1
|
Chen D, Yao H, Qiu X, Xu L, Ou Y, Xin J, Lu S, Li M, Geng Y, Zhang Y, Hu M, Ren Z, Wu JQ. Purification, Structural Analysis and Bioactivity of Pueraria montana Polysaccharide. Foods 2025; 14:1359. [PMID: 40282761 PMCID: PMC12026434 DOI: 10.3390/foods14081359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Pueraria montana is a medicinal and edible plant widely distributed in Asia. It has antipyretic, analgesic, and anti-inflammatory properties. In this study, a novel polysaccharide (PMPS-A1) was obtained through purification, and its biological activity was investigated. Structural analysis revealed that PMPS-A1 was composed of fructose and glucose, with a molecular weight of 12168 Da. The main chain structure was →1)-β-D-Fruf-(2→, →4)-α-D-Glcp-(1→, →4)-α-D-Glcp-(1→, and →4)-α-D-Glcp-(1→. The branched chain of α-D-Glcp-(1→3)-α-D-Glcp-(1→ and α-D-Glcp-(1→ connected to the O-3 and O-6 positions of residue →3,4)-α-D-Glcp-(1→ and →4,6)-α-D-Glcp-(1→, respectively. In vitro, PMPS-A1 had a favorable scavenging ability of the hydroxyl radical and 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical and downregulated the expression of interleukin-6 and nitric oxide in lipopolysaccharide-induced RAW264.7 macrophages. In addition, the Caenorhabditis model assay demonstrated that PMPS-A1 decreased the buildup of lipofuscin and reactive oxygen species. Overall, these results enhance our knowledge of the chemical composition and bioactivity of a Pueraria montana polysaccharide and point to the potential use of PMPS-A1 for antioxidant and anti-aging qualities, providing a theoretical basis for the medicinal and edible application of Pueraria montana polysaccharide.
Collapse
Affiliation(s)
- Dandan Chen
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China;
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Hongliang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Xiang Qiu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Lang Xu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Yanghui Ou
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Jianghui Xin
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Shengjia Lu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Mengjie Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Yan Geng
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Yali Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Minxiu Hu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.Y.); (X.Q.); (L.X.); (Y.O.); (J.X.); (S.L.); (M.L.); (Y.G.); (Y.Z.); (M.H.)
| | - Zhiming Ren
- JiangMen Industrial Technology Research Institute, Guangdong Academy of Sciences, Jiangmen 529020, China
| | - Jia-Qiang Wu
- School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, China;
| |
Collapse
|
2
|
Qiao M, Xue T, Zhu Y, Yang J, Hu J. Polysaccharides from Cistanche deserticola mitigate inflammatory bowel disease via modulating intestinal microbiota and SRC/EGFR/PI3K/AKT signaling pathways. Int J Biol Macromol 2025; 308:142452. [PMID: 40139591 DOI: 10.1016/j.ijbiomac.2025.142452] [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: 12/14/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Polysaccharides of Cistanche deserticola Ma (CDPS), with high safety and low toxicity have been reported to possess anti-inflammatory, immunomodulatory, antioxidant, anti-aging, anti-osteoporosis, antidepressant, intestinal flora regulatory and hepatoprotective properties. Nevertheless, the effects of CDPS on inflammatory bowel disease (IBD) and its underlying mechanisms have never been reported. To estimate its therapeutic potential on IBD, the extracted CDPS were characterized via utilizing a series of chemical, spectroscopic, and instrumental analyses, and the protective effects and mechanisms of CDPS in colitis mice was investigated. Our results indicated that CDPS were identified as acidic heteropolysaccharides. CDPS alleviated dextran sodium sulfate-induced IBD mice characterized by decreasing disease activity index, improving colon length and body weight, restoring histopathological lesions, inhibiting the expression of pro-inflammatory cytokine (IL-6, IL-1β, TNF-α) and MPO activity, elevating the expression of anti-inflammatory cytokine (IL-10) in colon tissue. The findings manifested CDPS could mitigate the inflammation of colon. Simultaneously, CDPS inhibited the expression of genes and proteins associated with SRC/EGFR/PI3K/AKT signaling pathways, and reduced the diversity and abundance of harmful gut microbiota, including Helicobacter, Bacteroides and Colidextribacter, while descending the relative abundance of Lachnospiraceae_NK4A136_group at genus level. In summary, this work elucidated that CDPS alleviates IBD symptoms via mitigating the inflammation of colon, and modulating intestinal microbiota and SRC/EGFR/PI3K/AKT signaling pathways. It underscores the promise of CDPS as a functional food ingredient or preventive drugs for IBD.
Collapse
Affiliation(s)
- Ming Qiao
- Department of Pharmacy, The First Affiliated Hospital, Xinjiang Medical University, Urumqi 830011, China; Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, China
| | - Taotao Xue
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Yi Zhu
- Department of Pharmacy, The First Affiliated Hospital, Xinjiang Medical University, Urumqi 830011, China; Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, China
| | - Jianhua Yang
- Department of Pharmacy, The First Affiliated Hospital, Xinjiang Medical University, Urumqi 830011, China; Xinjiang Key Laboratory of Clinical Drug Research, Urumqi 830011, China
| | - Junping Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
3
|
Xue TT, Zheng DX, Hou Q, Wen LM, Wang BJ, Geng RY, Wang QQ, Dai W, Tian LY, He SQ, Yang JH, Hu JP. Optimization of Extraction Process, Structural Characterization, and Antioxidant and Hypoglycemic Activity Evaluation of Polysaccharides From the Medicinal and Edible Plant: Cistanche deserticola Ma. PHYTOCHEMICAL ANALYSIS : PCA 2025. [PMID: 39853820 DOI: 10.1002/pca.3512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/09/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025]
Abstract
INTRODUCTION Cistanche deserticola Ma (CD), an edible and medicinal plant native to Xinjiang, Inner Mongolia, and Gansu in China, is rich in bioactive polysaccharides known for their health-promoting properties. The polysaccharides of C. deserticola (CDPs) have been shown to possess a range of beneficial activities, including immunomodulatory, anti-aging, antioxidant, and anti-osteoporosis effects. OBJECTIVE This study seeks to identify the optimal conditions for extracting CDPs using hot water. Additionally, it aims to evaluate their chemical properties, antioxidant activity, hypoglycemic effects, and cytotoxicity. The findings will provide a theoretical foundation for the potential use of CDPs in functional foods and pharmaceuticals. METHODOLOGY The study employed response surface methodology to optimize the hot water extraction conditions for CDPs. The extracted CDPs were characterized using a range of chemical, spectroscopic, and instrumental analyses. Furthermore, their antioxidant activity, hypoglycemic effects, and cytotoxicity were evaluated through relevant assays to assess their potential health benefits. RESULTS Under optimal conditions, the yield of CDPs was 45.85% ± 1.91%. CDPs were identified as acidic heteropolysaccharides with a wide molecular weight distribution, ranging from 0.3 to 128.2 kDa. They were composed primarily of glucose (51.21%), arabinose (32.86%), galactose (17.88%), and smaller amounts of galacturonic acid (4.66%), rhamnose (1.85%), mannose (1.32%), glucosamine hydrochloride (1.08%), and xylose (0.56%). Antioxidant assays demonstrated that CDPs exhibited significant free radical scavenging activity, metal ion chelation, and reducing power. Additionally, CDPs inhibited α-glucosidase and α-amylase in vitro through a mixed-type mechanism, as well as static fluorescence quenching. Cytotoxicity assays showed that CDPs were nontoxic to L02 and AML12 cells. CONCLUSION This study offers a theoretical foundation for the potential use of CDPs in functional foods and pharmaceuticals and provides valuable insights for the development of new antioxidant and hypoglycemic agents from natural sources.
Collapse
Affiliation(s)
- Tao-Tao Xue
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Dong-Xuan Zheng
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Qiang Hou
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Li-Mei Wen
- Department of Pharmacy, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China
| | - Bao-Juan Wang
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
- Department of Pharmacy, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China
| | - Ruo-Yu Geng
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Qian-Qian Wang
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Wu Dai
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Li-Ying Tian
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Sheng-Qi He
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Jian-Hua Yang
- Department of Pharmacy, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China
| | - Jun-Ping Hu
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi, China
| |
Collapse
|
4
|
Zhang Y, Zhang Y, Li S, Liu C, Liang J, Nong Y, Chen M, Sun R. Quaternity method for integrated screening, separation, extraction optimization, and bioactivity evaluation of acetylcholinesterase inhibitors from Sophora flavescens Aiton. PHYTOCHEMICAL ANALYSIS : PCA 2025; 36:52-67. [PMID: 38957046 DOI: 10.1002/pca.3415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Sophora flavescens Aiton (Fabaceae), a ubiquitous plant species in Asia, contains a wide range of pharmacologically active compounds, such as flavonoids, with potential anti-Alzheimer's disease (anti-AD) effects. OBJECTIVES The objective of the study is to develop a quaternity method for the screening, isolation, extraction optimization, and activity evaluation of acetylcholinesterase (AChE)-inhibiting compounds from S. flavescens to realize high-throughput screening of active substances in traditional Chinese medicine and to provide experimental data for the development of anti-AD drugs. METHODS With AChE as the target molecule, affinity ultrafiltration and liquid chromatography-mass spectrometry were applied to screen for potential inhibitors of the enzyme in S. flavescens. Orthogonal array experiments combined with the multi-objective Non-Dominated Sorting Genetic Algorithm III was used for the first time to optimize the process for extracting the active substances. Enzyme inhibition kinetics and molecular docking studies were performed to verify the potential anti-AD effects of the active compounds. RESULTS Five AChE-inhibiting compounds were identified: kushenol I, kurarinone, sophoraflavanone G, isokurarinone, and kushenol E. These were successfully separated at purities of 72.88%, 98.55%, 96.86%, 96.74%, and 95.84%, respectively, using the n-hexane/ethyl acetate/methanol/water (4.0/5.0/4.0/5.0, v/v/v/v), n-hexane/ethyl acetate/methanol/water (5.0/5.0/6.0/4.0, v/v/v/v), and n-hexane/ethyl acetate/methanol/water (4.9/5.1/5.7/4.3, v/v/v/v) mobile phase systems. Enzyme inhibition kinetics revealed that kushenol E had the best inhibitory effect. CONCLUSION This study elucidates the mechanism of action of five active AChE inhibitors in S. flavescens and provides a theoretical basis for the screening and development of anti-AD and other therapeutic drugs.
Collapse
Affiliation(s)
- Yutong Zhang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Yuchi Zhang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Sainan Li
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Chunming Liu
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Jiaqi Liang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Yuyu Nong
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Ming Chen
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Ruijun Sun
- Central Laboratory, Changchun Normal University, Changchun, China
| |
Collapse
|
5
|
Liao J, Zhao W, Zhang Y, Zou Z, Zhang Q, Chen D, Du B, Li P. Dendrobium officinale Kimura et Migo polysaccharide ameliorated DNFB-induced atopic dermatitis in mice associated with suppressing MAPK/NF-κB/STAT3 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118677. [PMID: 39121927 DOI: 10.1016/j.jep.2024.118677] [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: 06/11/2024] [Revised: 07/28/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dendrobium officinale Kimura et Migo as a valuable Chinese medicine has been used in China for more than 2000 years. Its main active components, polysaccharide (DOP), has been reported to have various pharmacological effects, including anti-inflammatory, antioxidant and alleviating AD effects. However, the precise mechanism underlying its therapeutic effect in AD remains largely unclear. AIM OF THE STUDY The present study sought to assess the efficacy of DOP and elucidate its intricate mechanisms in ameliorating DNFB-induced AD. MATERIALS AND METHODS Mice were sensitized with DNFB and treated with DOP application for 14 days. Treatment effects were assessed using dermatitis scores, ear thickness and scratching frequency. Epidermal thickness, mast cells and CD4+ T cells infiltration were detected by using H&E, toluidine blue staining and immunofluorescence staining respectively. Serum histamine (HIS), immunoglobulin E (IgE), thymic stromal lymphopoietin (TSLP), skin SOD, MDA, GHS, CAT, inflammatory cytokines (TNF-α, IFN-γ, IL-1β, IL-4, IL-5, IL-13) and chemokine (MIP-α, MDC, MCP-1) levels were quantify by ELISA and immunohistochemistry. Additionally, qPCR and Western blot analyses were performed to assess genes and proteins expression associated with MAPK/NF-κB/STAT3 signaling pathway. RESULTS The results indicated that DOP effectively mitigated AD-like skin lesions in mice through multiple pathways. It reduced epidermal thickness, ear thickness and scratching frequency in AD mice. Additionally, DOP mitigated inflammatory responses by decreasing the levels of inflammatory factors, as well as reducing serum levels of IgE, HIS, and TSLP. Moreover, DOP inhibited infiltration of mast cells and CD4+ T cells, suppressed the expression of skin chemokines such as MDC, MCP-1, and MIP-α, and enhanced filaggrin content in AD mice. Furthermore, DOP significantly boosted antioxidant capacity, as well as significantly reduced the expression of JAK1, STAT3, NF-κB p65, IκBα, ERK1/2, and p38 proteins and phosphorylated proteins such as p-JAK1, p-STAT3, p-NF-κB p65, p-IκBα, p-ERK1/2, and p-p38. CONCLUSIONS These findings suggested that DOP has significant anti-AD activity, primarily through reducing inflammatory responses, improving antioxidant capacity, repairing the skin barrier, and down-regulating key genes and proteins in MAPK/NF-κB/STAT3 signaling pathway, and that this study may provide valuable insights into the development of innovative therapies for the treatment of AD.
Collapse
Affiliation(s)
- Jingru Liao
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Wenjun Zhao
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Yuwei Zhang
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Zebin Zou
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Qilin Zhang
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Dongqiu Chen
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China; Hua an Tang Biotech Group Co., Ltd., Guangzhou, 510000, China
| | - Bing Du
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China
| | - Pan Li
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510640, China.
| |
Collapse
|
6
|
Meng R, Wan ZW, Yang HY, Wu L, Yaseen A, Li B, Zhang X. Ultrasonic-Assisted Extraction, Structural Characteristics, and Activity of Polysaccharides From Gymnocladus chinensis Baill. Chem Biodivers 2024:e202402097. [PMID: 39617719 DOI: 10.1002/cbdv.202402097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/09/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024]
Abstract
In this study, the response surface method (RSM) was used to determine the optimal parameters of ultrasonic-assisted extraction of Gymnocladus chinensis Baill. polysaccharides (GCBPs). The optimal extraction conditions were as follows: a liquid-solid ratio of 50.4 mL/g, an extraction temperature of 60°C, an extraction time of 43 min, and an extraction power of 240 W. Under these conditions, the extraction rate of GCBP reached 50.1%. Three fractions of GCBP-40, GCBP-60, and GCBP-80 with sugar contents of 64.33%, 59.16%, and 59.46% were obtained by different concentrations of ethanol purification. All the three fractions consisted of six monosaccharides with different ratios, namely, Man, Rha, Gal, Glu, Ara, and Xyl. From Congo red test, it is confirmed that all three fractions could have a triple helix structure. In addition, the antioxidant and hypoglycemic experiments showed that all three components had excellent antioxidant activity (2,2-diphenyl-1-picrylhydrazyl [DPPH] free-radical scavenging activity, 2,2-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) [ABTS] free-radical scavenging activity, and OH- free-radical scavenging activity) and hypoglycemic activity (α-glucosidase inhibitory activity).
Collapse
Affiliation(s)
- Rui Meng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Zhi-Wen Wan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Hua-Yuan Yang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Lv Wu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Aftab Yaseen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bin Li
- Guizhou Ecological Energy Vocational College, No. 1, Caiguan Road, Yunyan District, Guiyang, Guiyang, China
| | - Xia Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| |
Collapse
|
7
|
Liu M, Wang C, Zhang H, Guo H, Kang L, Li H, Li K. A systematic review on polysaccharides from Morinda officinalis How: Advances in the preparation, structural characterization and pharmacological activities. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118090. [PMID: 38521432 DOI: 10.1016/j.jep.2024.118090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/06/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Morinda officinalis How is called "Ba-Ji-Tian" in Traditional Chinese Medicine (TCM), which belongs to the genus Rubiaceae and is widely used for medicinal purposes in China and other eastern Asian countries. Morinda officinalis How polysaccharides (MOPs) are one of the key bioactive components, and have a variety of biological activities, such as antioxidation, antifatigue, enhanced immunity, antiosteoporosis, ect. AIM OF THE REVIEW This review is aimed at providing comprehensive information of the latest preparation technologies, structural characterization, and pharmacological effects of MOPs. A more in-depth research on the structure and clinical pharmacology of the MOPs was explored. It could lay a foundation for further investigate the pharmacological activities and guide the safe clinical practice of MOPs. MATERIALS AND METHODS The Web of Science, PubMed, Scifinder, Google Scholar, CNKI, Wanfang database, and other online database are used to search and collect the literature on extraction and separation methods, structural characterization, and pharmacological activities of MOPs publisher from 2004 to 2023. The key words are "Morinda officinalis polysaccharides", "extraction", "isolation", "purification" and "pharmacological effects". RESULTS Morinda officinalis has been widely used in tonifying the kidney yang since ancient times, and is famous for one of the "Four Southern Medicines" in China for the treatment of depression, osteoporosis, rheumatoid arthritis, infertility, fatigue and Alzheimer's disease. The active ingredients of Morinda officinalis that have been researched on the treatment of depression and osteoporosis are mostly polysaccharides and oligosaccharides. The content of polysaccharides varies with different methods of extraction, separation and purification. MOPs have a wide range of pharmacological effects, including antioxidant, antifatigue, immunomodulatory, antiosteoporosis, and regulation of spermatogenesis activities. These pharmacological properties lay a foundation for the treatment of oxidative stress, osteoporosis, spermatogenic dysfunction, immunodeficiency, inflammation and other diseases with MOPs. CONCLUSIONS At present, MOPs have been applied in the treatment of skeletal muscle atrophy, varicocele, osteoporosis, because of its effects of enhancing immunity, improving reproduction and antioxidant. However, the structure-activity relationship of these effects are still not clear. The more deeply study could be conducted on the MOPs in the future. The toxicology and clinical pharmacology, as well as mechanism of action of MOPs were also needed to deeply studied and clarified. This paper could lay the foundation for the application and safety of MOPs in multifunctional foods and drugs.
Collapse
Affiliation(s)
- Mengyun Liu
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, PR China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China
| | - Chen Wang
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, PR China
| | - Hongwei Zhang
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, PR China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China
| | - Hui Guo
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, PR China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China
| | - Le Kang
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, PR China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China
| | - Hongwei Li
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, PR China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China.
| | - Kai Li
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, PR China; Henan Research Center for Special Processing Technology of Chinese Medicine, Zhengzhou, 450046, PR China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, PR China.
| |
Collapse
|
8
|
Wang R, Ruan X, Chen J, Deng L, Zhou W, Shuai X, Liang R, Dai T. Physicochemical Characterization and Biological Properties of Polysaccharides from Alpiniae oxyphyllae Fructus. Polymers (Basel) 2024; 16:1705. [PMID: 38932054 PMCID: PMC11207487 DOI: 10.3390/polym16121705] [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: 05/13/2024] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Polysaccharides (AOPs) were extracted from Alpiniae oxyphyllae fructus using three distinct methods: hot water (AOP-HW), hydrochloric acid (AOP-AC), and NaOH/NaBH4 (AOP-AL). This study systematically investigated and compared the physicochemical properties, structural characteristics, antioxidant activities, and α-amylase inhibitory activities of the extracted polysaccharides. Among the three AOPs, AOP-AC exhibited the highest yield (13.76%) and neutral sugar content (80.57%), but had the lowest molecular weight (121.28 kDa). Conversely, AOP-HW had the lowest yield (4.54%) but the highest molecular weight (385.42 kDa). AOP-AL was predominantly composed of arabinose (28.42 mol%), galacturonic acid (17.61 mol%), and galactose (17.09 mol%), while glucose was the major sugar in both AOP-HW (52.31 mol%) and AOP-AC (94.77 mol%). Functionally, AOP-AL demonstrated superior scavenging activities against DPPH, hydroxyl, and ABTS radicals, whereas AOP-AC exhibited the strongest inhibitory effect on α-amylase. These findings indicate that the extraction solvent significantly influences the physicochemical and biological properties of AOPs, thus guiding the selection of appropriate extraction methods for specific applications. The results of this study have broad implications for industries seeking natural polysaccharides with antioxidant and enzymatic inhibitory properties.
Collapse
Affiliation(s)
- Risi Wang
- School of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China;
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xinmei Ruan
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Jun Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Lizhen Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Wei Zhou
- Key Laboratory of Tropical Crop Products Processing of Ministry of Agriculture and Rural Affairs, Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China
| | - Xixiang Shuai
- Key Laboratory of Tropical Fruit Biology of Ministry of Agriculture and Rural Affairs, South Subtropical Crop Research Institute, China Academy of Tropical Agricultural Sciences, Zhanjiang 524091, China
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Ruihong Liang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Taotao Dai
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| |
Collapse
|