AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.4 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Study on the interaction between β-carotene and gut microflora using an in vitro fermentation model

Zhixian Lia,bZhuqing Daia,b( )Enjuan Shia,bPeng WancGuijie ChencZhongyuan ZhangbYayuan XubRuichang GaoaXiaoxiong ZengcDajing Lia,b( )
School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Show Author Information

Abstract

β-Carotene, a typical non-oxygenated carotenoid, is the most efficient source of retinol (VA). The low bio-availability of β-carotene lead to large accumulation in colon; however, the relationship between β-carotene and gut microflora remains unclear. This study intends to explore the interaction between β-carotene and gut microflora using an in vitro fermentation model. After 24 h fermentation, the degradation rate of β-carotene was (64.28 ± 6.23)%, which was 1.46 times that of the group without gut microflora. Meanwhile, the production of VA was nearly 2 times that of the group without gut microflora, indicating that the gut microflora can metabolize β-carotene into VA. β-Carotene also influences the production of short-chain fatty acids (SCFAs), the production of total SCFAs in 0.5 mg/mL β-carotene (BCM) group was (44.00 ± 1.16) mmol/L, which was 2.26 times that of the blank control (BLK) group. Among them, the production of acetic acid in BCM group was (19.06 ± 0.82) mmol/L, which was 2.64 time that of the BLK group. Furthermore, β-carotene significantly affected the structure and composition of gut microflora, increasing the abundance of Roseburia, Parasutterella and Lachnospiraceae, and decreasing the abundance of Dialister, Collinsella and Enterobacter (P < 0.05). This study provides a new way to understand how β-carotene works in human body with gut microflora.

References

[1]

B.P. Arathi, P.R.R. Sowmya, K. Vijay, et al., Metabolomics of carotenoids: the challenges and prospects: a review, Trends Food Sci. Tech. 45 (2015) 105-117. https://doi.org/10.1016/j.tifs.2015.06.003.

[2]

R. Álvarez, A.J. Meléndez-Martínez, I.M. Vicario, et al., Carotenoid and vitamin A contents in biological fluids and tissues of animals as an effect of the diet: a review, Food Rev. Int. 31 (2015) 319-340. https://doi.org/10.1080/87559129.2015.1015139.

[3]

G. Maiani, M.J.P. Castón, G. Catasta, et al., Carotenoids: actual knowledge on food sources, intakes, stability and bioavailability and their protective role in humans, Mol. Nutr. Food Res. 53(Suppl 2) (2009) 194-218. https://doi.org/10.1002/mnfr.200800053.

[4]

T. Grune, G. Lietz, A. Palou, et al., β-Carotene is an important vitamin a source for humans, J. Nutr. 140 (2010) 2268S-2285S. https://doi.org/10.3945/jn.109.119024.

[5]

C. Sy, B. Gleize, O. Dangles, et al., Effects of physicochemical properties of carotenoids on their bioaccessibility, intestinal cell uptake, and blood and tissue concentrations, Mol. Nutr. Food Res. 56 (2012) 1385-1397. https://doi.org/10.1002/mnfr.201200041.

[6]

E. Hedrén, V. Diaz, U. Svanberg, Estimation of carotenoid accessibility from carrots determined by an in vitro digestion method, Eur. J. Clin. Nutr. 56 (2002) 425-430. https://doi.org/10.1038/sj.ejcn.1601329.

[7]

I. Sekirov, S.L. Russell, L.C.M. Antunes, et al., Gut microbiota in health and disease, Physiol. Rev. 90 (2010) 859-904. https://doi.org/10.1152/physrev.00045.2009.

[8]

L.M. Cox, S. Yamanishi, J. Sohn, et al., Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences, Cell 158 (2014) 705-721. https://doi.org/10.1016/j.cell.2014.05.052.

[9]

M.V. Selma, J.C. Espín, F.A. Tomás-Barberán, Interaction between phenolics and gut microbiota: role in human health, J. Agr. Food Chem. 57 (2009) 6485-6501. https://doi.org/10.1021/jf902107d.

[10]

T. Ozdal, D.A. Sela, J. Xiao, et al., The reciprocal interactions between polyphenols and gut microbiota and effects on bioaccessibility, Nutrients 8 (2016) 78. https://doi.org/10.3390/nu8020078.

[11]

F. Cardona, C. Andrés-Lacueva, S. Tulipani, et al., Benefits of polyphenols on gut microbiota and implications in human health, J. Nutr. Biochem. 24 (2013) 1414-1422. https://doi.org/10.1016/j.jnutbio.2013.05.001.

[12]

P. Grolier, P. Borel, C. Duszka, et al., The bioavailability of alpha- and betacarotene is affected by gut microflora in the rat, Brit. J. Nutr. 80 (1998) 199- 204. https://doi.org/10.1017/S0007114598001111.

[13]

F. Jalal, M.C. Nesheim, Z. Agus, et al., Serum retinol concentrations in children are affected by food sources of beta-carotene, fat intake, and anthelmintic drug treatment, Am. J. Clin. Nutr. 68 (1998) 623-629. https://doi.org/10.1093/ajcn/68.3.623.

[14]

T. Bohn, C. Desmarchelier, L.O. Dragsted, et al., Host-related factors explaining interindividual variability of carotenoid bioavailability and tissue concentrations in humans, Mol. Nutr. Food Res. 61 (2017) 1600685. https://doi.org/10.1002/mnfr.201600685.

[15]

I. Goñi, J. Serrano, F. Saura-Calixto, Bioaccessibility of beta-carotene, lutein, and lycopene from fruits and vegetables, J. Agr. Food Chem. 54 (2006) 5382-5387. https://doi.org/10.1021/jf0609835.

[16]

J. Serrano, I. Goñi, F. Saura-Calixto, Determination of beta-carotene and lutein available from green leafy vegetables by an in vitro digestion and colonic fermentation method, J. Agr. Food Chem. 53 (2005) 2936-2940. https://doi.org/10.1021/jf0480142.

[17]

A. Kaulmann, C.M. André, Y.J. Schneider, et al., Carotenoid and polyphenol bioaccessibility and cellular uptake from plum and cabbage varieties, Food Chem. 197 (2016) 325-332. https://doi.org/10.1016/j.foodchem.2015.10.049.

[18]

Z. Djuric, C.M. Bassis, M.A. Plegue, et al., Colonic mucosal bacteria are associated with inter-individual variability in serum carotenoid concentrations, J. Acad. Nutr. Diet. 118 (2018) 606-616. https://doi.org/10.1016/j.jand.2017.09.013.

[19]

H. Liu, M. Liu, X. Fu, et al., Astaxanthin prevents alcoholic fatty liver disease by modulating mouse gut microbiota, Nutrients 10 (2018) 1298. https://doi.org/10.3390/nu10091298.

[20]

V. Tyssandier, E. Reboul, J.F. Dumas, et al., Processing of vegetable-borne carotenoids in the human stomach and duodenum, Am. J. Physiol-Gastr. L. 284 (2003) 913-923. https://doi.org/10.1152/ajpgi.00410.2002.

[21]

D. Chen, G. Chen, Y. Ding, et al., Polysaccharides from the flowers of tea (Camellia sinensis L.) modulate gut health and ameliorate cyclophosphamide-induced immunosuppression, J. Funct. Foods 61 (2019) 1756-4646. https://doi.org/10.1016/j.jff.2019.103470.

[22]

B.D. Stéphanie, S. Maha, R. Mathieu, et al., Digestive stability of xanthophylls exceeds that of carotenes as studied in a dynamic in vitro gastrointestinal system, J. Nutr. 139 (2009) 876-883. https://doi.org/10.3945/jn.108.103655.

[23]

L. Tian, J. Scholte, K. Borewicz, et al., Effects of pectin supplementation on the fermentation patterns of different structural carbohydrates in rats, Mol. Nutr. Food Res. 60 (2016) 2256-2266. https://doi.org/10.1002/mnfr.201600149.

[24]

B. Zhao, J. Wu, J. Li, et al., Lycopene alleviates DSS-induced colitis and behavioral disorders via mediating microbes-gut-brain axis balance, J. Agr. Food Chem. (2020) 3963-3975. https://doi.org/10.1021/acs.jafc.0c00196.

[25]

T. Bohn, C. Desmarchelier, L.O. Dragsted, et al., Host-related factors explaining interindividual variability of carotenoid bioavailability and tissue concentrations in humans, Mol. Nutr. Food Res. 61 (2017). https://doi.org/10.1002/mnfr.201600685.

[26]

L. Zhou, W. Wang, J. Huang, et al., In vitro extraction and fermentation of polyphenols from grape seeds (Vitis vinifera) by human intestinal microbiota, Food Funct. 7 (2016) 1959-1967. https://doi.org/10.1039/c6fo00032k.

[27]

M.H. Liang, J. Zhu, J.G. Jiang, Carotenoids biosynthesis and cleavage related genes from bacteria to plants, Crit. Rev. Food Sci. 58 (2017) 2314- 2333. https://doi.org/10.1080/10408398.2017.1322552.

[28]

S. Raghuvanshi, V. Reed, W.S. Blaner, et al., Cellular localization of β-carotene-15, 15'- oxygenase-1 (BCO1) and β-carotene-9', 10'-oxygenase-2 (BCO2) in rat liver and intestine, Arch. Biochem. Biophys. 572 (2015) 19- 27. https://doi.org/10.1016/j.abb.2014.12.024.

[29]

P. Wan, Y. Peng, G. Chen, et al., Dicaffeoylquinic acids from Ilex kudingcha attenuate dextran sulfate sodium-induced colitis in C57BL/6 mice in association with the modulation of gut microbiota, J.Funct. Foods 61 (2019). https://doi.org/10.1016/j.jff.2019.103468.

[30]

D. Ma, P. Forsythe, J. Bienenstock, Live Lactobacillus rhamnosus is essential for the inhibitory effect on tumor necrosis factor alphainduced interleukin-8 expression, Infect. Immun. 72 (2004) 5308-5314. https://doi.org/10.1128/IAI.72.9.5308-5314.2004.

[31]

S. Fukuda, H. Toh, K. Hase, et al., Bifidobacteria can protect from enteropathogenic infection through production of acetate, Nature 469 (2011) 543-547. https://doi.org/10.1038/nature09646.

[32]

A.L. Glasser, J. Boudeau, N. Barnich, et al., Adherent invasive Escherichia coli strains from patients with Crohn's disease survive and replicate within macrophages without inducing host cell death, Infect. Immun. 69 (2001) 5529-5537. https://doi.org/10.1128/IAI.69.9.5529-5537.2001.

[33]

R. Knight, A. Vrbanac, B.C. Taylor, et al., Best practices for analysing microbiomes, Nat. Rev. Microbiol. 16 (2018) 410-422. https://doi.org/10.1038/s41579-018-0029-9.

[34]

P.B. Eckburg, E.M. Bik, C.N. Bernstein, et al., Diversity of the human intestinal microbial flora, Science 308 (2005) 1635-1638. https://doi.org/10.1126/science.1110591.

[35]

M.L. Leth, M. Ejby, C. Workman, et al., Differential bacterial capture and transport preferences facilitate co-growth on dietary xylan in the human gut, Nat. Microbiol. 3 (2018) 570-580. https://doi.org/10.1038/s41564-018-0132-8.

[36]

K. Kasahara, K.A. Krautkramer, E. Org, et al., Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model, Nat. Microbiol. 3 (2018) 1461-1471. https://doi.org/10.1038/s41564-018-0272-x.

[37]

R.Y. Tito, H. Cypers, M. Joossens, et al., Brief report: dialister as a microbial marker of disease activity in spondyloarthritis, Arthritis Rheumatol. 69 (2017) 114-121. https://doi.org/10.1002/art.39802.

Food Science and Human Wellness
Pages 1369-1378
Cite this article:
Li Z, Dai Z, Shi E, et al. Study on the interaction between β-carotene and gut microflora using an in vitro fermentation model. Food Science and Human Wellness, 2023, 12(4): 1369-1378. https://doi.org/10.1016/j.fshw.2022.10.030

768

Views

75

Downloads

16

Crossref

16

Web of Science

16

Scopus

0

CSCD

Altmetrics

Received: 08 October 2021
Revised: 29 October 2021
Accepted: 25 November 2021
Published: 18 November 2022
© 2023 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

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

Return