Detection of Increased Plasma Interleukin-6 Levels and Prevalence of Prevotella copri and Bacteroides vulgatus in the Feces of Type 2 Diabetes Patients

Aline Zazeri Leite, Nathália de Campos Rodrigues, Marina Ignácio Gonzaga, João Carlos Cicogna Paiolo, Carolina Arantes de Souza, Nadine Aparecida Vicentini Stefanutto, Wellington Pine Omori, Daniel Guariz Pinheiro, João Luiz Brisotti, Euclides Matheucci Junior, Vânia Sammartino Mariano, Gislane Lelis Vilela de Oliveira, Aline Zazeri Leite, Nathália de Campos Rodrigues, Marina Ignácio Gonzaga, João Carlos Cicogna Paiolo, Carolina Arantes de Souza, Nadine Aparecida Vicentini Stefanutto, Wellington Pine Omori, Daniel Guariz Pinheiro, João Luiz Brisotti, Euclides Matheucci Junior, Vânia Sammartino Mariano, Gislane Lelis Vilela de Oliveira

Abstract

Intestinal dysbiosis and metabolic endotoxemia have been associated with metabolic disorders, such as obesity, insulin resistance, and type 2 diabetes (T2D). The main goal of the present study was to evaluate the intestinal dysbiosis in Brazilian T2D patients and correlate these data with inflammatory cytokines and lipopolysaccharides (LPS) plasma concentrations. This study was approved by the Ethics Committees from Barretos Cancer Hospital and all individuals signed the informed consent form. Stool samples were required for DNA extraction, and the V3/V4 regions of bacterial 16S were sequenced using an Illumina platform. Peripheral blood was used to quantify inflammatory cytokines and plasma LPS concentrations, by CBA flex and ELISA, respectively. Statistical analyses were performed using Mann-Whitney and Spearman's tests. Analysis of variance, diversity indexes, and analysis of alpha- and beta-diversity were conducted using an annotated Operational Taxonomic Unit table. This study included 20 patients and 22 controls. We observed significant differences (P < 0.01) in the microbiota composition (beta-diversity) between patients and controls, suggesting intestinal dysbiosis in Brazilian T2D patients. The prevalent species found in patients' feces were the Gram-negatives Prevotella copri, Bacteroides vulgatus, Bacteroides rodentium, and Bacteroides xylanisolvens. The proinflammatory interleukin-6 (IL-6) was significantly increased (P < 0.05) in patients' plasma and LPS levels were decreased. We find correlations between the proinflammatory interferon-gamma with Gram-negatives Bacteroides and Prevotella species, and a positive correlation between the LPS levels and P. copri reads. The P. copri and B. vulgatus species were associated with insulin resistance in previous studies. In this study, we suggested that the prevalence of Gram-negative species in the gut and the increased plasma IL-6 in patients could be linked to low-grade inflammation and insulin resistance. In conclusion, the P. copri and B. vulgatus species could represent an intestinal microbiota signature, associated with T2D development. Furthermore, the identification of these Gram-negative bacteria, and the detection of inflammatory markers, such as increased IL-6, could be used as diabetes predictive markers in overweight, obese and in genetically predisposed individuals to develop T2D.

Keywords: dietary habits; inflammatory cytokines; interleukin-6; intestinal microbiota; metabolic endotoxemia; type 2 diabetes.

Figures

Figure 1
Figure 1
Alpha and beta-diversity in gut microbiota from type 2 diabetes (T2D) patients. (A,B) Rarefaction curves comparing the species richness, Chao1, and observed Operational Taxonomic Unit (OTU) numbers. (C,D) PcoA plots with weighted and unweighted UniFrac metric with Bonferroni’s correction.
Figure 2
Figure 2
Relative abundances of bacterial taxa in the feces of T2D patients. Prevalent phyla (A), classes (B), orders (C), families (D), genera (E), and species (F). Bars represent the reads percentages found in metagenomic analyses.
Figure 3
Figure 3
Negative correlation between the Ruminococcaceae reads with the HbA1C percentages. Statistical analyses were performed using the Spearman’s test. Significance was set at P < 0.05.
Figure 4
Figure 4
Cytokine profile in type 2 diabetes patients and control subjects. Plasma concentrations of IL-4 (A), interleukin-6 (IL-6) (B), IL-10 (C), IL-17A (D), interferon-gamma (IFN-γ) (E), and tumor necrosis factor (TNF) (F). Statistical analyses were performed by Mann–Whitney test. Significance was set at P < 0.05.
Figure 5
Figure 5
Correlations between the proinflammatory cytokines with relative abundances of bacterial taxa. Correlations found among interferon-gamma (IFN-γ) and Firmicutes phylum (A), Clostridia class (B), Bacteroides genus (C), and Prevotella genus (D). Positive correlation found between IL-17A with Enterobacteriaceae family (E). Positive correlation found between interleukin-6 (IL-6) with Negativicutes class (F). Statistical analyses were performed by Spearman’s test. Significance was set at P < 0.05.
Figure 6
Figure 6
Lipopolysaccharide (LPS) concentrations and the correlations with relative abundances of bacterial taxa. Plasma levels of the LPS in type 2 diabetes patients and controls (A). Negative correlation found between the LPS with Proteobacteria phylum (B). Positive correlation found between the LPS with Prevotella genus (C). Statistical analyses were performed using the Mann–Whitney and the Spearman’s test. Significance was set at P < 0.05.

References

    1. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol (2011) 11:98–107.10.1038/nri2925
    1. International Diabetes Federation. International Diabetes Federation Atlas. 7th ed Brussels: Karakas Print; (2015).
    1. Tremaroli V, Bäckhed F. Functional interactions between the gut microbiota and host metabolism. Nature (2012) 489:242–9.10.1038/nature11552
    1. Van Olden C, Groen AK, Nieuwdorp M. Role of the intestinal microbiome in lipid and glucose metabolism in diabetes mellitus. Clin Ther (2015) 37:1172–7.10.1016/j.clinthera.2015.03.008
    1. Kau AL, Ahern PP, Griffin NW, Goodman AL, Goerdon JL. Human nutrition, the gut microbiome and the immune system. Nature (2011) 474:327–36.10.1038/nature10213
    1. Kamada N, Seo SU, Chen GY, Núñez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol (2013) 13:321–35.10.1038/nri3430
    1. Karczewski J, Poniedzialek B, Adamski Z, Rzymski P. The effects of the microbiota on the host immune system. Autoimmunity (2014) 47:494–504.10.3109/08916934.2014.938322
    1. Hooper LV, Macpherson AJ. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol (2010) 10:159–69.10.1038/nri2710
    1. Walsh CJ, Guinane CM, Hill C, Ross RP, O’Toole PW, Cotter PD. In silico identification of bacteriocin gene clusters in the gastrointestinal tract, based on the Human Microbiome Project’s reference genome database. BMC Microbiol (2015) 15:183.10.1186/s12866-015-0515-4
    1. Doré J, Blottière H. The influence of diet on the gut microbiota and its consequences for health. Curr Opin Biotechnol (2015) 32:195–9.10.1016/j.copbio.2015.01.002
    1. Littman DR, Pamer EG. Role of the commensal microbiota in normal and pathogenic host immune responses. Cell Host Microbe (2011) 10:311–23.10.1016/j.chom.2011.10.004
    1. Renz H, Brandtzaeg P, Hornef M. The impact of perinatal immune development on mucosal homeostasis and chronic inflammation. Nat Rev Immunol (2012) 12(1):9–23.10.1038/nri3112
    1. Allin KH, Nielsen T, Pedersen O. Mechanisms in endocrinology: gut microbiota in patients with type 2 diabetes mellitus. Eur J Endocrinol (2015) 172:R167–77.10.1530/EJE-14-0874
    1. Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature (2016) 535:75–84.10.1038/nature18848
    1. Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature (2016) 535:65–74.10.1038/nature18847
    1. Petersen C, Round JL. Defining dysbiosis and its influence on host immunity and disease. Cell Microbiol (2014) 16:1024–33.10.1111/cmi.12308
    1. Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermúdez-Humarán LG, et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Mol Med (2011) 3:559–72.10.1002/emmm.201100159
    1. Manco M, Putignani L, Bottazzo GF. Gut microbiota, lipopolysaccharides, and innate immunity in the pathogenesis of obesity and cardiovascular risk. Endocr Rev (2010) 31:817–44.10.1210/er.2009-0030
    1. Cani PD, Delzenne NM. The role of the gut microbiota in energy metabolism and metabolic disease. Curr Pharm Des (2009) 15:1546–58.10.2174/138161209788168164
    1. Chawla A, Nguyen KD, Goh YP. Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol (2011) 11:738–49.10.1038/nri3071
    1. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes (2008) 57:1470–81.10.2337/db07-1403
    1. Caricilli AM, Saad MJ. The role of gut microbiota on insulin resistance. Nutrients (2013) 5:829–51.10.3390/nu5030829
    1. Pickup JC, Mattock MB, Chusney GD, Burt D. NIDDM as a disease of the innate immune system: association of acute-phase reactants and interleukin-6 with metabolic syndrome X. Diabetologia (1997) 40:1286–92.10.1007/s001250050822
    1. Jayashree B, Bibin YS, Prabhu D, Shanthirani CS, Gokulakrishnan K, Lakshmi BS, et al. Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Mol Cell Biochem (2014) 388:203–10.10.1007/s11010-013-1911-4
    1. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature (2012) 490:55–60.10.1038/nature11450
    1. Everard A, Cani PD. Diabetes, obesity and gut microbiota. Best Pract Res Clin Gastroenterol (2013) 27:73–83.10.1016/j.bpg.2013.03.007
    1. Sommer F, Anderson JM, Bharti R, Raes J, Rosenstiel P. The resilience of the intestinal microbiota influences health and disease. Nat Rev Microbiol (2017).10.1038/nrmicro.2017.58
    1. Fay MP, Proschan MA. Wilcoxon-Mann-Whitney or t-test? On assumptions for hypothesis tests and multiple interpretations of decision rules. Stat Surv (2010) 4:1–39.10.1214/09-SS051
    1. Bonett DG, Wright TA. Sample size requirements for estimating Pearson, Kendall and Spearman correlations. Psychometrika (2000) 65:23–8.10.1007/BF02294183
    1. Masella AP, Bartram AK, Truszkowski JM, Brown DG, Neufeld JD. PANDAseq: paired-end assembler for Illumina sequences. BMC Bioinformatics (2012) 13:31.10.1186/1471-2105-13-31
    1. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J (2011) 17:10–2.10.14806/ej.17.1.200
    1. Schmieder R, Edwards R. Quality control and preprocessing of metagenomic datasets. Bioinformatics (2011) 27:863–4.10.1093/bioinformatics/btr026
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods (2010) 7:335–6.10.1038/nmeth.f.303
    1. De Souza RS, Okura VK, Armanhi JS, Jorrín B, Lozano N, da Silva MJ, et al. Unlocking the bacterial and fungal communities assemblages of sugarcane microbiome. Sci Rep (2016) 6:28774.10.1038/srep28774
    1. Edgar RC. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res (2004) 32:1792–7.10.1093/nar/gkh340
    1. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods (2013) 10:996–8.10.1038/nmeth.2604
    1. Cole JR, Chai B, Farris RJ, Wang Q, Kulam SA, McGarrell DM, et al. The Ribosomal Database Project (RDP-II): sequences and tools for high-throughput rRNA analysis. Nucleic Acids Res (2005) 33:D294–6.10.1093/nar/gki038
    1. Schloss PD, Westcott SL, Rvabin T, Hall JR, Hartmann M, Hollister EB, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol (2009) 75:7537–41.10.1128/AEM.01541-09
    1. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, et al. Diversity of the human intestinal microbial flora. Science (2005) 308:1635–8.10.1126/science.1110591
    1. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science (2009) 326:1694–7.10.1126/science.1177486
    1. Sommer F, Bäckhed F. The gut microbiota – masters of host development and physiology. Nat Rev Microbiol (2013) 11:227–38.10.1038/nrmicro2974
    1. Biedermann L, Rogler G. The intestinal microbiota: its role in health and disease. Eur J Pediatr (2015) 174:151–67.10.1007/s00431-014-2476-2
    1. Wu H, Tremaroli V, Bäckhed F. Linking microbiota to human diseases: a systems biology perspective. Trends Endocrinol Metab (2015) 26:758–70.10.1016/j.tem.2015.09.011
    1. Haro C, Montes-Borrego M, Rangel-Zúñiga OA, Alcalá-Díaz JF, Gómez-Delgado F, Pérez Martínez P, et al. Two healthy diets modulate gut microbial community improving insulin sensitivity in a human obese population. J Clin Endocrinol Metab (2016) 101:233–42.10.1210/jc.2015-3351
    1. Janssen AW, Kersten S. The role of gut microbiota in metabolic health. FASEB J (2015) 29:3111–23.10.1096/fj.14-269514
    1. Palau-Rodriguez M, Tulipani S, Queipo-Ortuño MI, Urpi-Sarda M, Tinahones FJ, Andres-Lacueva C. Metabolomic insights into the intricate gut microbial-host interaction in the development of obesity and type 2 diabetes. Front Microbiol (2016) 6:1151.10.3389/fmicb.2015.01151
    1. Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bavless AJ, et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe (2015) 17:662–71.10.1016/j.chom.2015.03.005
    1. Canabi RB, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol (2011) 17:1519–28.10.3748/wjg.v17.i12.1519
    1. Wexler HM. Bacteroides: the good, the bad, and the nitty-gritty. Clin Microbiol Rev (2007) 20:593–621.10.1128/CMR.00008-07
    1. Larsen JM. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology (2017) 151:363–74.10.1111/imm.12760
    1. Crelly SJ, McTernan PG, Kusminski CM, Fisher FM, Da Silva NF, Khanolkar M, et al. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab (2007) 292:E740–7.10.1152/ajpendo.00302.2006
    1. Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BA, et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature (2016) 535:376–81.10.1038/nature18646
    1. Larsen N, Vogensen FK, van den Berg FW, Nielsen DS, Andreasen AS, Pedersen BK, et al. Gut microbiota in human adults with type 2 differs from non-diabetic adults. PLoS One (2010) 5:e9085.10.1371/journal.pone.0009085
    1. La Reau AJ, Meier-Kolthoff JP, Suen G. Sequence-based analysis of the genus Ruminococcus resolves its phylogeny and reveals strong host association. Microb Genom (2016) 2:e000099.10.1099/mgen.0.000099
    1. Greer RL, Dong X, Moraes ACF, Zielke RA, Fernandes GR, Peremyslova E, et al. Akkermansia muciniphila mediates negative effects of IFN-γ on glucose metabolism. Nat Commun (2016) 7:13329.10.1038/ncomms13329
    1. Wong N, Fam BC, Cempako GR, Steinberg GR, Walder K, Kay TW, et al. Deficiency in interferon-gamma results in reduced body weight and better glucose tolerance in mice. Endocrinology (2011) 152:3690–9.10.1210/en.2011-0288
    1. Rocha VZ, Folco EJ, Sukhova G, Shimizu K, Gotsman I, Vernon AH, et al. Interferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive immunity in obesity. Circ Res (2008) 103:467–76.10.1161/CIRCRESAHA.108.177105
    1. O’Rourke RW, White AE, Metcalf MD, Winters BR, Diggs BS, Zhu X, et al. Systemic inflammation and insulin sensitivity in obese IFN-γ knockout mice. Metabolism (2012) 61:1152–61.10.1016/j.metabol.2012.01.018
    1. Moya-Pérez A, Neef A, Sanz Y. Bifidobacterium pseudocatenulatum CECT 7765 reduces obesity-associated inflammation by restoring the lymphocyte-macrophage balance and gut microbiota structure in high-fat diet-fed mice. PLoS One (2015) 10:e0126976.10.1371/journal.pone.0126976
    1. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, et al. Selective increases of bifidobacteria in gut microflora improve high fat-diet-induced diabetes in mice through a mechanism associated with endotoxemia. Diabetologia (2007) 50:2374–83.10.1007/s00125-007-0791-0

Source: PubMed

3
Abonneren