gut microbiota differs in composition and functionality ... · cyte lysate assay (qcl-1000; lonza...

11
Gut Microbiota Differs in Composition and Functionality Between Children With Type 1 Diabetes and MODY2 and Healthy Control Subjects: A Case-Control Study Diabetes Care 2018;41:23852395 | https://doi.org/10.2337/dc18-0253 OBJECTIVE Type 1 diabetes is associated with compositional differences in gut microbiota. To date, no microbiome studies have been performed in maturity-onset diabetes of the young 2 (MODY2), a monogenic cause of diabetes. Gut microbiota of type 1 diabetes, MODY2, and healthy control subjects was compared. RESEARCH DESIGN AND METHODS This was a case-control study in 15 children with type 1 diabetes, 15 children with MODY2, and 13 healthy children. Metabolic control and potential factors mod- ifying gut microbiota were controlled. Microbiome composition was determined by 16S rRNA pyrosequencing. RESULTS Compared with healthy control subjects, type 1 diabetes was associated with a signicantly lower microbiota diversity, a signicantly higher relative abundance of Bacteroides, Ruminococcus, Veillonella, Blautia, and Streptococcus genera, and a lower relative abundance of Bidobacterium, Roseburia, Faecalibacterium, and Lachnospira. Children with MODY2 showed a signicantly higher Prevotella abun- dance and a lower Ruminococcus and Bacteroides abundance. Proinammatory cytokines and lipopolysaccharides were increased in type 1 diabetes, and gut per- meability (determined by zonulin levels) was signicantly increased in type 1 diabetes and MODY2. The PICRUSt analysis found an increment of genes related to lipid and amino acid metabolism, ABC transport, lipopolysaccharide biosynthesis, arachidonic acid metabolism, antigen processing and presentation, and chemokine signaling pathways in type 1 diabetes. CONCLUSIONS Gut microbiota in type 1 diabetes differs at taxonomic and functional levels not only in comparison with healthy subjects but fundamentally with regard to a model of nonautoimmune diabetes. Future longitudinal studies should be aimed at eval- uating if the modulation of gut microbiota in patients with a high risk of type 1 diabetes could modify the natural history of this autoimmune disease. 1 Pediatric Endocrinology, Hospital Materno- Infantil, M ´ alaga, Spain 2 Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of M ´ alaga, Virgen de la Victoria Uni- versity Hospital, Universidad de M ´ alaga, M ´ alaga, Spain 3 Centro de Investigaci ´ on Biom´ edica en Red (CIBER) de Fisiopatolog´ ıa de la Obesidad y Nutrici´ on, In- stituto Salud Carlos III, Madrid, Spain Corresponding author: Jos´ e Carlos Fern ´ andez- Garc´ ıa, [email protected]. Received 5 February 2018 and accepted 26 August 2018. This article contains Supplementary Data online at http://care.diabetesjournals.org/lookup/suppl/ doi:10.2337/dc18-0253/-/DC1. I.L.-G. and L.S.-A. contributed equally to this work. © 2018 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for prot, and the work is not altered. More infor- mation is available at http://www.diabetesjournals .org/content/license. Isabel Leiva-Gea, 1 Lidia S´ anchez-Alcoholado, 2 Beatriz Mart´ ın-Tejedor, 1 Daniel Castellano-Castillo, 2,3 Isabel Moreno-Indias, 2,3 Antonio Urda-Cardona, 1 Francisco J. Tinahones, 2,3 Jos´ e Carlos Fern ´ andez-Garc´ ıa, 2,3 and Mar´ ıa Isabel Queipo-Ortu~ no 2,3 Diabetes Care Volume 41, November 2018 2385 PATHOPHYSIOLOGY/COMPLICATIONS

Upload: others

Post on 20-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

Gut Microbiota Differs inComposition and FunctionalityBetween Children With Type 1Diabetes and MODY2 and HealthyControl Subjects: A Case-ControlStudyDiabetes Care 2018;41:2385–2395 | https://doi.org/10.2337/dc18-0253

OBJECTIVE

Type 1 diabetes is associated with compositional differences in gut microbiota. Todate, no microbiome studies have been performed in maturity-onset diabetesof the young 2 (MODY2), a monogenic cause of diabetes. Gut microbiota oftype 1 diabetes, MODY2, and healthy control subjects was compared.

RESEARCH DESIGN AND METHODS

This was a case-control study in 15 children with type 1 diabetes, 15 children withMODY2, and 13 healthy children. Metabolic control and potential factors mod-ifying gut microbiota were controlled. Microbiome composition was determinedby 16S rRNA pyrosequencing.

RESULTS

Compared with healthy control subjects, type 1 diabetes was associated with asignificantly lowermicrobiota diversity, a significantly higher relative abundance ofBacteroides, Ruminococcus, Veillonella, Blautia, and Streptococcus genera, and alower relative abundance of Bifidobacterium, Roseburia, Faecalibacterium, andLachnospira. Children with MODY2 showed a significantly higher Prevotella abun-dance and a lower Ruminococcus and Bacteroides abundance. Proinflammatorycytokines and lipopolysaccharides were increased in type 1 diabetes, and gut per-meability (determined by zonulin levels) was significantly increased in type 1diabetes andMODY2. The PICRUSt analysis found an increment of genes related tolipid and amino acid metabolism, ABC transport, lipopolysaccharide biosynthesis,arachidonic acidmetabolism, antigen processing and presentation, and chemokinesignaling pathways in type 1 diabetes.

CONCLUSIONS

Gut microbiota in type 1 diabetes differs at taxonomic and functional levels not onlyin comparison with healthy subjects but fundamentally with regard to a model ofnonautoimmune diabetes. Future longitudinal studies should be aimed at eval-uating if the modulation of gut microbiota in patients with a high risk of type 1diabetes could modify the natural history of this autoimmune disease.

1Pediatric Endocrinology, Hospital Materno-Infantil, Malaga, Spain2ClinicalManagement Unit of Endocrinology andNutrition, Laboratory of the Biomedical ResearchInstitute of Malaga, Virgen de la Victoria Uni-versityHospital,UniversidaddeMalaga,Malaga,Spain3Centro de Investigacion Biomedica en Red (CIBER)de Fisiopatologıa de la Obesidad y Nutricion, In-stituto Salud Carlos III, Madrid, Spain

Corresponding author: Jose Carlos Fernandez-Garcıa, [email protected].

Received 5 February 2018 and accepted 26August 2018.

This article contains Supplementary Data onlineat http://care.diabetesjournals.org/lookup/suppl/doi:10.2337/dc18-0253/-/DC1.

I.L.-G. and L.S.-A. contributed equally to thiswork.

© 2018 by the American Diabetes Association.Readers may use this article as long as the workis properly cited, the use is educational and notfor profit, and the work is not altered. More infor-mation is available at http://www.diabetesjournals.org/content/license.

Isabel Leiva-Gea,1

Lidia Sanchez-Alcoholado,2

Beatriz Martın-Tejedor,1

Daniel Castellano-Castillo,2,3

Isabel Moreno-Indias,2,3

Antonio Urda-Cardona,1

Francisco J. Tinahones,2,3

Jose Carlos Fernandez-Garcıa,2,3 andMarıa Isabel Queipo-Ortu~no2,3

Diabetes Care Volume 41, November 2018 2385

PATH

OPHYSIO

LOGY/CO

MPLIC

ATIO

NS

Page 2: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

Type 1 diabetes is amultifactorial immune-mediated disease characterized by theprogressive loss of insulin-producing b-cells in the islets of Langerhans in thepancreas. The causes that lead to theappearance of type 1 diabetes have notyet been fully identified, with geneticfactors playing a major role; however,environmental factors are also closelylinked, such as birth delivery mode (1),diet in early life (cow milk proteins orgluten-containing cereals) (2), and wide-spread usage of antibiotics (3), all fac-tors closely related to gut microbiota.Recent studies have associated the

microbiome with the development oftype 1 diabetes; animal studies havedemonstrated a close link between in-testinal microbiota and type 1 diabetesin BioBreeding diabetes-prone rats (4)and in nonobese diabetic mice (5). Also,in children with type 1 diabetes, auto-immune positivity has been related tochanges in microbiota composition (6,7).In a previous study, we found that in

comparison with healthy control sub-jects, children with type 1 diabetes pre-sented large significant differences inthe relative abundance of predominantphyla, families, and genera (8). Poten-tially, the altered microbiota profile intype 1 diabetes may be associated withalterations in the gut immune system,such as increased gut permeability (9).Recent studies have shown that com-mensal bacteria are crucial for the ma-turing and functioning of the mucosalimmune system. Moreover, an impairedintegrity of the intestinal barrier withan increase in permeability has beendescribed in both animal models andhuman type 1 diabetes studies (10).Therefore, given that intestinal microbesmay affect intestinal permeability, intes-tinal ecology could also play a crucial rolein the development of type 1 diabetes(11). On the other hand, zonulin, a phys-iological modulator of intercellular tightjunctions, increases gut permeability andmacromolecule absorption, and previousstudies claim a role for zonulin as a capitalregulator of intestinal barrier function inthe genesis of metabolic disorders (12).Maturity-onset diabetes of the young

(MODY) is a genetic form of diabetes thataccounts for 1–2% of all diabetes casesin Europe and is associated with specificloss-of-function mutations with charac-teristic phenotypes (13). The most com-mon presentation of MODY is MODY2,

caused by a heterozygous inactivatingmutation in the glucokinase (GCK) gene(14). To date, no studies have evaluatedthe gut microbiome structure in MODY2patients. Nevertheless,MODY2 is a highlyattractive model to assess the relation ofgut microbiota with type 1 diabetes, asMODY2 is not normally associated withobesity, a glycemic control similar totype 1 diabetes can be achieved, and,importantly, its cause is not of autoim-mune origin (15).

We hypothesize that if the fecal micro-biota in type 1 diabetes differs from thatof MODY2, the gut microbiota profilecould constitute a novel associated riskfactor for the type 1 diabetes autoim-mune process. On the contrary, if thefecal microbiota were similar and differ-ent from the microbiota of healthy con-trol subjects, this would indicate thatdifferences in intestinal microbiota couldbe attributed to hyperglycemia per se.

Therefore, the aim of this case-controlstudy is to evaluate the gut microbiotaprofile, functional capacity, low-gradeinflammation, and gut permeability be-tween patients with type 1 diabetes andMODY2 and healthy control subjects.

RESEARCH DESIGN AND METHODS

This study was a case-control study, in-cluding 15 children with type 1 diabetes,15 children with MODY2, and 13 healthycontrol children, all under 18 years old,of Caucasian origin and with the samegeographical location.

Type 1 diabetes was diagnosed accord-ing to the criteria of the American Di-abetes Association (16) and the positivityof at least two persistent, confirmed anti-islet autoantibodies (anti-insulin autoan-tibodies, GAD autoantibodies, or tyrosinephosphatase autoantibodies). MODY2children were diagnosed by suggestiveclinical history, negative anti-islet auto-antibodies, and positive genetic testing.Healthy control subjects were childrenwith negative anti-islet autoantibodies,matched to children with type 1 diabetesand MODY2 for age, sex, race, BMI, modeof delivery, and duration of breastfeed-ing. In addition, patients with type 1diabetes and MODY2 were controlledby HbA1c levels. Patients with type 1diabetes were undergoing treatmentwith multiple doses of insulin, whereasMODY2 patients were drug naive. Exclu-sion criteria to participate in this studyincluded acute or chronic inflammatory

diseases or infectious diseases or un-dergoing treatment with antibiotics, pre-biotics, or probiotics or any other medicaltreatment that could potentially influ-ence intestinal microbiota 3 months be-fore inclusion.

The parents of all the participantscompleted a structured interview to ob-tain health status, lifestyle aspects, anddietary habits. Patients with type 1 di-abetes and MODY2 were instructed tofollow a standard diabetic diet, contain-ing 40–50% of calories from carbohy-drates, 20–30% from fat, and 20% fromprotein. Dietary intake patterns weredetermined from a food frequency ques-tionnaire.

The written informed consents of thechildren’s guardians or parents wereobtained. The sampling and experimen-tal processes were performed with theapproval of the local Ethics Committeeof the Regional Hospital of Malaga.

Laboratory MeasurementsSerum glucose, cholesterol, triglycerides,and interleukin-1b (IL-1b) and IL-10 cyto-kines were measured by ELISA as previ-ously described (17). Concentrationsof IL-6,IL-13, and tumor necrosis factor (TNF)-awerequantifiedbyELISAassay kits (ThermoFisher Scientific) in serum samples ac-cording to the instructions of the man-ufacturer. The detection limits were asfollows: 7.8–500 pg/mL for IL-6, 1.6–100pg/mL for IL-13, and 15.6–1,000 pg/mLfor TNF-a.

DNAExtraction, Pyrosequencingof 16SrRNA Sequences, and BioinformaticAnalysisStudy participants collected their fecalsamples in a sterile and hermeticallysealed receptacle provided by the re-search team. Fecal samples were col-lected in the morning of the day ofsample receipt and were immediatelyrefrigerated in household freezers andtransported to the laboratory during thefollowing 4 h. Frozen fecal samples weretransported with ice to avoid importantchanges of temperature that might causebacterial DNA degradation and weresubsequently stored at 280°C in thelaboratory until analysis. No DNA stabi-lizers were added to the fecal samples.

DNA was extracted from the fecalsamples using the QIAamp DNA StoolMini Kit (Qiagen, Hilden, Germany) ac-cording to the protocol of the manufac-turer. Amplification of genomic DNA was

2386 Gut Microbiota in Type 1 Diabetes and MODY2 Diabetes Care Volume 41, November 2018

Page 3: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

performed using bar-coded primers thattargeted the V2–V3 regions of the bac-terial 16S rRNA gene. Amplification, se-quencing,andbasicanalysiswereperformedusing a GS Junior 454 platform according tothe protocols of the manufacturer and aTitanium chemistry apparatus (Roche Ap-plied Science, Indianapolis, IN). The 454pyrosequencing data sets were analyzed byQuantitative Insights intoMicrobial Ecology(QIIME) 1.8.0 software as previously de-scribed (17). PICRUSt analysis was used topredict metagenome function by pickingoperational taxonomic units (OTUs) againstthe Greengenes database as previouslydescribed (17,18). The statistical analysiswas performed in R 3.3.3. P values werecorrected for multiple comparisons usingthe Benjamini-Hochberg method (17).

Intestinal PermeabilityThe plasma level of zonulin was deter-mined by ELISA using commercial kits (Im-munodiagnostik AG, Bensheim, Germany).The detection limit was 0.22 ng/mL.

Limulus Amebocyte Lysate AssaysSerum concentrations of lipopolysacchar-ides (LPS) were measured by endotoxinassay, based on a Limulus amebocyte ex-tract with a chromogenic Limulus amebo-cyte lysate assay (QCL-1000; Lonza GroupLtd.) according to the instructions of themanufacturer. The sensitivity limit for theassay was 0.02 endotoxin units (EU)/mL.

Statistical AnalysisGiven the exploratory nature of this study(no previous studies evaluating the differ-ences in gut microbiota between type 1diabetes, MODY2, and healthy controlsubjects), a priori sample size estimationwas not performed.The relative abundances of each OTU

(taxa) were compared by a Wilcoxonsigned rank test with a continuity cor-rection using the Explicet software pack-age specifically addressed to analyzemicrobiome data. All the resulting Pvalues were then adjusted for multiplecomparisons via the Benjamini-Hochbergfalse discovery rate (FDR) correction(FDR-corrected P value of ,0.05). a- andb-diversities were achieved by QIIME,a-diversity using a nonparametric Stu-dent t test using a default number ofMonte Carlo permutations of 999, andb-diversity with the analysis of similaritiesstatistical method with 99 permutations.Differences in the clinical characteristicsbetween two groups were analyzed using

Mann-Whitney U test, and differencesamong the three groups were ana-lyzed using the Kruskal-Wallis test withBonferroni post hoc test. The Spearmancorrelation coefficient was calculated toestimate the linear correlations betweenvariables. A multiple linear regressionanalysis was performed to identify whichbacteria taxa were independent predic-tors for serum zonulin, LPS, inflammatorymediators, andHbA1c levels in each studygroup. Values were considered to bestatistically significant when P # 0.05.

RESULTS

Diet and Anthropometric andBiochemical MeasurementsAll study participants had a similar physicalactivity and dietary profile. No significantdifferences in the consumption patternsof wheat, rice, vegetables, fish, or meatwere found between study groups.

The main clinical and biochemical char-acteristics of the study groups are shownin Table 1. As expected, glucose and HbA1clevels were significantly higher in chil-dren with type 1 diabetes and MODY2 (nodifferences between them) when com-pared with healthy children. No othersignificant differences were observed, in-cluding breastfeeding time or mode ofdelivery. However, children with type 1diabetes had significantly higher levels ofIL-1b, IL-6, TNF-a, and LPS and signifi-cantly lower levels of IL-10 and IL-13 thanMODY2 and healthy control subjects.

Characterization of Fecal MicrobiotaPyrosequencingThe Chao index (community richness) ofeach group suggested a similar bacterialrichness in the fecal samples from thethree study groups (199.426 52.26 type1 diabetes, 195.08 6 41.62 MODY2,218.65 6 51.05 healthy control sub-jects; P . 0.05). Despite a significantlylower Shannon index (microbiota diver-sity) in type 1 diabetes (4.766 0.42) andMODY2 (4.786 0.47) in comparisonwithhealthy control subjects (5.16 6 0.33)(P , 0.05), no differences in a-diversitywere found between type 1 diabetesand MODY2 (P = 0.850).

Regarding the b-diversity of gut micro-biota, weighted UniFrac principal coor-dinates analysis (PCoA) showed thatchildren with type 1 diabetes had adifferent pattern of clustering whencompared with MODY2 and healthycontrol subjects (P = 0.03 and P = 0.02,

respectively) (Fig. 1A and B). Neverthe-less, analysis of similarities with permu-tations revealed no significant differencesbetween MODY2 and healthy controlsubjects (P = 0.27), as demonstrated bythe two principal component scores, whichaccounted for 14.50% and 26.34% oftotal variation (Fig. 1C).

Taxonomy-Based Comparisons ofFecalMicrobiota at thePhylum, Family,and Genus LevelThe dominant phyla of all groups wereBacteroidetes and Firmicutes, followedby Proteobacteria and Actinobacteria.Individuals with type 1 diabetes showeda significant increase in the abundanceof Bacteroidetes (64.69% type 1 diabe-tes, 39.85%MODY2, 49.85% healthy con-trol subjects; P , 0.001, FDR-adjustedP , 0.001) and a significant decrease inthe abundance of Firmicutes (19.60%type 1 diabetes, 31.15% MODY2, 29.69%healthy control subjects; P, 0.001, FDR-adjusted P , 0.001) and Actinobacteria(1.02% type 1 diabetes, 2.27% MODY2,2.82% healthy control subjects; P = 0.003,FDR-adjusted P = 0.007) when comparedwith MODY2 and healthy control subjects.The frequency of Bacteroidetes was sig-nificantly lower in MODY2 than in healthycontrol subjects (FDR-adjusted P, 0.001).Proteobacteria was significantly lower intype 1 diabetes when compared withhealthy control subjects (1.68% type 1diabetes, 3.06% healthy control subjects;P, 0.001, FDR-adjusted P, 0.001), butnot regarding MODY2 (1.68% type 1 di-abetes, 1.80% MODY2; P = 0.699, FDR-adjusted P . 0.05). The remainder of thebacterial population belonged to five otherphyla that had a relative abundance,1%(Supplementary Fig. 1 and Supplemen-tary Data). In addition, the Firmicutes-to-Bacteroidetes ratio was significantlylower in type 1 diabetes than in MODY2and healthy control subjects (0.30% type1 diabetes, 0.76% MODY2, 0.58% healthycontrol subjects; P, 0.001, FDR-adjustedP , 0.001).

A total of 16 families were identifiedamong the fecal samples analyzed.Within the Bacteroidetes, two differ-ent families were significantly higher intype 1 diabetes when compared withMODY2 and healthy control subjects:Bacteroidaceae (71.48% type 1 diabetes,55.43% MODY2, 59.62% healthy controlsubjects; P , 0.001, FDR-adjusted P ,0.001) and Rikenellaceae (15.26% type 1

care.diabetesjournals.org Leiva-Gea and Associates 2387

Page 4: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

diabetes, 7.01%MODY2, 11.19% healthycontrol subjects; P, 0.001, FDR-adjustedP, 0.001). Prevotellaceae (2.65% type 1diabetes, 15.41% MODY2, 1.25% healthycontrol subjects; P, 0.001, FDR-adjustedP = 0.003) was significantly higher in type1 diabetes than in healthy control sub-jects, and Prevotellaceae was significantlyhigher in MODY2 than in type 1 diabetes(P , 0.001, FDR-adjusted P , 0.001). Inthe Firmicutes, another three familieswere significantly higher in type 1 di-abetes compared with MODY2 andhealthy control subjects: Ruminococcaceae(38.19% type 1 diabetes, 25.74% MODY2,30.84% healthy control subjects; P ,0.001, FDR-adjusted P , 0.001),Veillonellaceae (31.94% type 1 diabetes,20.33% MODY2, 18.03% healthy controlsubjects; P , 0.001, FDR-adjusted P =0.003), and Streptococcaceae (1.93% type1 diabetes, 0.96%MODY2, 0.56% healthycontrol subjects; P, 0.001, FDR-adjustedP = 0.004). No significant differences atthe Firmicutes family level were foundbetween MODY2 and healthy controlsubjects except in the abundance ofRuminococcaceae (P,0.001, FDR-adjustedP , 0.001). In healthy children, onlyLachnospiraceae (22.1% type 1 diabetes,27.95% MODY2, 42.0% healthy control

subjects; P = 0.002, FDR-adjusted P =0.015) was significantly higher in compar-ison with type 1 diabetes and MODY2. InActinobacteria, a significant enrichmentof Bifidobacteriaceae (2.71% type 1 di-abetes, 4.50% MODY2, 5.68% healthycontrol subjects; P = 0.004, FDR-adjusted P = 0.017) was found in healthycontrol subjects when compared withMODY2 and type 1 diabetes. Finally, forthe Proteobacteria families, a significantincrease of Enterobacteriaceae (25.20%type1diabetes, 15.03%MODY2, 13.04%healthy control subjects; P = 0.006, FDR-adjusted P = 0.03) was found in type 1diabetes when compared with MODY2and healthy control subjects, but nosignificant differences were found forAlcaligenaceae (62.18% type 1 diabetes,49.03% MODY2, 57.07% healthy controlsubjects; P = 0.019, FDR-adjusted P =0.267) (Supplementary Fig. 2 and Supple-mentary Data).

Twelve genera were differentiallyabundant between study groups. Forthe Bacteroidetes genera, the type 1diabetes group was significantly en-riched with sequences attributed to thegenus Bacteroides (72.21% type 1 di-abetes, 52.41%MODY2, 58.45% healthycontrol subjects; P , 0.001, FDR-

adjusted P , 0.001). Prevotella wassignificantly increased in MODY2 andtype 1 diabetes when compared withhealthy control subjects (1.95% type 1diabetes, 8.32% MODY2, 1.42% healthycontrol subjects; P , 0.001, FDR-adjusted P = 0.005). Regarding Firmi-cutes, the relative abundances of fourgenera were significantly higher in type1 diabetes than in MODY2 and healthycontrol subjects: Ruminococcus (17.19%type 1 diabetes, 5.74% MODY2, 8.85%healthy control subjects; P , 0.001, FDR-adjusted P = 0.002), Blautia (15.50%type 1 diabetes, 5.73% MODY2, 3.74%healthy control subjects; P, 0.001, FDR-adjusted P = 0.003), Veillonella (21.59%type 1 diabetes, 12.33% MODY2, 7.20%healthy control subjects; P, 0.001, FDR-adjusted P = 0.006), and Streptococcus(4.86% type 1 diabetes, 2.64% MODY2,1.47% healthy control subjects; P = 0.003,FDR-adjusted P = 0.028). In addition, fourgenera were significantly lower in type 1diabetes andMODY2 than in healthy con-trol subjects: Lachnospira (5.34% type 1diabetes, 7.15% MODY2, 15.25% healthycontrol subjects; P, 0.001, FDR-adjustedP = 0.012), Roseburia (1.35% type 1 dia-betes, 4.16%MODY2, 6.99% healthy con-trol subjects; P , 0.001, FDR-adjusted

Table 1—Baseline anthropometric and biochemical variables

Healthy controlsubjects (n = 13)

Type 1 diabetes(n = 15)

MODY2(n = 15) P

Male/female, n 7/6 7/8 7/8

Vaginal delivery/cesarean section, n 8/5 10/5 10/5

Age (years) 12.25 6 2.92 12.56 6 3.59 13.06 6 3.20 0.654

BMI (kg/m2) 17.35 6 1.82 17.89 6 2.01 18.23 6 1.90 0.430

Age of onset of diabetes (years) 7.35 6 1.76 6.91 6 1.40 0.455

Duration of diabetes (years) 5.68 6 1.84 6.10 6 1.97 0.551

Breastfeeding time (months) 6.58 6 2.32 6.41 6 2.81 6.54 6 3.2 0.911

Birth weight (kg) 3.19 6 0.45 3.28 6 0.38 3.22 6 0.55 0.249

Weight (kg) 37.35 6 9.0 38.32 6 8.92 36.91 6 7.72 0.765

HbA1c (%) 4.47 6 0.21a 6.26 6 0.38b 6.11 6 0.33b 0.001

HbA1c (mmol/mol) 25.3 6 2.3a 44.9 6 4.2b 43.3 6 3.6b 0.001

Triglycerides (mg/dL) 52.67 6 9.43 53.50 6 10.15 53.88 6 9.88 0.843

Cholesterol (mg/dL) 153.88 6 14.64 153.62 6 16.87 154.5 6 17.9 0.920

IL-1b (pg/mL) 83.21 6 28.25b 119.41 6 27.12a 89.45 6 25.31b 0.004

IL-10 (pg/mL) 126.67 6 9.87b 81.03 6 9.97a 121.28 6 5.46b 0.001

IL-6 (pg/mL) 85.71 6 6.28b 109.89 6 6.50a 88.98 6 7.49b 0.001

TNF-a (pg/mL) 164.31 6 16.78b 373.46 6 90.65a 169.54 6 18.3b 0.001

IL-13 (pg/mL) 50.15 6 8.37b 22.83 6 5.47a 45.46 6 7.31b 0.001

LPS (EU/mL) 0.49 6 0.10b 1.10 6 0.40a 0.56 6 0.38b 0.001

Values are presented as means6 SD unless otherwise specified. P value was based on Kruskal-Wallis test. Mann-Whitney U test was used to compare theclinical characteristics between two groups. Different superscript letters (a,b) next to values in a row indicate that the means of the different groupsare significantly different (P , 0.05, Bonferroni post hoc test).

2388 Gut Microbiota in Type 1 Diabetes and MODY2 Diabetes Care Volume 41, November 2018

Page 5: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

P = 0.015), Anaerostipes (2.15% type 1diabetes, 2.97% MODY2, 5.79% healthycontrol subjects; P, 0.001, FDR-adjustedP = 0.023), and Faecalibacterium (4.21%type 1 diabetes, 8.08% MODY2, 13.26%healthy control subjects; P, 0.001, FDR-adjusted P = 0.004). In Actinobacteria, onlythe Bifidobacterium was significantly in-creased in healthy control subjects whencompared with type 1 diabetes (1.93%type 1 diabetes, 6.75% healthy controlsubjects; P , 0.001, FDR-adjusted P =0.017), but not regarding MODY2 (6.07%MODY2, 6.75% healthy control subjects;P = 0.503, FDR-adjusted P. 0.05). Finally,

in the Proteobacteria genera, Sutterellaand Enterobacter were significantly in-creased in type 1 diabetes when com-pared with MODY2 and healthy controlsubjects (61.30% type 1 diabetes, 49.32%MODY2, 57.07% healthy control subjects[P = 0.002, FDR-adjusted P = 0.027] and16.18% type 1 diabetes, 8.25% MODY2,6.99% healthy control subjects [P, 0.001,FDR-adjusted P = 0.003], respectively).No significant differences betweenstudy groups were found in the rela-tive abundance of Desulfovibrio, Hae-mophilus, and Bilophila (FDR-adjustedP . 0.05) (Supplementary Fig. 3 and

Supplementary Data). A cladogramshowing differences in fecal microbiotabetween study groups is shown inFig. 2.

Serum Zonulin Levels in Type 1Diabetes, MODY2, and HealthyControl SubjectsPatients with MODY2 showed higherserum zonulin levels (4.80 6 1.41 ng/mgprotein) when compared with patientswith type 1 diabetes (3.946 1.44 ng/mgprotein, P = 0.02) and healthy controlsubjects (3.216 1.24 ng/mg protein, P,0.001). Significant differences in serum

Figure 1—Clustering of fecal bacterial communities according to the different study groups by PCoA using weighted UniFrac distances. Each pointcorresponds to a community coded according to the child group. The percentage of variation explained by the plotted principal coordinates isindicated on the axes.A: Type 1 diabetes (red dots) vs. healthy control subjects (blue dots). B: Type 1 diabetes (blue dots) vs.MODY2 (red dots). C: MODY2(blue dots) vs. healthy control subjects (red dots). PC1, principal coordinate 1; PC2, principal coordinate 2.

care.diabetesjournals.org Leiva-Gea and Associates 2389

Page 6: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

zonulin levels were also found betweentype 1 diabetes and control subjects(P , 0.05).

The Relationship Between GutMicrobiota Composition, SerumZonulin, HbA1c, InflammatoryMediators, and LPSSeveral significant correlations betweenthe relative abundance of specific bac-teria at different taxa levels and serum

zonulin levels and HbA1c were foundin children with type 1 diabetes andMODY2, but not in healthy control sub-jects (Table 2). In addition, significantcorrelations between serum levels ofIL-1b, IL-6, TNF-a, IL-10, IL-13, and LPSand gut microbiota composition werefound in type 1 diabetes (SupplementaryTable 1).

The linear regression analysis includingall the bacterial groups showed that the

increase in Bacteroides (P = 0.002, b =0.995, r2=0.94)andVeillonella (P=0.021,b = 0.636, r2 = 0.93) and the decrease inFaecalibacterium (P = 0.041, b =20.671,r2 = 0.94) and Roseburia (P = 0.038,b =20.694, r2 = 0.93) in type 1 diabetesand the rise in Prevotella in MODY2 (P =0.002, b = 0.682, r2 = 0.90) were asso-ciatedwith the increment in serum zonulinlevels. On the other hand, the increasein the abundance of Blautia (P = 0.043,

Figure 2—Cladogram showing differentially abundant taxa of the fecalmicrobiota in type 1 diabetes,MODY2, and healthy controls. Linear discriminantanalysis effect size analysis (P , 0.05 for Kruskal-Wallis test) was used to validate the statistical significance and the effect size of the differentialabundances of taxa in the study groups. The diameter of each circle is proportional to its abundance.

2390 Gut Microbiota in Type 1 Diabetes and MODY2 Diabetes Care Volume 41, November 2018

Page 7: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

b = 0.469, r2 = 0.93) and the decrease inthe Firmicutes-to-Bacteroidetes ratio (P =0.001, b = 20.947, r2 = 0.91) in patientswith type 1 diabetes were associated withHbA1c levels, whereas in MODY2 patients,only the decrease in Ruminococcus (P =0.003, b = 20.877, r2 = 0.92) was asso-ciated with HbA1c levels. In addition, theincrease in the relative abundance ofBacteroides (P = 0.006, b = 0.991, r2 =0.95) and Veillonella (P = 0.012, b =0.825, r2 = 0.95) and the decrease inBifidobacterium (P = 0.039, b = 20.654,r2 = 0.94), Roseburia (P = 0.032,b = 20.675, r2 = 0.95), and Faecalibac-terium (P = 0.023, b =20.678, r2 = 0.94)in type 1 diabetes were associated withserum IL-1b levels. Finally, in type 1diabetes, the increase in the abundanceof Bacteroides (P = 0.007, b = 0.632, r2 =0.85) and the decrease in Roseburia (P =0.029, b = 20.518, r2 = 0.85) were asso-ciated with serum IL-6 and TNF-a levels,and the increase in Streptococcus (P =0.014, b = 0.616, r2 = 0.82) and thedecrease in Bifidobacterium (P = 0.009,b = 20.904, r2 = 0.82) were associatedwith serum IL-10 and IL-13 levels. Regard-ing the serum levels of LPS, only the in-crease in the abundance of Veillonella (P =0.006,b = 0.887, r2 = 0.83) was associatedwith the levels in type 1 diabetes.

Functional Differences in the GutMicrobiota of Study GroupsThe PICRUSt analysis indicated that genesfor energy (P = 0.015) and carbohydratemetabolism (P = 0.014) were significantlydepleted in type 1 diabetes in comparisonwith MODY2 and healthy control subjects.Nevertheless, lipid metabolism functions(P = 0.008), together with amino acidmetabolism functions (P = 0.013), wereoverrepresented in type 1 diabetes whencompared with the other groups. In

addition, pathways of lipid and carbohy-drate metabolism in type 1 diabetes had asignificant enrichment in the proportion ofgenes related to arachidonic acid andpropanoate metabolism (P = 0.002 andP = 0.003, respectively), in comparisonwith MODY2 and healthy subjects (Fig. 3).Metagenomic comparison between studygroups showed that gene families linkedto amino acid metabolism, such as aminoacid–relatedenzymes (P =0.010), arginineand proline metabolism (P = 0.006), andvaline, leucine, and isoleucine biosynthe-sis (P = 0.020), were significantly increasedin type 1 diabetes, whereas genes forglutationmetabolism (P = 0.005) were sig-nificantly depleted. Conversely, in MODY2and healthy control subjects, in compar-ison with type 1 diabetes, a significantincrease in genes related to glycolysis/gluconeogenesis (P=0.018),pentosephos-phate pathway (P = 0.015), and butanoatemetabolism (P = 0.023), as well as in en-ergy metabolism genes such as sulfur (P =0.014), nitrogen metabolism (P = 0.012),and oxidative phosphorylation (P = 0.018),was detected (Fig. 3).

Finally, when compared with MODY2and healthy control subjects, gut micro-biota from patients with type 1 diabeteswas significantly enriched with genes forantigen processing and presentation (P =0.010), chemokine signaling pathways(P = 0.001), LPS biosynthesis (P = 0.008),bacterial invasion of epithelial cells (P =0.017), and ABC transporters (P = 0.016)(Fig. 3).

CONCLUSIONS

In this study comparing the bacterial florain type 1 diabetes, MODY2, and healthycontrol subjects, we show that type 1diabetes is associated with different gutmicrobial composition and functionalprofiling, in comparison with MODY2

and healthy control subjects. Also, we re-port that gut permeability, determinedby serum zonulin levels, is significantly in-creased in MODY2 and type 1 diabeteswhen compared with healthy control sub-jects. Another key finding in this study is asignificantly lower diversity of the dom-inant bacterial community in type 1 di-abetes and MODY2 when compared withhealthy control subjects.

The higher loss of diversity in patientswith type 1 diabetes when comparedwith healthy control subjects might berelated to the autoimmune process. In aprevious study evaluating type 1 diabetesmarkers, a lower microbial diversity wasfound in fecal samples of children withat least two positive disease-associatedautoantibodies than in samples fromautoantibody-negative children matchedfor age, sex, early feeding history, andHLA genotyping (19). Also, in longitudinalstudies from birth with children at risk fortype 1 diabetes, a decrease of microbialdiversity occurred just before the appear-ance of anti-islet cell antibodies and sub-sequent onset of type 1 diabetes (20).

We demonstrate that there are cleardifferences in the gut microbiota profileof type 1 diabetes, MODY2, and healthycontrol subjects, given that in the OTU-based PCoA plot analysis, patients withtype 1 diabetes clustered separately whencompared with MODY2 patients andhealthy control subjects. Accordingly, chil-dren with type 1 diabetes showed a signi-ficant increase in the relative abundanceof Bacteroides, Ruminococcus, Veillonella,Blautia, Enterobacter, and Streptococcusgenera and a decrease in the relativeabundance of Bifidobacterium, Roseburia,Faecalibacterium, and Lachnospira, whencompared with MODY2 and healthy con-trol subjects. On the other hand, MODY2was related to a significant increase in

Table 2—Correlation between gut microbiota composition and serum zonulin and HbA1c levels in children with type 1diabetes and MODY2

Zonulin HbA1c

Type 1 diabetes MODY2 Type 1 diabetes MODY2

Ruminococcus 20.162 (P = 0.55) 20.540 (P = 0.036) Ruminococcus 20.362 (P = 0.18) 20.584 (P = 0.025)

Roseburia 20.320 (P = 0.031) 20.732 (P = 0.48) Firmicutes-to-Bacteroidetes ratio

20.561 (P = 0.029) 20.332 (P = 0.45)

Prevotella 0.560 (P = 0.37) 0.798 (P = 0.037) Blautia 0.559 (P = 0.038) 0.740 (P = 0.79)

Faecalibacterium 20.703 (P = 0.027) 20.547 (P = 0.49) Streptococcus 0.068 (P = 0.018) 0.441 (P = 0.12)

Bacteroides 0.739 (P = 0.004) 0.350 (P = 0.090)

Veillonella 0.570 (P = 0.033) 0.704 (P = 0.04)

Correlations are reported as Spearman r (r), and P values are given in parentheses. Statistical significance was set at a P value of ,0.05.

care.diabetesjournals.org Leiva-Gea and Associates 2391

Page 8: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

Prevotella abundance and a significantdecrease in Ruminococcus and Bacter-oides, when compared with type 1 di-abetes and healthy control subjects.Despite the great variability of intestinal

microbiota in type 1 diabetes (regardlessof ethnicity, age, and geography), mostpublished studies (including the presentone) identify Bacteroides (acetate- andpropionate-producing bacteria) as the

main genus leading to type 1 diabetes–associated dysbiosis (21,22).

Bacteroides, a Gram-negative bacte-rium, could contribute to chronic inflam-mation by the impairment of the barrier

Figure 3—Predicted functional composition of metagenomes based on 16S rRNA gene sequencing data of type 1 diabetes (T1DM), MODY2, and healthycontrol subjects. Heat map of differentially abundant Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways identified in the three study groups.The values of color in the heat map represent the normalized relative abundance of KEGG pathways.

2392 Gut Microbiota in Type 1 Diabetes and MODY2 Diabetes Care Volume 41, November 2018

Page 9: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

function of the epithelial cell layer (23).In our study, subjectswith type1diabetes,when compared with MODY2 andhealthy control subjects, presented sig-nificantly higher levels of LPS and proin-flammatory cytokines IL-1b, IL-6, andTNF-a along with a significant decreasein anti-inflamatory citokines IL-10 andIL-13. Moreover, the significant correla-tions found between gut microbiota andhost serum levels of LPS and cytokinesindicate that the significant depletion ofFaecalibacterium, Roseburia, and Bifidobac-terium, reported to exert anti-inflammatoryeffects and strengthen gut barrier function(through cytokine production modulationand butyrate production, respectively)(24,25), together with the significant in-crease of Veillonella (a lactate-utilizingand propionate-producing bacteria withproinflammatory capacity) and Bacter-oides, could raise paracellular permeabil-ity and low-grade inflammation in type 1diabetes (26,27). This situation could al-low luminal antigens to escape from thegut and promote islet-directed autoim-mune responses. Therefore, the lowerabundance of anti-inflammatory bacteriaunable to regulate epithelial integritycould be associated with the intestinalimmune activation in type 1 diabetes(28,29).On the other hand, we have found a

significant increase in the relative abun-dance of Prevotella in MODY2 whencompared with type 1 diabetes andhealthy control subjects. Given thatPrevotella is an important succinateproducer and mucin degrader bacteria,this could suggest a lack of mucin on theintestinal epithelial layer of MODY2subjects, disturbing the protection ofthe host mucosal surfaces. Moreover,Prevotella-produced succinate is a bac-terial metabolite that leads to inhibitionof hepatic glucose output and improvesglycemic control and energy metabolismthrough the activation of intestinal glu-coneogenesis (30). Accordingly, Spegelet al. (13) suggested that in MODY2,despite the shift in insulin secretion, met-abolic control remains intact probably dueto the existence of compensatory mech-anisms external to b-cells. Therefore, wesuggest that the significant increase inthe abundance of Prevotella found inMODY2 patients could be related to themaintenance of glycemic control.In addition, we have found in type 1

diabetes a significant positive correlation

between zonulin levels and the relativeabundance of Bacteroides and Veillonella,and a significant negative correlationwithFaecalibacterium and Roseburia, as wellas a significant positive correlation be-tween zonulin and Prevotella in patientswith MODY2. Zonulin regulates intestinalpermeability by modulating intercellulartight junctions. A possible mechanism isbased on bacterial antigens and micro-organism toxins that may be sensed bymolecules related to epithelial cell tightjunctions like zonulin, altering their ac-tivity and consequently increasing gutpermeability and bacterial translocation(21,31). However, zonulin levels alone donot show impaired epithelial integrity,and in accordance with other authors,the impaired epithelial integrity foundin the patients with type 1 diabetes mightbe caused by the binding of Veillonella (alactate-utilizing bacteria) to immaturecells in colonic crypts able to fermentglucose to lactate, which are pushed tothe luminal surface and form poor tightjunctions (32). Also, in type 1 diabetes,there is an increased intestinal permeabil-ity that may affect absorption of antigenscapable of attacking and damaging pan-creatic b-cells (33–35). Thus, the signif-icant increase in gut permeability maybe an important player in the develop-ment of type 1 diabetes. Moreover,some authors have stated that a leakygut could be involved in the develop-ment of type 1 diabetes complications,as high serum LPS activity has beenassociated with features of metabolicsyndrome, visceral fat mass, and theprogression of kidney disease in type 1diabetes (36–38).

On the other hand, we have shownthat the relative abundance of Blautiaand the Firmicutes-to-Bacteroidetes ra-tio were positively correlated with HbA1cin type 1 diabetes, and in MODY2, HbA1clevels were negatively correlated withthe abundance of Ruminococcus. Butyrate-producing intestinal bacteria such as Ru-minococcus and Blautia could play animportant role inbloodglucose regulationand lipid metabolism, as shown by fecaltransplantation studies (39). Some au-thors have reported that Blautia is pos-itively correlated with serum glucose,HbA1c levels, and the number of type 1diabetes autoantibodies, suggesting thatBlautiamight influence the developmentof type 1 diabetes through the regulationof T-cell differentiation (20,40).

Also, we report a significantly lowerFirmicutes-to-Bacteroidetes ratio in type1 diabetes and a negative correlation be-tween this ratio and HbA1c. This has beenpreviously demonstrated by our group(8) and by other authors who havereported a decline in Firmicutes andan increase in Bacteroidetes abundanceover time in the gut microbiome until thedevelopment of type 1 diabetes (41).

The PICRUSt analysis demonstratedthat several microbial functions weresignificantly over- or underrepresentedbetween study groups, due to importantdifferences in bacteria composition.Thus, when compared with healthy con-trol subjects and MODY2, gut microbiotain type 1 diabetes showed a depletedabundance of genes involved in meta-bolic pathways such as carbohydrate andenergy metabolism. Conversely, therewas an increase in genes related to lipidand amino acid metabolism, ABC trans-port, LPS biosynthesis, arachidonic acidmetabolism, antigen processing andpresentation, and chemokine signalingpathways related to inflammation andimmune response. The relative abun-dance of genes associated with a givenpathway may indicate an increased met-abolic capacity of the gut microbiota withregard to this pathway. Interestingly, ahigher level of arachidonic acid metab-olism (inflammatory intermediate) intype 1 diabetes gut microbiota mightbe the result of a growth of proinflam-matory pathobionts in the gut (42).

Our study has certain limitations butalso some important strengths. The lim-itations include the relatively small sam-ple size (mainly caused by the very lowprevalence ofMODY2 in children), whichmay not be enough for detecting differ-ences between low-abundancemicrobesthat may be of relevance or to assessoverall differences between type 1 dia-betes and MODY2. Another limitation isthe inherent nature of the study, a cross-sectional design, where only an associ-ation andnot a cause canbe inferred, andwhere potential changes of the gut mi-crobioma from a healthy state to a gutpattern potentially boosting autoimmu-nity in type 1 diabetes were not evaluated.Finally, although PICRUSt gives functionalinformation of potential importance, it is alimitation compared with shotgun meta-genomics analysis. On the other hand,the strengths of our study lie in thecareful design, the inclusion of patients

care.diabetesjournals.org Leiva-Gea and Associates 2393

Page 10: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

with MODY2, the well-matched cohorts(including age, mode of delivery, breast-feeding time, antibiotics use, BMI, andglycemic levels), and the next-generationsequencing of the microbiome.In conclusion, our data suggest that

gut microbiota in type 1 diabetes not onlydiffers at the taxonomic level regardingMODY2 and healthy subjects but also atthe functional level, involving differentmetabolic pathways.Type 1 diabetes was characterized by

a less diverse gutmicrobiota profile, withBacteroidetes dominating at the phylumlevel and an increased proportion ofproinflammatory bacteria. Therefore, thetype 1 diabetes gut microbiota profilewas associated with impaired epithelialintegrity, low-grade inflammation, andautoimmune response. On the other hand,gut microbiota in MODY2 was charac-terized by a dominance of succinate-producer and mucin-degrading bacteria,potentially modulating glucose metabo-lism in the intestine of the host and in-fluencing systemic energy homeostasis.Our results provide evidence of a dif-

ferent microbiota profile and function-ality in children with type 1 diabetes, notonly in comparison with healthy subjectsbut fundamentally with regard to amodel of nonautoimmune diabetes,suggesting a potential role of gut micro-biota in the autoimmune process in-volved in type 1 diabetes. Given thatmost studies to date have shown thatintestinal microbiota, rather than beinginvolved in the initiation of the diseaseprocess of type 1 diabetes, might be in-volved in the progression from b-cellautoimmunity to the clinical disease (43),future longitudinal studies should beaimed at evaluating if the modulation ofgut microbiota in patients with a high riskof type 1 diabetes could modify the nat-ural history of this autoimmune disease.

Acknowledgments. The research group be-longs to the Centro de Investigacion Biomedicaen Red (CIBER, CB06/03/0018) of the Institutode Salud Carlos III.Funding. This study was supported by theMinisterio de Educacion, Cultura y Deporte(FPU13/04211 to D.C.-C.), the Miguel ServetType I program from the Instituto de Salud CarlosIII (cofoundedby theFondoEuropeodeDesarrolloRegional) (CP16/00163 to I.M.-I. and CP13/00065to M.I.Q.-O.), and the Servicio Andaluz de Salud(research contract B-0033-2014 to J.C.F.-G.).Duality of Interest. No potential conflicts ofinterest relevant to this article were reported.

Author Contributions. I.L.-G. conceived thestudy, developed the experimental design, ac-quired and selected all samples used in this study,compiled the database, performed statisticalanalysis and data interpretation, and wrote themanuscript and provided critical revision. L.S.-A.performed laboratory assays and wrote the man-uscript. B.M.-T. acquired and selected all samplesused in this study, compiled the database, per-formed statistical analysis and data interpretation,and wrote the manuscript and provided criticalrevision. D.C.-C. performed laboratory assays,compiled the database, and performed statisticalanalysis anddata interpretation. I.M.-I. performedlaboratory assays, compiled the database, per-formed statistical analysis and data interpretation,and wrote the manuscript. A.U.C. acquired andselected all samples used in this study, compiledthe database, and performed statistical analysisand data interpretation. F.J.T. conceived the study,developed the experimental design, and wrote themanuscript and provided critical revision. J.C.F.-G.conceived the study, developed the experimentaldesign, compiled the database, performed statis-tical analysis anddata interpretation, andwrote themanuscript and provided critical revision.M.I.Q.-O.conceived the study, developed the experimentaldesign, acquired and selected all samples used inthis study, performed laboratory assays, compiledthe database, performed statistical analysis anddata interpretation, and wrote the manuscript andprovided critical revision. All authors read and ap-proved the final manuscript. J.C.F.-G. and M.I.Q.-O.are the guarantors of this work and, as such, had fullaccess to all the data in the study and take re-sponsibility for the integrity of the data and theaccuracy of the data analysis.

References1. Khashan AS, Kenny LC, Lundholm C, KearneyPM, Gong T, Almqvist C. Mode of obstetricaldelivery and type 1 diabetes: a sibling designstudy. Pediatrics 2014;134:e806–e8132. Rewers M, Ludvigsson J. Environmental riskfactors for type 1 diabetes. Lancet 2016;387:2340–23483. Hansen CH, Krych L, Nielsen DS, et al. Earlylife treatment with vancomycin propagatesAkkermansia muciniphila and reduces diabetesincidence in the NOD mouse. Diabetologia 2012;55:2285–22944. Roesch LF, Lorca GL, Casella G, et al. Culture-independent identification of gut bacteria cor-relatedwith the onset of diabetes in a rat model.ISME J 2009;3:536–5485. Kriegel MA, Sefik E, Hill JA, Wu HJ, Benoist C,Mathis D. Naturally transmitted segmented fil-amentous bacteria segregate with diabetes pro-tection in nonobese diabetic mice. Proc NatlAcad Sci U S A 2011;108:11548–115536. Vaarala O. Is the origin of type 1 diabetes inthe gut? Immunol Cell Biol 2012;90:271–2767. He C, Shan Y, Song W. Targeting gut micro-biota as a possible therapy for diabetes. Nutr Res2015;35:361–3678. MurriM, Leiva I, Gomez-Zumaquero JM, et al.Gut microbiota in children with type 1 diabetesdiffers from that in healthy children: a case-control study. BMC Med 2013;11:469. Vaarala O. Leaking gut in type 1 diabetes. CurrOpin Gastroenterol 2008;24:701–706

10. Bosi E, Molteni L, Radaelli MG, et al. In-creased intestinal permeability precedes clinicalonset of type 1 diabetes. Diabetologia 2006;49:2824–282711. Mathis D, Benoist C. The influence of themicrobiota on type-1 diabetes: on the thresholdof a leap forward in our understanding. ImmunolRev 2012;245:239–24912. Zak-Gołab A, Kocełak P, Aptekorz M, et al.Gut microbiota, microinflammation, metabolic pro-file, and zonulin concentration in obese and normalweight subjects. Int J Endocrinol 2013;2013:67410613. SpegelP, EkholmE,TuomiT,GroopL,MulderH, Filipsson K. Metabolite profiling reveals nor-mal metabolic control in carriers of mutations inthe glucokinase gene (MODY2). Diabetes 2013;62:653–66114. Nyunt O, Wu JY, McGown IN, et al. Inves-tigating maturity onset diabetes of the young.Clin Biochem Rev 2009;30:67–7415. Fajans SS, Bell GI. MODY: history, genetics,pathophysiology, and clinical decision making.Diabetes Care 2011;34:1878–188416. American Diabetes Association. Classificationand diagnosis of diabetes. Sec. 2. In Standards ofMedical Care in Diabetesd2017. Diabetes Care2017;40(Suppl. 1):S11–S2417. Sanchez-Alcoholado L, Castellano-Castillo D,Jordan-Martınez L, et al. Role of gutmicrobiota oncardio-metabolic parameters and immunity in cor-onary artery disease patients with and without type-2diabetes mellitus. Front Microbiol 2017;8:193618. Bhute S, Pande P, Shetty SA, et al. Molecularcharacterization and meta-analysis of gut mi-crobial communities illustrate enrichment ofPrevotella and Megasphaera in Indian subjects.Front Microbiol 2016;7:66019. de Goffau MC, Luopajarvi K, Knip M, et al.Fecal microbiota composition differs betweenchildren with b-cell autoimmunity and thosewithout. Diabetes 2013;62:1238–124420. Kostic AD, Gevers D, Siljander H, et al.;DIABIMMUNE Study Group. The dynamics of thehuman infant gut microbiome in developmentand in progression toward type 1 diabetes. CellHost Microbe 2015;17:260–27321. Mejıa-Leon ME, Petrosino JF, Ajami NJ,Domınguez-Bello MG, de la Barca AM. Fecal mi-crobiota imbalance in Mexican children withtype 1 diabetes. Sci Rep 2014;4:381422. Brown CT, Davis-Richardson AG, Giongo A,et al. Gut microbiome metagenomics analysissuggests a functional model for the develop-ment of autoimmunity for type 1 diabetes. PLoSOne 2011;6:e2579223. Tlaskalova-Hogenova H, Stepankova R,Kozakova H, et al. The role of gut microbiota(commensal bacteria) and the mucosal barrierin the pathogenesis of inflammatory and auto-immune diseases and cancer: contribution ofgerm-free and gnotobiotic animal models of hu-mandiseases. CellMol Immunol2011;8:110–12024. Sokol H, Pigneur B, Watterlot L, et al. Fae-calibacterium prausnitzii is an anti-inflammatorycommensal bacterium identified by gut micro-biota analysis of Crohn disease patients. Proc NatlAcad Sci U S A 2008;105:16731–1673625. Tamanai-Shacoori Z, Smida I, Bousarghin L,et al. Roseburia spp.: a marker of health? Fu-ture Microbiol 2017;12:157–17026. Bischoff SC, Barbara G, Buurman W, et al.Intestinal permeability–a new target for disease

2394 Gut Microbiota in Type 1 Diabetes and MODY2 Diabetes Care Volume 41, November 2018

Page 11: Gut Microbiota Differs in Composition and Functionality ... · cyte lysate assay (QCL-1000; Lonza Group Ltd.) according to the instructions of the manufacturer. The sensitivity limit

prevention and therapy. BMC Gastroenterol2014;14:18927. Burger-van PaassenN, Vincent A, Puiman PJ,et al. The regulation of intestinal mucin MUC2expression by short-chain fatty acids: implicationsfor epithelial protection. Biochem J 2009;420:211–21928. LiN,HatchM,Wasserfall CH, et al. Butyrate andtype 1 diabetes mellitus: can we fix the intestinalleak? J Pediatr Gastroenterol Nutr 2010;51:414–41729. Vaarala O. Gut microbiota and type 1 di-abetes. Rev Diabet Stud 2012;9:251–25930. DeVadderF,Kovatcheva-DatcharyP,ZitounC,Duchampt A, Backhed F, Mithieux G. Microbiota-produced succinate improves glucose homeostasisvia intestinal gluconeogenesis. Cell Metab 2016;24:151–15731. Vaarala O. Human intestinal microbiota andtype 1 diabetes. Curr Diab Rep 2013;13:601–60732. Lopez CA, Miller BM, Rivera-Chavez F, et al.Virulence factors enhance Citrobacter roden-tium expansion through aerobic respiration.Science 2016;353:1249–1253

33. Watts T, Berti I, Sapone A, et al. Role of theintestinal tight junction modulator zonulin in thepathogenesis of type I diabetes in BB diabetic-prone rats. Proc Natl Acad Sci U S A 2005;102:2916–292134. Gulden E, Wong FS, Wen L. The gut micro-biota and type 1 diabetes. Clin Immunol 2015;159:143–15335. Lee AS, Gibson DL, Zhang Y, Sham HP, VallanceBA, Dutz JP. Gut barrier disruption by an entericbacterial pathogen accelerates insulitis in NODmice. Diabetologia 2010;53:741–74836. Nymark M, Pussinen PJ, Tuomainen AM,ForsblomC, Groop PH, LehtoM; FinnDiane StudyGroup. Serum lipopolysaccharide activity is as-sociated with the progression of kidney diseasein Finnish patients with type 1 diabetes. Diabe-tes Care 2009;32:1689–169337. Lassenius MI, Pietilainen KH, Kaartinen K,et al.; FinnDiane StudyGroup. Bacterial endotoxinactivity in human serum is associated with dysli-pidemia, insulin resistance, obesity, and chronicinflammation. Diabetes Care 2011;34:1809–1815

38. Lassenius MI, Ahola AJ, Harjutsalo V,Forsblom C, Groop PH, Lehto M. Endotoxinsare associated with visceral fat mass in type 1diabetes. Sci Rep 2016;6:3888739. Puddu A, Sanguineti R, Montecucco F,Viviani GL. Evidence for the gut microbiotashort-chain fatty acids as key pathophysiologicalmolecules improving diabetes. Mediators In-flamm 2014;2014:16202140. Qi CJ, Zhang Q, Yu M, et al. Imbalance offecal microbiota at newly diagnosed type 1 di-abetes in Chinese children. Chin Med J (Engl)2016;129:1298–130441. Giongo A, Gano KA, Crabb DB, et al. Towarddefining the autoimmune microbiome for type1 diabetes. ISME J 2011;5:82–9142. Candela M, Biagi E, Soverini M, et al. Mod-ulation of gut microbiota dysbioses in type 2diabetic patients by macrobiotic Ma-Pi 2 diet.Br J Nutr 2016;116:80–9343. KnipM, Siljander H. The role of the intestinalmicrobiota in type 1 diabetes mellitus. Nat RevEndocrinol 2016;12:154–167

care.diabetesjournals.org Leiva-Gea and Associates 2395