PDF (3.2 MB)
Collect
Submit Manuscript
Research Article | Open Access | Online First

Potential of Dendrobium officinale oligosaccharides to alleviate chronic colitis by modulating inflammation and gut microbiota

Dong-Cheng Shi1,2,Pei-Yi Wang1,2,Lei Xu1,2Hua Zhu1,2Wang-Yue Zhang1,2Qi-Yong Wu1,2Ting-Ting Bu1,2Bao-Ming Tian1,2,3Pei-Long Sun1,2,3Ming Cai1,2,3()
College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
Key Laboratory of Food Macromolecular Resources Processing Technology Research (Zhejiang University of Technology), China National Light Industry, China
Eco-Industrial Innovation Institute ZJUT, Longyou, 324400, China

† These authors contributed equally to this work.

Show Author Information

Highlights

(1) DOO indicates significant alleviation of chronic colitis symptoms in a DSS-induced mouse model.

(2) DOO reduced pro-inflammatory cytokines and downregulated the NF-κB signaling pathway.

(3) DOO altered gut microbiota composition in colitis mice, promoting beneficial bacteria.

(4) DOO increased the secretion of short-chain fatty acids (acetate, propionate, butyrate).

Graphical Abstract

View original image Download original image
This study focused on Dendrobium officinale oligosaccharides (DOO), a novel prebiotic from Dendrobium officinale, to explore its potential application in the treatment of inflammatory bowel disease (IBD). A DSS-induced chronic colitis mouse model was used to explore the effect of DOO on the improvement of UC symptoms and investigated the possible mechanisms. In terms of the underlying indicators, DOO had an ameliorating effect on the symptoms of weight loss, colon shortening, and bloody stool caused by colitis. DOO alleviated colonic inflammation injury and reduced the levels of pro-inflammatory cytokines by inhibiting the NF-κB signaling pathway. Additionally, DOO positively modulated the gut microbiota structure and increased the secretion of SCFAs to maintain intestinal homeostasis in colitis mice. These findings highlight the potential of DOO as a promising prebiotic for UC treatment.

Abstract

Dendrobium officinale oligosaccharides (DOO), as a novel prebiotic, offer promising therapeutic potential for the treatment of inflammatory diseases. In this study, the alleviating effect of DOO on dextran sulfate sodium (DSS)-induced chronic colitis in mice was investigated. DOO treatment relieved the main symptoms of weight loss, colon shortening, and bloody stool caused by colitis. DOO ameliorated histopathological colon tissue damage and reduced levels of pro-inflammatory factors (TNF-α, IL-6, IL-1β). Western blot results suggested that DOO downregulated the phosphorylation levels of key proteins in the NF-κB signaling pathway, including IKKα/β, IκBα and p65. 16S rRNA sequencing suggested that DOO altered the diversity and composition of gut microbiota in DSS-induced mice. It promoted the intestinal colonization of beneficial bacteria such as Dubosiella, Lactobacillus and Alistipes, and inhibited the abnormal overgrowth of Akkermansia. Furthermore, DOO increased the secretion of short-chain fatty acids (SCFAs) such as acetic acid, propionic acid, and butyric acid to maintain intestinal homeostasis. These findings suggest the potential of DOO as a therapeutic agent for ulcerative colitis (UC).

References

[1]

Hu, S., Bourgonje, A. R., Gacesa, R., et al. Mucosal host-microbe interactions associate with clinical phenotypes in inflammatory bowel disease. Nature Communications, 2024, 15: 1470. https://doi.org/10.1038/s41467-024-45855-2

[2]

de Souza, H. S. P., Fiocchi, C. Immunopathogenesis of IBD: current state of the art. Nature Reviews Gastroenterology & Hepatology, 2016, 13: 13–27. https://doi.org/10.1038/nrgastro.2015.186

[3]

de Lange, K. M., Moutsianas, L., Lee, J. C., et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nature Genetics, 2017, 49: 256–261. https://doi.org/10.1038/ng.3760

[4]

Hu, J., Mei, Y., Zhang, H., et al. Ameliorative Effect of an Acidic polysaccharide from Phellinus Linteus on ulcerative colitis in a DSS-induced mouse model. International Journal of Biological Macromolecules, 2024, 265: 130959. https://doi.org/10.1016/j.ijbiomac.2024.130959

[5]

Lee, M., Kim, Y.-S., Lim, S., et al. Protein stabilization of ITF2 by NF-κB prevents colitis-associated cancer development. Nature Communications, 2023, 14: 2363. https://doi.org/10.1038/s41467-023-38080-w

[6]

Franzosa, E. A., Sirota-Madi, A., Avila-Pacheco, J., et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nature Microbiology, 2019, 4: 293–305. https://doi.org/10.1038/s41564-018-0306-4

[7]

Huttenhower, C., Kostic, A. D., Xavier, R. J. Inflammatory bowel disease as a model for translating the microbiome. Immunity, 2014, 40: 843–854. https://doi.org/10.1016/j.immuni.2014.05.013

[8]

Hong, D., Kim, H. K., Yang, W., et al. Integrative analysis of single-cell RNA-seq and gut microbiome metabarcoding data elucidates macrophage dysfunction in mice with DSS-induced ulcerative colitis. Communications Biology, 2024, 7: 1–14. https://doi.org/10.1038/s42003-024-06409-w

[9]

Hu, Y., Chen, Z., Xu, C., et al. Disturbances of the Gut microbiota and microbiota-derived metabolites in inflammatory bowel disease. Nutrients, 2022, 14: 5140. https://doi.org/10.3390/nu14235140

[10]

Noor, N. M., Raine, T. Innovations to improve the efficiency of phase II IBD clinical trials. Nature Reviews Gastroenterology & Hepatology, 2023, 20: 555–556. https://doi.org/10.1038/s41575-023-00805-z

[11]

Chen, Y., Sui, X., Wang, Y., et al. Preparation, structural characterization, biological activity, and nutritional applications of oligosaccharides. Food Chemistry: X, 2024, 22: 101289. https://doi.org/10.1016/j.fochx.2024.101289

[12]

Chen, X., Hu, J., Yang, J., et al. Human milk oligosaccharide 2′-fucosyllactose alleviates DSS-induced ulcerative colitis via improving intestinal barrier function and regulating gut microbiota. Food Bioscience, 2024, 59: 104162. https://doi.org/10.1016/j.fbio.2024.104162

[13]

Liu, N., Wang, H., Yang, Z., et al. The role of functional oligosaccharides as prebiotics in ulcerative colitis. Food & Function, 2022, 13: 6875–6893. https://doi.org/10.1039/D2FO00546H

[14]

Wei, X., Yu, L., Zhang, C., et al. Prebiotic activity of chitooligosaccharides and their ability to alleviate necrotizing enterocolitis in newborn rats. Carbohydrate Polymers, 2023, 299: 120156. https://doi.org/10.1016/j.carbpol.2022.120156

[15]

Cheng, J., Dang, P.-P., Zhao, Z., et al. An assessment of the Chinese medicinal Dendrobium industry: Supply, demand and sustainability. Journal of Ethnopharmacology, 2019, 229: 81–88. https://doi.org/10.1016/j.jep.2018.09.001

[16]
Zeng, X., Tang, S., Dong, X., et al. Analysis of metagenome and metabolome disclosed the mechanisms of Dendrobium officinale polysaccharide on DSS-induced ulcerative colitis-affected mice. International Journal of Biological Macromolecules, 2024 , 134229. https://doi.org/10.1016/j.ijbiomac.2024.134229
[17]

Cheng, H., Xu, L., Zhu, H., et al. Structural characterization of oligosaccharide from Dendrobium officinale and its properties in vitro digestion and fecal fermentation. Food Chemistry, 2024, 460: 140511. https://doi.org/10.1016/j.foodchem.2024.140511

[18]

Cai, M., Zhu, H., Xu, L., et al. Structure, anti-fatigue activity and regulation on gut microflora in vivo of ethanol-fractional polysaccharides from Dendrobium officinale. International Journal of Biological Macromolecules, 2023, 234: 123572. https://doi.org/10.1016/j.ijbiomac.2023.123572

[19]

Wang, P., Cai, M., Yang, K., et al. Phenolics from Dendrobium officinale leaf ameliorate dextran sulfate sodium-induced chronic colitis by regulating gut microbiota and intestinal barrier. Journal of Agricultural and Food Chemistry, 2023, 71: 16630–16646. https://doi.org/10.1021/acs.jafc.3c05339

[20]
Li, J., Tao, W., Zhou, W., et al. Dendrobium officinale polysaccharides alleviated DSS-induced ulcerative colitis in mice through remolding gut microbiota to regulate purine metabolism. Journal of Functional Foods, 2024 , 119: 106336. https://doi.org/10.1016/j.jff.2024.106336
[21]

Xie, Q., Sun, Y., Cao, L., et al. Antifatigue and antihypoxia activities of oligosaccharides and polysaccharides from Codonopsis pilosula in mice. Food & Function, 2020, 11: 6352–6362. https://doi.org/10.1039/D0FO00468E

[22]

Shao, X., Sun, C., Tang, X., et al. Anti-inflammatory and intestinal microbiota modulation properties of Jinxiang garlic ( Allium Sativum L.) polysaccharides toward dextran sodium sulfate-induced colitis. Journal of Agricultural and Food Chemistry, 2020, 68: 12295–12309. https://doi.org/10.1021/acs.jafc.0c04773

[23]

Tian, B., Jiang, Y., Liu, R., et al. Positive effects of extracellular polysaccharides from Paecilomyces hepiali on immune-enhancing properties by regulating gut microbiota in cyclophosphamide-induced mice. International Journal of Biological Macromolecules, 2024, 274: 133390. https://doi.org/10.1016/j.ijbiomac.2024.133390

[24]

Feng, Y., Chen, S., Song, Y., et al. A novel Sagittaria sagittifolia L. polysaccharides mitigate DSS-induced colitis via modulation of gut microbiota and MAPK/NF-κB signaling pathways. International Journal of Biological Macromolecules, 2024, 254: 127835. https://doi.org/10.1016/j.ijbiomac.2023.127835

[25]

Raka, R. N., Xiao, J., Wu, H., et al. Pingyin rose essential oil restores intestinal barrier integrity in DSS-induced mice colitis model. Food Research International, 2023, 164: 112362. https://doi.org/10.1016/j.foodres.2022.112362

[26]

Tian, B., Liu, R., Xu, T., et al. Modulating effects of Hericium erinaceus polysaccharides on the immune response by regulating gut microbiota in cyclophosphamide-treated mice. Journal of the Science of Food and Agriculture, 2023, 103: 3050–3064. https://doi.org/10.1002/jsfa.12404

[27]

Xia, Q., Zhao, Q., Zhu, H., et al. Physicochemical characteristics of Ganoderma lucidum oligosaccharide and its regulatory effect on intestinal flora in vitro fermentation. Food Chemistry: X, 2022, 15: 100421. https://doi.org/10.1016/j.fochx.2022.100421

[28]
Zhang, Y., Liu, Y., Luo, J., et al. Rheum tanguticum polysaccharide alleviates DSS-induced ulcerative colitis and regulates intestinal microbiota in mice. Food Bioscience, 2023 , 53: 102788. https://doi.org/10.1016/j.fbio.2023.102788
[29]

Han, H., Gao, M., Wang, F., et al. Protective effects of patchouli alcohol against DSS-induced ulcerative colitis. Scientific Reports, 2024, 14: 16745. https://doi.org/10.1038/s41598-024-66259-8

[30]

Xie, X. T., Zheng, L. X., Duan, H. M., et al. Structural characteristics of Gracilaria lemaneiformis oligosaccharides and their alleviation of dextran sulphate sodium-induced colitis by modulating the gut microbiota and intestinal metabolites in mice. Food & Function, 2021, 12: 8635–8646. https://doi.org/10.1039/D1FO01201K

[31]

Wei, C., Yao, L., Zhang, Y., et al. Structural characterization of peach gum polysaccharide and its effects on the regulation of DSS-induced acute colitis. International Journal of Biological Macromolecules, 2023, 225: 1224–1234. https://doi.org/10.1016/j.ijbiomac.2022.11.183

[32]

Dong, T., Li, C., Wang, X., et al. Ainsliadimer a selectively inhibits IKKα/β by covalently binding a conserved cysteine. Nature Communications, 2015, 6: 6522. https://doi.org/10.1038/ncomms7522

[33]

Yu, H., Lin, L., Zhang, Z., et al. Targeting NF-κB pathway for the therapy of diseases: mechanism and clinical study. Signal Transduction and Targeted Therapy, 2020, 5: 1–23. https://doi.org/10.1038/s41392-020-00312-6

[34]

Lu, K., Zhou, J., Deng, J., et al. Periplaneta Americana oligosaccharides exert anti-inflammatory activity through immunoregulation and modulation of gut microbiota in acute colitis mice model. Molecules, 2021, 26: 1718. https://doi.org/10.3390/molecules26061718

[35]

Tan, Z., Zhang, Q., Zhao, R., et al. A comparative study on the effects of different sources of carboxymethyl poria polysaccharides on the repair of DSS-induced colitis in mice. International Journal of Molecular Sciences, 2023, 24: 9034. https://doi.org/10.3390/ijms24109034

[36]

Kim, G., Jang, M., Hwang, I., et al. Radish sprout alleviates DSS-induced colitis via regulation of NF-kB signaling pathway and modifying gut microbiota. Biomedicine & Pharmacotherapy, 2021, 144: 112365. https://doi.org/10.1016/j.biopha.2021.112365

[37]

Zheng, J., Ahmad, A. A., Yang, C., et al. Orally administered Lactobacillus rhamnosus CY12 alleviates DSS-induced colitis in mice by restoring the intestinal barrier and inhibiting the TLR4-MyD88-NF-κB pathway via intestinal microbiota modulation. Journal of Agricultural and Food Chemistry, 2024, 72: 9102–9116. https://doi.org/10.1021/acs.jafc.3c07279

[38]

Liu, W., Zhang, Y., Qiu, B., et al. Quinoa whole grain diet compromises the changes of gut microbiota and colonic colitis induced by dextran Sulfate sodium in C57BL/6 mice. Scientific Reports, 2018, 8: 14916. https://doi.org/10.1038/s41598-018-33092-9

[39]

Belzer, C., Chia, L. W., Aalvink, S., et al. Microbial metabolic networks at the mucus layer lead to diet-independent butyrate and vitamin B12 production by intestinal symbionts. mBio, 2017, 8: e00770–17. https://doi.org/10.1128/mbio.00770-17

[40]

Nagalingam, N. A., Kao, J. Y., Young, V. B. Microbial ecology of the murine gut associated with the development of dextran sodium sulfate-induced colitis. Inflammatory Bowel Diseases, 2011, 17: 917–926. https://doi.org/10.1002/ibd.21462

[41]
Qu, S., Zheng, Y., Huang, Y., et al. Excessive consumption of mucin by over-colonized Akkermansia muciniphila promotes intestinal barrier damage during malignant intestinal environment. Frontiers in Microbiology, 2023 , 14. https://doi.org/10.3389/fmicb.2023.1111911
[42]
Wu, H., Rao, Q., Ma, G. C., et al. Effect of triptolide on dextran sodium sulfate-induced ulcerative colitis and gut microbiota in mice. Frontiers in Pharmacology, 2020 , 10. https://doi.org/10.3389/fphar.2019.01652
[43]
Zhang, Y., Tu, S., Ji, X., et al. Dubosiella newyorkensis modulates immune tolerance in colitis via the L-lysine-activated AhR-IDO1-Kyn pathway. Nature Communications, 2024 , 15: 1333. https://doi.org/10.1038/s41467-024-45636-x
[44]

Rosser, E. C., Piper, C. J. M., Matei, D. E., et al. Microbiota-derived metabolites suppress arthritis by amplifying aryl-hydrocarbon receptor activation in regulatory B cells. Cell Metabolism, 2020, 31: 837–851.e10. https://doi.org/10.1016/j.cmet.2020.03.003

[45]

Lucas, S., Omata, Y., Hofmann, J., et al. Short-chain fatty acids regulate systemic bone mass and protect from pathological bone loss. Nature Communications, 2018, 9: 55. https://doi.org/10.1038/s41467-017-02490-4

[46]

Fernando, M. R., Saxena, A., Reyes, J. L., et al. Butyrate enhances antibacterial effects while suppressing other features of alternative activation in IL-4-induced macrophages. American Journal of Physiology-Gastrointestinal and Liver Physiology, 2016, 310: G822–G831. https://doi.org/10.1152/ajpgi.00440.2015

[47]
Mohebali, N., Weigel, M., Hain, T., et al. Faecalibacterium prausnitzii, Bacteroides faecis and Roseburia intestinalis attenuate clinical symptoms of experimental colitis by regulating Treg/Th17 cell balance and intestinal barrier integrity. Biomedicine & Pharmacotherapy, 2023 , 167: 115568. https://doi.org/10.1016/j.biopha.2023.115568
[48]

Holmberg, S. M., Feeney, R. H., Prasoodanan P. K., V., et al. The gut commensal blautia maintains colonic mucus function under low-fiber consumption through secretion of short-chain fatty acids. Nature Communications, 2024, 15: 3502. https://doi.org/10.1038/s41467-024-47594-w

[49]

Harris, H. C., Morrison, D. J., Edwards, C. A. Impact of the source of fermentable carbohydrate on SCFA production by human gut microbiota in vitro - a systematic scoping review and secondary analysis. Critical Reviews in Food Science and Nutrition, 2021, 61: 3892–3903. https://doi.org/10.1080/10408398.2020.1809991

Food & Medicine Homology
Cite this article:
Shi D-C, Wang P-Y, Xu L, et al. Potential of Dendrobium officinale oligosaccharides to alleviate chronic colitis by modulating inflammation and gut microbiota. Food & Medicine Homology, 2024, https://doi.org/10.26599/FMH.2025.9420077
Metrics & Citations  
Article History
Copyright
Rights and Permissions
Return