Oyster Protein Hydrolysate Alleviates Cadmium Toxicity by Restoring Cadmium-Induced Intestinal Damage and Gut Microbiota Dysbiosis in Mice via Its Abundance of Methionine, Tyrosine, and Glutamine

WANG Jing Wen FANG Zhi Jia LI Yong Bin HUANG Lin Ru SUN Li Jun LIU Ying WANG Ya Ling LIAO Jian Meng

WANG Jing Wen, FANG Zhi Jia, LI Yong Bin, HUANG Lin Ru, SUN Li Jun, LIU Ying, WANG Ya Ling, LIAO Jian Meng. Oyster Protein Hydrolysate Alleviates Cadmium Toxicity by Restoring Cadmium-Induced Intestinal Damage and Gut Microbiota Dysbiosis in Mice via Its Abundance of Methionine, Tyrosine, and Glutamine[J]. Biomedical and Environmental Sciences, 2022, 35(7): 669-673. doi: 10.3967/bes2022.088
Citation: WANG Jing Wen, FANG Zhi Jia, LI Yong Bin, HUANG Lin Ru, SUN Li Jun, LIU Ying, WANG Ya Ling, LIAO Jian Meng. Oyster Protein Hydrolysate Alleviates Cadmium Toxicity by Restoring Cadmium-Induced Intestinal Damage and Gut Microbiota Dysbiosis in Mice via Its Abundance of Methionine, Tyrosine, and Glutamine[J]. Biomedical and Environmental Sciences, 2022, 35(7): 669-673. doi: 10.3967/bes2022.088

doi: 10.3967/bes2022.088

Oyster Protein Hydrolysate Alleviates Cadmium Toxicity by Restoring Cadmium-Induced Intestinal Damage and Gut Microbiota Dysbiosis in Mice via Its Abundance of Methionine, Tyrosine, and Glutamine

Funds: This work was supported by the National Natural Science Foundation of China [No. 32172215 and 31701706]; the Program for Scientific Research Start-up Funds of the Guangdong Ocean University [No. R17102]; Characteristic Innovation Project of Guangdong Province [No. 2018KTSCX089]; and Guangdong Basic and Applied Basic Research Foundation [No. 2019A1515010809, 2021A1515012443]
More Information
    Author Bio:

    WANG Jing Wen, female, born in 1995, PhD in progress, majoring in toxic effects and mitigation mechanisms of the heavy metal cadmium

    Corresponding author: FANG Zhi Jia, Tel/Fax: 86-759-2396027, E-mail: fzj4437549@163.comLIU Ying, Tel/Fax: 86-759-2396027, E-mail: 406662864@qq.com
  • Figure  1.  The influence of OPs on weight gain (A), feed intake (B), and relative organ weight (C) in Cd-treated mice. Serum inflammatory cytokine, LPS (D) and TNF-α (E), concentrations were determined by enzyme-linked immunosorbent assay. *P < 0.05 and **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group. LPS, lipopolysaccharide

    Figure  2.  The effects of OPs on Cd-induced histological changes in mouse intestines. (A) Representative histological image sections of the jejunum (magnification at 100× and 200×; (B) villus height; (C) crypt depth; **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group.

    S1.  Gut mRNA levels of tight junction genes (occludin, ZO-1, and plakophilin-1) determined by quantitative real-time PCR. ZO-1 (zonula occludens-1). **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group.

    Figure  3.  Comparing diversity and relative abundance of gut microbial communities between mouse groups. (A) Chao index for richness; (B) Ace index for richness; (C) Shannon index for diversity; (D) Gut microbiome patterns by principal component analysis. (E) Relative abundance of significantly altered gut bacteria taxa classified at the phylum level. (F) Circos graph representing gut microbiota composition at the genus level. (G) Microbial abundance heatmap at the genus level. *P < 0.05 and **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group.

    S2.  Effects of amino acids on the relative abundance of key gut microbiota (A–D: Prevotella, Parabacteroides, Oscillospira, and Flexispira) in mice. A (Alanine); C (Cysteine); D (Aspartic acid); E (Glutamic acid); F (Phenylalanine); G (Glycine) ; H (Histidine); I (Isoleucine); K (Lysine); L (leucine); M (Methionine); N (Asparagine); P (Proline); Q (Glutamine); R (Arginine); S (Serine); T (Threonine); V (Valine); W (Tryptophan); Y (Tyrosine). **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group.

    S1.   Animal experimental protocol

    GroupsSamples7 days treatments
    Controln = 6Mice were given drinking water
    Cdn = 6Drinking water containing 100 mg/L of CdCl2
    Cd+EDTAn = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 100 mg/kg per day of EDTA
    Cd+Milkn = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 100 mg/kg per day of milk
    Cd+L-OPsn = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 100 mg/kg per day of OPs
    Cd+H-OPsn = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 300 mg/kg per day of OPs
    下载: 导出CSV

    S2.   Animal experimental protocol

    GroupSamples7 days Treatments
    Controln = 6Mice were given drinking water
    Cdn = 6Drinking water containing 100 mg/L of CdCl2
    Cd+Ala(A)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Ala
    Cd+Cys(C)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Cys
    Cd+Asp(D)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Asp
    Cd+Glu(E)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Glu
    Cd+Phe(F)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Phe
    Cd+Gly(G)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Gly
    Cd+His(H)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of His
    Cd+Ile(I)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Ile
    Cd+Lys(K)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Lys
    Cd+Leu(L)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Leu
    Cd+Met(M)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Met
    Cd+Asn(N)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Asn
    Cd+Pro(P)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Pro
    Cd+Gln(Q)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Gln
    Cd+Arg(R)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Arg
    Cd+Ser(S)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Ser
    Cd+Thr(T)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Thr
    Cd+Val(V)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Val
    Cd+Trp(W)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Trp
    Cd+Tyr(Y)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Tyr
    下载: 导出CSV

    S3.   Primer sequences of target genes

    Gene nameProduct length (bp)Primer sequence (5´–3´)
    ZO-1192Forward: CTGCACGATTTCTGCTTCCTC
    Reverse: CCTCCATTGCTGTGCTAGTGA
    Occludin252Forward: CTAAATTGGCATCCAGCCCAG
    Reverse: TCCTTTCCACTCGGGCTCA
    Plakophilin-1274Forward: ACTATGACTGCCCACTCCCT
    Reverse: CCTGATTCCCGCACCACATC
    β-actin101Forward: TGTCCACCTTCCAGCAGATGT
    Reverse: AGCTCAGTAACAGTCCGCCTAGA
    下载: 导出CSV

    S4.   PICTUSt predicted KEGG pathways of the gut microbial community in different groups

    Function typeKEGG pathwaysRelative abundance (%)
    ControlCdCd+EDTACd+MilkCd+L-OPsCd+H-OPs
    MetabolismAmino Acid Metabolism7.209.23*8.658.848.127.94#
    Carbohydrate Metabolism7.208.78*8.32#8.34#7.87#7.73#
    Xenobiotics Biodegradation and Metabolism7.637.237.427.547.787.72
    Lipid Metabolism8.899.128.948.978.838.81
    Energy Metabolism4.244.94*4.42#4.46#4.34#4.32#
    Glycan Biosynthesis and Metabolism5.085.34*5.285.295.185.14
    Metabolism of Cofactors and Vitamins5.935.965.885.925.76#5.74#
    Metabolism of Terpenoids and Polyketides4.664.894.774.824.744.72
    Metabolism of Other Amino Acids3.814.244.184.144.083.97#
    Biosynthesis of Other Secondary Metabolites5.085.175.145.165.125.10
    Human DiseasesInfectious Diseases0.430.410.440.450.420.46
    Neurodegenerative Diseases0.240.320.250.260.230.22
    Genetic Information ProcessingTranslation3.824.18*3.84#3.923.88#3.83#
    Folding, Sorting and Degradation2.963.44*3.21#3.353.18#3.16#
      Note. *Significant difference from the control group at P < 0.05. #Significant difference from the Cd-exposed group at P < 0.05.
    下载: 导出CSV

    S5.   Composition and contents of amino acids of OPs and milk (μg/g)

    Amino acidsContents in milkContents in OPs
    Ala#3.45 ± 0.141499.88 ± 119.08
    CysND24.24 ± 10.71
    Asp0.47 ± 0.08136.22 ± 10.71
    Glu13.98 ± 0.011267.78 ± 72.61
    Phe*#2.27 ± 0.051652.84 ± 176.83
    GlyNDND
    His3.83 ± 0.171463.54 ± 86.01
    Ile*#3.86 ± 0.058964.73 ± 489.29
    Lys*6.17 ± 0.11598.17 ± 301.07
    Leu*#4.91 ± 0.11991.21 ± 26.48
    Met*#0.88 ± 0.014899.66 ± 148.11
    Asn6.84 ± 0.02529.08 ± 63.47
    Pro#3.18 ± 0.081409.75 ± 32.95
    Gln1.61 ± 0.042327.62 ± 96.69
    Arg1.46 ± 0.02ND
    Ser0.51 ± 0.01288.53 ± 35.01
    Thr*0.32 ± 0.02820.61 ± 43.03
    Val*#8.89 ± 0.26877.9 ± 134.03
    Trp1.69 ± 0.031142.69 ± 28.61
    Tyr#6.12 ± 0.2710236.49 ± 270.07
    Total amino acids70.55 ± 0.9139131.02 ± 2083.37
    Essential amino acid27.35 ± 0.3718805.14+1265.91
    Hydrophobic amino acids33.60 ± 0.8430532.49 ± 1343.91
      Note. *Essential amino acid. #Hydrophobic amino acids.
    下载: 导出CSV
  • [1] Chunhabundit R. Cadmium exposure and potential health risk from foods in contaminated area, Thailand. Toxicol Res, 2016; 32, 65−72.
    [2] Rani A, Kumar A, Lal A, et al. Cellular mechanisms of cadmium-induced toxicity: a review. Int J Environ Health Res, 2014; 24, 378−99.
    [3] He XW, Qi ZD, Hou H, et al. Structural and functional alterations of gut microbiome in mice induced by chronic cadmium exposure. Chemosphere, 2020; 246, 125747.
    [4] Assefa S, Köhler G. Intestinal microbiome and metal toxicity. Curr Opin Toxicol, 2020; 19, 21−7.
    [5] Yang JS, Chen W, Sun Y, et al. Effects of cadmium on organ function, gut microbiota and its metabolomics profile in adolescent rats. Ecotoxicol Environ Saf, 2021; 222, 112501.
    [6] Xiang XW, Zheng HZ, Wang R, et al. Ameliorative effects of peptides derived from oyster (Crassostrea gigas) on immunomodulatory function and gut microbiota structure in cyclophosphamide-treated mice. Mar Drugs, 2021; 19, 456.
    [7] Zhai QX, Wang G, Zhao JX, et al. Protective effects of Lactobacillus plantarum CCFM8610 against chronic cadmium toxicity in mice indicate routes of protection besides intestinal sequestration. Appl Environ Microbiol, 2014; 80, 4063−71.
    [8] Zhang XY, Peng ZL, Zheng HN, et al. The potential protective effect and possible mechanism of peptides from oyster (Crassostrea hongkongensis) hydrolysate on triptolide-induced testis injury in male mice. Mar Drugs, 2021; 19, 566.
    [9] Liu L, Dong WX, Wang SN, et al. Deoxycholic acid disrupts the intestinal mucosal barrier and promotes intestinal tumorigenesis. Food Funct, 2018; 9, 5588−97.
    [10] Huang LR, Fang ZJ, Gao J, et al. Protective role of L-threonine against cadmium toxicity in Saccharomyces cerevisiae. J Basic Microbiol, 2021; 61, 339−50.
    [11] Chen WY, Wang M, Zhang JW, et al. Acrolein disrupts tight junction proteins and causes endoplasmic reticulum stress-mediated epithelial cell death leading to intestinal barrier dysfunction and permeability. Am J Pathol, 2017; 187, 2686−97.
    [12] Zhao YC, Xie B, Gao J, et al. Dietary supplementation with sodium sulfate improves rumen fermentation, fiber digestibility, and the plasma metabolome through modulation of rumen bacterial communities in steers. Appl Environ Microbiol, 2020; 86, e01412−20.
  • [1] YU Jin Hai, HU Guo Liang, GUO Xiao Quan, CAO Hua Bin, XIA Zhao Fei, AMIN Buhe.  Quercetin Alleviates Lipopolysaccharide-Induced Cardiac Inflammation via Inhibiting Autophagy and Programmed Cell Death . Biomedical and Environmental Sciences, 2024, 37(1): 54-70. doi: 10.3967/bes2024.006
    [2] MENG Li Ying, TAO Wen Fu, LI Jing, ZHU Min, ZHONG De Bin, ZHOU Jing, QIN Xue, WEI Rong Guo.  Effects of Bisphenol A and Its Substitute, Bisphenol F, on the Gut Microbiota in Mice . Biomedical and Environmental Sciences, 2024, 37(1): 19-30. doi: 10.3967/bes2024.003
    [3] WEN Si Hui, LI Lu Bei, HUANG Hui Dan, XIE Ying, LUO Lei.  Metallothionein 1E Alleviates Cadmium-induced Renal Cytotoxicity through Promoting Mitochondrial Functional Recovery . Biomedical and Environmental Sciences, 2024, 37(1): 117-121. doi: 10.3967/bes2024.011
    [4] Dina Tucovic, Jelena Kulas, Ivana Mirkov, Dusanka Popovic, Lidija Zolotarevski, Marta Despotovic, Milena Kataranovski, Aleksandra Popov Aleksandrov.  Oral Cadmium Intake Enhances Contact Allergen-induced Skin Reaction in Rats . Biomedical and Environmental Sciences, 2022, 35(11): 1038-1050. doi: 10.3967/bes2022.132
    [5] LU Wei, SONG Yan Hua, XIA Yong, ZHANG Shi Xin, YAN Jun, CAI De Lei, XU Cai Ju.  Subacute Toxicity of High-Dose Resveratrol and its Effect on Gut Microbiota in Healthy Male and Female Sprague-Dawley Rats . Biomedical and Environmental Sciences, 2021, 34(7): 587-591. doi: 10.3967/bes2021.082
    [6] YANG Xue Hua, WU Yue Zhang, XIAO Kang, GAO Li Ping, CHEN Dong Dong, DONG Xiao Ping, SHI Qi.  Preparation of PrP-specific Polyclonal Antibody via Immunization of PRNP-knockout Mice with Recombinant Human PrP Protein . Biomedical and Environmental Sciences, 2020, 33(7): 493-501. doi: 10.3967/bes2020.066
    [7] WU Qian, WANG Lei, CHEN Yu Wei, ZHANG Hu, FANG Min, WU Yong Ning, GONG Zhi Yong.  Comparison of the Absorption and Distribution of Cadmium via Cadmium Chloride and Cadmium-contaminated Rice in Rats . Biomedical and Environmental Sciences, 2020, 33(2): 141-144. doi: 10.3967/bes2020.019
    [8] CHEN Zhi Jian, CHEN Jia Xing, WU Li Kang, LI Bi Yun, TIAN Ya Feng, XIAN Min, HUANG Zi Pei, YU Ri An.  Induction of Endoplasmic Reticulum Stress by Cadmium and Its Regulation on Nrf2 Signaling Pathway in Kidneys of Rats . Biomedical and Environmental Sciences, 2019, 32(1): 1-10. doi: 10.3967/bes2019.001
    [9] ZHU Shu Yun, JIANG Ning, TU Jie, YANG Jing, ZHOU Yue.  Antioxidant and Anti-aging Activities of Silybum Marianum Protein Hydrolysate in Mice Treated with D-galactose . Biomedical and Environmental Sciences, 2017, 30(9): 623-631. doi: 10.3967/bes2017.083
    [10] Effect of the Gut Microbiota on Obesity and ItsUnderlying Mechanisms:an Update . Biomedical and Environmental Sciences, 2015, 28(11): 839-847. doi: 10.3967/bes2015.117
    [11] LI Shu Gang, DING Yu Song, NIU Qiang, XU Shang Zhi, PANG Li Juan, MA Ru Lin, JING Ming Xia, FENG Gang Ling, LIU Jia Ming, GUO Shu Xia.  Grape Seed Proanthocyanidin Extract Alleviates Arsenic-induced Oxidative Reproductive Toxicity in Male Mice . Biomedical and Environmental Sciences, 2015, 28(4): 272-280. doi: 10.3967/bes2015.038
    [12] KE Shen, KE Qin Mei, JIA Wen Jing, CHENG Xi Yu, LI Hao, ZHANG Jie Ying, LUO Hui Fang, HE Jin Sheng, CHEN Zhi Nan.  Benchmark Dose Estimation for Cadmium-Induced Renal Effects Based on a Large Sample Population from Five Chinese Provinces . Biomedical and Environmental Sciences, 2015, 28(5): 383-387. doi: 10.3967/bes2015.054
    [13] WANG Lin, LIN Shu Qian, HE Yuan Long, LIU Gang, WANG Zhen Yong.  Protective Effects of Quercetin on Cadmium-induced Cytotoxicity in Primary Cultures of Rat Proximal Tubular Cells . Biomedical and Environmental Sciences, 2013, 26(4): 258-267. doi: 10.3967/0895-3988.2013.04.004
    [14] GONG-LIANG LIU, JU-FANG WANG, ZHI-YONG LI, SHI-ZHONG LIANG, XIAO-NING WANG.  Immunoassay for Cadmium Detection and Quantification . Biomedical and Environmental Sciences, 2009, 22(3): 188-193.
    [15] MILENA KATARANOVSKI, SRDJA JANKOVI(C), DRAGAN KATARANOVSKI, JELENA (S)TOSI(C), DESA BOGOJEVI(C).  Gender Differences in Acute Cadmium-Induced Systemic Inflammation in Rats . Biomedical and Environmental Sciences, 2009, 22(1): 1-7.
    [16] RIAN YU, LIN-GFEI HE, XUE-MIN CHEN.  Effects of Cadmium on Hepatocellular DNA Damage, Proto-Oncogene Expression and Apoptosis in Rats . Biomedical and Environmental Sciences, 2007, 20(2): 146-153.
    [17] XIU-LI CHANG, TAI-YI JIN, YUAN-FEN ZHOU.  Metallothionein 1 Isoform Gene Expression Induced by Cadmium in Human Peripheral Blood Lymphocytes . Biomedical and Environmental Sciences, 2006, 19(2): 104-109.
    [18] JUE CHEN, Tai-Yi JIN.  Influence of Isoflavones on Cadmium-induced Adverse Effects in Vascular Endothelial Cells (ECV 304) . Biomedical and Environmental Sciences, 2005, 18(3): 146-152.
    [19] XIAO PING, JIA Xiao-dong, ZHONG Wei-jian, JIN XI-PENG, GUNNAR NORDBERG.  Restorative Effects of Zinc and Selenium on Cadmium-induced Kidney Oxidative Damage in Rats . Biomedical and Environmental Sciences, 2002, 15(1): 67-74.
    [20] TANG LING-FANG, YANG Yong-nian, CHEN YAN-MENG, ZHANG Zhen-ling, SONG LING, FENG ZHU-YING.  Influences of Chloropazine, Nimodipine and Their Combination on the Toxic Effects of Cadmium in Liver and Kidney of Mice . Biomedical and Environmental Sciences, 1999, 12(3): 214-221.
  • 22061Supplementary Materials.pdf
  • 加载中
图(5) / 表ll (5)
计量
  • 文章访问数:  699
  • HTML全文浏览量:  293
  • PDF下载量:  89
  • 被引次数: 0
出版历程
  • 收稿日期:  2022-02-17
  • 录用日期:  2022-06-12
  • 刊出日期:  2022-07-27

Oyster Protein Hydrolysate Alleviates Cadmium Toxicity by Restoring Cadmium-Induced Intestinal Damage and Gut Microbiota Dysbiosis in Mice via Its Abundance of Methionine, Tyrosine, and Glutamine

doi: 10.3967/bes2022.088
    基金项目:  This work was supported by the National Natural Science Foundation of China [No. 32172215 and 31701706]; the Program for Scientific Research Start-up Funds of the Guangdong Ocean University [No. R17102]; Characteristic Innovation Project of Guangdong Province [No. 2018KTSCX089]; and Guangdong Basic and Applied Basic Research Foundation [No. 2019A1515010809, 2021A1515012443]
    作者简介:

    WANG Jing Wen, female, born in 1995, PhD in progress, majoring in toxic effects and mitigation mechanisms of the heavy metal cadmium

    通讯作者: FANG Zhi Jia, Tel/Fax: 86-759-2396027, E-mail: fzj4437549@163.comLIU Ying, Tel/Fax: 86-759-2396027, E-mail: 406662864@qq.com

English Abstract

WANG Jing Wen, FANG Zhi Jia, LI Yong Bin, HUANG Lin Ru, SUN Li Jun, LIU Ying, WANG Ya Ling, LIAO Jian Meng. Oyster Protein Hydrolysate Alleviates Cadmium Toxicity by Restoring Cadmium-Induced Intestinal Damage and Gut Microbiota Dysbiosis in Mice via Its Abundance of Methionine, Tyrosine, and Glutamine[J]. Biomedical and Environmental Sciences, 2022, 35(7): 669-673. doi: 10.3967/bes2022.088
Citation: WANG Jing Wen, FANG Zhi Jia, LI Yong Bin, HUANG Lin Ru, SUN Li Jun, LIU Ying, WANG Ya Ling, LIAO Jian Meng. Oyster Protein Hydrolysate Alleviates Cadmium Toxicity by Restoring Cadmium-Induced Intestinal Damage and Gut Microbiota Dysbiosis in Mice via Its Abundance of Methionine, Tyrosine, and Glutamine[J]. Biomedical and Environmental Sciences, 2022, 35(7): 669-673. doi: 10.3967/bes2022.088
  • As a typical heavy metal contaminant in food, cadmium (Cd) easily enters the human food chain via crop production, and therefore, threatens human health[1]. Intestinal absorption is the principal route for Cd entry into the body, and therefore, intestinal tissue become the primary target for Cd toxicity[2]. Previous studies showed that acute Cd exposure damaged intestinal tight junctions, resulting in gut barrier destruction and alterations in gut microbiota[3]. Thus, the effective prevention of Cd-induced intestinal damage has become a major research challenge.

    The gut microbiota is considered an important target of Cd, and therefore, affects gut microbiota diversity[4]. A previous study reported that Cd exposure significantly altered the mouse gut microbiome and significantly decreased microbial diversity in the gut[5]. However, it is unclear how to efficiently alleviate the adverse effects of Cd.

    Recent studies indicated that oyster protein hydrolysates (OPs) exhibited various biological and pharmacological functions, including maintaining the gut microbiota and intestinal barrier[6]. However, the effects of OPs on Cd-induced intestinal damage and gut microbiota dysbiosis are unknown. In view of the potential protective effects of bioactive oyster proteins against Cd, we investigated the alleviating effect of OPs on intestinal damage and gut microbiota in Cd-exposed mice.

    We obtained 168 specific pathogen-free (SPF) mice (Kunming strain, 25–35 g) from Changsha Tianqin Biotechnology (Changsha, China), and maintained animals at the Guangdong Ocean University Animal Center under a controlled light (12 h light and dark) and temperature (25 °C) environment. Mice were supplied standard commercial mouse food and water. Animal studies were approved by the Guangdong Ocean University Animal Welfare Committee (Approval No. GDOU-LAE-2020-009). A completely randomized treatment study design was used.

    For toxicity tests, 36 mice of similar weight were randomly divided into six groups (n = 6/group). The experimental design is shown in Supplementary Table S1 (available in www.besjournal.com). Grouping schemes and treatments were based on previous studies[7,8]. After 7 days of continuous treatment, mice were humanely sacrificed using ether anesthesia, and heart, liver, spleen, lung, and kidney organs removed and stored at −80 °C for further analysis. The intestines were excised, washed in saline solution, and fixed in 10% formalin for histopathological studies.

    Table S1.  Animal experimental protocol

    GroupsSamples7 days treatments
    Controln = 6Mice were given drinking water
    Cdn = 6Drinking water containing 100 mg/L of CdCl2
    Cd+EDTAn = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 100 mg/kg per day of EDTA
    Cd+Milkn = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 100 mg/kg per day of milk
    Cd+L-OPsn = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 100 mg/kg per day of OPs
    Cd+H-OPsn = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 300 mg/kg per day of OPs

    To evaluate the protective effects of amino acids on critical gut microbiota in-Cd exposed mice, 132 SPF mice were randomly divided into 22 groups (n = 6/group). The experimental design is shown in Supplementary Table S2 (available in www.besjournal.com).

    Table S2.  Animal experimental protocol

    GroupSamples7 days Treatments
    Controln = 6Mice were given drinking water
    Cdn = 6Drinking water containing 100 mg/L of CdCl2
    Cd+Ala(A)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Ala
    Cd+Cys(C)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Cys
    Cd+Asp(D)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Asp
    Cd+Glu(E)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Glu
    Cd+Phe(F)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Phe
    Cd+Gly(G)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Gly
    Cd+His(H)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of His
    Cd+Ile(I)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Ile
    Cd+Lys(K)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Lys
    Cd+Leu(L)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Leu
    Cd+Met(M)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Met
    Cd+Asn(N)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Asn
    Cd+Pro(P)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Pro
    Cd+Gln(Q)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Gln
    Cd+Arg(R)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Arg
    Cd+Ser(S)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Ser
    Cd+Thr(T)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Thr
    Cd+Val(V)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Val
    Cd+Trp(W)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Trp
    Cd+Tyr(Y)n = 6Drinking water containing 100 mg/L of CdCl2 and orally administered with 40 mmol/kg per day of Tyr

    OPs were prepared as previously described[8]. The oyster (Crassostrea hongkongensis) of Hong Kong, China was homogenized in distilled water (1:3 w/w) and ground three times at 4,000 ×g for 5 min. The homogenate was centrifuged (4 ℃) at 12,000 ×g for 20 min. Homogenized oyster was hydrolyzed with a neutral protease (Pangbo Biotech, Nanning, China, 2 × 105 U/g) at a protease/substrate ratio of 2.0% (w/v) (pH 7.0). After a 4 h incubation with the neutral protease at 50 ℃, the protease was inactivated at 100 ℃ for 15 min, and the enzymatic hydrolysate centrifuged at 15,000 ×g at 4 ℃ for 20 min to generate a supernatant. Samples were freeze-dried (FD-551, EYELA, Tokyo, Japan) for subsequent studies.

    To assess the alleviating effects of OPs on Cd-induced intestinal damage, serum tumor necrosis factor alpha (TNF-α) and lipopolysaccharide (LPS) were tested as indicators of intestinal damage. The epithelial barrier absorbs nutrients and effectively prevents the invasion of pathogens and LPS[9]. Intestinal damage was observed by microscopy after samples were stained in hematoxylin and eosin (H&E). Gut barrier related gene (ZO-1, occludin, and plakophilin-1, primer sequences are shown in Supplementary Table S3, available in www.besjournal.com) levels were measured by quantitative real-time PCR (qRT-PCR). β-actin was used as an internal reference housekeeping gene. qRT-PCR was performed on a CFX Real-Time System (CFX96, Bio-Rad, Hercules, CA, USA). All samples were analyzed in triplicate, and the 2−ΔΔCt method was used to analyze gene expression levels.

    Table S3.  Primer sequences of target genes

    Gene nameProduct length (bp)Primer sequence (5´–3´)
    ZO-1192Forward: CTGCACGATTTCTGCTTCCTC
    Reverse: CCTCCATTGCTGTGCTAGTGA
    Occludin252Forward: CTAAATTGGCATCCAGCCCAG
    Reverse: TCCTTTCCACTCGGGCTCA
    Plakophilin-1274Forward: ACTATGACTGCCCACTCCCT
    Reverse: CCTGATTCCCGCACCACATC
    β-actin101Forward: TGTCCACCTTCCAGCAGATGT
    Reverse: AGCTCAGTAACAGTCCGCCTAGA

    To assess the alleviating effects of OPs on Cd-induced gut microbiota dysbiosis using 16S rRNA sequencing, we used 515F (5'-GTGCCAGCMGCCGCGGTAA-3') and 806R (5'-GGACTACHVGGGTWTCTAAT-3') primer sequences, and sequencing data were used to identify species. Sequence data analyses were mainly performed in QIIME and R packages (v3.2.0). Operating taxonomic unit (OTU) α-diversity indices (Chao, ACE, and Shannon) were calculated in QIIME. We also performed β-diversity analyses to investigate structural variations in microbial communities across samples using UniFrac distance metrics. PICRUSt was used to predict functional profiles in microbial communities according to Kyoto Encyclopedia of Genes and Genomes (KEGG) database functional categories. To identify key amino acids used by OPs in modulating gut microbiota, the effects of each amino acid on key gut microbiota were evaluated using 16S rRNA sequencing.

    The amino acid composition and content of OPs and milk were measured by liquid chromatography with tandem mass spectrometry (LC-MS/MS) using the AB SCIEX QTRAP® 4000 System as previously described[10].

    Data were expressed as the mean ± standard deviation. JMP Pro 13 was used for statistical analyses. A P < 0.05 value was considered statistically significant.

    We observed that Cd-exposure exerted adverse effects on the growth performance of mice (weight gain and feed intake). Additionally, OPs significantly reduced increases in relative weight of the liver and intestine (jejunum) in Cd-exposed mice (Figure 1A–C).

    Figure 1.  The influence of OPs on weight gain (A), feed intake (B), and relative organ weight (C) in Cd-treated mice. Serum inflammatory cytokine, LPS (D) and TNF-α (E), concentrations were determined by enzyme-linked immunosorbent assay. *P < 0.05 and **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group. LPS, lipopolysaccharide

    Next, we tested the alleviating effects of OPs on Cd-induced intestine injury. OP administration markedly reduced serum TNF-α and LPS levels in Cd-exposed mice (Figure 1D, E). Microscopic examinations showed that OPs restored villus height and crypt depth to approximate control group levels (Figure 2A–C). Thus, OPs exerted significant protective effects against Cd-induced intestinal damage. High serum LPS levels are usually caused by increased barrier permeability and broken tight junctions[11]. Cd significantly inhibited occludin and ZO-1 mRNA expression levels, and as expected, OPs restored this expression. No significant changes in mRNA expression levels were observed for plakophilin-1 (Supplementary Figure S1, available in www.besjournal.com).

    Figure 2.  The effects of OPs on Cd-induced histological changes in mouse intestines. (A) Representative histological image sections of the jejunum (magnification at 100× and 200×; (B) villus height; (C) crypt depth; **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group.

    Figure S1.  Gut mRNA levels of tight junction genes (occludin, ZO-1, and plakophilin-1) determined by quantitative real-time PCR. ZO-1 (zonula occludens-1). **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group.

    We further tested the alleviating effects of OPs on gut microbiota dysbiosis in Cd-exposed mice. Richness indices (Chao and Ace) and the diversity index (Shannon) were significantly recovered in a dose-dependent manner after OP supplementation to Cd-exposed mice (Figure 3A–C). Principal component analysis showed that the control group was well separated from the Cd-treatment group. These results suggested OPs may alleviate Cd toxicity by effectively restoring gut microbiota richness and diversity in Cd-exposed mice. OPs supplementation restored Firmicutes and Proteobacteria abundance and inhibited the increase of Bacteroidetes by Cd. At the genus level, when compared with the control group, Prevotella and Parabacteroides were significantly increased in the Cd group. In contrast, Oscillospira and Flexispira were significantly decreased in the Cd group, while the OP supplementation group had the best recovery (Figure 3D–F). These data demonstrated that OPs putatively alleviated intestinal damage and barrier function by reversing gut microbiota dysbiosis in Cd-exposed mice.

    Figure 3.  Comparing diversity and relative abundance of gut microbial communities between mouse groups. (A) Chao index for richness; (B) Ace index for richness; (C) Shannon index for diversity; (D) Gut microbiome patterns by principal component analysis. (E) Relative abundance of significantly altered gut bacteria taxa classified at the phylum level. (F) Circos graph representing gut microbiota composition at the genus level. (G) Microbial abundance heatmap at the genus level. *P < 0.05 and **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group.

    We then investigated the modulating mechanisms of OPs on gut microbiota dysbiosis in Cd-exposed mice and compared gut microbiota functional profiles between groups using PICRUSt computation analysis. As shown in Supplementary Table S4 (available in www.besjournal.com), most enriched KEGG pathways were associated with metabolic function. Amongst pathways, the relative proportion of metabolic pathways, such as amino acid metabolism, carbohydrate metabolism, energy metabolism, and glycan biosynthesis and metabolism were significantly up-regulated (Welch's t-test, P < 0.05) after exposure to Cd. In a previous study, acute Cd-exposure potentially induced stress responses in gut microbiota and up-regulated pathways associated with amino acid metabolism to supplement the amino acids that Cd had consumed, such as glutathione[3]. Prevotella was closely associated with amino acid metabolism and its growth was affected by amino acids such as aspartate.

    Table S4.  PICTUSt predicted KEGG pathways of the gut microbial community in different groups

    Function typeKEGG pathwaysRelative abundance (%)
    ControlCdCd+EDTACd+MilkCd+L-OPsCd+H-OPs
    MetabolismAmino Acid Metabolism7.209.23*8.658.848.127.94#
    Carbohydrate Metabolism7.208.78*8.32#8.34#7.87#7.73#
    Xenobiotics Biodegradation and Metabolism7.637.237.427.547.787.72
    Lipid Metabolism8.899.128.948.978.838.81
    Energy Metabolism4.244.94*4.42#4.46#4.34#4.32#
    Glycan Biosynthesis and Metabolism5.085.34*5.285.295.185.14
    Metabolism of Cofactors and Vitamins5.935.965.885.925.76#5.74#
    Metabolism of Terpenoids and Polyketides4.664.894.774.824.744.72
    Metabolism of Other Amino Acids3.814.244.184.144.083.97#
    Biosynthesis of Other Secondary Metabolites5.085.175.145.165.125.10
    Human DiseasesInfectious Diseases0.430.410.440.450.420.46
    Neurodegenerative Diseases0.240.320.250.260.230.22
    Genetic Information ProcessingTranslation3.824.18*3.84#3.923.88#3.83#
    Folding, Sorting and Degradation2.963.44*3.21#3.353.18#3.16#
      Note. *Significant difference from the control group at P < 0.05. #Significant difference from the Cd-exposed group at P < 0.05.

    As major components of OPs, amino acids have key roles modulating gut microbiota dysbiosis in Cd-exposed mice. To identify such amino acids, we evaluated the effects of each amino acid on key gut microbiota (Prevotella, Parabacteroides, Oscillospira, and Flexispira) using 16S rRNA sequencing. Of the amino acids which potentially regulated gut microbiota, tyrosine (Y), methionine (M), and glutamine (Q) exhibited extremely significant (P < 0.01) effects in restoring the relative abundance of key gut microbiota in Cd-exposed mice (Supplementary Figure S2, available in www.besjournal.com). The amino acid composition of OPs and milk was analyzed by LC-MS/MS. As shown (Supplementary Table S5 available in www.besjournal.com), OPs were particularly rich in free tyrosine (10,236.49 ± 270.07 μg/g), isoleucine (8,964.73 ± 489.29 μg/g), methionine (4,899.66 ± 148.11 μg/g), glutamine (2,327.62 ± 96.69 μg/g), and threonine (820.61 ± 43.03 μg/g), and levels were approximately 1000-fold higher than in milk. A previous study reported that methionine decreased the relative abundances of Prevotella but increased the relative abundances of Flexispira in steers[12]. Thus, OPs may reduce Cd-induced stress responses, down-regulate pathways associated with amino acid metabolism in gut microbial communities, and decrease Prevotella abundance and other related gut microbiota by providing amino acids.

    Figure S2.  Effects of amino acids on the relative abundance of key gut microbiota (A–D: Prevotella, Parabacteroides, Oscillospira, and Flexispira) in mice. A (Alanine); C (Cysteine); D (Aspartic acid); E (Glutamic acid); F (Phenylalanine); G (Glycine) ; H (Histidine); I (Isoleucine); K (Lysine); L (leucine); M (Methionine); N (Asparagine); P (Proline); Q (Glutamine); R (Arginine); S (Serine); T (Threonine); V (Valine); W (Tryptophan); Y (Tyrosine). **P < 0.01; significant difference from the control group. #P < 0.05 and ##P < 0.01; significant difference from the Cd-treated group.

    Table S5.  Composition and contents of amino acids of OPs and milk (μg/g)

    Amino acidsContents in milkContents in OPs
    Ala#3.45 ± 0.141499.88 ± 119.08
    CysND24.24 ± 10.71
    Asp0.47 ± 0.08136.22 ± 10.71
    Glu13.98 ± 0.011267.78 ± 72.61
    Phe*#2.27 ± 0.051652.84 ± 176.83
    GlyNDND
    His3.83 ± 0.171463.54 ± 86.01
    Ile*#3.86 ± 0.058964.73 ± 489.29
    Lys*6.17 ± 0.11598.17 ± 301.07
    Leu*#4.91 ± 0.11991.21 ± 26.48
    Met*#0.88 ± 0.014899.66 ± 148.11
    Asn6.84 ± 0.02529.08 ± 63.47
    Pro#3.18 ± 0.081409.75 ± 32.95
    Gln1.61 ± 0.042327.62 ± 96.69
    Arg1.46 ± 0.02ND
    Ser0.51 ± 0.01288.53 ± 35.01
    Thr*0.32 ± 0.02820.61 ± 43.03
    Val*#8.89 ± 0.26877.9 ± 134.03
    Trp1.69 ± 0.031142.69 ± 28.61
    Tyr#6.12 ± 0.2710236.49 ± 270.07
    Total amino acids70.55 ± 0.9139131.02 ± 2083.37
    Essential amino acid27.35 ± 0.3718805.14+1265.91
    Hydrophobic amino acids33.60 ± 0.8430532.49 ± 1343.91
      Note. *Essential amino acid. #Hydrophobic amino acids.

    In conclusion, OPs may alleviate Cd toxicity by restoring Cd-induced gut microbiota dysbiosis via its abundance of the free amino acids tyrosine, methionine, and glutamine.

    Conflicts of Interest The authors declare none.

参考文献 (12)
补充材料:
22061Supplementary Materials.pdf

目录

    /

    返回文章
    返回