1
|
He H, Sun S, Xu W, Zhang M. Network Pharmacology Followed by Experimental Validation to Explore the Mechanism of Stigmasterol in Sangbaipi Decoction Regulating PI3K/Akt Signaling to Alleviate Acute Exacerbation of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:1819-1834. [PMID: 39140079 PMCID: PMC11319098 DOI: 10.2147/copd.s459814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Abstract
Purpose Sangbaipi decoction (SBPD), a traditional Chinese medicine (TCM) prescription, has been widely used to treat acute exacerbation of chronic obstructive pulmonary disease (AECOPD), while the underlying pharmacological mechanism remains unclear due to the complexity of composition. Methods A TCM-active ingredient-drug target network of SBPD was constructed utilizing the TCM-Systems-Pharmacology database. AECOPD-relevant proteins were gathered from Gene Cards and the Online-Mendelian-Inheritance-in-Man database. Protein-protein interaction, GO and KEGG enrichment analyses of the targets from the intersection of SBPD and AECOPD targets were performed to identify the core signaling pathway, followed by molecular docking verification of its interaction with active ingredients. The network pharmacology results were checked using in-vivo experiments. To induce AECOPD, rats were exposure to combined tobacco smoke and lipopolysaccharide (LPS). Then rats underwent gavage with stigmasterol (SM) after successful modeling. The involvement of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling was investigated using its inhibitor, LY294002. Lung function and histopathology were examined. The levels of inflammatory cytokines in the lung and serum were assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), Western blot and/or Enzyme-linked immunosorbent assay (ELISA). Results SM was recognized as an active ingredient of SBPD and stably bound to Akt1. SM improved lung function and histological abnormalities, concomitant with suppressed PI3K/Akt signaling, downregulated lung and serum Interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α) levels and serum transforming growth factor-β (TGF-β) levels and upregulated lung and serum Interleukin 10 (IL-10) levels in AECOPD rats. In AECOPD rats, LY294002 restored lung function, and it also improved lung histological abnormalities and inflammation, which was found to be potentiated by SM. Conclusion SM targets PI3K/Akt signaling to reduce lung injury and inflammation in AECOPD rats.
Collapse
Affiliation(s)
- Haidong He
- Department of Pulmonary and Critical Care Medicine, Tongde Hospital of Zhejiang Province, Hangzhou City, Zhejiang Province, People’s Republic of China
| | - Shuihua Sun
- Department of Medical Oncology, Tongde Hospital of Zhejiang Province, Hangzhou City, Zhejiang Province, People’s Republic of China
| | - Weihua Xu
- Department of Pulmonary and Critical Care Medicine, Tongde Hospital of Zhejiang Province, Hangzhou City, Zhejiang Province, People’s Republic of China
| | - Mingwan Zhang
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou City, Zhejiang Province, People’s Republic of China
| |
Collapse
|
2
|
Cui C, Fan Z. Exploring the mechanism of Liang Xue Wu Hua Tang in the treatment of rosacea via network pharmacology and molecular docking. Medicine (Baltimore) 2024; 103:e38705. [PMID: 38941423 PMCID: PMC11466150 DOI: 10.1097/md.0000000000038705] [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: 12/22/2023] [Accepted: 06/05/2024] [Indexed: 06/30/2024] Open
Abstract
Rosacea is a chronic and recurrent inflammatory skin disease affecting the center of the face that causes burning and itching sensations and changes in aesthetics. Liang Xue Wu Hua Tang (LXWHT) is a classic herbal formulation that is efficacious and has been widely used in the clinical treatment of rosacea; however, the pharmacological mechanisms remain unclear. The aim of the present study was to investigate the mechanism of action of LXWHT using network pharmacology and molecular docking. The Traditional Chinese Medicine System Pharmacology database was searched to identify the active ingredients and pharmacological targets of LXWHT, and the GeneCard, Disgenet, and Gene Expression Omnibus databases were applied to screen rosacea-related targets. Cytoscape software was used to visualize the protein-protein interaction network, and network topology analysis was used to identify core targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed for the core targets. Molecular docking simulations and visualization were performed using Maestro and PyMOL, respectively. A total of 43 active compounds and 28 potential targets for LXWHT treatment of rosacea were selected for analysis. The Gene Ontology/Kyoto Encyclopedia of Genes and Genomes results indicated that LXWHT may exert therapeutic effects on rosacea by intervening in immune pathways including tumor necrosis factor pathway, interleukin-17 pathways, and Toll-like receptor signaling pathways. Chemokine ligand 2, interferon-γ, interleukin-1ß, peroxisome proliferator-activated receptor-γ, and matrix metallopeptidase 9 may be the core therapeutic target. Quercetin, stigmasterol, kaempferol, beta-sitosterol, luteolin, beta-carotene, baicalein, acetin, and isorhamnetin were predicted to be the key active ingredients. LXWHT may exert therapeutic effects in the treatment of rosacea by modulating immunity and angiogenesis, laying the foundation for further research.
Collapse
Affiliation(s)
- Can Cui
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhu Fan
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
El Omari N, Bakrim S, Khalid A, Abdalla AN, Iesa MAM, El Kadri K, Tang SY, Goh BH, Bouyahya A. Unveiling the molecular mechanisms: dietary phytosterols as guardians against cardiovascular diseases. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:27. [PMID: 38722432 PMCID: PMC11082103 DOI: 10.1007/s13659-024-00451-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
Until recently, the main pharmaceuticals used to control cholesterol and prevent cardiovascular disease (CVD) were statin-related drugs, known for their historical side effects. Therefore, there is growing interest in exploring alternatives, such as nutritional and dietary components, that could play a central role in CVD prevention. This review aims to provide a comprehensive understanding of how natural phytosterols found in various diets combat CVDs. We begin with a description of the overall approach, then we explore in detail the different direct and indirect mechanisms that contribute to reducing cardiovascular incidents. Phytosterols, including stigmasterol, β-sitosterol, ergosterol, and fucosterol, emerge as promising molecules within nutritional systems for protection against CVDs due to their beneficial effects at different levels through direct or indirect cellular, subcellular, and molecular mechanisms. Specifically, the mentioned phytosterols exhibit the ability to diminish the generation of various radicals, including hydroperoxides and hydrogen peroxide. They also promote the activation of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione, while inhibiting lipid peroxidation through the activation of Nrf2 and Nrf2/heme oxygenase-1 (HO-1) signaling pathways. Additionally, they demonstrate a significant inhibitory capacity in the generation of pro-inflammatory cytokines, thus playing a crucial role in regulating the inflammatory/immune response by inhibiting the expression of proteins involved in cellular signaling pathways such as JAK3/STAT3 and NF-κB. Moreover, phytosterols play a key role in reducing cholesterol absorption and improving the lipid profile. These compounds can be used as dietary supplements or included in specific diets to aid control cholesterol levels, particularly in individuals suffering from hypercholesterolemia.
Collapse
Affiliation(s)
- Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, 80000, Agadir, Morocco
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, 45142, Jazan, Saudi Arabia.
- Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Center for Research, P. O. Box 2404, Khartoum, Sudan.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, 21955, Makkah, Saudi Arabia
| | - Mohamed A M Iesa
- Department of Physiology, Al Qunfudah Medical College, Umm Al Qura University, Mecca, Saudi Arabia
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, 10106, Rabat, Morocco
| | - Siah Ying Tang
- Department of Chemical Engineering, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, 47500, Bandar Sunway, Malaysia.
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, 47500, Sunway City, Malaysia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, 10106, Rabat, Morocco.
| |
Collapse
|
4
|
Goswami M, Jaswal S, Gupta GD, Kumar Verma S. A Comprehensive Update on Phytochemistry, Analytical Aspects, Medicinal Attributes, Specifications and Stability of Stigmasterol. Steroids 2023; 196:109244. [PMID: 37137454 DOI: 10.1016/j.steroids.2023.109244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Phytosterols are bioactive substances naturally found in plant cell membranes, and their chemical structure is comparable to cholesterol found in mammalian cells. They are widely distributed in plant foods like olive oil, nuts, seeds, and legumes. Amongst the variety of phytosterols, stigmasterol is the vital compound found abundantly in plants. Numerous hormones, including estrogen, progesterone, corticoids and androgen, are synthesized by stigmasterol. Multiple in-vitro and in-vivo investigations have shown that stigmasterol has various biological effects, including antioxidant, anticancer, antidiabetic, respiratory diseases, and lipid-lowering effects. Experimental research on stigmasterol provides indisputable proof that this phytosterol has the potential to be employed in supplements used to treat the illnesses mentioned above. This substance has a high potential, making it a noteworthy medication in the future. Although several researchers have investigated this phytosterol to assess its prospective qualities, it has not yet attained therapeutic levels, necessitating additional clinical studies. This review offers a comprehensive update on stigmasterol, including chemical framework, biosynthesis, synthetic derivatives, extraction and isolation, analytical aspects, pharmacological profile, patent status, clinical trials, stability and specifications as per regulatory bodies.
Collapse
Affiliation(s)
- Megha Goswami
- Department of Pharmacognosy, ISF College of Pharmacy, Moga-142 001 (Punjab), India
| | - Shalini Jaswal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga-142 001 (Punjab), India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142 001 (Punjab), India
| | - Sant Kumar Verma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga-142 001 (Punjab), India.
| |
Collapse
|
5
|
Miya GM, Oriola AO, Payne B, Cuyler M, Lall N, Oyedeji AO. Steroids and Fatty Acid Esters from Cyperus sexangularis Leaf and Their Antioxidant, Anti-Inflammatory and Anti-Elastase Properties. Molecules 2023; 28:molecules28083434. [PMID: 37110668 PMCID: PMC10141076 DOI: 10.3390/molecules28083434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Cyperus sexangularis (CS) is a plant in the sedges family (Cyperaceae) that grows abundantly in swampy areas. The leaf sheath of plants in the Cyperus genus are mostly used domestically for mat making, while they are implicated for skin treatment in traditional medicine. The plant was investigated for its phytochemical contents as well as its antioxidant, anti-inflammatory and anti-elastase properties. The n-hexane and dichloromethane leaf extracts were chromatographed on a silica gel column to afford compounds 1-6. The compounds were characterized by nuclear magnetic resonance spectroscopy and mass spectrometry. The inhibitory effect of each compound against 2,2-diphenyl-1-picrylhydrazyl (DPPH), nitric oxide (NO) and ferric ion radicals were determined by standard in vitro antioxidant methods. The in vitro anti-inflammatory response was measured using egg albumin denaturation (EAD) assay, while the anti-elastase activity of each compound in human keratinocyte (HaCaT) cells was also monitored. The compounds were characterized as three steroidal derivatives, stigmasterol (1), 17-(1-methyl-allyl)-hexadecahydro-cyclopenta[a]phenanthrene (2) and β-sitosterol (3), dodecanoic acid (4) and two fatty acid esters, ethyl nonadecanoate (5) and ethyl stearate (6). Stigmasterol (1) exhibited the best biological properties, with IC50 of 38.18 ± 2.30 µg/mL against DPPH, 68.56 ± 4.03 µg/mL against NO and 303.58 ± 10.33 µAAE/mg against Fe3+. At 6.25 µg/mL, stigmasterol inhibited EAD by 50%. This activity was lower when compared to diclofenac (standard), which demonstrated 75% inhibition of the protein at the same concentration. Compounds 1, 3, 4 and 5 showed comparable anti-elastase activity with an IC50 ≥ 50 µg/mL, whereas the activity of ursolic acid (standard) was double fold with an IC50 of 24.80 ± 2.60 µg/mL when compared to each of the compounds. In conclusion, this study has identified three steroids (1-3), one fatty acid (4), and two fatty acid esters (5 and 6) in C. sexangularis leaf for the first time. The compounds showed considerable antioxidant, anti-inflammatory and anti-elastase properties. Thus, the findings may serve as a justification for the folkloric use of the plant as a local skin ingredient. It may also serve to validate the biological role of steroids and fatty acid compounds in cosmeceutical formulations.
Collapse
Affiliation(s)
- Gugulethu Mathews Miya
- Department of Chemical and Physical Sciences, Faculty of Natural Sciences, Walter Sisulu University, Mthatha 5117, South Africa
| | - Ayodeji Oluwabunmi Oriola
- Department of Chemical and Physical Sciences, Faculty of Natural Sciences, Walter Sisulu University, Mthatha 5117, South Africa
| | - Bianca Payne
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Marizé Cuyler
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Namrita Lall
- Department of Plant and Soil Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Adebola Omowunmi Oyedeji
- Department of Chemical and Physical Sciences, Faculty of Natural Sciences, Walter Sisulu University, Mthatha 5117, South Africa
| |
Collapse
|
6
|
Kim J, Kim K. Elucidating the potential pharmaceutical mechanism of Gyejibokryeong-hwan on rosacea using network analysis. Medicine (Baltimore) 2023; 102:e33023. [PMID: 36862896 PMCID: PMC9981404 DOI: 10.1097/md.0000000000033023] [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: 03/04/2023] Open
Abstract
Rosacea is a chronic erythematous disease with telangiectasia that affects the central area of the face. However, because of the ambiguity in the pathophysiology of rosacea, its treatment has not been clearly elucidated; therefore, new therapeutic options need to be developed. Gyejibokryeong-hwan (GBH) is widely used in clinical practice for various blood circulation disorders, including hot flushes. Therefore, we explored the potential pharmaceutical mechanism of GBH on rosacea and investigated the therapeutic points exclusive to GBH through comparative analysis with chemical drugs recommended in 4 guidelines for rosacea based on network analysis. The active compounds in GBH were identified, and the proteins targeted by these compounds and the genes related to rosacea were searched. Additionally, the proteins targeted by the guideline drugs were also searched to compare their effects. And the pathway/term analysis of common genes was conducted. Ten active compounds were obtained for rosacea. There were 14 rosacea-related genes targeted by GBH, with VEGFA, TNF, and IL-4, which were suggested as core genes. The pathway/term analysis of the 14 common genes revealed that GBH could potentially act on rosacea via 2 pathways: the "interleukin 17 signaling pathway" and the "neuroinflammatory response." Comparison and analysis of the protein targets between GBH and guideline drugs revealed that only GBH separately acts on the "vascular wound healing pathway." GBH has the potential to act on IL-17 signaling pathway, neuroinflammatory response and vascular wound healing pathway. Further studies are needed to determine the potential mechanism of GBH in rosacea.
Collapse
Affiliation(s)
- Jundong Kim
- Department of Ophthalmology, Otorhinolaryngology and Dermatology of Korean Medicine, Graduate School of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Kyuseok Kim
- Department of Ophthalmology, Otorhinolaryngology and Dermatology of Korean Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
- *Correspondence: Kyuseok Kim, Department of Ophthalmology, Otorhinolaryngology and Dermatology of Korean Medicine, Kyung Hee University College of Korean Medicine, Kyung Hee University Medical Center, Seoul, 02447, Republic of Korea (e-mail address: )
| |
Collapse
|
7
|
Liu X, Yang Y, Li Y, Zhang Q, Wang J, Guo J, Song Z, Liu Z, Zhang Y, Song X. Network Pharmacology-Based Approach for Investigating the Role of Xanthii Fructus in Treatment of Allergic Rhinitis. Chem Biodivers 2023; 20:e202200785. [PMID: 36855022 DOI: 10.1002/cbdv.202200785] [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: 08/21/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/02/2023]
Abstract
Xanthii Fructus (XF) has been used for treatment of allergic rhinitis (AR), but its pharmacological mechanism of action remains unclear. We aimed to explore the potential mechanism of XF in treatment of AR by using a network pharmacology approach combined with in vivo verification experiments in this study. We identified 945 AR-related pathogenic genes, 11 active components in XF and 178 targets of those active components by corresponding databases. Finally, 54 targets of active components from XF in treatment of AR were identified by the Protein-protein interaction (PPI) network, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, among which Tumor Necrosis Factor (TNF), Mitogen-activated Protein Kinase 3 (MAPK3), Prostaglandin G/H Synthase 2 (PTGS2), Epidermal Growth Factor Receptor (EGFR) showed strongest interactions. The molecular docking analysis showed that moupinamide could bind to EGFR at LEU704 and LEU703, and PTGS2 at TRP387; 24-Ethylcholest-4-en-3-one was identified to bind to MAPK3 at THR347. The validation of quantitative real-time reverse transcription PCR (RT-PCR) showed that XF decreased the levels of MAPK3, PTGS2, and EGFR expression in the nasal mucosa from AR mice gavaged with an XF water decoction. Meanwhile, the levels of interleukin (IL)-4, IL-5 and IL-13were also decreased after the treatment of XF by Enzyme-linked immunosorbent assay (ELISA). Our results provide the pharmacological mechanism and possible intervention targets of XF in treatment of AR.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Yujuan Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Yumei Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Qiang Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Jianwei Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Jing Guo
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Zheying Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Zhen Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Yu Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, No. 20, East Road, Zhifu District, Yantai, 264000, China.,Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, 264000, Yantai, China
| |
Collapse
|
8
|
An in silico and in vitro integrated analysis method to reveal the curative mechanisms and pharmacodynamic substances of Bufei granule on chronic obstructive pulmonary disease. Mol Divers 2023; 27:103-123. [PMID: 35266101 DOI: 10.1007/s11030-022-10404-w] [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: 09/01/2021] [Accepted: 02/07/2022] [Indexed: 02/08/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory disease with high disability and mortality. Clinical studies have shown that the Traditional Chinese Medicine Bufei Granule (BFG) has conspicuous effects on relieving cough and improving lung function in patients with COPD and has a reliable effect on the treatment of COPD, whereas the therapeutic mechanism is vague. In the present study, the latent bronchodilators and mechanism of BFG in the treatment of COPD were discussed through the method of network pharmacology. Then, the molecular docking and molecular dynamics simulation were performed to calculate the binding efficacy of corresponding compounds in BFG to muscarinic receptor. Finally, the effects of BFG on bronchial smooth muscle were validated by in vitro experiments. The network pharmacology results manifested the anti-COPD effect of BFG was mainly realized via restraining airway smooth muscle contraction, activating cAMP pathways and relieving oxidative stress. The results of molecular docking and molecular dynamics simulation showed alpinetin could bind to cholinergic receptor muscarinic 3. The in vitro experiment verified both BFG and alpinetin could inhibit the levels of CHRM3 and acetylcholine and could be potential bronchodilators for treating COPD. This study provides an integrating network pharmacology method for understanding the therapeutic mechanisms of traditional Chinese medicine, as well as a new strategy for developing natural medicines for treating COPD.
Collapse
|
9
|
Selected Seeds as Sources of Bioactive Compounds with Diverse Biological Activities. Nutrients 2022; 15:nu15010187. [PMID: 36615843 PMCID: PMC9823554 DOI: 10.3390/nu15010187] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Seeds contain a variety of phytochemicals that exhibit a wide range of biological activities. Plant-derived compounds are often investigated for their antioxidant, anti-inflammatory, immunomodulatory, hypoglycemic, anti-hypercholesterolemic, anti-hypertensive, anti-platelet, anti-apoptotic, anti-nociceptive, antibacterial, antiviral, anticancer, hepatoprotective, or neuroprotective properties. In this review, we have described the chemical content and biological activity of seeds from eight selected plant species-blackberry (Rubus fruticosus L.), black raspberry (Rubus coreanus Miq.), grape (Vitis vinifera L.), Moringa oleifera Lam., sea buckthorn (Hippophae rhamnoides L.), Gac (Momordica cochinchinensis Sprenger), hemp (Cannabis sativa L.), and sacha inchi (Plukenetia volubilis L). This review is based on studies identified in electronic databases, including PubMed, ScienceDirect, and SCOPUS. Numerous preclinical, and some clinical studies have found that extracts, fractions, oil, flour, proteins, polysaccharides, or purified chemical compounds isolated from the seeds of these plants display promising, health-promoting effects, and could be utilized in drug development, or to make nutraceuticals and functional foods. Despite that, many of these properties have been studied only in vitro, and it's unsure if their effects would be relevant in vivo as well, so there is a need for more animal studies and clinical trials that would help determine if they could be applied in disease prevention or treatment.
Collapse
|
10
|
Zhang X, Wang J, Zhu L, Wang X, Meng F, Xia L, Zhang H. Advances in Stigmasterol on its anti-tumor effect and mechanism of action. Front Oncol 2022; 12:1101289. [PMID: 36578938 PMCID: PMC9791061 DOI: 10.3389/fonc.2022.1101289] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
Stigmasterol is a phytosterol derived from multiple herbaceous plants such as herbs, soybean and tobacco, and it has received much attention for its various pharmacological effects including anti-inflammation, anti-diabetes, anti-oxidization, and lowering blood cholesterol. Multiple studies have revealed that stigmasterol holds promise as a potentially beneficial therapeutic agent for malignant tumors because of its significant anti-tumor bioactivity. It is reported that stigmasterol has anti-tumor effect in a variety of malignancies (e.g., breast, lung, liver and ovarian cancers) by promoting apoptosis, inhibiting proliferation, metastasis and invasion, and inducing autophagy in tumor cells. Mechanistic study shows that stigmasterol triggers apoptosis in tumor cells by regulating the PI3K/Akt signaling pathway and the generation of mitochondrial reactive oxygen species, while its anti-proliferative activity is mainly dependent on its modulatory effect on cyclin proteins and cyclin-dependent kinase (CDK). There have been multiple mechanisms underlying the anti-tumor effect of stigmasterol, which make stigmasterol promising as a new anti-tumor agent and provide insights into research on its anti-tumor role. Presently, stigmasterol has been poorly understood, and there is a paucity of systemic review on the mechanism underlying its anti-tumor effect. The current study attempts to conduct a literature review on stigmasterol for its anti-tumor effect to provide reference for researchers and clinical workers.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiayun Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Zhu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuezhen Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Feifei Meng
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Xia
- Department of Pathology, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Hairong Zhang, ; Lei Xia,
| | - Hairong Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Jinan, China,*Correspondence: Hairong Zhang, ; Lei Xia,
| |
Collapse
|
11
|
Ni B, Song X, Shi B, Wang J, Sun Q, Wang X, Xu M, Cao L, Zhu G, Li J. Research progress of ginseng in the treatment of gastrointestinal cancers. Front Pharmacol 2022; 13:1036498. [PMID: 36313365 PMCID: PMC9603756 DOI: 10.3389/fphar.2022.1036498] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/03/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer has become one of the major causes of human death. Several anticancer drugs are available; howeve their use and efficacy are limited by the toxic side effects and drug resistance caused by their continuous application. Many natural products have antitumor effects with low toxicity and fewer adverse effects. Moreover, they play an important role in enhancing the cytotoxicity of chemotherapeutic agents, reducing toxic side effects, and reversing chemoresistance. Consequently, natural drugs are being applied as potential therapeutic options in the field of antitumor treatment. As natural medicinal plants, some components of ginseng have been shown to have excellent efficacy and a good safety profile for cancer treatment. The pharmacological activities and possible mechanisms of action of ginseng have been identified. Its broad range of pharmacological activities includes antitumor, antibacterial, anti-inflammatory, antioxidant, anti-stress, anti-fibrotic, central nervous system modulating, cardioprotective, and immune-enhancing effects. Numerous studies have also shown that throuth multiple pathways, ginseng and its active ingredients exert antitumor effects on gastrointestinal (GI) tract tumors, such as esophageal, gastric, colorectal, liver, and pancreatic cancers. Herein, we introduced the main components of ginseng, including ginsenosides, polysaccharides, and sterols, etc., and reviewed the mechanism of action and research progress of ginseng in the treatment of various GI tumors. Futhermore, the pathways of action of the main components of ginseng are discussed in depth to promote the clinical development and application of ginseng in the field of anti-GI tumors.
Collapse
Affiliation(s)
- Baoyi Ni
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaotong Song
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bolun Shi
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia Wang
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Qianhui Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinmiao Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Manman Xu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luchang Cao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | | | - Jie Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jie Li,
| |
Collapse
|
12
|
Bakrim S, Benkhaira N, Bourais I, Benali T, Lee LH, El Omari N, Sheikh RA, Goh KW, Ming LC, Bouyahya A. Health Benefits and Pharmacological Properties of Stigmasterol. Antioxidants (Basel) 2022; 11:1912. [PMID: 36290632 PMCID: PMC9598710 DOI: 10.3390/antiox11101912] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 07/30/2023] Open
Abstract
Stigmasterol is an unsaturated phytosterol belonging to the class of tetracyclic triterpenes. It is one of the most common plant sterols, found in a variety of natural sources, including vegetable fats or oils from many plants. Currently, stigmasterol has been examined via in vitro and in vivo assays and molecular docking for its various biological activities on different metabolic disorders. The findings indicate potent pharmacological effects such as anticancer, anti-osteoarthritis, anti-inflammatory, anti-diabetic, immunomodulatory, antiparasitic, antifungal, antibacterial, antioxidant, and neuroprotective properties. Indeed, stigmasterol from plants and algae is a promising molecule in the development of drugs for cancer therapy by triggering intracellular signaling pathways in numerous cancers. It acts on the Akt/mTOR and JAK/STAT pathways in ovarian and gastric cancers. In addition, stigmasterol markedly disrupted angiogenesis in human cholangiocarcinoma by tumor necrosis factor-α (TNF-α) and vascular endothelial growth factor receptor-2 (VEGFR-2) signaling down-regulation. The association of stigmasterol and sorafenib promoted caspase-3 activity and down-regulated levels of the anti-apoptotic protein Bcl-2 in breast cancer. Antioxidant activities ensuring lipid peroxidation and DNA damage lowering conferred to stigmasterol chemoprotective activities in skin cancer. Reactive oxygen species (ROS) regulation also contributes to the neuroprotective effects of stigmasterol, as well as dopamine depletion and acetylcholinesterase inhibition. The anti-inflammatory properties of phytosterols involve the production of anti-inflammatory cytokines, the decrease in inflammatory mediator release, and the inhibition of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Stigmasterol exerts anti-diabetic effects by reducing fasting glucose, serum insulin levels, and oral glucose tolerance. Other findings showed the antiparasitic activities of this molecule against certain strains of parasites such as Trypanosoma congolense (in vivo) and on promastigotes and amastigotes of the Leishmania major (in vitro). Some stigmasterol-rich plants were able to inhibit Candida albicans, virusei, and tropicalis at low doses. Accordingly, this review outlines key insights into the pharmacological abilities of stigmasterol and the specific mechanisms of action underlying some of these effects. Additionally, further investigation regarding pharmacodynamics, pharmacokinetics, and toxicology is recommended.
Collapse
Affiliation(s)
- Saad Bakrim
- Molecular Engineering, Biotechnologies and Innovation Team, Geo-Bio-Environment Engineering and Innovation Laboratory, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Nesrine Benkhaira
- Laboratory of Microbial Biotechnology and Bioactive Molecules, Department of Biology, Faculty of Sciences and Techniques, University Sidi Mohamed Ben Abdellah, Fez 1975, Morocco
| | - Ilhame Bourais
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Taoufiq Benali
- Environment and Health Team, Polydisciplinary Faculty of Safi, Cadi Ayyad University, Sidi Bouzid B.P. 4162, Morocco
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Ryan A. Sheikh
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Long Chiau Ming
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| |
Collapse
|
13
|
Coadministration of Stigmasterol and Dexamethasone (STIG+DEX) Modulates Steroid-Resistant Asthma. Mediators Inflamm 2022; 2022:2222270. [PMID: 36060927 PMCID: PMC9433298 DOI: 10.1155/2022/2222270] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/25/2022] [Accepted: 07/24/2022] [Indexed: 11/18/2022] Open
Abstract
Airway inflammation in asthma is managed with anti-inflammatory steroids such as dexamethasone (DEX). However, about 20% of asthmatics do not respond to this therapy and are classified as steroid-resistant. Currently, no effective therapy is available for steroid-resistant asthma. This work therefore evaluated the effect of a plant sterol, stigmasterol (STIG), and stigmasterol-dexamethasone combination (STIG+DEX) in LPS-ovalbumin-induced steroid-resistant asthma in Guinea pigs. To do this, the effect of drugs on inflammatory features such as airway hyperreactivity and histopathology of lung tissue was evaluated. Additionally, the possible pathway of drug action was assessed by measuring events such neutrophil levels, oxidative and nitrative stress, and histone deacetylase 2 (HDAC2) and interleukin 17 (IL-17) levels. STIG alone did not affect inflammatory features, although it caused some changes in the molecular events associated with steroid-resistant asthma. However, STIG+DEX caused significant modulation of inflammatory features by protecting against destruction of lung tissue. The modulation of inflammatory features was associated with significant inhibition of neutrophilia and oxidative and nitrative stress, decrease in HDAC2, and increase in IL-17 levels that are usually associated with steroid-resistant asthma. Our findings show that although STIG and DEX individually do not protect against steroid-resistant asthma, their coadministration results in significant modulation of inflammatory features and the associated molecular events that lead to steroid-resistant asthma.
Collapse
|
14
|
Gao L, Zhang C, Li Q, Peng X, Shima G, Cao H, Hao P, Li C, Zhang Z. Network Pharmacology and Experimental Analyses of the Mechanism of Analgesic and Glucose Intolerance Through Glucocorticoid Signaling in C57 Mice Treated with Water Extract of Prunella vulgaris L. Spica. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221111032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The aim of this study was to confirm the anti-inflammatory effect and explore the adverse effects and underlying mechanisms of Prunella vulgaris L., which has been extensively used for hundreds of years in East Asia. Network pharmacology studies predicted that glucocorticoids (GCs), GC-targeting molecules, and brain-derived neurotrophic factor (BDNF) were intensively involved in the anti-inflammation and glucose intolerance. To attest the effects and underlying mechanisms, C57 male mice were randomly divided into 5 groups, control (C), dexamethasone (Dex), water extract of P. vulgaris (PE 35 or 70 mg), and PE (70 mg) + mifepristone (PEM). After a 3-week treatment, acetic acid-induced writhing and hot plate tests confirmed the peripheral and central analgesic effects, respectively. Plasma GCs and BDNF were significantly increased. Coincidently, plasma pro-inflammatory cytokines, including IL1β, IL6, and IL10, were decreased by PE treatment, which were blocked by the application of mifepristone ( P < 0.5). Western blots confirmed GC receptor (GR) translocation, and decreased cyclooxygenase 2 in the lumber spine by PE treatment. Food intake was impeded after a 4-week PE treatment, but the ratio of bodyweight gain to food intake was increased in a time-dependent manner. An intraperitoneal glucose tolerance test disclosed that PE treatment impaired glucose disposal in mice. Quantitative polymerase chain reaction (PCR) showed that hepatic GC-responsive genes such as GC-induced leucine zipper protein and glucose-6-phosphatase catalytic subunit 1 were up-regulated, and hypothalamic neuropeptide Y and agouti-related protein expressions were decreased by PE treatment. Hypothalamic BDNF was up-regulated, whereas hepatic BDNF was down-regulated. The regulation of these genes by PE was reversed by mifepristone administration. In conclusion, PE treatment plays analgesic and glucose regulation roles simultaneously through GC-induced signaling pathways, and P. vulgaris may provide a natural ligand of GR for the treatment of inflammation with glucose dysregulation.
Collapse
Affiliation(s)
- Li Gao
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Chaoyun Zhang
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Qiuying Li
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Xiaojuan Peng
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Guanghan Shima
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Hongwei Cao
- Tiandao Wines & Spirits Co., Ltd, Handan, Hebei, China
| | - Pengfei Hao
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Chao Li
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| | - Zhongming Zhang
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, Henan, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, Henan, China
| |
Collapse
|
15
|
Singla RK, Dhir V, Madaan R, Kumar D, Singh Bola S, Bansal M, Kumar S, Dubey AK, Singla S, Shen B. The Genus Alternanthera: Phytochemical and Ethnopharmacological Perspectives. Front Pharmacol 2022; 13:769111. [PMID: 35479320 PMCID: PMC9036189 DOI: 10.3389/fphar.2022.769111] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Ethnopharmacological relevance: The genus Alternanthera (Amaranthaceae) comprises 139 species including 14 species used traditionally for the treatment of various ailments such as hypertension, pain, inflammation, diabetes, cancer, microbial and mental disorders. Aim of the review: To search research gaps through critical assessment of pharmacological activities not performed to validate traditional claims of various species of Alternanthera. This review will aid natural product researchers in identifying Alternanthera species with therapeutic potential for future investigation. Materials and methods: Scattered raw data on ethnopharmacological, morphological, phytochemical, pharmacological, toxicological, and clinical studies of various species of the genus Alternanthera have been compiled utilizing search engines like SciFinder, Google Scholar, PubMed, Science Direct, and Open J-Gate for 100 years up to April 2021. Results: Few species of Alternanthera genus have been exhaustively investigated phytochemically, and about 129 chemical constituents related to different classes such as flavonoids, steroids, saponins, alkaloids, triterpenoids, glycosides, and phenolic compounds have been isolated from 9 species. Anticancer, antioxidant, antibacterial, CNS depressive, antidiabetic, analgesic, anti-inflammatory, and immunomodulator effects have been explored in the twelve species of the genus. A toxicity study has been conducted on 3 species and a clinical study on 2 species. Conclusions: The available literature on pharmacological studies of Alternanthera species reveals that few species have been selected based on ethnobotanical surveys for scientific validation of their traditional claims. But most of these studies have been conducted on uncharacterized and non-standardized crude extracts. A roadmap of research needs to be developed for the isolation of new bioactive compounds from Alternanthera species, which can emerge out as clinically potential medicines.
Collapse
Affiliation(s)
- Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Vivek Dhir
- Chitkara College of Pharmacy, Chitkara University Punjab, Rajpura, India
| | - Reecha Madaan
- Chitkara College of Pharmacy, Chitkara University Punjab, Rajpura, India
- *Correspondence: Bairong Shen, ; Reecha Madaan,
| | - Deepak Kumar
- Department of Health and Family Welfare, Civil Hospital, Rampura Phul, India
| | - Simranjit Singh Bola
- Akal College of Pharmacy and Technical Education, Mastuana Sahib, Sangrur, India
| | - Monika Bansal
- Akal College of Pharmacy and Technical Education, Mastuana Sahib, Sangrur, India
| | - Suresh Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | | | - Shailja Singla
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Bairong Shen, ; Reecha Madaan,
| |
Collapse
|
16
|
Phytochemical Characterization, Antioxidant Activity, and Cytotoxicity of Methanolic Leaf Extract of Chlorophytum Comosum (Green Type) (Thunb.) Jacq. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030762. [PMID: 35164026 PMCID: PMC8840168 DOI: 10.3390/molecules27030762] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/19/2022]
Abstract
Chlorophytum genus has been extensively studied due to its diverse biological activities. We evaluated the methanolic extract of leaves of Chlorophytum comosum (Green type) (Thunb.) Jacques, the species that is less studied compared to C. borivilianum. The aim was to identify phytoconstituents of the methanolic extract of leaves of C. comosum and biological properties of its different fractions. Water fraction was analyzed with matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry. Nineteen compounds belonging to different chemical classes were identified in the methanolic extract of leaves of C. comosum (Green type) (Thunb.) Jacques. In addition to several fatty acids, isoprenoid and steroid compounds were found among the most abundant constituents. One of the identified compounds, 4'-methylphenyl-1C-sulfonyl-β-d-galactoside, was not detected earlier in Chlorophytum extracts. The water fraction was toxic to HeLa cells but not to Vero cells. Our data demonstrate that methanolic extract of leaves of C. comosum can be a valuable source of bioactive constituents. The water fraction of the extract exhibited promising antitumor potential based on a high ratio of HeLa vs. Vero cytotoxicity.
Collapse
|
17
|
Wen S, He L, Zhong Z, Zhao R, Weng S, Mi H, Liu F. Stigmasterol Restores the Balance of Treg/Th17 Cells by Activating the Butyrate-PPARγ Axis in Colitis. Front Immunol 2021; 12:741934. [PMID: 34691046 PMCID: PMC8526899 DOI: 10.3389/fimmu.2021.741934] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/21/2021] [Indexed: 12/20/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder with gut microbiota disequilibrium and regulatory T (Treg)/T helper 17 (Th17) immune imbalance. Stigmasterol, a plant-derived sterol, has shown anti-inflammatory effects. Our study aimed to identify the effects of stigmasterol on experimental colitis and the related mechanisms. Stigmasterol treatment restored the Treg/Th17 balance and altered the gut microbiota in a dextran sodium sulfate (DSS)-induced colitis model. Transplantation of the faecal microbiota of stigmasterol-treated mice significantly alleviated inflammation. Additionally, stigmasterol treatment enhanced the production of gut microbiota-derived short-chain fatty acids (SCFAs), particularly butyrate. Next, human naïve CD4+ T cells sorted from IBD patients were cultured under Treg- or Th17-polarizing conditions; butyrate supplementation increased the differentiation of Tregs and decreased Th17 cell differentiation. Mechanistically, butyrate activated peroxisome proliferator-activated receptor gamma (PPARγ) and reprogrammed energy metabolism, thereby promoting Treg differentiation and inhibiting Th17 differentiation. Our results demonstrate that butyrate-mediated PPARγ activation restores the balance of Treg/Th17 cells, and this may be a possible mechanism, by which stigmasterol attenuates IBD.
Collapse
Affiliation(s)
- Shuting Wen
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Long He
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuotai Zhong
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Runyuan Zhao
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Senhui Weng
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hong Mi
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengbin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
18
|
Sampath SJP, Rath SN, Kotikalapudi N, Venkatesan V. Beneficial effects of secretome derived from mesenchymal stem cells with stigmasterol to negate IL-1β-induced inflammation in-vitro using rat chondrocytes-OA management. Inflammopharmacology 2021; 29:1701-1717. [PMID: 34546477 DOI: 10.1007/s10787-021-00874-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/01/2021] [Indexed: 12/24/2022]
Abstract
Osteoarthritis (OA) is the most prevalent joint disease predominantly characterized by inflammation which drives cartilage destruction. Mesenchymal stem cells-condition medium (MSC-CM) or the secretome is enriched with bioactive factors and possesses anti-inflammatory and regenerative effects. The present study aimed at evaluating the effects of combining MSC-conditioned medium with stigmasterol compared with the individual treatments in alleviating interleukin-1 beta (IL-1β)-induced inflammation in rat chondrocytes. Stigmasterol is a phytosterol exhibiting anti-inflammatory effects. IL-1β (10 ng/ml) was used to induce inflammation and mimic OA in-vitro in primary rat articular chondrocytes. The IL-1β-stimulated chondrocytes were treated with MSC-CM, stigmasterol, and a combination of MSC-CM and stigmasterol for 24 h. Cell viability was measured using MTT assay. Protein expression of inducible nitric oxide synthase (iNOS), interleukin-6 (IL-6), collagen II (COL2A1) and matrix metalloproteinase (MMP)-13 were evaluated by immunofluorescence. Gene expression levels of MMP-3, MMP-13 and A Disintegrin-like and Metalloproteinases with Thrombospondin Motifs (ADAMTS)-5 were measured using qRT-PCR. NF-κB signaling pathway was studied using western blotting. A significant reduction in the expression of iNOS, IL-6, MMP-3, MMP-13 and ADAMTS-5, and a significant increase in COL2A1 expression was observed in the rat chondrocytes across all the treatment groups. However, the combination treatment of MSC-CM and stigmasterol remarkably reversed the IL-1β-induced pro-inflammatory/pro-catabolic responses to near normal levels comparable to the control group. The combination treatment (MSC-CM + stigmasterol) elicited a superior anti-inflammatory/anti-catabolic effect by inhibiting the IL-1β-induced NF-κB activation evidenced by the negligible phosphorylation of p65 and IκBα subunits, thereby emphasizing the benefit of the combination therapy over the individual treatments.
Collapse
Affiliation(s)
- Samuel Joshua Pragasam Sampath
- Stem Cell Research Laboratory, Department of Cell and Molecular Biology, National Institute of Nutrition, Indian Council of Medical Research, Tarnaka, Hyderabad, 500007, Telangana, India
| | - Subha Narayan Rath
- Regenerative Medicine and Stem Cells Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, 502285, Telangana, India
| | - Nagasuryaprasad Kotikalapudi
- Stem Cell Research Laboratory, Department of Cell and Molecular Biology, National Institute of Nutrition, Indian Council of Medical Research, Tarnaka, Hyderabad, 500007, Telangana, India
| | - Vijayalakshmi Venkatesan
- Stem Cell Research Laboratory, Department of Cell and Molecular Biology, National Institute of Nutrition, Indian Council of Medical Research, Tarnaka, Hyderabad, 500007, Telangana, India.
| |
Collapse
|
19
|
Heliotropium indicum L.: From Farm to a Source of Bioactive Compounds with Therapeutic Activity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9965481. [PMID: 34158818 PMCID: PMC8187075 DOI: 10.1155/2021/9965481] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/11/2021] [Indexed: 01/13/2023]
Abstract
This study aimed to summarize the available data on the ethnomedicinal and phytopharmacological activities of Heliotropium indicum L. based on database reports. For this purpose, an up-to-date literature search was carried out in the Google Scholar, Scopus, Springer Link, Web of Science, ScienceDirect, ResearchGate, PubMed, Chem Spider, Elsevier, BioMed Central, and patent offices (e.g., USPTO, CIPO, NPI, Google patents, and Espacenet) for the published materials. The findings suggest that the plant contains many important phytochemicals, including pyrrolizidine alkaloids, indicine, echinitine, supinine, heleurine, heliotrine, lasiocarpine, acetyl indicine, indicinine, indicine N-oxide, cynoglossine, europine N-oxide, heleurine N-oxide, heliotridine N-oxide, heliotrine N-oxide, heliotrine, volatile oils, triterpenes, amines, and sterols. Scientific reports revealed that the herb showed antioxidant, analgesic, antimicrobial, anticancer, antituberculosis, antiplasmodial, anticataract, antifertility, wound healing, antiinflammatory, antinociceptive, antihyperglycemic, anthelmintic, diuretic, antitussive, antiglaucoma, antiallergic, and larvicidal activity. In conclusion, in vitro studies with animal models seem to show the potential beneficial effects of H. indicum against a wide variety of disorders and as a source of phytotherapeutic compounds. However, clinical studies are necessary to confirm the effects observed in animal models, determine the toxicity of the therapeutic dose and isolate the truly bioactive components.
Collapse
|
20
|
Antioxidant, Anti-Inflammatory, and Inhibition of Acetylcholinesterase Potentials of Cassia timoriensis DC. Flowers. Molecules 2021; 26:molecules26092594. [PMID: 33946788 PMCID: PMC8125573 DOI: 10.3390/molecules26092594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022] Open
Abstract
Despite being widely used traditionally as a general tonic, especially in South East Asia, scientific research on Cassia timoriensis, remains scarce. In this study, the aim was to evaluate the in vitro activities for acetylcholinesterase (AChE) inhibitory potential, radical scavenging ability, and the anti-inflammatory properties of different extracts of C. timoriensis flowers using Ellman’s assay, a DPPH assay, and an albumin denaturation assay, respectively. With the exception of the acetylcholinesterase activity, to the best of our knowledge, these activities were reported for the first time for C. timoriensis flowers. The phytochemical analysis confirmed the existence of tannins, flavonoids, saponins, terpenoids, and steroids in the C. timoriensis flower extracts. The ethyl acetate extract possessed the highest phenolic and flavonoid contents (527.43 ± 5.83 mg GAE/g DW and 851.83 ± 10.08 mg QE/g DW, respectively) as compared to the other extracts. In addition, the ethyl acetate and methanol extracts exhibited the highest antioxidant (IC50 20.12 ± 0.12 and 34.48 ± 0.07 µg/mL, respectively), anti-inflammatory (92.50 ± 1.38 and 92.22 ± 1.09, respectively), and anti-AChE (IC50 6.91 ± 0.38 and 6.40 ± 0.27 µg/mL, respectively) activities. These results suggest that ethyl acetate and methanol extracts may contain bioactive compounds that can control neurodegenerative disorders, including Alzheimer’s disease, through high antioxidant, anti-inflammatory, and anti-AChE activities.
Collapse
|
21
|
Network Pharmacology Analysis of Traditional Chinese Medicine Formula Shuang Di Shou Zhen Tablets Treating Nonexudative Age-Related Macular Degeneration. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6657521. [PMID: 33815556 PMCID: PMC8012120 DOI: 10.1155/2021/6657521] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 12/14/2022]
Abstract
Objective To analyze the pharmacological mechanism of the treatment of dry age-related macular degeneration (dry AMD) based on a network pharmacological approach of Shuang Di Shou Zhen Tablets (SDSZT) and to provide a new reference for the current lack of effective treatment of dry AMD. Methods The main chemical constituents and their targets of Rehmanniae Radix Praeparata, Ligustrum lucidum, Mori Fructus, Paeonia albiflora, Rhizoma Dioscoreae, Alisma orientale, Schisandra chinensis, Radix Polygoni Multiflori Preparata, Ophiopogon japonicus, and Radix Rehmanniae were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and Traditional Chinese Medicine Integrated Database (TCMID). The active ingredients of traditional Chinese medicine were screened according to Absorption, Distribution, Metabolism, and Excretion (ADME), the gene names of the targets of each active ingredient were obtained from the Uniprot database, the main targets of dry AMD were obtained from GeneCards and DisGeNET database, and the protein interaction analysis was performed on the String database. The Metascape database was used to analyze the “drug-component-target” and the biological processes and networks involved, and then, Cytoscape 3.8.1 was used to construct the “ SDSZT component-dry AMD target-pathway” network. Results The main active ingredients of SDSZT for dry AMD treatment are quercetin, kaempferol, luteolin, β-glutamine, β-carotene, etc. And, the core targets are RAC-alpha serine/threonine-protein kinase (AKT1), prostaglandin G/H synthase 1 (PTGS1), tumor necrosis factor (TNF), transcription factor AP-1 (JUN), apoptosis regulator Bcl-2 (BCL2), caspase-3 (CASP3), phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit gamma isoform (PIK3CG), androgen receptor (AR), apoptosis regulator BAX (BAX), etc. The biological pathways for the treatment of age-related macular degeneration by SDSZT mainly act on pathways in cancer, fluid shear stress and atherosclerosis, and TNF signaling pathway, and the main function of SDSZT is to regulate intracellular cytokine receptor binding. Conclusion This study initially reveals the multiconstituent, multitarget, and multipathway mechanism of action of SDSZT in the treatment of dry AMD and provides the basis for the clinical application of SDSZT.
Collapse
|
22
|
Jansen C, Baker JD, Kodaira E, Ang L, Bacani AJ, Aldan JT, Shimoda LMN, Salameh M, Small-Howard AL, Stokes AJ, Turner H, Adra CN. Medicine in motion: Opportunities, challenges and data analytics-based solutions for traditional medicine integration into western medical practice. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113477. [PMID: 33098971 PMCID: PMC7577282 DOI: 10.1016/j.jep.2020.113477] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/23/2020] [Accepted: 10/13/2020] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional pharmacopeias have been developed by multiple cultures and evaluated for efficacy and safety through both historical/empirical iteration and more recently through controlled studies using Western scientific paradigms and an increasing emphasis on data science methodologies for network pharmacology. Traditional medicines represent likely sources of relatively inexpensive drugs for symptomatic management as well as potential libraries of new therapeutic approaches. Leveraging this potential requires hard evidence for efficacy that separates science from pseudoscience. MATERIALS AND METHODS We performed a review of non-Western medical systems and developed case studies that illustrate the epistemological and practical translative barriers that hamper their transition to integration with Western approaches. We developed a new data analytics approach, in silico convergence analysis, to deconvolve modes of action, and potentially predict desirable components of TM-derived formulations based on computational consensus analysis across cultures and medical systems. RESULTS Abstraction, simplification and altered dose and delivery modalities were identified as factors that influence actual and perceived efficacy once a medicine is moved from a non-Western to Western setting. Case studies on these factors highlighted issues with translation between non-Western and Western epistemologies, including those where epistemological and medicinal systems drive markets that can be epicenters for zoonoses such as the novel Coronavirus. The proposed novel data science approach demonstrated the ability to identify and predict desirable medicinal components for a test indication, pain. CONCLUSIONS Relegation of traditional therapies to the relatively unregulated nutraceutical industry may lead healthcare providers and patients to underestimate the therapeutic potential of these medicines. We suggest three areas of emphasis for this field: First, vertical integration and embedding of traditional medicines into healthcare systems would subject them to appropriate regulation and evidence-based practice, as viable integrative implementation mode. Second, we offer a new Bradford-Hill-like framework for setting research priorities and evaluating efficacy, with the goal of rescuing potentially valuable therapies from the nutraceutical market and discrediting those that are pseudoscience. Third, data analytics pipelines offer new capacity to generate new types of TMS-inspired medicines that are rationally-designed based on integrated knowledge across cultures, and also provide an evaluative framework against which to test claims of fidelity and efficacy to TMS made for nutraceuticals.
Collapse
Affiliation(s)
- C Jansen
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawai'i, USA.
| | - J D Baker
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawai'i, USA.
| | - E Kodaira
- Medicinal Plant Garden, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, 252-0373, Kanagawa, Japan.
| | - L Ang
- Undergraduate Program in Biology, Chaminade University, Honolulu, Hawai'i, USA.
| | - A J Bacani
- Undergraduate Program in Biology, Chaminade University, Honolulu, Hawai'i, USA.
| | - J T Aldan
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawai'i, USA; Graduate Program in Public Health, Eastern Washington University, Spokane, WA, USA.
| | - L M N Shimoda
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawai'i, USA.
| | - M Salameh
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawai'i, USA.
| | | | - A J Stokes
- Laboratory of Experimental Medicine, John A. Burns School of Medicine, Honolulu, Hawai'i, USA; Hawai'i Data Science Institute, University of Hawai'i at Manoa, Honolulu, Hawai'i, USA; The Adra Institute, Boston, MA, USA.
| | - H Turner
- Laboratory of Immunology and Signal Transduction, Chaminade University, Honolulu, Hawai'i, USA; The Adra Institute, Boston, MA, USA.
| | - C N Adra
- The Adra Institute, Boston, MA, USA.
| |
Collapse
|
23
|
Network Pharmacology Strategy to Investigate the Pharmacological Mechanism of Siwu Decoction on Primary Dysmenorrhea and Molecular Docking Verification. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021. [DOI: 10.1155/2021/6662247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective. To study the pharmacological mechanisms of Siwu decoction (SWD) on primary dysmenorrhea (PDM) and verify with molecular docking. Methods. The Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) was utilized to acquire the active compounds and their corresponding target genes. The GeneCards database was utilized in the search for target genes that were associated with PDM. The intersection genes from the active target genes of SWD and those associated with PDM represented the active target genes of SWD that act on PDM. The Gene Ontology (GO) function enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were both carried out by RGUI 3.6.1 and Cytoscape 3.6.0 software. Cytoscape was also utilized for creating a compound-target network, and a protein-protein interaction (PPI) network was created through the STRING database. Molecular docking simulations of the macromolecular protein target receptors and their corresponding compounds were performed using AutoDockTool 1.5.6 and AutoDock Vina software. Results. We identified 14 active compounds as well as 97 active target genes of SWD by using the TCMSP. We compared the 97 active target genes of SWD to the 299 target genes related to PDM, and 23 active target genes for SWD that act on PDM which correlated with 11 active compounds were detected. The compound-target network as well as the PPI network were created, in addition to selecting the most essential compounds and their targets in order to create a key compound-target network. The most essential compounds were kaempferol, beta-sitosterol, stigmasterol, and myricanone. The key targets were AKT1, PTGS2, ESR1, AHR, CASP3, and PGR. Lastly, molecular docking was used to confirm binding of the target with its corresponding compound. Conclusion. The pharmacological mechanisms of SWD that act on PDM were investigated, and the active compounds in the SWD for treating PDM were further verified.
Collapse
|
24
|
Li X, Lin H, Wang Q, Cui L, Luo H, Luo L. Chemical composition and pharmacological mechanism of shenfu decoction in the treatment of novel coronavirus pneumonia (COVID-19). Drug Dev Ind Pharm 2020; 46:1947-1959. [PMID: 33054436 DOI: 10.1080/03639045.2020.1826510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Shenfu decoction has outstanding curative effects in the treatment of COVID-19. This study aimed to explore the material basis and molecular mechanism of Shenfu Decoction through network pharmacology and molecular mechanisms, to provide a research basis for clinical medication and clues for subsequent research. METHODS The active components and targets of Shenfu decoction were searched in the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), and the COVID-19-associated genes were collected using the Gene Cards platform. The target protein-protein interaction network map was constructed by mapping two genes, and the 'drug-active ingredient-target' network was constructed using Cytoscape software. The Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment of the mapping targets were analyzed. RESULT Based on Traditional Chinese medicine, Shenfu Decoction can take effect in the lung, spleen, kidney and heart. Considering oral bioavailability (OB) ≥ 30% and drug-like (DL) ≥ 0.18 as the standard, 43 active compounds were screened and 114 Shenfu decoction action targets were collected. The key targets were CASP3, MAPK8, PTGS2, IL1B, PPARG, ICAM1, IFNG, RELA, NOS2, NOS3, HMOX1, CASP8, STAT1, and TGFB1. According to the standard of p < .05, GO function was enriched in 108 biological processes, 16 cell processes and 27 molecular processes. Sixty-three signaling pathways were enriched by KEGG, which can be divided into four types: viral infection pathways, signal pathways, biological process pathways and different disease pathways. The comparison of negative and positive prescriptions further reflects the positive effect of Shenfu decoction against COVID-19. Finally, the effective ingredients with the high degree were molecular docked with Mpro, Rdrp and Spro proteins to further confirm the intervention effect of Shenfu Decoction on COVID-19. CONCLUSION Shenfu decoction played an important role in regulating the anti-virus process, regulating immunity, inhibiting inflammation and regulating apoptosis through the interrelated regulation mechanism of multi-components and multi-targets, to treat patients with severe COVID-19.
Collapse
Affiliation(s)
- Xiaoling Li
- Animal Experiment Center, Guangdong Medical University, Zhanjiang, China
| | - Haowen Lin
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Qu Wang
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, China
| | - Hui Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
| |
Collapse
|
25
|
Masuelli L, Benvenuto M, Focaccetti C, Ciuffa S, Fazi S, Bei A, Miele MT, Piredda L, Manzari V, Modesti A, Bei R. Targeting the tumor immune microenvironment with "nutraceuticals": From bench to clinical trials. Pharmacol Ther 2020; 219:107700. [PMID: 33045254 DOI: 10.1016/j.pharmthera.2020.107700] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2020] [Indexed: 02/06/2023]
Abstract
The occurrence of immune effector cells in the tissue microenvironment during neoplastic progression is critical in determining tumor growth outcomes. On the other hand, tumors may also avoid immune system-mediated elimination by recruiting immunosuppressive leukocytes and soluble factors, which coordinate a tumor microenvironment that counteracts the efficiency of the antitumor immune response. Checkpoint inhibitor therapy results have indicated a way forward via activation of the immune system against cancer. Widespread evidence has shown that different compounds in foods, when administered as purified substances, can act as immunomodulators in humans and animals. Although there is no universally accepted definition of nutraceuticals, the term identifies a wide category of natural compounds that may impact health and disease statuses and includes purified substances from natural sources, plant extracts, dietary supplements, vitamins, phytonutrients, and various products with combinations of functional ingredients. In this review, we summarize the current knowledge on the immunomodulatory effects of nutraceuticals with a special focus on the cancer microenvironment, highlighting the conceptual benefits or drawbacks and subtle cell-specific effects of nutraceuticals for envisioning future therapies employing nutraceuticals as chemoadjuvants.
Collapse
Affiliation(s)
- Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Monica Benvenuto
- Saint Camillus International University of Health and Medical Sciences, via di Sant'Alessandro 8, 00131 Rome, Italy; Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy; Department of Human Science and Promotion of the Quality of Life, San Raffaele University Rome, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Sara Ciuffa
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Sara Fazi
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Arianna Bei
- Medical School, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Lucia Piredda
- Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Vittorio Manzari
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy; CIMER, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
26
|
Abd Rani NZ, Kumolosasi E, Jasamai M, Jamal JA, Lam KW, Husain K. In vitro anti-allergic activity of Moringa oleifera Lam. extracts and their isolated compounds. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:361. [PMID: 31829185 PMCID: PMC6907282 DOI: 10.1186/s12906-019-2776-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/27/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Moringa oleifera Lam. is a commonly used plant in herbal medicine and has various reported bioactivities such as antioxidant, antimicrobial, anticancer and antidiabetes. It is rich in nutrients and polyphenols. The plant also has been traditionally used for alleviating allergic conditions. This study was aimed to examine the anti-allergic activity of M. oleifera extracts and its isolated compounds. METHOD M. oleifera leaves, seeds and pods were extracted with 80% of ethanol. Individual compounds were isolated using a column chromatographic technique and elucidated based on the nuclear magnetic resonance (NMR) and electrospray ionisation mass spectrometry (ESIMS) spectral data. The anti-allergic activity of the extracts, isolated compounds and ketotifen fumarate as a positive control was evaluated using rat basophilic leukaemia (RBL-2H3) cells for early and late phases of allergic reactions. The early phase was determined based on the inhibition of beta-hexosaminidase and histamine release; while the late phase was based on the inhibition of interleukin (IL-4) and tumour necrosis factor (TNF-α) release. RESULTS Two new compounds; ethyl-(E)-undec-6-enoate (1) and 3,5,6-trihydroxy-2-(2,3,4,5,6-pentahydroxyphenyl)-4H-chromen-4-one (2) together with six known compounds; quercetin (3), kaempferol (4), β-sitosterol-3-O-glucoside (5), oleic acid (6), glucomoringin (7), 2,3,4-trihydroxybenzaldehyde (8) and stigmasterol (9) were isolated from M. oleifera extracts. All extracts and the isolated compounds inhibited mast cell degranulation by inhibiting beta-hexosaminidase and histamine release, as well as the release of IL-4 and TNF-α at varying levels compared with ketotifen fumarate. CONCLUSION The study suggested that M. oleifera and its isolated compounds potentially have an anti-allergic activity by inhibiting both early and late phases of allergic reactions.
Collapse
Affiliation(s)
- Nur Zahirah Abd Rani
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Endang Kumolosasi
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Malina Jasamai
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Jamia Azdina Jamal
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Kok Wai Lam
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Khairana Husain
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
A Network Pharmacology Approach to Uncover the Molecular Mechanisms of Herbal Formula Kang-Bai-Ling for Treatment of Vitiligo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3053458. [PMID: 31781265 PMCID: PMC6875403 DOI: 10.1155/2019/3053458] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/15/2019] [Accepted: 10/04/2019] [Indexed: 12/19/2022]
Abstract
Background Kang-bai-ling (KBL), a Chinese patent medicine, has been demonstrated as an effective therapy for vitiligo in China. However, the pharmacological mechanisms of KBL have not been completely elucidated. Methods In this study, the potential multicomponent, multitarget, and multipathway mechanism of KBL against vitiligo was clarified by using network pharmacology-based strategy. In brief, potential targets of KBL were collected based on TCMSP databases, followed by network establishment concerning the interactions of potential targets of KBL with well-known therapeutic targets of vitiligo by using protein-protein interaction (PPI) data. As a result, key nodes with higher level of seven topological parameters, including “degree centrality (DC),” “betweenness centrality (BC),” “closeness centrality (CC),” “eigenvector centrality (EC),” “network centrality (NC),” and “local average connectivity (LAC)” were identified as the main targets in the network, followed by subsequent incorporation into the ClueGO for GO and KEGG signaling pathway enrichment analysis. Results In accordance with the topological importance, a total of 23 potential targets of KBL on vitiligo were identified as main hubs. Additionally, enrichment analysis suggested that targets of KBL on vitiligo were mainly clustered into multiple biological processes (associated with DNA translation, lymphocyte differentiation and activation, steroid biosynthesis, autoimmune and systemic inflammatory reaction, neuron apoptosis, and vitamin deficiency) and related pathways (TNF, JAK-STAT, ILs, TLRs, prolactin, and NF-κB), indicating the underlying mechanisms of KBL on vitiligo. Conclusion In this work, we successfully illuminated the “multicompounds, multitargets” therapeutic action of KBL on vitiligo by using network pharmacology. Moreover, our present outcomes might shed light on the further clinical application of KBL on vitiligo treatment.
Collapse
|