Journal Home > Volume 13 , Issue 2

Hyperuricemia is a metabolic disorder caused by abnormal purine metabolism, resulting in abnormally high serum uric acid. In this study, a novel Levilactobacillus brevis PDD-5 isolated from salty vegetables was verified with the function of alleviating hyperuricemia. The relevant effects of L. brevis PDD-5 in lowering uric acid were analyzed by in vitro and in vivo experiments. The results showed that the L. brevis PDD-5 has (68.86 ± 15.46)% of inosine uptake capacity and (95.75 ± 3.30)% of guanosine uptake capacity in vitro. Oral administration of L. brevis PDD-5 to hyperuricemia rats reduced uric acid, creatinine, and urea nitrogen in serum, as well as decreased inosine and guanosine levels in the intestinal contents of rats. Analysis of relevant markers in the kidney by ELISA kits revealed that L. brevis PDD-5 alleviated oxidative stress and inflammation. Moreover, the gene expression of uric acid transporter 1 (URAT1) and glucose transporter 9 (GLUT9) was down-regulated, and the gene expression of organic anion transporter 1 (OAT1) was up-regulated after treatment with L. brevis PDD-5. Western blot analysis showed that L. brevis PDD-5 alleviated hyperuricemia-induced kidney injury through the NLRP3 pathway. These findings suggest that L. brevis PDD-5 can lower uric acid, repair kidney damage, and also has the potential to prevent uric acid nephropathy.


menu
Abstract
Full text
Outline
Electronic supplementary material
About this article

A novel strain of Levilactobacillus brevis PDD-5 isolated from salty vegetables has beneficial effects on hyperuricemia through anti-inflammation and improvement of kidney damage

Show Author's information Jue Xua,bMaolin Tua,bXiankang Fana,bYuxing GuocTao ZhangcXiaoqun Zenga,bZhendong Caia,bZhen Wua,bDaodong Pana,b,( )
State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China
Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China
Department of Food Science and Technology, School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China

Peer review under responsibility of Tsinghua University Press.

Abstract

Hyperuricemia is a metabolic disorder caused by abnormal purine metabolism, resulting in abnormally high serum uric acid. In this study, a novel Levilactobacillus brevis PDD-5 isolated from salty vegetables was verified with the function of alleviating hyperuricemia. The relevant effects of L. brevis PDD-5 in lowering uric acid were analyzed by in vitro and in vivo experiments. The results showed that the L. brevis PDD-5 has (68.86 ± 15.46)% of inosine uptake capacity and (95.75 ± 3.30)% of guanosine uptake capacity in vitro. Oral administration of L. brevis PDD-5 to hyperuricemia rats reduced uric acid, creatinine, and urea nitrogen in serum, as well as decreased inosine and guanosine levels in the intestinal contents of rats. Analysis of relevant markers in the kidney by ELISA kits revealed that L. brevis PDD-5 alleviated oxidative stress and inflammation. Moreover, the gene expression of uric acid transporter 1 (URAT1) and glucose transporter 9 (GLUT9) was down-regulated, and the gene expression of organic anion transporter 1 (OAT1) was up-regulated after treatment with L. brevis PDD-5. Western blot analysis showed that L. brevis PDD-5 alleviated hyperuricemia-induced kidney injury through the NLRP3 pathway. These findings suggest that L. brevis PDD-5 can lower uric acid, repair kidney damage, and also has the potential to prevent uric acid nephropathy.

Keywords: Lactic acid bacteria, Hyperuricemia, Purine nucleoside, Uric acid nephropathy

References(49)

[1]

A. Dehghan, A. Köttgen, Q. Yang, et al., Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study, Lancet 372 (2008) 1953-1961. https://doi.org/10.1016/s0140-6736(08)61343-4.

[2]

R.P. Obermayr, C. Temml, G. Gutjahr, et al., Elevated uric acid increases the risk for kidney disease, J. Am. Soc. Nephrol. 19 (2008) 2407-2413. https://doi.org/10.1681/asn.2008010080.

[3]

A. Dehghan, A. Köttgen, Q. Yang, et al., Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study, Lancet 372 (2008) 1953-1961. https://doi.org/10.1016/S0140-6736(08)61343-4.

[4]

Q. Luo, X. Xia, B. Li, et al. Serum uric acid and cardiovascular mortality in chronic kidney disease: a meta-analysis, BMC Nephrol. 20 (2019) 18. https://doi.org/10.1186/s12882-018-1143-7.

[5]

H.K. Choi, K. Atkinson, E.W. Karlson, et al., Purine-rich foods, dairy and protein intake, and the risk of gout in men, New Engl. J. Med. 350 (2004) 1093-1103. https://doi.org/10.1056/NEJMoa035700.

[6]

M.H. Pillinger, B.F. Mandell, Therapeutic approaches in the treatment of gout, Semin. Arthritis Rheum. 50 (2020) S24-S30. https://doi.org/10.1016/j.semarthrit.2020.04.010.

[7]

H. Shuwen, D. Kefeng, Intestinal phages interact with bacteria and are involved in human diseases, Gut Microbes. 14 (2022) 2113717. https://doi.org/10.1080/19490976.2022.2113717.

[8]

L. Gan, W.H. Xu, Y. Xiong, et al., Probiotics: their action against pathogens can be turned around, Sci. Rep. 11 (2021) 13247. https://doi.org/10.1038/s41598-021-91542-3.

[9]

Y. Xiao, C. Zhang, X. Zeng, et al., Microecological treatment of hyperuricemia using Lactobacillus from pickles, BMC Microbiol. 20 (2020) 195. https://doi.org/10.1186/s12866-020-01874-9.

[10]

N. Yamada, C. Saito-Iwamoto, M. Nakamura, et al., Lactobacillus gasseri PA-3 uses the purines IMP, inosine and hypoxanthine and reduces their absorption in rats, Microorganisms 5 (2017) 10. https://doi.org/10.3390/microorganisms5010010.

[11]

H. Wang, L. Mei, Y. Deng, et al., Lactobacillus brevis DM9218 ameliorates fructose-induced hyperuricemia through inosine degradation and manipulation of intestinal dysbiosis, Nutrition 62 (2019) 63-73. https://doi.org/10.1016/j.nut.2018.11.018.

[12]

A.T. Vieira, I. Galvao, F.A. Amaral, et al., Oral treatment with Bifidobacterium longum 51A reduced inflammation in a murine experimental model of gout, Benef. Microbes. 6 (2015) 799-806. https://doi.org/10.3920/BM2015.0015.

[13]

A. de Moreno de LeBlanc, C. Matar, E. Farnworth, et al., Study of cytokines involved in the prevention of a murine experimental breast cancer by Kefir, Cytokine 34 (2006) 1-8. https://doi.org/10.1016/j.cyto.2006.03.008.

[14]

G. Wang, Q. Si, S. Yang, et al., Lactic acid bacteria reduce diabetes symptoms in mice by alleviating gut microbiota dysbiosis and inflammation in different manners, Food Funct. 11 (2020) 5898-5914. https://doi.org/10.1039/C9FO02761K.

[15]

P.L. Conway, S.L. Gorbach, B.R. Goldin, Survival of lactic acid bacteria in the human stomach and adhesion to intestinal cells, J. Dairy Sci. 70 (1987) 1-12. https://doi.org/10.3168/jds.S0022-0302(87)79974-3.

[16]

J. He, W. Wang, Z. Wu, et al., Effect of Lactobacillus reuteri on intestinal microbiota and immune parameters: involvement of sex differences, J. Funct. Foods 53 (2019) 36-43. https://doi.org/10.1016/j.jff.2018.12.010.

[17]

U. Schillinger, F.K. Lücke, Antibacterial activity of Lactobacillus sake isolated from meat, Appl. Environ. Microb. 55 (1989) 1901-1906. https://doi.org/10.1128/aem.55.8.1901-1906.1989.

[18]

M. Rosenberg, D. Gutnick, E. Rosenberg, Adherence of bacteria to hydrocarbons: a simple method for measuring cell-surface hydrophobicity, FEMS Microbiol. Lett. 9 (1980) 29-33. https://doi.org/10.1111/j.1574-6968.1980.tb05599.x.

[19]

H.V. Herck, V. Baumans, C. Brandt, et al., Orbital sinus blood sampling in rats as performed by different animal technicians: the influence of technique and expertise, Lab. Anim.-UK 32 (1998) 377-386. https://doi.org/10.1177/0023677217741332.

[20]

R. Chavez, D.J. Fraser, T. Bowen, et al., Kidney ischaemia reperfusion injury in the rat: the EGTI scoring system as a valid and reliable tool for histological assessment, J. Histology Histopathology 3 (2016) 1. https://doi.org/10.7243/2055-091X-3-1.

[21]

R. van der Westen, J. Sjollema, R. Molenaar, et al., Floating and tether-coupled adhesion of bacteria to hydrophobic and hydrophilic surfaces, Langmuir 34 (2018) 4937-4944. https://doi.org/10.1021/acs.langmuir.7b04331.

[22]

P.F. Pérez, Y. Minnaard, E.A. Disalvo, et al., Surface properties of bifidobacterial strains of human origin, Appl. Environ. Microb. 64 (1998) 21-26. https://doi.org/10.1002/abio.370180412.

[23]

D. Vedder, W. Walrabenstein, M. Heslinga, et al., Dietary interventions for gout and effect on cardiovascular risk factors: a systematic review, Nutrients 11 (2019) 2955. https://doi.org/10.3390/nu11122955.

[24]

L.M. Salati, C.J. Gross, L.M. Henderson, et al., Absorption and metabolism of adenine, adenosine-5’-monophosphate, adenosine and hypoxanthine by the isolated vascularly perfused rat small intestine, J. Nutr. 114 (1984) 753-760. https://doi.org/10.1093/jn/114.4.753.

[25]

E.B.M. Daliri, B.H. Lee, New perspectives on probiotics in health and disease, Food Sci. Hum. Well. 4 (2015) 56-65. https://doi.org/10.1016/j.fshw.2015.06.002.

[26]

J. Wang, Y. Chen, H. Zhong, et al., The gut microbiota as a target to control hyperuricemia pathogenesis: potential mechanisms and therapeutic strategies, Crit. Rev. Food Sci. 62 (2022) 3979-3989. https://doi.org/10.1080/10408398.2021.1874287.

[27]

C. Huang, M. Zheng, Y. Huang, et al., The effect of purine content on sensory quality of pork, Meat Sci. 172 (2021) 108346. https://doi.org/10.1016/j.meatsci.2020.108346.

[28]

Y. Lee, P. Werlinger, J.W. Suh, et al., Potential probiotic Lacticaseibacillus paracasei MJM60396 prevents hyperuricemia in a multiple way by absorbing purine, suppressing xanthine oxidase and regulating urate excretion in mice, Microorganisms 10 (2022) 851. https://doi.org/10.3390/microorganisms10050851.

[29]

T.H. Nguyen, Y. Kim, J.S. Kim, et al., Evaluating the cryoprotective encapsulation of the lactic acid bacteria in simulated gastrointestinal conditions, Biotechnol. Bioprocess E. 25 (2020) 287-292. https://doi.org/10.1007/s12257-019-0406-x.

[30]

J.H. Jung, S.J. Kim, J.Y. Lee, et al., Multifunctional properties of Lactobacillus plantarum strains WiKim83 and WiKim87 as a starter culture for fermented food, Food Sci. Nutr. 7 (2019) 2505-2516. https://doi.org/10.1002/fsn3.1075.

[31]

V. Santarmaki, Y. Kourkoutas, G. Zoumpopoulou, et al., Survival, intestinal Mucosa Adhesion, and immunomodulatory potential of Lactobacillus plantarum strains, Curr. Microbiol. 74 (2017) 1061-1067. https://doi.org/10.1007/s00284-017-1285-z.

[32]

K. McDowell, P. Welsh, K.F. Docherty, et al., Dapagliflozin reduces uric acid concentration, an independent predictor of adverse outcomes in DAPA-HF, Eur. J. Heart Fail 24 (2022) 1066-1076. https://doi.org/10.1002/ejhf.2433.

[33]

J. Cao, Q. Liu, H. Hao, et al., Lactobacillus paracasei X11 ameliorates hyperuricemia and modulates gut microbiota in mice, Front. Immunol. 13 (2022) 940228. https://doi.org/10.3389/fimmu.2022.940228.

[34]

E. Altermann, W.M. Russell, M.A. Azcarate Peril, et al., Complete genome sequence of the probiotic lactic acid bacterium Lactobacillus acidophilus NCFM, Proc. Natl. Acad. Sci. U.S.A. 102 (2005) 3906-3912. https://doi.org/10.1073/pnas.0409188102.

[35]
Lesinurad/Allopurinol (duzallo) for gout-associated hyperuricemia, JAMA 319 (2018) 188-189. https://doi.org/10.1001/jama.2017.20189.
DOI
[36]

C.T. Lee, L.C. Chang, C.W. Liu, et al., Negative correlation between serum uric acid and kidney URAT1 mRNA expression caused by resveratrol in rats, Mol. Nutr. Food Res. 61 (2017) 1601030. https://doi.org/10.1002/mnfr.201601030.

[37]

G. Chen, M.L. Tan, K.K. Li, et al., Green tea polyphenols decreases uric acid level through xanthine oxidase and renal urate transporters in hyperuricemic mice, J. Ethnopharmacol. 175 (2015) 14-20. https://doi.org/10.1016/j.jep.2015.08.043.

[38]

C. Grant, U. Oh, K. Yao, et al., Dysregulation of TGF-beta signaling and regulatory and effector T-cell function in virus-induced neuroinflammatory disease, Blood 111 (2008) 5601-5609. https://doi.org/10.1182/blood-2007-11-123430.

[39]

G. Wang, H. Tang, Y. Zhang, et al., The intervention effects of Lactobacillus casei LC2W on Escherichia coli O157:H7-induced mouse colitis, Food Sci. Hum. Well. 9 (2020) 289-294. https://doi.org/10.1016/j.fshw.2020.04.008.

[40]

Y. Wu, Z. Ye, P. Feng, et al., Limosilactobacillus fermentum JL-3 isolated from “Jiangshui” ameliorates hyperuricemia by degrading uric acid, Gut Microbes. 13 (2021) 1897211. https://doi.org/10.1080/19490976.2021.1897211.

[41]

G. Agirman, K.B. Yu, E.Y. Hsiao, Signaling inflammation across the gutbrain axis, Science 374 (2021) 1087-1092. https://doi.org/10.1126/science.abi6087.

[42]

I. Calabuig, M. Gómez-Garberí, M. Andrés, Gout is prevalent but underregistered among patients with cardiovascular events: a field study, Front. Med. 7 (2020) 560. https://doi.org/10.3389/fmed.2020.00560.

[43]

Y. Zhou, M. Zhao, Z. Pu, et al., Relationship between oxidative stress and inflammation in hyperuricemia: analysis based on asymptomatic young patients with primary hyperuricemia, Medicine 97 (2018) e13108. https://doi.org/10.1097/MD.0000000000013108.

[44]

J. Huo, Y. Ming, H. Li, et al., The protective effects of peptides from Chinese baijiu on AAPH-induced oxidative stress in HepG2 cells via Nrf2-mediated signaling pathway, Food Sci. Hum. Well. 11 (2022) 1527-1538. https://doi.org/10.1016/j.fshw.2022.06.010.

[45]

T. Hu, R. Chen, Y. Qian, et al., Antioxidant effect of Lactobacillus fermentum HFY02-fermented soy milk on D-galactose-induced aging mouse model, Food Sci. Hum. Well. 11 (2022) 1362-1372. https://doi.org/10.1016/j.fshw.2022.04.036.

[46]

T. Nutmakul, A review on benefits of quercetin in hyperuricemia and gouty arthritis, Saudi. Pharm. J. 30 (2022) 918-926. https://doi.org/10.1016/j.jsps.2022.04.013.

[47]

Z.R. Sun, H.R. Liu, D. Hu, et al., Ellagic acid exerts beneficial effects on hyperuricemia by inhibiting xanthine oxidase and NLRP3 inflammasome activation, J. Agr. Food Chem. 69 (2021) 12741-12752. https://doi.org/10.1021/acs.jafc.1c05239.

[48]

M. Kanbay, A. Ozkara, Y. Selcoki, et al., Effect of treatment of hyperuricemia with allopurinol on blood pressure, creatinine clearence, and proteinuria in patients with normal renal functions, Int. Urol. Nephrol. 39 (2007) 1227-1233. https://doi.org/10.1007/s11255-007-9253-3.

[49]

R.J. Fontana, Y.J. Li, E. Phillips, et al., Allopurinol hepatotoxicity is associated with human leukocyte antigen Class I alleles, Liver Int. 41 (2021) 1884-1893. https://doi.org/10.1111/liv.14903.

File
fshw-13-2-898_ESM.docx (109 KB)
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 11 July 2022
Revised: 27 September 2022
Accepted: 26 October 2022
Published: 25 September 2023
Issue date: March 2024

Copyright

© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

Acknowledgements

Acknowledgements

The authors thank the National Natural Science Foundation of China (31972048; 32272339) and the National Key R & D Program of China (2021YFD2100104) for financial support.

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return