1
|
Singh P, Borkar M, Doshi G. Network pharmacology approach to unravel the neuroprotective potential of natural products: a narrative review. Mol Divers 2025:10.1007/s11030-025-11198-3. [PMID: 40279084 DOI: 10.1007/s11030-025-11198-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 04/13/2025] [Indexed: 04/26/2025]
Abstract
Aging is a slow and irreversible biological process leading to decreased cell and tissue functions with higher risks of multiple age-related diseases, including neurodegenerative diseases. It is widely accepted that aging represents the leading risk factor for neurodegeneration. The pathogenesis of these diseases involves complex interactions of genetic mutations, environmental factors, oxidative stress, neuroinflammation, and mitochondrial dysfunction, which complicate treatment with traditional mono-targeted therapies. Network pharmacology can help identify potential gene or protein targets related to neurodegenerative diseases. Integrating advanced molecular profiling technologies and computer-aided drug design further enhances the potential of network pharmacology, enabling the identification of biomarkers and therapeutic targets, thus paving the way for precision medicine in neurodegenerative diseases. This review article delves into the application of network pharmacology in understanding and treating neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and spinal muscular atrophy. Overall, this article emphasizes the importance of addressing aging as a central factor in developing effective disease-modifying therapies, highlighting how network pharmacology can unravel the complex biological networks associated with aging and pave the way for personalized medical strategies.
Collapse
Affiliation(s)
- Pankaj Singh
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai Campus, V. M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Maheshkumar Borkar
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V. M. Road, Vile Parle (W), Mumbai, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mithibai Campus, V. M. Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
2
|
Baranowska-Wójcik E, Gajowniczek-Ałasa D, Pawlikowska-Pawlęga B, Szwajgier D. The Potential Role of Phytochemicals in Alzheimer's Disease. Nutrients 2025; 17:653. [PMID: 40004981 PMCID: PMC11858096 DOI: 10.3390/nu17040653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterised by memory loss and cognitive disorders. The disease has been related to the presence of so-called senile plaques forming due to the buildup of amyloid β in the hippocampus. The AD therapies developed to date continue to prove insufficient, while long-term exposure to synthetic drugs tends to lead to serious side effects, which is why potential herbal treatments are generally preferable to conventional drug regimens and, as such, have been under considerable research scrutiny in recent years. There are a number of herbs, e.g., lavender Ginkgo biloba, that are already commonly employed in alleviating the symptoms of certain neurological disorders. In light of the above, the aim of the following paper is to discuss the importance of medicinal herbs, their neuroprotective properties, and their mechanisms of activity. The article presents a review of the identified therapeutic properties of phytomedicines that exhibit strong anti-Alzheimer's disease (AD) activity.
Collapse
Affiliation(s)
- Ewa Baranowska-Wójcik
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences, Skromna Street 8, 20-704 Lublin, Poland; (D.G.-A.); (D.S.)
| | - Dorota Gajowniczek-Ałasa
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences, Skromna Street 8, 20-704 Lublin, Poland; (D.G.-A.); (D.S.)
| | - Bożena Pawlikowska-Pawlęga
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland;
| | - Dominik Szwajgier
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences, Skromna Street 8, 20-704 Lublin, Poland; (D.G.-A.); (D.S.)
| |
Collapse
|
3
|
Jain S, Murmu A, Patel S. Elucidating the therapeutic mechanism of betanin in Alzheimer's Disease treatment through network pharmacology and bioinformatics analysis. Metab Brain Dis 2024; 39:1175-1187. [PMID: 38995496 DOI: 10.1007/s11011-024-01385-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
Betanin, a natural compound with anti-inflammatory and antioxidant properties, has shown promise in mitigating Alzheimer's disease (AD) by reducing amyloid plaque production. Employing network pharmacology, this study aimed to elucidate betanin's therapeutic mechanism in AD treatment. Through integrated analyses utilizing SwissTargetPrediction, STITCH, BindingDB, Therapeutic Target Database (TTD), and OMIM databases, potential protein targets of betanin in AD were predicted. Gene ontology analysis facilitated the identification of 49 putative AD targets. Subsequent gene enrichment and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway analysis revealed associations between these targets and AD. Network pharmacology techniques and molecular docking aided in prioritizing essential genes, with APP, CASP7, ITPR1, CASP8, CASP3, ITPR3, and NF-KB1 emerging as top candidates. The results provide novel insights into betanin's therapeutic efficacy, shedding light on its potential clinical application in AD treatment. By targeting key genes implicated in AD pathology, betanin demonstrates promise as a valuable addition to existing therapeutic strategies. This holistic approach emphasizes the relevance of network pharmacology and bioinformatics analysis in understanding natural chemical disease therapy processes.
Collapse
Affiliation(s)
- Smita Jain
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Kishangarh, India
| | - Ankita Murmu
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Kishangarh, India
| | - Saraswati Patel
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 602105, India.
| |
Collapse
|
4
|
Ruan X, Zhang X, Liu L, Zhang J. Mechanism of Xiaoyao San in treating non-alcoholic fatty liver disease with liver depression and spleen deficiency: based on bioinformatics, metabolomics and in vivo experiments. J Biomol Struct Dyn 2024; 42:5128-5146. [PMID: 37440274 DOI: 10.1080/07391102.2023.2231544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
Xiaoyao san (XYS) plays an important role in treatment of non-alcoholic fatty liver disease (NAFLD) with liver stagnation and spleen deficiency, but its specific mechanism is still unclear. This study aimed to investigate the material basis and mechanism by means of network pharmacology, metabolomics, systems biology and molecular docking methods. On this basis, NAFLD rat model with liver stagnation and spleen deficiency was constructed and XYS was used to intervene, and liver histopathology, biochemical detection, enzyme-linked immunosorbent assay, quantitative PCR assay and western blotting were used to further verify the mechanism. Through the above research methods, network pharmacology study showed that there were 94 targets in total for XYS in the treatment of NAFLD. Metabolomics study showed that NAFLD with liver depression and spleen deficiency had a total of 73 differential metabolites. Systems biology found that PTGS2 and PPARG were the core targets; Quercetin, kaempferol, naringenin, beta-sitosterol and stigmasterol were the core active components; AA, cAMP were the core metabolites. And molecular docking showed that the core active components can act well on the key targets. Animal experiments showed that XYS could improve liver histopathology, increase 5HT and NA, decrease INS and FBG, improve blood lipids and liver function, decrease AA, increase cAMP, down-regulate PTGS2, up-regulate PPARG, and decrease PGE2 and 15d-PGJ2. In conclusion, XYS might treat NAFLD with liver depression and spleen deficiency by down-regulating PTGS2, up-regulating PPARG, reducing AA content, increasing cAMP, improving insulin resistance, affecting glucose and lipid metabolism, inhibiting oxidative stress and inflammatory response.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Xiaofeng Ruan
- School of Acupuncture - Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaoming Zhang
- School of Acupuncture - Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Liming Liu
- School of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Liver Medicine, Hubei No.3 People's Hospital of Jianghan University, Wuhan, China
| | - Jianjun Zhang
- School of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- Department of Liver Medicine, Hubei No.3 People's Hospital of Jianghan University, Wuhan, China
| |
Collapse
|
5
|
Zhang Q, Wang F, Liu J, Li J, Zhang W, Na S, Lu J, Wang Y. Integration of transcriptomics and metabolomics reveals toxicological mechanisms of ZhuRiHeng drop pill in the 180-day repeated oral toxicity study. Front Pharmacol 2024; 15:1333167. [PMID: 38560353 PMCID: PMC10978746 DOI: 10.3389/fphar.2024.1333167] [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: 11/04/2023] [Accepted: 02/09/2024] [Indexed: 04/04/2024] Open
Abstract
Background: ZhuRiHeng Drop Pill (ZRH) is a traditional Mongolian medicinal preparation. Despite its long history of use for the treatment of coronary heart disease, there have been few toxicological studies of the safety profile of ZRH. Purpose: In order to comprehensively elucidate the underlying mechanisms behind the observed toxicity of ZRH on rat livers in the 180-day repeated oral toxicity study, we conducted a comprehensive analysis by integrating transcriptomic and metabolomic data. Methods: High-resolution mass spectrometry was conducted to evaluate the constituents of ZRH. For the acute oral toxicity study, mice were administered a dose of 32 g/(kg·d) of ZRH, while rats were instead orally administered 0.934, 1.868, or 3.736 g/(kg·d) of ZRH over a 180-day period in a 180-day repeated oral toxicity study. Conventional index and organ weights/histology were then monitored to detect any potential ZRH treatment-related toxicity. To identify key genes and metabolites involved in ZRH toxicological processes, we performed transcriptomic and metabolomic analyses of liver tissue upon ZRH treatment using RNA-seq techniques, qPCR and liquid chromatography-mass spectrometry analyses. Results: A total of 60 compounds in ZRH were identified and speculated in positive and negative ion modes. Mice in the acute toxicity study exhibited no signs of ZRH-related toxicity. In a protracted oral toxicity investigation spanning 180 days, discernible elevations in liver ratios were noted in both male and female rats across all three dose cohorts, relative to the control group (p < 0.05 or p < 0.01). Upon subjecting to ZRH treatment, our transcriptomic and qPCR analyses unveiled notable upregulation of crucial genes, exemplified by Abcb1b and Cyp2b2, known for theirs involvement in liver drug transport and metabolism function. Furthermore, our untargeted metabolomic analysis provided supplementary insights, revealing significant regulation in pyrimidine metabolism, as well as alanine, aspartate, and glutamate metabolism pathways. Conclusion: Our study unveils a panoramic understanding of the temporal, dosage-specific, and gene dimensions surrounding the metabolic and transcriptional shifts induced by ZRH exposure. As we peer into the future, recommendations emerge for further exploration, encompassing aspects such as time dynamics, dosage considerations, and gene-centric avenues to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Qian Zhang
- Inner Mongolia Key Laboratory of Chinese and Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Fang Wang
- Inner Mongolia Key Laboratory of Chinese and Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Jing Liu
- Inner Mongolia Key Laboratory of Chinese and Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Jun Li
- Inner Mongolia Key Laboratory of Chinese and Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Wei Zhang
- Inner Mongolia Key Laboratory of Chinese and Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Shengsang Na
- Inner Mongolia Key Laboratory of Chinese and Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Jingkun Lu
- Inner Mongolia Key Laboratory of Chinese and Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Yuewu Wang
- Inner Mongolia Key Laboratory of Chinese and Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
6
|
Preethy H A, Venkatakrishnan YB, Ramakrishnan V, Krishnan UM. A network pharmacological approach for the identification of potential therapeutic targets of Brahmi Nei - a complex traditional Siddha formulation. J Biomol Struct Dyn 2024:1-24. [PMID: 38459935 DOI: 10.1080/07391102.2024.2322612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/19/2024] [Indexed: 03/11/2024]
Abstract
Brahmi Nei (BN), a traditional Indian polyherbal formulation has been described in classical texts for the treatment of anxiety and depression, as well as to fortify the immune system. The individual herbs of BN have been used for treatment of wide range of disorders including cognition, inflammation, skin ailments and cancer etc., This diverse basket of therapeutic activity suggests that BN may possess therapeutic benefits to other disorders. So, the present study aims to identify the potential therapeutic targets of BN using a network pharmacological approach to comprehend the multi target action of its multiple phytoconstituents. We have employed Randić Index for the first time to calculate the contribution score of module segregated targets towards diseases. Our results suggests that BN targets could also be effective in other diseases such as lysosomal storage disorders, respiratory disorders etc., apart from neurological disorders. The key targets with highest topological measures of Targets-(Pathway)-Targets network were identified as potential therapeutic targets of BN. And the top hit target PTGS2, a gene encoding for cyclooxygenase-2 was further evaluated using molecular docking, molecular dynamic simulation and in vitro studies. Our findings open up new therapeutic facets for BN that can be explored systematically in future.
Collapse
Affiliation(s)
- Agnes Preethy H
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur, India
- School of Chemical & Biotechnology (SCBT), SASTRA Deemed University, Thanjavur, India
| | | | | | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur, India
- School of Chemical & Biotechnology (SCBT), SASTRA Deemed University, Thanjavur, India
- School of Arts, Sciences, Humanities & Education (SASHE), SASTRA Deemed University, Thanjavur, India
| |
Collapse
|
7
|
Zhou L, Yang C, Liu Z, Chen L, Wang P, Zhou Y, Yuan M, Zhou LT, Wang X, Zhu LQ. Neuroprotective effect of the traditional decoction Tian-Si-Yin against Alzheimer's disease via suppression of neuroinflammation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117569. [PMID: 38086513 DOI: 10.1016/j.jep.2023.117569] [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: 07/18/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is the most prevalent neurodegenerative disease among old adults. As a traditional Chinese medicine, the herbal decoction Tian-Si-Yin consists of Morinda officinalis How. and Cuscuta chinensis Lam., which has been widely used to nourish kidney. Interestingly, Tian-Si-Yin has also been used to treat dementia, depression and other neurological conditions. However, its therapeutic potential for neurodegenerative diseases such as AD and the underlying mechanisms remain unclear. AIM OF THE STUDY To evaluate the therapeutic effect of the herbal formula Tian-Si-Yin against AD and to explore the underlying mechanisms. MATERIALS AND METHODS The N2a cells treated with amyloid β (Aβ) peptide or overexpressing amyloid precursor protein (APP) were used to establish cellular models of AD. The in vivo anti-AD effects were evaluated by using Caenorhabditis elegans and 3 × Tg-AD mouse models. Tian-Si-Yin was orally administered to the mice for 8 weeks at a dose of 10, 15 or 20 mg/kg/day, respectively. Its protective role on memory deficits of mice was examined using the Morris water maze and fear conditioning tests. Network pharmacology, proteomic analysis and ultra-high performance liquid chromatography-mass spectrometry/mass spectrometry (UHPLC-MS/MS) were used to explore the underlying molecular mechanisms, which were further investigated by Western blotting and immunohistochemistry. RESULTS Tian-Si-Yin was shown to improve cell viability of Aβ-treated N2a cells and APP-expressing N2a-APP cells. Tian-Si-Yin was also found to reduce ROS level and extend lifespan of transgenic AD-like C. elegans model. Oral administration of Tian-Si-Yin at medium dose was able to effectively rescue memory impairment in 3 × Tg mice. Tian-Si-Yin was further shown to suppress neuroinflammation by inhibition of glia cell activation and downregulation of inflammatory cytokines, diminishing tau phosphoralytion and Aβ deposition in the mice. Using UHPLC-MS/MS and network pharmacology technologies, 17 phytochemicals from 68 components of Tian-Si-Yin were identified as potential anti-AD components. MAPK1, BRAF, TTR and Fyn were identified as anti-AD targets of Tian-Si-Yin from network pharmacology and mass spectrum. CONCLUSIONS This study has established the protective effect of Tian-Si-Yin against AD and demonstrates that Tian-Si-Yin is capable of improving Aβ level, tau pathology and synaptic disorder by regulating inflammatory response.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Chunqing Yang
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zhiqiang Liu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Linlin Chen
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, PR China
| | - Ping Wang
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, PR China
| | - Yuan Zhou
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, PR China
| | - Mei Yuan
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, PR China
| | - Lan-Ting Zhou
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, PR China; Neuroscience and Brainscience Institute of Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China.
| | - Xueren Wang
- Department of Anesthesiology, Shanxi Bethune Hospital, Taiyuan, PR China; Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Laboratory of Neurological Disorders of the Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
8
|
He C, Yu W, Yang M, Li Z, Yu J, Zhong D, Deng S, Song Z, Cheng S. Qi Fu Yin ameliorates neuroinflammation through inhibiting RAGE and TLR4/NF-κB pathway in AD model rats. Aging (Albany NY) 2023; 15:13239-13264. [PMID: 38006400 DOI: 10.18632/aging.205238] [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: 07/10/2023] [Accepted: 10/23/2023] [Indexed: 11/27/2023]
Abstract
The purpose of this study is to investigate the therapeutic effect of Qi Fu Yin (QFY) on Alzheimer's disease (AD) both computationally and experimentally. Network pharmacology analysis and molecular docking were conducted to identify potential targets and signaling pathways involved in QFY treating AD. Streptozotocin-induced AD rat model was used to verify important targets and predicted pathways. The components of QFY were identified using liquid chromatography-tandem mass spectrometry. The results indicate that the potential targets of QFY are highly enriched for anti-inflammatory pathways. Molecular docking analysis revealed stable structures formed between QFY's active compounds, including stigmasterol, β-sitosterol, and isorhamnetin, and the identified targets. In vivo, QFY improved cognitive memory in AD rats and reduced the mRNA expression levels of toll-like receptor 4 (TLR4), the receptor for advanced glycation end products (AGER), and the inflammatory factors interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in the brains of AD rats. Furthermore, QFY effectively reduced nuclear translocation of nuclear factor-kappa B (NF-κB) and inhibited NF-κB and microglia activation. In conclusion, QFY can ameliorate neuroinflammation in AD model rats, partly via the inhibition of TLR4 and RAGE/NF-κB pathway and microglia activation, thereby enhancing learning and memory in AD model rats.
Collapse
Affiliation(s)
- Chunxiang He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Wenjing Yu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Miao Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Ze Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jingping Yu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Baoshan College of Traditional Chinese Medicine, Baoshan, Yunnan 678000, China
| | - Dayuan Zhong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, Guangdong 528000, China
| | - Sisi Deng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhenyan Song
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Shaowu Cheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
9
|
Zheng Y, Zhang X, Zhang R, Wang Z, Gan J, Gao Q, Yang L, Xu P, Jiang X. Inflammatory signaling pathways in the treatment of Alzheimer's disease with inhibitors, natural products and metabolites (Review). Int J Mol Med 2023; 52:111. [PMID: 37800614 PMCID: PMC10558228 DOI: 10.3892/ijmm.2023.5314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023] Open
Abstract
The intricate nature of Alzheimer's disease (AD) pathogenesis poses a persistent obstacle to drug development. In recent times, neuroinflammation has emerged as a crucial pathogenic mechanism of AD, and the targeting of inflammation has become a viable approach for the prevention and management of AD. The present study conducted a comprehensive review of the literature between October 2012 and October 2022, identifying a total of 96 references, encompassing 91 distinct pharmaceuticals that have been investigated for their potential impact on AD by inhibiting neuroinflammation. Research has shown that pharmaceuticals have the potential to ameliorate AD by reducing neuroinflammation mainly through regulating inflammatory signaling pathways such as NF‑κB, MAPK, NLRP3, PPARs, STAT3, CREB, PI3K/Akt, Nrf2 and their respective signaling pathways. Among them, tanshinone IIA has been extensively studied for its anti‑inflammatory effects, which have shown significant pharmacological properties and can be applied clinically. Thus, it may hold promise as an effective drug for the treatment of AD. The present review elucidated the inflammatory signaling pathways of pharmaceuticals that have been investigated for their therapeutic efficacy in AD and elucidates their underlying mechanisms. This underscores the auspicious potential of pharmaceuticals in ameliorating AD by impeding neuroinflammation.
Collapse
Affiliation(s)
| | | | - Ruifeng Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Ziyu Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Jiali Gan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Qing Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Lin Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Pengjuan Xu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Xijuan Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| |
Collapse
|
10
|
Jiang S, Borjigin G, Sun J, Li Q, Wang Q, Mu Y, Shi X, Li Q, Wang X, Song X, Wang Z, Yang C. Identification of Uncaria rhynchophylla in the Potential Treatment of Alzheimer's Disease by Integrating Virtual Screening and In Vitro Validation. Int J Mol Sci 2023; 24:15457. [PMID: 37895137 PMCID: PMC10607254 DOI: 10.3390/ijms242015457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Uncaria rhynchophylla (Gouteng in Chinese, GT) is the main medicine in many traditional recipes in China. It is commonly used to alleviate central nervous system (CNS) disorders, although its mechanism in Alzheimer's disease is still unknown. This study was designed to predict and validate the underlying mechanism in AD treatment, thus illustrating the biological mechanisms of GT in treating AD. In this study, a PPI network was constructed, KEGG analysis and GO analysis were performed, and an "active ingredient-target-pathway" network for the treatment of Alzheimer's disease was constructed. The active ingredients of GT were screened out, and the key targets were performed by molecular docking. UHPLC-Q-Exactive Orbitrap MS was used to screen the main active ingredients and was compared with the network pharmacology results, which verified that GT did contain the above ingredients. A total of targets were found to be significantly bound up with tau, Aβ, or Aβ and tau through the network pharmacology study. Three SH-SY5Y cell models induced by okadaic acid (OA), Na2S2O4, and H2O2 were established for in vitro validation. We first found that GT can reverse the increase in the hyperphosphorylation of tau induced by OA to some extent, protecting against ROS damage. Moreover, the results also indicated that GT has significant neuroprotective effects. This study provides a basis for studying the potential mechanisms of GT in the treatment of AD.
Collapse
Affiliation(s)
- Shuang Jiang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Gilwa Borjigin
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Jiahui Sun
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Qi Li
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Qianbo Wang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Yuanqiu Mu
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Xuepeng Shi
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Qian Li
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Xiaotong Wang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Xiaodan Song
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
| | - Zhibin Wang
- Key Laboratory of Chinese Materia Medica, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Chunjuan Yang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (S.J.); (G.B.); (J.S.); (Q.L.); (Q.W.); (Y.M.); (X.S.); (Q.L.); (X.W.); (X.S.)
- Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
11
|
Kim JW, Geum JH, Ha WB, Woo HJ, Han YH, Park SH, Lee JH. The efficacy, effectiveness, and safety of Kyung-ok-ko: A narrative review. Medicine (Baltimore) 2022; 101:e31311. [PMID: 36397335 PMCID: PMC9666190 DOI: 10.1097/md.0000000000031311] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Kyung-ok-ko (KOK), a traditional medicinal formula in East Asia, has been recently studied across various fields. However, comprehensive reviews of clinical applications of KOK targeting clinical and experimental studies are lacking. Therefore, the application of KOK is being limited to the range of tonic medicines. To overcome this limitation, we aim to investigate the effectiveness, mechanism, and safety of KOK to obtain evidence regarding its effects in clinical applications. We searched for clinical and experimental articles in 11 databases (PubMed, Cochrane Library, Excerpta Medica dataBASE, China National Knowledge Infrastructure, Google Scholar, Research Information Sharing Service, Oriental Medicine Advanced Searching Integrated System, Koreanstudies Information Service System, Korean Medical Database, DBpia, and ScienceON). We selected 54 studies based on the inclusion criteria. Three clinical studies used KOK for a consumptive disease and health promotion. Fifty-one experimental studies reported the antioxidant activity, neuroprotective activity, anticancer effect, anti-inflammatory activity, immunological activity, growth promotion, impacts on cardiovascular system diseases, gastrointestinal system diseases, respiratory system diseases, and metabolic bone disease, hepatoprotective function, and antifatigue function of KOK, which were considered effective and safe in consumptive, chronic, metabolic, inflammatory, and immune diseases. We identified the effectiveness of KOK in the treatment of a wide range of diseases. However, further clinical studies are warranted in the future.
Collapse
Affiliation(s)
- Ji-Woo Kim
- Chuna Manual Medicine Research Group, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
| | - Ji-Hye Geum
- Chuna Manual Medicine Research Group, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
- Department of Rehabilitation Medicine of Korean Medicine, Iksan-Jeil Korean Medicine Hospital, Iksan, Republic of Korea
- Department of Korean Medicine Rehabilitation, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
| | - Won-Bae Ha
- Chuna Manual Medicine Research Group, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
- Department of Korean Medicine Rehabilitation, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
| | - Hyeon-Jun Woo
- Chuna Manual Medicine Research Group, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
- Department of Korean Medicine Rehabilitation, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
| | - Yun-Hee Han
- Chuna Manual Medicine Research Group, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
- Department of Korean Medicine Rehabilitation, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
| | - Shin-Hyeok Park
- Chuna Manual Medicine Research Group, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
- Department of Acupuncture and Moxibustion Medicine, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
| | - Jung-Han Lee
- Chuna Manual Medicine Research Group, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
- Department of Korean Medicine Rehabilitation, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
- Research Center of Traditional Korean Medicine, College of Korean Medicine, Won-Kwang University, Iksan, Republic of Korea
- *Correspondence: Jung-Han Lee, Department of Korean Medicine Rehabilitation, College of Korean Medicine, Won-Kwang University, 895 Muwang-ro, Iksan, Jeollabuk-do 54538, Republic of Korea (e-mail: )
| |
Collapse
|
12
|
Yang MQ, Chen C, Mao YF, Li Y, Zhong X, Yu YD, Xue YT, Song YM. Application of network pharmacology and molecular docking approach to explore active compounds and potential pharmacological mechanisms of Aconiti Lateralis Radix Praeparata and Lepidii Semen Descurainiae Semen for treatment of heart failure. Medicine (Baltimore) 2022; 101:e30102. [PMID: 35984130 PMCID: PMC9387970 DOI: 10.1097/md.0000000000030102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Heart failure (HF) is the end stage of the development of heart disease, whose prognosis is poor. The previous research of our team indicated that the formulae containing Aconiti Lateralis Radix Praeparata and Lepidii Semen Descurainiae Semen (ALRP-LSDS) could inhibit myocardial hypertrophy, inhibit cardiomyocyte apoptosis, delay myocardial remodeling (REM), and improve the prognosis of patients with HF effectively. In order to explore the mechanism of ALRP-LSDS for the treatment of HF, a combined approach of network pharmacology and molecular docking was conducted. METHODS Public database TCMSP was used to screen the active compounds of ALRP-LSDS. The targets of screened active compounds were obtained from the TCMSP database and predicted using the online analysis tool PharmMapper. The targets of HF were obtained from 6 databases including GeneCards, OMIM, DrugBank, TTD, PharmGKB, and DisGeNET. Protein-protein interaction and enrichment analysis were performed, respectively, by STRING and Metascape online tools after merging the targets of active compounds and HF. Cytoscape software was used to conduct networks. Finally, molecular docking was performed by Vina to verify the correlation between key targets and active compounds. RESULTS Final results indicated that the active compounds including β-sitosterol, isorhamnetin, quercetin, kaempferol, and (R)-norcoclaurine, the targets including AKT1, CASP3, and MAPK1 might be the main active compounds and key targets of ALRP-LSDS for the treatment of HF separately. The binding ability of AKT1 to the main active compounds was better compared with the other 2 key targets, which means it might be more critical. The pathways including AGE-RAGE signaling pathway in diabetic complications, Pathways in cancer, and Fluid shear stress and atherosclerosis might play important roles in the treatment of HF with ALRP-LSDS. In general, ALRP-LSDS could inhibit cardiomyocyte apoptosis, delay REM, and improve cardiac function through multicompound, multitarget, and multipathway, which contributes to the treatment of HF. CONCLUSIONS Based on the combined approach of network pharmacology and molecular docking, this study screened out the main active compounds, key targets, and main pathways of ALRP-LSDS for the treatment of HF, and revealed its potential mechanisms, providing a theoretical basis for further research.
Collapse
Affiliation(s)
- Meng-Qi Yang
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cong Chen
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi-Fei Mao
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xia Zhong
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi-Ding Yu
- College for Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi-Tao Xue
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Yi-Tao Xue, Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250024, China (e-mail: )
| | - Yong-Mei Song
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Investigating the Mechanisms of Pollen Typhae in the Treatment of Diabetic Retinopathy Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5728408. [PMID: 35024051 PMCID: PMC8747905 DOI: 10.1155/2022/5728408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To explore the main bioactive compounds and investigate the underlying mechanism of Pollen Typhae (PT) against diabetic retinopathy (DR) by network pharmacology and molecular docking analysis. METHODS Bioactive ingredients and the target proteins of PT were obtained from TCMSP, and the related target genes were acquired from the SwissTargetPrediction database. The target genes of DR were obtained from GeneCards, TTD database, DisGeNET database, and DrugBank. The compound-target interaction network was established based on Cytoscape 3.7.2. The protein-protein interaction (PPI) network was constructed via STRING database and Cytoscape 3.7.2. Gene ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were visualized through DAVID database and Bioinformatics. Ingredient-gene-pathway network analysis was conducted to further screen the ingredients, target proteins, and pathways closely related to the biological mechanism on PT for DR, and molecular docking analysis was performed by SYBYL-X 2.1.1 software. Finally, the mechanism and underlying targets of PT in the treatment of DR were predicted. RESULTS A total of 8 compounds and 171 intersection targets were obtained based on the online network database. 7 main compounds were screened from compound-target network, and 53 targets including the top six key targets (PTGS2, AKT1, VEGFA, MAPK3, TNF, and EGFR) were further acquired from PPI analysis. The 53 key targets covered 80 signaling pathways, among which PI3K-Akt signaling pathway, focal adhesion, Rap1 signaling pathway, VEGF signaling pathway, and HIF-1 signaling pathway were closely connected with the biological mechanism involved in the alleviation of DR by PT. Ingredient-gene-pathway network shows that AKTI, EGFR, and VEGFA were core genes, kaempferol and isorhamnetin were pivotal ingredients, and VEGF signaling pathway and Rap1 signaling pathway were closely involved in anti-DR. The docking results indicated that five main compounds (arachidonic acid, isorhamnetin, quercetin, kaempferol, and (2R)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one) had good binding activity with EGFR and AKT1 targets. CONCLUSION The active ingredients in PT may regulate the levels of inflammatory factors, suppress the oxidative stress, and inhibit the proliferation, migration, and invasion of retinal pericytes by acting on PTGS2, AKT1, VEGFA, MAPK3, TNF, and EGFR targets through VEGF signaling pathway, PI3K-Akt signaling pathway, Rap1 signaling pathway, and HIF-1 signaling pathway to play a therapeutic role in diabetic retinopathy.
Collapse
|
14
|
Network Pharmacology and Molecular Docking Analysis on Molecular Targets and Mechanisms of Buyang Huanwu Decoction in the Treatment of Ischemic Stroke. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8815447. [PMID: 33727944 PMCID: PMC7937485 DOI: 10.1155/2021/8815447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/01/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Methods The bioactive components and potential targets of BHD were screened by TCMSP, BATMAN-TCM, ETCM, and SymMap databases. Besides, compounds that failed to find the targets from the above databases were predicted through STITCH, SwissTargetPrediction, and SEA. Moreover, six databases were searched to mine targets of IS. The intersection targets were obtained and analyzed by GO and KEGG enrichment. Furthermore, BHD-IS PPI network, compound-target network, and herb-target-pathway network were constructed by Cytoscape 3.6.0. Finally, AutoDock was used for molecular docking verification. Results A total of 235 putative targets were obtained from 59 active compounds in BHD. Among them, 62 targets were related to IS. PPI network showed that the top ten key targets were IL6, TNF, VEGFA, AKT1, etc. The enrichment analysis demonstrated candidate BHD targets were more frequently involved in TNF, PI3K-Akt, and NF-kappa B signaling pathway. Network topology analysis showed that Radix Astragali was the main herb in BHD, and the key components were quercetin, beta-sitosterol, kaempferol, stigmasterol, etc. The results of molecular docking showed the active components in BHD had a good binding ability with the key targets. Conclusions Our study demonstrated that BHD exerted the effect of treating IS by regulating multitargets and multichannels with multicomponents through the method of network pharmacology and molecular docking.
Collapse
|
15
|
Comparative metabolism study on chlorogenic acid, cryptochlorogenic acid and neochlorogenic acid using UHPLC-Q-TOF MS coupled with network pharmacology. Chin J Nat Med 2021; 19:212-224. [PMID: 33781455 DOI: 10.1016/s1875-5364(21)60023-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Indexed: 11/21/2022]
Abstract
Chlorogenic acid (5-CQA), neochlorogenic acid (3-CQA), and cryptochlorogenic acid (4-CQA), usually simultaneously exist in many traditional Chinese medicines (TCMs). However, insufficient attentions have been paid to the comparative metabolism study on these three isomeric constituents with similar effects on anti-inflammation until now. In this study, a novel strategy was established to perform comparative analysis of their metabolic fates in rats and elucidate the pharmacological mechanism of anti-inflammation. Firstly, diagnostic product ions (DPIs) deduced from the representative reference standards were adopted to rapidly screen and characterize the metabolites in rat plasma, urine and faeces using UHPLC-Q-TOF MS. Subsequently, Network pharmacology was utilized to elucidate their anti-inflammatory mechanism. Consequently, a total of 73 metabolites were detected and characterized, including 50, 47 and 43 metabolites for 5-CQA, 4-CQA and 3-CQA, orderly. Moreover, the network pharmacology study indicated that these three isomeric constituents and their major metabolites with similar in vivo metabolic pathways exerted anti-inflammatory effects through co-owned 20 biological processes, which involved 10 major signal pathways and 159 potential targets. Our study shed light on the similarities and differences of the metabolic profiling and anti-inflammatory activity among these three isomeric constituents and set an example for the further researches on the active mechanism of isomeric constituents existing in TCMs based on comparative metabolism study.
Collapse
|
16
|
Jian GH, Su BZ, Zhou WJ, Xiong H. Application of network pharmacology and molecular docking to elucidate the potential mechanism of Eucommia ulmoides- Radix Achyranthis Bidentatae against osteoarthritis. BioData Min 2020; 13:12. [PMID: 32874205 PMCID: PMC7456016 DOI: 10.1186/s13040-020-00221-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Osteoarthritis is a disabling disease, which seriously affects the quality of life of patients. Increasing evidence has indicated that Chinese herbal medicine including Eucommia ulmoides (EU) and Radix Achyranthis Bidentatae (RAB) have potential in the treatment of osteoarthritis, and this is associated with their multi-target and multi-link action characteristics. Although their potential anti-arthritic activity has been reported, the exact mechanism of EU-RAB action in osteoarthritis remains unexplored. Therefore, this study explores the mechanism of EU-RAB against osteoarthritis using network pharmacology and molecular docking technology. METHODS Public databases including TCMSP、BATMAN-TCM、OMIM and Genecards were used to predict the bioactive ingredients and putative targets of EU-RAB against osteoarthritis. Enrichment analysis was performed to expound the biological functions and associated pathways of the hub targets. Cytoscape software was used to construct a "compounds-targets-pathways" network for elucidating the comprehensive molecular mechanism of EU-RAB against osteoarthritis. Molecular docking was used to verify the correlation between the main active ingredients and hub targets. RESULTS Network pharmacological analysis of EU-RAB in the treatment of osteoarthritis, identified 50 active ingredients including quercetin, kaempferol, wogonin, and baicalein with important biological effect. A total of 68 key targets were screened, including IL-6, EGFR, MAPK8, etc., and they were found to be enriched in a series of signaling pathways, such as apoptosis, TNF, MAPK, PI3K/AKT, and IL-17 signaling pathways. Moreover, molecular docking analysis showed that the main ingredients were tightly bound to the core targets, further confirming the anti-arthritic effects. CONCLUSION Based on network pharmacology and molecular docking analysis, the present study provides insights into the potential mechanism of EU-RAB in osteoarthritis after successfully screening for associated key target genes and signaling pathways. These findings further provide a theoretical basis for further pharmacological research into the potential mechanism of EU-RAB in osteoarthritis.
Collapse
Affiliation(s)
- Gong-hui Jian
- Hunan University of Chinese Medicine, Changsha, Hunan Province People’s Republic of China
| | - Bing-zhu Su
- Hunan University of Chinese Medicine, Changsha, Hunan Province People’s Republic of China
| | - Wen-jia Zhou
- Hunan University of Chinese Medicine, Changsha, Hunan Province People’s Republic of China
| | - Hui Xiong
- Hunan University of Chinese Medicine, Changsha, Hunan Province People’s Republic of China
| |
Collapse
|
17
|
Deciphering the Molecular Targets and Mechanisms of HGWD in the Treatment of Rheumatoid Arthritis via Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7151634. [PMID: 32908565 PMCID: PMC7471805 DOI: 10.1155/2020/7151634] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022]
Abstract
Background Huangqi Guizhi Wuwu Decoction (HGWD) has been applied in the treatment of joint pain for more than 1000 years in China. Currently, most physicians use HGWD to treat rheumatoid arthritis (RA), and it has proved to have high efficacy. Therefore, it is necessary to explore the potential mechanism of action of HGWD in RA treatment based on network pharmacology and molecular docking methods. Methods The active compounds of HGWD were collected, and their targets were identified from the Traditional Chinese Medicine Systems Pharmacology Database (TCMSP) and DrugBank database, respectively. The RA-related targets were retrieved by analyzing the differentially expressed genes between RA patients and healthy individuals. Subsequently, the compound-target network of HGWD was constructed and visualized through Cytoscape 3.8.0 software. Protein-protein interaction (PPI) network was constructed to explore the potential mechanisms of HGWD on RA using the plugin BisoGenet of Cytoscape 3.8.0 software. Gene ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed in R software (Bioconductor, clusterProfiler). Afterward, molecular docking was used to analyze the binding force of the top 10 active compounds with target proteins of VCAM1, CTNNB1, and JUN. Results Cumulatively, 790 active compounds and 1006 targets of HGWD were identified. A total of 4570 differentially expressed genes of RA with a p value <0.05 and |log 2(fold change)| > 0.5 were collected. Moreover, 739 GO entries of HGWD on RA were identified, and 79 pathways were screened based on GO and KEGG analysis. The core target gene of HGWD in RA treatment was JUN. Other key target genes included FOS, CCND1, IL6, E2F2, and ICAM1. It was confirmed that the TNF signaling pathway and IL-17 signaling pathway are important pathways of HGWD in the treatment of RA. The molecular docking results revealed that the top 10 active compounds of HGWD had a strong binding to the target proteins of VCAM1, CTNNB1, and JUN. Conclusion HGWD has important active compounds such as quercetin, kaempferol, and beta-sitosterol, which exert its therapeutic effect on multiple targets and multiple pathways.
Collapse
|