Hostname: page-component-5c6d5d7d68-wtssw Total loading time: 0 Render date: 2024-08-14T08:24:35.715Z Has data issue: false hasContentIssue false

The ameliorative and neuroprotective effects of dietary fibre on hyperuricaemia mice: a perspective from microbiome and metabolome

Published online by Cambridge University Press:  03 June 2024

Yu Wang
Affiliation:
Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, People’s Republic of China University of Chinese Academy of Sciences, Beijing, People’s Republic of China
Fengping Miao
Affiliation:
Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, People’s Republic of China
Jun Wang
Affiliation:
Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, People’s Republic of China University of Chinese Academy of Sciences, Beijing, People’s Republic of China
Maokun Zheng
Affiliation:
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
Feng Yu
Affiliation:
Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, People’s Republic of China
Yuetao Yi*
Affiliation:
Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, People’s Republic of China Shandong Saline-Alkali Land Modern Agriculture Company, Dongying, People’s Republic of China
*
*Corresponding author: Yuetao Yi, email ytyi@yic.ac.cn

Abstract

The effect of single dietary fibre (DF) on lowering uric acid (UA) level has been reported in the literature. However, the potential protective mechanism of DF against potassium oxybate-induced hyperuricaemia (HUA), as modelled by prophylactic administration, remains unclear. The data demonstrate that DF significantly decreased serum and cerebral tissue UA concentrations, inhibited xanthine oxidase expression and activity in the liver and reduced levels of creatinine and urea nitrogen in the serum. Additionally, it mitigated the deposition of amyloid-β in cerebral tissue. Correlation analysis showed that DF modulated the Toll-like receptor 4/NF-κB signalling pathway, attenuating oxidative stress and inflammatory responses in HUA mice. Additionally, DF helps to maintain the composition of the gut microbiota, reducing harmful Desulfovibrio and enriching beneficial Akkermansia and Ruminococcus populations. The results of the faecal metabolomics analysis indicate that DF facilitates the regulation of metabolic pathways involved in oxidative stress and inflammation. These pathways include pyrimidine metabolism, tryptophan metabolism, nucleotide metabolism and vitamin B6 metabolism. Additionally, the study found that DF has a preventive effect on anxiety-like behaviour induced by HUA. In summary, DF shows promise in mitigating HUA and cognitive deficits, primarily by modulating gut microbiota and metabolites.

Type
Research Article
Copyright
© The Author(s), 2024. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Rai, SK, Fung, TT, Lu, N, et al. (2017) The Dietary Approaches to Stop Hypertension (DASH) diet, Western diet, and risk of gout in men: prospective cohort study. BMJ 357, j1794.Google Scholar
Kyu, HH, Abate, D, Abate, KH, et al. (2018) Global, regional, and national disability-adjusted life-years (DALYs) for 359 diseases and injuries and healthy life expectancy (HALE) for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392, 18591922.Google Scholar
Rakoff-Nahoum, S, Foster, KR & Comstock, LE (2016) The evolution of cooperation within the gut microbiota. Nature 533, 255259.Google Scholar
Lv, Q, Xu, D, Zhang, X, et al. (2020) Association of hyperuricemia with immune disorders and intestinal barrier dysfunction. Front Physiol 11, 524236.Google Scholar
Yin, H, Liu, N & Chen, J (2022) The role of the intestine in the development of hyperuricemia. Front Immunol 13, 845684.Google Scholar
Zhao, F, Zhao, Z, Man, D, et al. (2023) Changes in gut microbiota structure and function in gout patients. Food Biosci 54, 102912.Google Scholar
Tang, DH, Ye, YS, Wang, CY, et al. (2017) Potassium oxonate induces acute hyperuricemia in the tree shrew (tupaia belangeri chinensis). Exp Anim 66, 209216.Google Scholar
Martínez-Nava, GA, Méndez-Salazar, EO, Vázquez-Mellado, J, et al. (2023) The impact of short-chain fatty acid–producing bacteria of the gut microbiota in hyperuricemia and gout diagnosis. Clin Rheumatol 42, 203214.Google Scholar
Dang, K, Zhang, N, Gao, H, et al. (2023) Influence of intestinal microecology in the development of gout or hyperuricemia and the potential therapeutic targets. Int J Rheum Dis 26, 19111922.Google Scholar
Ju, YH, Bhalla, M, Hyeon, SJ, et al. (2022) Astrocytic urea cycle detoxifies Aβ-derived ammonia while impairing memory in Alzheimer’s disease. Cell Metab 34, 11041120.e1108.Google Scholar
Khaled, Y, Abdelhamid, AA, Al-Mazroey, H, et al. (2023) Higher serum uric acid is associated with poorer cognitive performance in healthy middle-aged people: a cross-sectional study. Intern Emerg Med 18, 17011709.Google Scholar
Li, L, Yan, S, Liu, S, et al. (2023) In-depth insight into correlations between gut microbiota and dietary fiber elucidates a dietary causal relationship with host health. Food Res Int 172, 113133.Google Scholar
Zarrinpar, A, Chaix, A, Yooseph, S, et al. (2014) Diet and feeding pattern affect the diurnal dynamics of the gut microbiome. Cell Metab 20, 10061017.Google Scholar
Fu, J, Zheng, Y, Gao, Y, et al. (2022) Dietary fiber intake and gut microbiota in human health. Microorganisms 10, 2507.Google Scholar
Tawfick, MM, Xie, H, Zhao, C, et al. (2022) Inulin fructans in diet: role in gut homeostasis, immunity, health outcomes and potential therapeutics. Int J Biol Macromol 208, 948961.Google Scholar
Zhu, Q, Yu, L, Li, Y, et al. (2022) Association between dietary fiber intake and hyperuricemia among Chinese adults: analysis of the China adult chronic disease and nutrition surveillance (2015). Nutrients 14, 1433.Google Scholar
Sun, Y, Sun, J, Zhang, P, et al. (2019) Association of dietary fiber intake with hyperuricemia in U.S. adults. Food Funct 10, 49324940.Google Scholar
Guo, Y, Yu, Y, Li, H, et al. (2021) Inulin supplementation ameliorates hyperuricemia and modulates gut microbiota in Uox-knockout mice. Eur J Nutr 60, 22172230. 2020/10/27 ed.Google Scholar
Khazaie, S, Jafari, M, Heydari, J, et al. (2019) Modulatory effects of vitamin C on biochemical and oxidative changes induced by acute exposure to diazinon in rat various tissues: prophylactic and therapeutic roles. J Anim Physiol Anim Nutr (Berl) 103, 16191628.Google Scholar
Takashi, K (2021) Modification of dietary habits for prevention of gout in Japanese people: gout and macronutrient Intake. Am J Health Res 9, 128142.Google Scholar
Handley, RR, Reid, SJ, Brauning, R, et al. (2017) Brain urea increase is an early Huntington’s disease pathogenic event observed in a prodromal transgenic sheep model and HD cases. Proc Natl Acad Sci USA 114, E11293E11302.Google Scholar
Mijailovic, NR, Vesic, K & Borovcanin, MM (2022) The influence of serum uric acid on the brain and cognitive dysfunction. Front Psychiatry 13, 828476.Google Scholar
Liu, X, Lv, Q, Ren, H, et al. (2020) The altered gut microbiota of high-purine-induced hyperuricemia rats and its correlation with hyperuricemia. PeerJ 8, e8664.Google Scholar
Sun, Y, Xu, D, Zhang, G, et al. (2024) Wild-type Escherichia coli Nissle 1917 improves hyperuricemia by anaerobically degrading uric acid and maintaining gut microbiota profile of mice. J Funct Foods 112, 105935.Google Scholar
Wang, H, Shen, Q, Fu, Y, et al. (2023) Effects on diabetic mice of consuming lipid extracted from foxtail millet (Setaria italica): gut microbiota analysis and serum metabolomics. J Agric Food Chem 71, 1007510086.Google Scholar
Yuan, L, Zhao, J, Liu, Y, et al. (2024) Multiomics analysis revealed the mechanism of the anti-diabetic effect of Salecan. Carbohydr Polym 327, 121694.Google Scholar
Wu, Y, Wang, Y, Hu, A, et al. (2022) Lactobacillus plantarum-derived postbiotics prevent Salmonella-induced neurological dysfunctions by modulating gut-brain axis in mice. Front Nutr 9, 946096.Google Scholar
Yan, S, Chen, J, Zhu, L, et al. (2022) Oryzanol attenuates high fat and cholesterol diet-induced hyperlipidemia by regulating the gut microbiome and amino acid metabolism. J Agric Food Chem 70, 64296443.Google Scholar
Kaakoush, NO (2015) Insights into the role of erysipelotrichaceae in the human host. Front Cell Infect Microbiol 5, 84.Google Scholar
Downes, J, Dewhirst, FE, Tanner, ACR, et al. (2013) Description of Alloprevotella rava gen. nov., sp. nov., isolated from the human oral cavity, and reclassification of Prevotella tannerae Moore et al. 1994 as Alloprevotella tannerae gen. nov., comb. nov. Int J Syst Evol Microbiol 63, 12141218.Google Scholar
Meng, Y, Hu, Y, Wei, M, et al. (2023) Amelioration of hyperuricemia by Lactobacillus acidophilus F02 with uric acid -lowering ability via modulation of NLRP3 inflammasome and gut microbiota homeostasis. J Funct Foods 111, 105903.Google Scholar
Wang, R, Hu, B, Ye, C, et al. (2022) Stewed rhubarb decoction ameliorates adenine-induced chronic renal failure in mice by regulating gut microbiota dysbiosis. Front Pharmacol 13, 842720.Google Scholar
Pan, L, Han, P, Ma, S, et al. (2020) Abnormal metabolism of gut microbiota reveals the possible molecular mechanism of nephropathy induced by hyperuricemia. Acta Pharm Sin B 10, 249261.Google Scholar
Zhang, Y, Chen, S, Yuan, M, et al. (2022) Gout and diet: a comprehensive review of mechanisms and management. Nutrients 14, 3525.Google Scholar
Srivastava, A, Kaze, AD, McMullan, CJ, et al. (2018) Uric acid and the risks of kidney failure and death in individuals with CKD. Am J Kidney Dis 71, 362370.Google Scholar
Chilappa, CS, Aronow, WS, Shapiro, D, et al. (2010) Gout and hyperuricemia. Compr Ther 36, 313.Google Scholar
Leblanc, M, Garred, LJ, Cardinal, J, et al. (1998) Catabolism in critical illness: estimation from urea nitrogen appearance and creatinine production during continuous renal replacement therapy. Am J Kidney Dis 32, 444453.Google Scholar
Chaudhary, P, Janmeda, P, Docea, AO, et al. (2023) Oxidative stress, free radicals and antioxidants: potential crosstalk in the pathophysiology of human diseases. Front Chem 11, 1158198.Google Scholar
Li, Q, Lin, L, Zhang, C, et al. (2024) The progression of inorganic nanoparticles and natural products for inflammatory bowel disease. J Nanobiotechnol 22, 17.Google Scholar
Tomasello, G, Mazzola, M, Leone, A, et al. (2016) Nutrition, oxidative stress and intestinal dysbiosis: influence of diet on gut microbiota in inflammatory bowel diseases. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 160, 461466.Google Scholar
Bian, M, Wang, J, Wang, Y, et al. (2020) Chicory ameliorates hyperuricemia via modulating gut microbiota and alleviating LPS/TLR4 axis in quail. Biomed Pharmacother 131, 110719.Google Scholar
Li, X, Chen, Y, Gao, X, et al. (2021) Antihyperuricemic effect of green alga Ulva lactuca ulvan through regulating urate transporters. J Agric Food Chem 69, 1122511235.Google Scholar
Oshima, S, Shiiya, S & Nakamura, Y (2019) Serum uric acid -lowering effects of combined glycine and tryptophan treatments in subjects with mild hyperuricemia: a randomized, double-blind, placebo-controlled, crossover study. Nutrients 11, 564.Google Scholar
Zhang, K, Qin, X, Qiu, J, et al. (2023) Desulfovibrio desulfuricans aggravates atherosclerosis by enhancing intestinal permeability and endothelial TLR4/NF-κB pathway in Apoe (-/-) mice. Genes Dis 10, 239253.Google Scholar
Fang, J, Wang, H, Zhou, Y, et al. (2021) Slimy partners: the mucus barrier and gut microbiome in ulcerative colitis. Exp Mol Med 53, 772787.Google Scholar
Murros, KE, Huynh, VA, Takala, TM, et al. (2021) Desulfovibrio bacteria are associated with Parkinson’s disease. Front Cell Infect Microbiol 11, 652617.Google Scholar
Huynh, VA, Takala, TM, Murros, KE, et al. (2023) Desulfovibrio bacteria enhance α-synuclein aggregation in a Caenorhabditis elegans model of Parkinson’s disease. Front Cell Infect Microbiol 13, 1181315.Google Scholar
Li, LL, Ma, YH, Bi, YL, et al. (2021) Serum uric acid may aggravate Alzheimer’s disease risk by affecting amyloidosis in cognitively intact older adults: the CABLE study. J Alzheimers Dis 81, 389401.Google Scholar
Xu, Y, Yang, Y, Li, B, et al. (2022) Dietary methionine restriction improves gut microbiota composition and prevents cognitive impairment in D-galactose-induced aging mice. Food Funct 13, 1289612914.Google Scholar
Sun, Y, Koyama, Y & Shimada, S (2022) Inflammation from peripheral organs to the brain: how does systemic inflammation cause neuroinflammation? Front Aging Neurosci 14, 903455.Google Scholar
Nettleton, JE, Klancic, T, Schick, A, et al. (2021) Prebiotic, probiotic, and synbiotic consumption alter behavioral variables and intestinal permeability and microbiota in BTBR mice. Microorganisms 9, 1833.Google Scholar
Hsiao, EY, McBride, SW, Hsien, S, et al. (2013) Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 155, 14511463.Google Scholar
Inoue, R, Sakaue, Y, Kawada, Y, et al. (2019) Dietary supplementation with partially hydrolyzed guar gum helps improve constipation and gut dysbiosis symptoms and behavioral irritability in children with autism spectrum disorder. J Clin Biochem Nutr 64, 217223.Google Scholar
Valdivielso, JM, Eritja, À, Caus, M, et al. (2020) Glutamate-gated NMDA receptors: insights into the function and signaling in the kidney. Biomolecules 10, 1051.Google Scholar
Wang, R, Halimulati, M, Huang, X, et al. (2023) Sulforaphane-driven reprogramming of gut microbiome and metabolome ameliorates the progression of hyperuricemia. J Adv Res 52, 1928.Google Scholar
Olajide, OJ & Chapman, CA (2021) Amyloid-β (1–42) peptide induces rapid NMDA receptor-dependent alterations at glutamatergic synapses in the entorhinal cortex. Neurobiol Aging 105, 296309.Google Scholar
Wu, GM, Du, CP & Xu, Y (2023) Oligomeric Aβ25–35 induces the tyrosine phosphorylation of PSD-95 by SrcPTKs in rat hippocampal CA1 subfield. Int J Neurosci 133, 888895.Google Scholar
Oshima, S, Shiiya, S & Nakamura, Y (2019) Combined supplementation with glycine and tryptophan reduces purine-induced serum uric acid elevation by accelerating urinary uric acid excretion: a randomized, single-blind, placebo-controlled, crossover study. Nutrients 11, 2562.Google Scholar
Han, EH, Lim, MK, Lee, SH, et al. (2018) Synergic effect in the reduction of serum uric acid level between ethanol extract of Aster glehni and vitamin B(6). Food Sci Biotechnol 27, 14391444.Google Scholar
Zhang, Y & Qiu, H (2018) Folate, vitamin B6 and vitamin B12 intake in relation to hyperuricemia. J Clin Med 7, 210.Google Scholar
Wang, L, Li, X, Montazeri, A, et al. (2023) Phenome-wide association study of genetically predicted B vitamins and homocysteine biomarkers with multiple health and disease outcomes: analysis of the UK Biobank. Am J Clin Nutr 117, 564575.Google Scholar
Mesripour, A, Alhimma, F & Hajhashemi, V (2019) The effect of vitamin B6 on dexamethasone-induced depression in mice model of despair. Nutr Neurosci 22, 744749.Google Scholar
Theofylaktopoulou, D, Ulvik, A, Midttun, Ø, et al. (2014) Vitamins B2 and B6 as determinants of kynurenines and related markers of interferon-γ-mediated immune activation in the community-based Hordaland Health Study. Br J Nutr 112, 10651072.Google Scholar
Wang, J, Chen, Y, Zhong, H, et al. (2022) The gut microbiota as a target to control hyperuricemia pathogenesis: potential mechanisms and therapeutic strategies. Crit Rev Food Sci Nutr 62, 39793989.Google Scholar
Supplementary material: File

Wang et al. supplementary material

Wang et al. supplementary material
Download Wang et al. supplementary material(File)
File 2.7 MB