Industry Promotion, National Institute for Korean Medicine Development, Geongsan 38540, Republic of Korea
| | - Gah-Hyun Lim
- Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Dong Hee Kim
- Department of Pathology, College of Korean Medicine, Daejeon University, Daejeon 34520, Republic of Korea
| | - Jin-Young Yang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
- Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
- Correspondence: ; Tel.: +82-51-510-2286; Fax: +82-51-581-2962
| |
Collapse
|