Detection of Alterations in the Gut Microbiota and Intestinal Permeability in Patients With Hashimoto Thyroiditis

Leonardo César de Freitas Cayres, Larissa Vedovato Vilela de Salis, Guilherme Siqueira Pardo Rodrigues, André van Helvoort Lengert, Ana Paula Custódio Biondi, Larissa Donadel Barreto Sargentini, João Luiz Brisotti, Eleni Gomes, Gislane Lelis Vilela de Oliveira, Leonardo César de Freitas Cayres, Larissa Vedovato Vilela de Salis, Guilherme Siqueira Pardo Rodrigues, André van Helvoort Lengert, Ana Paula Custódio Biondi, Larissa Donadel Barreto Sargentini, João Luiz Brisotti, Eleni Gomes, Gislane Lelis Vilela de Oliveira

Abstract

Hashimoto thyroiditis (HT) is the most common autoimmune disease worldwide, characterized by chronic inflammation and circulating autoantibodies against thyroid peroxidase and thyroglobulin. Patients require hormone replacement with oral levothyroxine, and if untreated, they can develop serious adverse health effects and ultimately death. There is a lot of evidence that the intestinal dysbiosis, bacterial overgrowth, and increased intestinal permeability favor the HT development, and a thyroid-gut axis has been proposed, which seems to impact our entire metabolism. Here, we evaluated alterations in the gut microbiota in Brazilian patients with HT and correlated this data with dietary habits, clinical data, and systemic cytokines and zonulin concentrations. Stool samples from 40 patients with HT and 53 controls were analyzed using real-time PCR, the serum cytokine levels were evaluated by flow cytometry, zonulin concentrations by ELISA, and the dietary habits were recorded by a food frequency questionnaire. We observed a significant increase (p < 0.05) in the Bacteroides species and a decrease in Bifidobacterium in samples of patients with HT. In addition, Lactobacillus species were higher in patients without thyroid hormone replacement, compared with those who use oral levothyroxine. Regarding dietary habits, we demonstrated that there are significant differences in the consumption of vegetables, fruits, animal-derived proteins, dairy products, saturated fats, and carbohydrates between patients and control group, and an inverse correlation between animal-derived protein and Bacteroides genus was detected. The microbiota modulation by diet directly influences the inflammatory profile due to the generated microbiota metabolites and their direct or indirect action on immune cells in the gut mucosa. Although there are no differences in systemic cytokines in our patients with HT, we detected increased zonulin concentrations, suggesting a leaky gut in patients with HT. These findings could help understand the development and progression of HT, while further investigations to clarify the underlying mechanisms of the diet-microbiota-immune system axis are still needed.

Keywords: Hashimoto thyroiditis; autoimmune disease; dietary habits; gut microbiota; inflammatory cytokines; intestinal dysbiosis; intestinal permeability.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Cayres, de Salis, Rodrigues, Lengert, Biondi, Sargentini, Brisotti, Gomes and de Oliveira.

Figures

Figure 1
Figure 1
Relative expression units (REU) of the intestinal microbiota found in stool samples from patients with Hashimoto thyroiditis (HT) and healthy controls (CTRL). (A)Bacteroides, (B)Bifidobacterium, (C)Clostridium coccoides, (D)Clostridium coccoides-Eubacterium-rectale, (E)Clostridium leptum, (F)Lactobacillus, (G)Prevotella, and (H)Roseburia species. Bars represent the median with interquartile range of REU per 200 mg of stool.
Figure 2
Figure 2
Spearmans' correlation between the relative expression units (REU) of the intestinal microbiota and clinical data. (A) REU of Clostridium coccoides and Clostridium coccoides-Eubacterium-rectale with TSH concentrations, (B) REU of Roseburia with FT4 levels, (C) REU of Clostridium coccoides with disease duration, and (D) REU of Lactobacillus species in treated patients (LT4) and patients with no hormone replacement (NR).
Figure 3
Figure 3
Cytokine profile in HT patients and control subjects (CTRL). Serum concentrations of (A) IL-2, (B) IL-4, (C) IL-6, (D) IL-10, (E) IL-17A, (F) IFN-γ, and (G) TNF. Statistical analyses were performed by Mann-Whitney. Significance was set at P < 0.05.
Figure 4
Figure 4
Zonulin concentrations and correlations with systemic cytokine levels. (A) Serum zonulin concentrations in HT patients and controls (CTRL), (B) Correlations among zonulin levels with IFN-γ and IL-2 concentrations.

References

    1. Caturegli P, De Remigis A, Rose NR. Hashimoto thyroiditis: clinical and diagnostic criteria. Autoimmun Rev. (2014) 13:391–7. 10.1016/j.autrev.2014.01.007
    1. Fröhlich E, Wahl R. Thyroid autoimmunity: role of anti-thyroid antibodies in thyroid and extra-thyroidal diseases. Front Immunol. (2017) 8:521. 10.3389/fimmu.2017.00521
    1. Ajjan RA, Weetman AP. The pathogenesis of Hashimoto's thyroiditis: further developments in our understanding. Horm Metab Res. (2015) 47:702–10. 10.1055/s-0035-1548832
    1. Lauritano EC, Bilotta AL, Gabrielli M, Scarpellini E, Lupascu A, Laginestra A, et al. . Association between hypothyroidism and small intestinal bacterial overgrowth. J Clin Endocrinol Metab. (2007) 92:4180–4. 10.1210/jc.2007-0606
    1. Chiovato L, Magri F, Carlé A. Hypothyroidism in context: where we've been and where we're going. Adv Ther. (2019) 36(Suppl. 2):47–58. 10.1007/s12325-019-01080-8
    1. Chaker L, Bianco AC, Jonklaas J, Peeters RP. Hypothyroidism. Lancet Lond Engl. (2017) 390:1550–62. 10.1016/S0140-6736(17)30703-1
    1. Hiromatsu Y, Satoh H, Amino N. Hashimoto's thyroiditis: history and future outlook. Horm Athens Greece. (2013) 12:12–8. 10.1007/BF03401282
    1. Garmendia Madariaga A, Santos Palacios S, Guillén-Grima F, Galofré JC. The incidence and prevalence of thyroid dysfunction in Europe: a meta-analysis. J Clin Endocrinol Metab. (2014) 99:923–31. 10.1210/jc.2013-2409
    1. Sgarbi JA, Teixeira PFS, Maciel LMZ, Mazeto GMFS, Vaisman M, Montenegro Junior RM, et al. . The Brazilian consensus for the clinical approach and treatment of subclinical hypothyroidism in adults: recommendations of the thyroid Department of the Brazilian Society of Endocrinology and Metabolism. Arq Bras Endocrinol Amp Metabol. (2013) 57:166–83. 10.1590/S0004-27302013000300003
    1. Loh HH, Lim LL, Yee A, Loh HS. Association between subclinical hypothyroidism and depression: an updated systematic review and meta-analysis. BMC Psychiatry. (2019) 19:12. 10.1186/s12888-018-2006-2
    1. Qin J, Yu Z, Guan H, Shi L, Liu Y, Zhao N, et al. . High thyroglobulin antibody levels increase the risk of differentiated thyroid carcinoma. Dis Markers. (2015) 2015:648670. 10.1155/2015/648670
    1. Jankovic B, Le KT, Hershman JM. Clinical review: Hashimoto's thyroiditis and papillary thyroid carcinoma: is there a correlation? J Clin Endocrinol Metab. (2013) 98:474–82. 10.1210/jc.2012-2978
    1. Prinzi N, Sorrenti S, Baldini E, De Vito C, Tuccilli C, Catania A, et al. . Association of thyroid diseases with primary extra-thyroidal malignancies in women: results of a cross-sectional study of 6,386 patients. PLoS ONE. (2015) 10:e0122958. 10.1371/journal.pone.0122958
    1. Taylor PN, Albrecht D, Scholz A, Gutierrez-Buey G, Lazarus JH, Dayan CM, et al. . Global epidemiology of hyperthyroidism and hypothyroidism. Nat Rev Endocrinol. (2018) 14:301–16. 10.1038/nrendo.2018.18
    1. Liontiris MI, Mazokopakis EE. A concise review of Hashimoto thyroiditis (HT) and the importance of iodine, selenium, vitamin D and gluten on the autoimmunity and dietary management of HT patients. Points that need more investigation. Hell J Nucl Med. (2017) 20:51–6. 10.1967/s002449910507
    1. Köhling HL, Plummer SF, Marchesi JR, Davidge KS, Ludgate M. The microbiota and autoimmunity: their role in thyroid autoimmune diseases. Clin Immunol. (2017) 183:63–74. 10.1016/j.clim.2017.07.001
    1. Fröhlich E, Wahl R. Microbiota and thyroid interaction in health and disease. Trends Endocrinol Metab TEM. (2019) 30:479–90. 10.1016/j.tem.2019.05.008
    1. Knezevic J, Starchl C, Tmava Berisha A, Amrein K. Thyroid-gut-axis: how does the microbiota influence thyroid function? Nutrients. (2020) 12:1769. 10.3390/nu12061769
    1. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. (2017) 17:219–32. 10.1038/nri.2017.7
    1. Meng S, Badrinarain J, Sibley E, Fang R, Hodin R. Thyroid hormone and the d-type cyclins interact in regulating enterocyte gene transcription. J Gastrointest Surg Off J Soc Surg Aliment Tract. (2001) 5:49–55. 10.1016/S1091-255X(01)80013-5
    1. Song Y, Zhao M, Zhang H, Zhang X, Zhao J, Xu J, et al. . Thyroid-stimulating hormone levels are inversely associated with serum total bile acid levels: a cross-sectional study. Endocr Pract Off J Am Coll Endocrinol Am Assoc Clin Endocrinol. (2016) 22:420–6. 10.4158/EP15844.OR
    1. Fenneman AC, Rampanelli E, Yin YS, Ames J, Blaser MJ, Fliers E, et al. . Gut microbiota and metabolites in the pathogenesis of endocrine disease. Biochem Soc Trans. (2020) 48:915–31. 10.1042/BST20190686
    1. Ishaq HM, Mohammad IS, Guo H, Shahzad M, Hou YJ, Ma C. Molecular estimation of alteration in intestinal microbial composition in Hashimoto's thyroiditis patients. Biomed Pharmacother Biomedecine Pharmacother. (2017) 95:865–74. 10.1016/j.biopha.2017.08.101
    1. Zhao F, Feng J, Li J, Zhao L, Liu Y, Chen H, et al. . Alterations of the gut microbiota in Hashimoto's thyroiditis patients. Thyroid Off J Am Thyroid Assoc. (2018) 28:175–86. 10.1089/thy.2017.0395
    1. Larsen N, Vogensen FK, van den Berg FWJ, Nielsen DS, Andreasen AS, Pedersen BK, et al. . Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE. (2010) 5:e9085. 10.1371/journal.pone.0009085
    1. Rodrigues GSP, Cayres LCF, Gonçalves FP, Takaoka NNC, Lengert AH, Tansini A, et al. . Detection of increased relative expression units of Bacteroides and Prevotella, and decreased Clostridium leptum in stool samples from Brazilian rheumatoid arthritis patients: a pilot study. Microorganisms. (2019) 7:413. 10.3390/microorganisms7100413
    1. Lerner A, Aminov R, Matthias T. Dysbiosis may trigger autoimmune diseases via inappropriate post-translational modification of host proteins. Front Microbiol. (2016) 7:84. 10.3389/fmicb.2016.00084
    1. de Oliveira GLV, Leite AZ, Higuchi BS, Gonzaga MI, Mariano VS. Intestinal dysbiosis and probiotic applications in autoimmune diseases. Immunology. (2017) 152:1–12. 10.1111/imm.12765
    1. De Luca F, Shoenfeld Y. The microbiome in autoimmune diseases. Clin Exp Immunol. (2019) 195:74–85. 10.1111/cei.13158
    1. Lerner A, Jeremias P, Matthias T. Gut-thyroid axis and celiac disease. Endocr Connect. (2017) 6:R52–8. 10.1530/EC-17-0021
    1. Mu Q, Kirby J, Reilly CM, Luo XM. Leaky gut as a danger signal for autoimmune diseases. Front Immunol. (2017) 8:598. 10.3389/fimmu.2017.00598
    1. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. (2016) 16:341–52. 10.1038/nri.2016.42
    1. Liu S, An Y, Cao B, Sun R, Ke J, Zhao D. The composition of gut microbiota in patients bearing Hashimoto's thyroiditis with euthyroidism and hypothyroidism. Int J Endocrinol. (2020) 2020:5036959. 10.1155/2020/5036959
    1. Su X, Zhao Y, Li Y, Ma S, Wang Z. Gut dysbiosis is associated with primary hypothyroidism with interaction on gut-thyroid axis. Clin Sci Lond Engl. (1979) (2020) 134:1521–35. 10.1042/CS20200475
    1. Wu F, Guo X, Zhang J, Zhang M, Ou Z, Peng Y. Phascolarctobacterium faecium abundant colonization in human gastrointestinal tract. Exp Ther Med. (2017) 14:3122–6. 10.3892/etm.2017.4878
    1. O'Callaghan A, van Sinderen D. Bifidobacteria and their role as members of the human gut microbiota. Front Microbiol. (2016) 7:925. 10.3389/fmicb.2016.00925
    1. Patole SK, Rao SC, Keil AD, Nathan EA, Doherty DA, Simmer KN. Benefits of Bifidobacterium breve M-16V supplementation in preterm neonates - a retrospective cohort study. PLoS ONE. (2016) 11:e0150775. 10.1371/journal.pone.0150775
    1. Saez-Lara MJ, Gomez-Llorente C, Plaza-Diaz J, Gil A. The role of probiotic lactic acid bacteria and bifidobacteria in the prevention and treatment of inflammatory bowel disease and other related diseases: a systematic review of randomized human clinical trials. BioMed Res Int. (2015) 2015:505878. 10.1155/2015/505878
    1. Azad MAK, Sarker M, Li T, Yin J. Probiotic species in the modulation of gut microbiota: an overview. BioMed Res Int. (2018) 2018:9478630. 10.1155/2018/9478630
    1. Qi H, Li Y, Yun H, Zhang T, Huang Y, Zhou J, et al. . Lactobacillus maintains healthy gut mucosa by producing L-Ornithine. Commun Biol. (2019) 2:171. 10.1038/s42003-019-0424-4
    1. Patil AD. Link between hypothyroidism and small intestinal bacterial overgrowth. Indian J Endocrinol Metab. (2014) 18:307–9. 10.4103/2230-8210.131155
    1. Yao Z, Zhao M, Gong Y, Chen W, Wang Q, Fu Y, et al. . Relation of gut microbes and L-thyroxine through altered thyroxine metabolism in subclinical hypothyroidism subjects. Front Cell Infect Microbiol. (2020) 10:495. 10.3389/fcimb.2020.00495
    1. Laursen MF, Bahl MI, Michaelsen KF, Licht TR. First foods and gut microbes. Front Microbiol. (2017) 8:356. 10.3389/fmicb.2017.00356
    1. Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. (2019) 16:35–56. 10.1038/s41575-018-0061-2
    1. Korem T, Zeevi D, Suez J, Weinberger A, Avnit-Sagi T, Pompan-Lotan M, et al. . Growth dynamics of gut microbiota in health and disease inferred from single metagenomic samples. Science. (2015) 349:1101–6. 10.1126/science.aac4812
    1. Kolodziejczyk AA, Zheng D, Elinav E. Diet-microbiota interactions and personalized nutrition. Nat Rev Microbiol. (2019) 17:742–53. 10.1038/s41579-019-0256-8
    1. Yap YA, Mariño E. An insight into the intestinal web of mucosal immunity, microbiota, and diet in inflammation. Front Immunol. (2018) 9:2617. 10.3389/fimmu.2018.02617
    1. Cândido FG, Valente FX, Grześkowiak ŁM, Moreira APB, Rocha DMUP, Alfenas R de CG. Impact of dietary fat on gut microbiota and low-grade systemic inflammation: mechanisms and clinical implications on obesity. Int J Food Sci Nutr. (2018) 69:125–43. 10.1080/09637486.2017.1343286
    1. Zou J, Chassaing B, Singh V, Pellizzon M, Ricci M, Fythe MD, et al. . Fiber-mediated nourishment of gut microbiota protects against diet-induced obesity by restoring IL-22-mediated colonic health. Cell Host Microbe. (2018) 23:41–53.e4. 10.1016/j.chom.2017.11.003
    1. Matt SM, Allen JM, Lawson MA, Mailing LJ, Woods JA, Johnson RW. Butyrate and dietary soluble fiber improve neuroinflammation associated with aging in mice. Front Immunol. (2018) 9:1832. 10.3389/fimmu.2018.01832
    1. Sepahi A, Liu Q, Friesen L, Kim CH. Dietary fiber metabolites regulate innate lymphoid cell responses. Mucosal Immunol. (2020) 15:1–14. 10.1038/s41385-020-0312-8
    1. Tabasi M, Eybpoosh S, Sadeghpour Heravi F, Siadat SD, Mousavian G, Elyasinia F, et al. . Gut microbiota and serum biomarker analyses in obese patients diagnosed with diabetes and hypothyroid disorder. Metab Syndr Relat Disord. (2020) 24:1–8. 10.21203/-46107/v1
    1. Küçükemre Aydin B, Yildiz M, Akgün A, Topal N, Adal E, Önal H. Children with Hashimoto's thyroiditis have increased intestinal permeability: results of a pilot study. J Clin Res Pediatr Endocrinol. (2020) 12:303–7. 10.4274/jcrpe.galenos.2020.2019.0186
    1. Fasano A. Zonulin and its regulation of intestinal barrier function: the biological door to inflammation, autoimmunity, and cancer. Physiol Rev. (2011) 91:151–75. 10.1152/physrev.00003.2008
    1. Fasano A. All disease begins in the (leaky) gut: role of zonulin-mediated gut permeability in the pathogenesis of some chronic inflammatory diseases. F1000Research. (2020) 9:1–12. 10.12688/f1000research.20510.1
    1. Drago S, El Asmar R, Di Pierro M, Grazia Clemente M, Tripathi A, Sapone A, et al. . Gliadin, zonulin and gut permeability: effects on celiac and non-celiac intestinal mucosa and intestinal cell lines. Scand J Gastroenterol. (2006) 41:408–19. 10.1080/00365520500235334
    1. El Asmar R, Panigrahi P, Bamford P, Berti I, Not T, Coppa GV, et al. . Host-dependent zonulin secretion causes the impairment of the small intestine barrier function after bacterial exposure. Gastroenterology. (2002) 123:1607–15. 10.1053/gast.2002.36578

Source: PubMed

3
S'abonner