• No results found

Allergy development is associated with consumption of breastmilk with a reduced microbial richness in the first month of life

N/A
N/A
Protected

Academic year: 2021

Share "Allergy development is associated with consumption of breastmilk with a reduced microbial richness in the first month of life"

Copied!
33
0
0

Loading.... (view fulltext now)

Full text

(1)

Allergy development is associated with

consumption of breastmilk with a reduced

microbial richness in the first month of life

Majda Dzidic, Alex Mira, Alejandro Artacho, Thomas Abrahamsson, Maria Jenmalm and Maria Carmen Collado

The self-archived postprint version of this journal article is available at Linköping University Institutional Repository (DiVA):

http://urn.kb.se/resolve?urn=urn:nbn:se:liu:diva-162862

N.B.: When citing this work, cite the original publication.

Dzidic, M., Mira, A., Artacho, A., Abrahamsson, T., Jenmalm, M., Carmen Collado, M., (2019), Allergy development is associated with consumption of breastmilk with a reduced microbial richness in the first month of life, Pediatric Allergy and Immunology. https://doi.org/10.1111/pai.13176

Original publication available at: https://doi.org/10.1111/pai.13176 Copyright: Wiley (12 months) http://eu.wiley.com/WileyCDA/

(2)

Allergy development is associated with consumption of breastmilk

1

with a reduced microbial richness in the first month of life

2

3

Majda Dzidica,b,c, PhD, Alex Mirab, PhD, Alejandro Artachob, BSc, Thomas R.

4

Abrahamssond, MD, PhD, Maria C. JenmalmcY, PhD, M. Carmen Collado1Y*, PhD

5 6 7

Short title: Breastmilk microbiota and infant’s allergy development 8

9

Affiliations: 10

a. Institute of Agrochemistry and Food Technology (IATA-CSIC), Department of 11

Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain 12

b. Department of Health and Genomics, Center for Advanced Research in Public Health, 13

FISABIO, Valencia, Spain; and CIBER-ESP, Madrid; Spain 14

c. Department of Clinical and Experimental Medicine, Division of Autoimmunity and Immune 15

Regulation, Linköping University, Linköping, Sweden 16

d. Department of Clinical and Experimental Medicine and Department of Pediatrics, Linköping 17

University, Linköping, Sweden 18 19 20 21 Correspondence: 22

*To whom correspondence may be addressed: 23

Address: Institute of Agrochemistry and Food Technology (IATA-CSIC), Department of 24

Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain; 25

mcolam@iata.csic.es 26

YShared senior authors

27 28

Funding: Alex Mira: Spanish Ministry of Economy and Competitiveness (grant no.

BIO2015-29

68711-R). Maria C. Jenmalm: The Swedish Research Council (2016-01698); the Swedish Heart 30

and Lung Foundation (20140321 and 20170365); the Cancer and Allergy Foundation. M. 31

Carmen Collado: European Research Council (ERC-starting grant 639226). 32

33

Conflict of interest: Maria C. Jenmalm has received funding for a clinical trial and honoraria 34

for lectures from BioGaia AB, as well as consultant fees and travel support from 35

Nutricia/Danone. 36

(3)

ABSTRACT 38

Background: Early colonization with a diverse microbiota seems to play a crucial role for 39

appropriate immune maturation during childhood, and breastmilk microbiota is one important 40

source of microbes for the infant, transferred together with maternal IgA antibodies. We 41

previously observed that allergy development during childhood was associated with aberrant 42

IgA responses to the gut microbiota already at 1 month of age, when the IgA antibodies are 43

predominantly maternally derived in breastfed infants. 44

Objective: To determine the microbial composition and IgA-coated bacteria in breastmilk in 45

relation to allergy development in children participating in an intervention trial with pre- and 46

postnatal Lactobacillus reuteri supplementation. 47

Methods: A combination of flow cytometric cell sorting and 16S rRNA gene sequencing was 48

used to characterize the bacterial recognition patterns by IgA in breastmilk samples collected 49

one-month post partum from 40 mothers whose children did or did not develop allergic and 50

asthmatic symptoms during the first 7 years of age. 51

Results: The milk fed to children developing allergic manifestations had significantly lower 52

bacterial richness, when compared to the milk given to children that remained healthy. Probiotic 53

treatment influenced the breastmilk microbiota composition. However, the proportions of IgA-54

coated bacteria, the total bacterial load and the patterns of IgA-coating were similar in 55

breastmilk between mothers of healthy children and those developing allergies. 56

Conclusion: Consumption of breastmilk with a reduced microbial richness in the first month 57

of life may play an important role in allergy development during childhood. 58

59 60 61

Keywords: Allergy, Breastmilk, IgA, microbiota, mother-infant transfer. 62

63 64 65

(4)

66

INTRODUCTION 67

Human breastmilk is considered to be an optimal nutritional source for the immature immune 68

system of the infant.1 Among the bioactive factors, breastmilk contains immunoglobulins, that

69

can be transferred to the offspring through breastfeeding.2 Although all immunoglobulin

70

isotypes can be encountered in breastmilk, secretory IgA (SIgA) is the dominating isotype and 71

considered most important due to its anti-inflammatory properties and important role in 72

defending the mucous membranes, thus regulating the binding and invasion of commensals and 73

pathogenic microorganisms.3 This passive immunization trough breastfeeding is crucial as

74

early production of secretory IgA in newborns is limited.3

75 76

The increasing prevalence of allergic diseases in affluent societies is hypothesized to be caused 77

by reduced intensity and diversity of microbial stimulation.4,5 In support of this theory, the gut

78

microbiota differs in composition and diversity during the first months of life in children who 79

later do or do not develop allergic disease.6–8 Breastmilk hosts a diverse array of microbiota

80

and potential probiotic bacteria, transferred together with maternal IgA antibodies9, likely

81

influencing the infant's developing mucosal immune system. 82

83

We have previously observed that allergy development during childhood was associated with 84

divergent patterns of IgA recognized bacteria in the gut already at 1 month of age, when the 85

IgA antibodies are predominantly maternally derived in breast-fed children.10 However, the

86

identities of the bacterial taxa targeted by IgA in the breastmilk and what role they may play in 87

immune and allergy development are unknown. In this study, we aimed to characterize the 88

composition and IgA-coating pattern of the breastmilk microbiota from mothers whose children 89

developed allergic symptoms during early childhood or stayed healthy. 90

(5)

91

METHODS 92

For detailed methods, experimental protocols and statistical analyses, see the Methods section 93

in this article’s Online Repository. 94

Study design 95

The subjects included in this study were part of a larger randomized double-blind trial in 96

Sweden, recruiting participants between 2001 and 2003, where the potential allergy preventive 97

effect of probiotic Lactobacillus reuteri ATCC 55730 in the infants with family history of 98

allergic disease was evaluated.11,12 The mothers were supplemented with L. reuteri during

99

pregnancy from postmenstrual week 36+0 to delivery and the infants continued with the same 100

treatment from day 1-2 of life until 12 months of age. Among the 184 mothers of children that 101

completed the 7-year follow up in the original study, breastmilk samples, at one month post 102

partum, from 24 mothers whose children did not and from 26 mothers whose children did 103

develop allergic disease during early childhood, were randomly selected for flow cytometry 104

based-sorting of IgA-coated bacteria in the current study. From these flow cytometry-sorted 105

samples, subsequent 16S rRNA gene characterization was performed on the IgA-coated and 106

IgA-free fractions of breastmilk bacteria from 20 mothers whose children did not and from 20 107

mothers whose children did develop allergic manifestations as well as from total, non-sorted 108

breastmilk samples from the same mothers. Selection of the samples used for 16S rRNA 109

sequencing in this study was based on the sample availability and required sample volume for 110

Illumina sequencing, and a clear allergy diagnosis (based on proven symptoms to allergy 111

provocation) of the child. Allergic disease included eczema (n=12), gastrointestinal allergy 112

(n=1), asthma (n=10), allergic rhinoconjunctitivis (n=14) and allergic urticaria (n=3). The 113

(6)

criteria of these diagnoses are described in detail in 11,12 and in the supplementary information

114

of this manuscript. The majority of the children included in the current study were exclusively 115

breastfed during the first month of life (93%). 116

There were no differences regarding potential confounders, such as sex, mode of delivery, birth 117

order, maternal atopy, breastfeeding, antibiotics, and probiotic supplementation, between the 118

infants who did or did not have allergic manifestations (Table I). 119

120

Total IgA levels were measured by ELISA in a study by Böttcher et al.13 The studies were

121

approved by the Regional Ethics Committee for Human Research in Linköping, Sweden (Dnr 122

99323, M122-31 and M171-07, respectively). 123

124

Sample preparation and flow cytometry-based sorting 125

The breastmilk samples were stained with goat anti-mouse IgA labelled with fluorescein 126

isothiocyanate (FITC), used as an isotype control corresponding to unspecific binding (Sigma; 127

reference SLBD9273), or with goat anti-human IgA labelled with FITC (Life Technologies; 128

reference A18782), according to the manufacturer’s instructions. The sorting of the bacterial 129

cells according to whether they were IgA-coated (IgA+) or IgA-free (IgA-) was performed with 130

the MoFlo XDP Cell Sorter (Beckman Coulter, Brea, Calif), according to the procedures of 131

Simon-Soro et al.14

132

DNA Extraction 133

DNA from sorted breastmilk bacteria, both IgA-coated and IgA-free, as well as the total milk 134

sample was isolated by using the MasterPure complete DNA and RNA Purification Kit 135

(Epicentre Biotechnologies, Madison, Wis), according to the manufacturer’s instructions. 136

(7)

16S rRNA gene amplification and sequencing 138

DNA from sorted bacterial fractions (in total 80) together with total non-sorted breastmilk 139

samples (in total 40) was used for PCR amplification and Illumina sequencing to describe the 140

bacterial composition of breastmilk. Sequences supporting the conclusions of this article are 141

publicly available at European Nucleotide Archive database (ENA) with accession number 142

PRJEB30065. 143

Sequence analysis 144

16S rRNA gene reads from the total milk samples were used in order to perform an accurate 145

filtering of the flow cytometry IgA-sorted fractions that, due to low bacterial yield, were more 146

susceptible to sequencing contaminations. This was done by eliminating OTUs in IgA sorted 147

fractions that were absent in corresponding total non-sorted milk samples. 148

149

For analyzing IgA-coating patterns, the IgA index score (calculated according the formula 150

log(IgA-coated(IgA+)/IgA-free(IgA-)) was used to describe the degree of mucosal immune 151

responsiveness to the microbiota. 152

Statistical analyses were performed in R version 3.2.2 and GraphPad Prism 6 (GraphPad 153

Software, San Diego, CA, USA, Version 6.1f), where p < 0.05 was considered significant. 154

155 156

RESULTS 157

IgA proportions in breastmilk 158

On average, approximately 40% of bacteria in breastmilk appeared to be IgA-coated. 159

Proportions of IgA-coated bacteria were similar in breastmilk samples of mothers whose 160

children did or did not develop allergic (Fig. 1A, p=0.567) and asthmatic symptoms (Fig. 1B) 161

(8)

during the first 7 years of life. Allergic disease included development of eczema, 162

gastrointestinal allergy, asthma, allergic rhinoconjunctitivis or allergic urticaria during the first 163

7 years of life. Moreover, no differences in proportions of breastmilk IgA-coated bacteria in 164

relation to allergy development (most commonly eczema), during the first 2 years of age, were 165

observed (data not shown). IgA proportions observed did not seem to be influenced by the total 166

IgA levels in breastmilk samples of these mothers (n=29; Spearman correlation test r=0.32, 167

p=0.095). The proportions of IgA-coated bacteria were lower in breastmilk from mothers 168

supplemented with probiotics during the last month of pregnancy, as compared with placebo 169

(p=0.04; Fig. S1A in this article’s Online Repository), and particularly in breastmilk of 170

probiotic supplemented mothers whose children stayed healthy (p=0.02; Fig. S1B). 171

172

Bacterial diversity, richness and density in total non-sorted milk samples and IgA-coated 173

fractions 174

The overall species richness (as determined by Chao1 index) in total non-sorted breastmilk 175

samples was significantly higher (p=0.02, Fig. 2A) in mothers with healthy children, although 176

the bacterial load (Fig. S2) and species diversity (Shannon index, Fig. 2B) were similar in 177

breastmilk samples of mothers whose children did/did not develop allergic manifestations. The 178

total species richness also tended to be higher in breastmilk from mothers whose children stayed 179

healthy than from mothers whose children developed asthmatic symptoms (p=0.066, Fig. 2A). 180

However, no significant differences between mothers of healthy and allergic subjects were 181

observed upon comparing the richness and diversity of the IgA-coated fractions in breastmilk 182

(Table SI). Additionally, no differences in bacterial load, species diversity or richness was 183

observed between the total non-sorted breastmilk from the mothers treated with probiotics and 184

placebo (data not shown). 185

Bacterial composition in allergy development and probiotic supplementation 186

(9)

Bacterial 16S rRNA gene sequencing, of the total non-sorted milk and IgA-coated/free 187

fractions, was performed in order to determine the milk microbial composition and to assess 188

IgA responses towards specific bacteria. After quality filtering and removal of chimeric 189

sequences, 40 total non-sorted breastmilk samples and 80 IgA separated fractions remained 190

with 2,000,107 and 1,525,770 high quality reads, respectively. Sequencing of total non-sorted 191

breastmilk samples resulted in an average of 48,987±2725 (SEM) reads per sample, while the 192

IgA separated breastmilk fractions had an average of 20,322±1660 (SEM) sequence reads per 193

sample. 194

The relative abundance of genera in total breastmilk, i.e. non-sorted samples, is presented in 195

Fig. 3A and 3B. Infants developing allergies during the first 7 years of life tended to have higher 196

abundance of the genera Enterococcus (p=0.01; adj. p-value=0.18; Fig. S3A) and Pseudomonas 197

(p=0.01; adj. p-value=0.18; Fig.S3B). The genus Enterococcus was found in significantly 198

higher abundance (p=0.001; adj. p-value= 0.02) in breastmilk given to children that developed 199

asthmatic symptoms, when compared to breastmilk given to the children that remained healthy 200

(Fig. 3B, Fig. S3A). No effect of the confounding factors, such as probiotic supplementation 201

during pregnancy, maternal atopy, sex and the delivery mode influenced the microbial 202

abundance between the groups, as determined with the MaAslin multivariate statistical 203

logarithm. 204

205

Microbiota composition patterns of total non-sorted breastmilk were significantly different 206

(p=0.04 Adonis testing) between mothers that received, or not, probiotic supplementation (Fig. 207

4A) with significantly increased relative abundance (p=0.03, Wilcoxon rank-sum test adjusted 208

p-value) of the genus Rothia in mothers treated with placebo. However, upon comparing IgA 209

pattern recognitions of bacteria in mothers who were treated, or not, with probiotics 210

supplementation during the last month of the pregnancy, no statistically significantly different 211

(10)

IgA responses could be observed for bacterial genera presented (Fig. 4B). 212

213

IgA responses towards milk microbiota in allergy development 214

Although the analysis of the relative abundance of dominant bacterial genera in breastmilk, and 215

the composition patterns of sorted IgA fractions (Fig. S4) were generally similar between the 216

IgA-coated and IgA-free fractions, upon considering the health status of the children, some 217

differences at genus level were observed when analyzing the bacterial targets of IgA responses, 218

represented as the IgA index here. The value of the IgA index can range from positive values, 219

reflecting genera found dominantly in the IgA positive fraction, to negative values (genera 220

found dominantly in the IgA negative fraction). However, no statistically significant differences 221

could be observed in IgA-coating patterns of breastmilk bacteria given to children that did and 222

did not developed allergic and/or asthmatic symptoms (Fig. 5A-5B). 223

(11)

DISCUSSION 225

The data presented in the current study demonstrate that total non-sorted breastmilk from 226

mothers whose children developed allergic symptoms during early childhood had lower 227

bacterial richness, when compared to milk fed to children staying healthy. Moreover, probiotic 228

supplementation during pregnancy modifies the breastmilk microbiota composition and the 229

proportion of IgA-coated bacteria. 230

The influence of breastmilk composition on later allergy development appears to be linked to 231

higher richness of bacterial species and not to the relative abundance of specific bacteria. 232

Previously, low total diversity of the gut and oral microbiota have been associated with atopy 233

and asthma development during early childhood.6,8,15,16 Moreover, aberrant IgA immune

234

responses towards gut microbiota, but not the proportions of IgA-coating of bacteria, in relation 235

to subsequent allergy development during childhood were observed as early as 1 month post 236

partum, in the same cohort of children.10 At this time point, the IgA antibodies are

237

predominantly maternally derived in exclusively breastfed children, as the levels of 238

endogenously produced IgA in the baby during this period is limited.17 Therefore, any divergent

239

responses observed at this time suggest that immunological interactions between mother and 240

infant may play an important role in subsequent immune development. However, the data in 241

the current study suggest that bacterial IgA responses in breastmilk does not correlate with that 242

observed in the gut of the breastfed infants. Likely, not only the proportions of IgA-coated 243

bacteria but also the composition and timing of bacterial colonization (including the 244

establishment of specific bacterial species, recognized by IgA or not) during early infancy, will 245

have an effect on the proper immune system education. Thus, further research is needed in order 246

to understand the exact function of vertical transmission of IgA-coated bacteria from mother to 247

breastfeeding child. 248

(12)

Streptococcus, Acinetobacter, Staphylococcus and Veillonella were the most commonly found 249

bacterial genera in milk samples used in this study. Furthermore, lactic acid bacteria 250

Lactococcus, Lactobacillus and Enterococcus as well as oral inhabitant Gemella were also 251

detected, in agreement with previous reports.18 Various studies have demonstrated that there is

252

a mother-to-infant transfer of bacterial genera including

253

Lactobacillus, Staphylococcus, Enterococcus and Bifidobacterium through breastfeeding.19–22

254

The constant intake during lactation of breastmilk bacteria leads to the establishment of an 255

intestinal microbiota that deeply impacts on the newborn’s immune maturation.23 An interesting

256

finding is the significantly higher levels of the lactic acid bacteria Enterococcus in breastmilk 257

fed to children developing asthma. Enterococcus is a common inhabitant of breastmilk 258

microbiota and one of the first microbes to colonize the infant gut after birth8,24, being more

259

abundant in the gut of atopic infants, from the same cohort, at 12 months of age.8 However,

260

whether our finding here reflects an overgrowth of Enterococcus due to the absence of 261

competing species, or whether Enterococcus is suppressing the growth of allergy protecting 262

bacteria remains to be addressed. 263

264

An important function of SIgA is immune exclusion, a mechanism where this antibody binds 265

to commensals, through its recognition of multiple antigenic epitopes on the surface, 266

encountered in gut lumen and prevents their attachment to mucosal barrier.25 Moreover, this

267

antibody can also promote bacterial adhesion to the mucosa, as shown in vitro and in vivo26,27,

268

thus enriching for the growth of particular microbial strains.28 In this study, we were not able

269

to observe significant differences in the patterns of IgA-binding in breastmilk fed to children 270

staying healthy and children developing allergies but the possibility that the true differences are 271

at the lower bacterial resolution (for instance species and even different strains), should not be 272

excluded. The fact that the majority of the mothers included in this study, and also in the original 273

(13)

study of this cohort, suffered from allergic disease, should be considered as it also may affect 274

both the IgA-coating patterns and the microbiota transferred to the child. Moreover, due to 275

sample availability, in this study we have examined solely a part of the total original cohort 276

which might limit the findings presented here. 277

IgA-coating proportions of breastmilk bacteria were higher in mothers treated with placebo, 278

compared to those treated with probiotic L. reuteri. As the mothers were treated with probiotics 279

until delivery, likely the changes in the microbial composition were more significant in 280

colostrum than in breastmilk samples extracted at one month post partum, as in the present 281

study. We have previously shown that Lactobacilli colonization was significantly increased in 282

colostrum of the mothers treated with L. reuteri, but not in samples obtained one month post 283

partum.29 This could suggest that part of the mother´s IgA responses, while treated with

284

probiotics, were directed towards L. reuteri, but that the ending of supplementation was 285

reflected in diminished targeting of this species and perhaps decreased proportions of IgA-286

coated microbiota. Additionally, microbiota patterns of total non-sorted milk were also 287

significantly different between mothers treated with probiotics and those with placebo, with the 288

genus Rothia (a common oral inhabitant associated with good oral health30) significantly

289

increased in the placebo group. However, the importance of these results and the detailed 290

mechanisms behind the relationship of probiotic supplement and the development of allergic 291

diseases during childhood needs to be further investigated, preferably in larger study cohorts 292

including mothers not suffering from allergic disease. 293

294

Together with the maternal intestinal and vaginal microorganisms that are ingested by the 295

neonate during the passage through the birth canal, breastmilk microbiota appears to contribute 296

to the initial microbial colonization in infants, thus having a pivotal role in modulating and 297

influencing the newborns’ immune system. The bacterial species that initially colonize the 298

(14)

mucosal surfaces likely define the ecosystem conditions which in turn will affect the 299

establishment of further co-colonization patterns. Also, milk microbes are transferred together 300

with maternal IgA antibodies, that may enable maintenance of a mutually beneficial 301

relationship with a diverse set of commensals, while protecting against pathogens. 302

303

In conclusion, consumption of breastmilk with a reduced microbial richness in the first month 304

of life correlates with an increased risk to develop allergy during childhood. In addition, our 305

data show that probiotic supplementation during pregnancy alters the breastmilk microbiota 306

composition and the proportion of IgA-coated bacteria. These findings open the possibility of 307

modulating breastmilk microbiota and its interaction with antibodies as a strategy to promote a 308

healthy microbial colonization with the purpose of reducing allergy risk. 309

(15)

REFERENCES 310

1. Walker A. Breast Milk as the Gold Standard for Protective Nutrients. J Pediatr. 2010 311

Feb;156(2):S3–7. 312

2. Ballard O, Morrow AL. Human milk composition: nutrients and bioactive factors. 313

Pediatr Clin North Am. 2013 Feb;60(1):49–74. 314

3. Newburg DS, Walker WA. Protection of the Neonate by the Innate Immune System of 315

Developing Gut and of Human Milk. Pediatr Res. 2007 Jan;61(1):2–8. 316

4. West CE, Jenmalm MC, Prescott SL. The gut microbiota and its role in the 317

development of allergic disease: a wider perspective. Clin Exp Allergy. 2015 318

Jan;45(1):43–53. 319

5. West CE, Dzidic M, Prescott SL, Jenmalm MC. Bugging allergy; role of pre-, pro- and 320

synbiotics in allergy prevention. Allergol Int. 2017;66(4). 321

6. Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm 322

MC. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin 323

Exp Allergy. 2014 Jun;44(6):842–50. 324

7. Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince C, Jernberg C, et al. 325

Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced 326

Th1 responses in infants delivered by Caesarean section. Gut. 2014 Apr;63(4):559–66. 327

8. Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm 328

MC. Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin 329

Immunol. 2012 Mar;129(2):434–40. 330

9. Jenmalm MC. The mother–offspring dyad: microbial transmission, immune 331

interactions and allergy development. J Intern Med. 2017 Jul;282(6):484–95. 332

10. Dzidic M, Abrahamsson TRTR, Artacho A, Björkstén B, Collado MCMC, Mira A, et 333

al. Aberrant IgA responses to the gut microbiota during infancy precede asthma and 334

(16)

allergy development. J Allergy Clin Immunol. 2017 Mar;139(3):1017-1025.e14. 335

11. Abrahamsson TR, Jakobsson T, Böttcher MF, Fredrikson M, Jenmalm MC, Björkstén 336

B, et al. Probiotics in prevention of IgE-associated eczema: A double-blind, 337

randomized, placebo-controlled trial. J Allergy Clin Immunol. 2007 May;119(5):1174– 338

80. 339

12. Abrahamsson TR, Jakobsson T, Björkstén B, Oldaeus G, Jenmalm MC. No effect of 340

probiotics on respiratory allergies: a seven-year follow-up of a randomized controlled 341

trial in infancy. Pediatr Allergy Immunol. 2013 Sep;24(6):556–61. 342

13. Böttcher MF, Abrahamsson TR, Fredriksson M, Jakobsson T, Björkstén B. Low breast 343

milk TGF-β2 is induced by Lactobacillus reuteri supplementation and associates with 344

reduced risk of sensitization during infancy. Pediatr Allergy Immunol. 2008 345

Aug;19(6):497–504. 346

14. Simón-Soro Á, D’Auria G, Collado MC, Džunková M, Culshaw S, Mira A. Revealing 347

microbial recognition by specific antibodies. BMC Microbiol. 2015 Jul;15(1):132. 348

15. Dzidic M, Abrahamsson T, Artacho A, Collado MC, Mira A, Jenmalm MC. Oral 349

microbiota maturation during the first 7 years of life in relation to allergy development. 350

Allergy. 2018 Mar;0(ja). 351

16. Azad MB, Konya T, Guttman DS, Field CJ, Sears MR, HayGlass KT, et al. Infant gut 352

microbiota and food sensitization: associations in the first year of life. Clin Exp 353

Allergy. 2015 Mar;45(3):632–43. 354

17. Tourneur E, Chassin C. Neonatal immune adaptation of the gut and its role during 355

infections. Clin Dev Immunol. 2013 May;2013:270301. 356

18. Fitzstevens JL, Smith KC, Hagadorn JI, Caimano MJ, Matson AP, Brownell EA. 357

Systematic Review of the Human Milk Microbiota. Nutr Clin Pract. 2016 Sep;32:354– 358

64. 359

(17)

19. Martín R, Langa S, Reviriego C, Jimínez E, Marín ML, Xaus J, et al. Human milk is a 360

source of lactic acid bacteria for the infant gut. J Pediatr [Internet]. 2003;143(6):754–8. 361

20. Soto A, Martín V, Jiménez E, Mader I, Rodríguez JM, Fernández L. Lactobacilli and 362

Bifidobacteria in Human Breast Milk: Influence of Antibiotherapy and Other Host and 363

Clinical Factors. J Pediatr Gastroenterol Nutr. 2014 Jul;59(1):78–88. 364

21. Martín V, Maldonado-Barragán A, Moles L, Rodriguez-Baños M, Campo R del, 365

Fernández L, et al. Sharing of Bacterial Strains Between Breast Milk and Infant Feces. 366

J Hum Lact. 2012 Jan;28(1):36–44. 367

22. Benito D, Lozano C, Jiménez E, Albújar M, Gómez A, Rodríguez JM, et al. 368

Characterization of Staphylococcus aureus strains isolated from faeces of healthy 369

neonates and potential mother-to-infant microbial transmission through breastfeeding. 370

FEMS Microbiol Ecol. 2015 Mar;91(3):fiv007–fiv007. 371

23. Houghteling PD, Walker WA. Why Is Initial Bacterial Colonization of the Intestine 372

Important to Infantsʼ and Childrenʼs Health? J Pediatr Gastroenterol Nutr. 2015 373

Mar;60(3):294–307. 374

24. Mshvildadze M, Neu J. The infant intestinal microbiome: friend or foe? Early Hum 375

Dev. 2010;86. 376

25. Hurley WL, Theil PK. Perspectives on immunoglobulins in colostrum and milk. 377

Nutrients. 2011;3(4):442–74. 378

26. Donaldson GP, Ladinsky MS, Yu KB, Sanders JG, Yoo BB, Chou WC, et al. Gut 379

microbiota utilize immunoglobulin A for mucosal colonization. Science (80- ) 380

[Internet]. 2018;360(6390):795–800. 381

27. McLoughlin K, Schluter J, Rakoff-Nahoum S, Smith AL, Foster KR. Host Selection of 382

Microbiota via Differential Adhesion. Cell Host Microbe. 2016;19(4):550–9. 383

28. Rogier EW, Frantz AL, Bruno MEC, Wedlund L, Cohen DA, Stromberg AJ, et al. 384

(18)

Secretory antibodies in breast milk promote long-term intestinal homeostasis by 385

regulating the gut microbiota and host gene expression. Proc Natl Acad Sci. 2014 386

Feb;111(8):3074–9. 387

29. Abrahamsson TR, Sinkiewicz G, Jakobsson T, Fredrikson M, Björkstén B. Probiotic 388

Lactobacilli in Breast Milk and Infant Stool in Relation to Oral Intake During the First 389

Year of Life. J Pediatr Gastroenterol Nutr. 2009;49(3). 390

30. Lundberg JO, Carlström M, Weitzberg E. Metabolic Effects of Dietary Nitrate in 391

Health and Disease. Cell Metab. 2018;28(1):9–22. 392

393 394

(19)

395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 Children Healthy (% [no.]) n=20 Developing allergic disease (% [no.]) n=20 P value* Developing asthma (% [no.]) n=10 P value* Girls 50(10) 60(12) 0.53 60(6) 0.71 Older siblings 40(8) 35(7) 1.00 30(3) 0.70 Caesarean delivery 20(4) 20(4) 1.00 20(2) 1.00 Furred pets 15(3) 10(2) 1.00 10(1) 1.00 Breastfeeding 1 month exclusive 90(18) 95(19) 1.00 90(9) 1.00 3 months exclusive 85(17) 70(14) 0.45 50(5) 0.08 12 months partial 20(4) 25(5) 1.00 0(0) 0.27 Maternal atopy 85(17) 80(16) 1.00 70(7) 0.37 Antibiotic treatment (1-12 m) 35(7) 30(6) 1.00 50(5) 0.46 Day care (1-12 m) 10(2) 5(1) 1.00 10(1) 1.00 Probiotics-L. reuteri 45(9) 55(11) 0.53 60(6) 0.70

Table I. Descriptive data of children compare in this study.

*The x2 test was used to detect potential differences in frequencies between groups, except when the

(20)

FIGURES 413 414 415 416 417 418 419

Figure 1. Proportions of IgA-coated breastmilk bacteria collected one-month post partum. A) 420

Proportion of breastmilk IgA-coated bacteria in mothers whose children stayed healthy (n=24, 421

circles) or developed allergic symptoms (n=26, triangles) during the first 7 years of life, as 422

determined by flow cytometry based-cell sorting (p=0.567). B) Proportion of breastmilk 423

bacteria coated to IgA at one month of age in children staying healthy (n=24) or developing 424

asthmatic symptoms (n=13) during the first 7 years of life (p=308). Medians and interquartile 425

ranges are indicated. 426 427 428 429 B A Healthy Allergic 0 20 40 60 80 100 Ig A b o u n d b a cte ria (% ) Healthy Asthmatic 0 20 40 60 80 100 Ig A b o u n d b a cte ria (% )

(21)

430

Figure 2. Bacterial richness (A) and diversity (B) at 16S rRNA OTU’s species level, as 431

described with Chao1 and Shannon indices, respectively, of the total non-sorted breastmilk 432

samples from mothers whose children stayed healthy up to 7 years of age (n=20), mothers 433

whose children developed allergic manifestations (n=20) and/or asthmatic symptoms (n=10). 434

Medians and interquartile ranges are indicated. * p-value <0.05, Mann-Whitney U test. 435

436

437

Figure 3. Microbiota composition of most dominant bacterial genera in total non-sorted milk 438

samples. A) The relative abundance (>0.5 % of the total) of dominant bacterial genera in 20 439

mothers whose children stayed healthy and in 20 mothers whose children developed allergic 440

manifestation during their first 7 years of life. B) The relative abundance of dominant bacterial 441

genera in breastmilk samples in 20 mothers whose children stayed healthy and 10 mothers 442

whose children developed asthmatic manifestation. 443

Healthy Allergic Asthmatic

0 1 2 3 4

Shannon diversity indices

Sp e c ie s d iv e rs ity

B

A

Healthy Allergic Asthmatic

0 200 400 600 800 1000 Sp e c ie s r ic h n e s s

*

B A Healthy Allergic 0 50 100 Re la tiv e a b u n d a n ce (%) Healthy Asthmatic 0 50 100 Re la tiv e a b u n d a n ce (%) Streptococcus Staphylococcus Veillonella Corynebacterium Acinetobacter Gemella Lactobacillus Lactococcus Rothia Pseudomonas Granulicatella Enterococcus Others Healthy Allergic 0 50 100 Re la tiv e a b u n d a n ce (%) Healthy Asthmatic 0 50 100 Re la tiv e a b u n d a n ce (%) Streptococcus Staphylococcus Veillonella Corynebacterium Acinetobacter Gemella Lactobacillus Lactococcus Rothia Pseudomonas Granulicatella Enterococcus Others

(22)

444

445

Figure 4. Breastmilk microbiota composition and IgA-coating patterns of the dominant genera 446

(>0.5% of total), in probiotic supplementation. A) Constrained correspondence analyses (CCA) 447

based on microbiota composition patterns (determined by 16S rRNA sequencing) in breastmilk 448

of mothers treated with probiotics L. reuteri or placebo. The percentage of variation explained 449

by constrained correspondence components is indicated on the axes. p value for CCA plots 450

were determined by Adonis (p=0.04) and indicate if the factor provided (in this case probiotics) 451

can significantly explain data variability. B) Plots represent IgA-coating patterns (defined by 452

IgA index, reflecting the ratio in IgA-coated and IgA-free breastmilk microbiota) to dominant 453

genera in breastmilk samples collected at one month post partum from mothers whose were 454

treated with probiotic L. reuteri or placebo. For a given genera, the value of the IgA index can 455

range from positive values, reflecting genera found dominantly in the IgA-coated fraction, to 456

negative values (genera found dominantly in the IgA-free fraction). nProbiotics=20, nPlacebo=20.

457 458 459 −4 −2 0 2 4 6 −4 −2 0 2 4

CCA p−value: 0.049 − ADONIS p−value: 0.04

CCA1 5.687% CA1 34.294% Probiotics ● ●●●●● ● ● ● ● ● ●● ● ● ● ● ● ● ● Placebo ● ●● ● ● ●● ● ● ● ● ● ● ● ● ● ● ● ● B A

(23)

460

Figure 5. IgA responses towards breastmilk microbiota. Plots represent IgA-coating patterns 461

to dominant genera (>0.5 % of total) from mothers whose children stay healthy (n= 20) 462

compared to children that develop allergic (A, n=20) or asthmatic (B, n=10) symptoms. For a 463

given genera, the value of the IgA index can range from positive values, reflecting genera found 464

dominantly in the coated fraction, to negative values (genera found dominantly in the IgA-465

free fraction). Means with SEs are indicated. Wilcoxon rank-sum test with FDR correction was 466

performed and no statistically significant differences were observed. 467

B A

(24)

3 4 METHODS 5 Study design 6

The subjects included in this study were part of a larger randomized double-blind trial in South-7

eastern Sweden, recruiting participants between 2001 and 2003, where the potential allergy 8

preventive effect of probiotic Lactobacillus reuteri ATCC 55730 in the infants with family 9

history of allergic disease was evaluated.1,2 The mothers were supplemented with L. reuteri

10

during pregnancy from postmenstrual week 36+0 to delivery and the infants from day 1-2 of 11

life until 12 months of age. Among the 184 mothers that completed the 7-year follow up in the 12

original study, breastmilk samples from 51 mothers were used for flow cytometry based-sorting 13

of IgA-coated bacteria in the current study. When comparing the IgA proportions in breastmilk 14

from mothers that were treated with probiotics or placebo, 27 and 24 samples were used for 15

these analyses. However, for comparisons of the breastmilk fed to children developing allergies 16

or staying healthy, we excluded one of the samples as this child had allergic symptoms, but not 17

sensitization, in order to get more accurate analyses. 18

19

Subsequently, 16S rRNA gene characterization was performed on the IgA-coated and IgA-free 20

fractions of breastmilk bacteria from 20 mothers whose children did not and from 20 mothers 21

whose children did develop allergic manifestations as well as from total, non-sorted breastmilk 22

samples from the same mothers. The selection of the samples used for 16S rRNA sequencing 23

in this study, was based on the sample availability and a clear allergy diagnosis (based on 24

proven symptoms to allergy provocation) of the child. There were no differences regarding 25

potential confounders, such as sex, mode of delivery, birth order, maternal atopy, breastfeeding, 26

antibiotics, and probiotic supplementation, between the infants who did or did not have allergic 27

(25)

(n=10), allergic rhinoconjunctitivis (n=14) and allergic urticaria (n=3). The criteria of these 29

diagnoses are described in detail in1,2. All children with allergic disease in the current study

30

were also sensitized (i.e. they had at least one positive skin prick test (evaluated at 6, 12 and 24 31

months and 7 years of age) and/or detectable circulating allergen specific-IgE antibodies 32

(assessed at 6, 12 and 24 months), while the healthy children were non-sensitized. Skin prick 33

tests were performed on the forearm with egg white, fresh skimmed cow milk and standardized 34

cat, dog, birch, peanut and timothy extracts at 6, 12, 24 months and 7 years of age (here even 35

mite (Der p)).2 Moreover, symptoms related to allergic disease, physical examination,

36

spirometry and measurement of fractional exhaled nitric oxid (FENO) were observed. Children

37

were diagnosed with allergy if they have had symptoms of and/or have been treated for the 38

actual allergic disease during the last twelve months. A diagnosis of gastrointestinal allergy 39

required vomiting, diarrhea, or systemic reaction after ingestion of a potentially allergenic food 40

and a confirmation by challenge, unless there was a clear history of a severe systemic 41

reaction.1,3 Asthma diagnosis was based on at least one of following two criteria: 1. Doctor

42

diagnosis and asthma symptoms and/or medication during the last twelve months; 2. Wheeze 43

or nocturnal cough and a positive reversibility test and/or pathological FENO value.1,3 All

44

asthmatic children were also included in the allergic group. For further details, please see the 45

publications from Abrahamsson T. et al.1–3

46 47

Breastmilk samples were collected one-month post partum by the mother at home. They were 48

immediately placed in the freezer and brought to the hospital and stored at −70°C within 3 days. 49

All the children included in the current study were exclusively breastfed during the first month 50

of life. Total IgA levels were measured by ELISA in a study by Böttcher et al.4

(26)

Linköping, Sweden (Dnr 99323, M122-31 and M171-07, respectively). An informed consent 53

was obtained from both parents before inclusion in the study. 54

55

Sample preparation and flow cytometry-based sorting 56

After thawing, the breastmilk fatty layer and whey were removed by centrifugations. The 57

resulting sample fraction was further suspended in sterile saline solution (autoclaved H2O; 58

NaCl Sodium Chloride 99.5% PA-ACS-ISO; Panreac, Barcelona, Spain; reference 59

131689.1211) with 5% BSA (Sigma-Aldrich, St Louis, Mo; reference A7030-100gr) to prevent 60

nonspecific antibody binding. The samples were stained with goat anti-mouse IgA labelled with 61

fluorescein isothiocyanate (FITC), used as an isotype control corresponding to unspecific 62

binding (Sigma; reference SLBD9273), or with goat anti-human IgA labelled with FITC (Life 63

Technologies; reference A18782), according to the manufacturer’s instructions. The sorting of 64

the bacterial cells according to whether they were IgA-coated or IgA-free was performed with 65

the MoFlo XDP Cell Sorter (Beckman Coulter, Brea, Calif), according to the procedures of 66

Simon- Soro et al.5

67 68

DNA Extraction 69

DNA from sorted breastmilk bacteria, both IgA+ and IgA-, as well as the total milk sample (2 70

ml) was isolated by using the MasterPure complete DNA and RNA Purification Kit (Epicentre 71

Biotechnologies, Madison, Wis), according to the manufacturer’s instructions, with a previous 72

glass bead beating (0.17 mm in diameter) and an additional enzymatic lysis step with lysozyme 73

(20 mg/mL, 378C, 30 minutes; Thermo-mixer comfort, Eppendorf, Hamburg, Germany), 74

lysostaphin (2000 units/mg protein, 37 °C, 60 min; Sigma-Aldrich, Madrid, Spain) and 75

mutanolysin (4000 units/mg protein, 37 °C, 60 min; Sigma-Aldrich). 76

(27)

DNA from sorted bacterial fractions (in total 80) together with total non-sorted breastmilk 78

samples (in total 40) was used for PCR amplification and Illumina sequencing to describe the 79

bacterial composition of breastmilk. Universal bacterial degenerate primers 8F—5ʹ-80

AGAGTTTGATCMTG GCTCAG-3ʹ and 926R—5ʹ-CCGTCAATTCMTTTRAGT- 3ʹ, which 81

encompass the hypervariable regions V1–V5 of 16S ribosomal RNA (rRNA) gene were used 82

for an initial amplification in order to increase the bacterial yield. Purification of PCR products 83

was completed using Nucleofast 96 PCR filter plates (Macherey-Nagel, Düren, Germany). 84

An Illumina amplicon library was performed following the 16S rRNA gene Metagenomic 85

Sequencing Library Preparation Illumina protocol (Part #15044223 Rev. A). The gene-specific 86

primer sequences used in this protocol were selected from Klindworth et al. 6, and target the

87

16S rRNA gene V3 and V4 regions, resulting in a single amplicon of approximately 460 bp. 88

After 16S rRNA gene amplification, the DNA was sequenced on a MiSeq Sequencer according 89

to manufacturer’s instructions (Illumina) using the 2 × 300 bp paired-end protocol. Sequences 90

supporting the conclusions of this article are publicly available at European Nucleotide Archive 91

database (ENA) with accession number PRJEB30065. 92

Total bacterial load 93

Total bacterial load (number of bacterial cells per ml breastmilk) was measured by quantitative 94

PCR. Amplifications were performed in duplicates on a LightCycler 480 Real-Time PCR 95

System (Roche Technologies) by using annealing temperatures of 60 °C. Each reaction mixture 96

of 10mL was composed of SYBR Green PCR Master Mix (Roche), 0.5 mL of the specific 97

primer (concentration 10 mmol/L), and 2 mL of DNA template. The universal forward and 98

reverse primers were 5ʹ-CGTGCCAGCAGCCGCGG-3ʹ and 5ʹ-

99

TGGACTACCAGGGTATCTAATCCTG-3ʹ, targeting a 293bp long region of the bacterial 100

(28)

calibrated standard curve. 102

Bioinformatics 103

The PRINSEQ program was used for a sequence quality assessment.8 Sequences of <250

104

nucleotides in length were discarded; sequence end-trimming was performed by cutting out 105

nucleotides with a mean quality of <30 in 20-bp windows. Chimeric 16S sequences were 106

filtered out using USEARCH program.9 OTUs were built at 97% of identity by using vsearch

107

program and Qiime modules (version qiime2-2017.12) were used for taxonomic annotation.10

108

In order to assign taxonomy up to species level to each OTU’s centroid, we classified them 109

using a naïve bayes classifier model previously fitted against the Green Genes database version 110

13.5. 111

112

16S rRNA gene reads from the total milk samples were used in order to perform an accurate 113

filtering of the flow cytometry IgA-sorted fractions that, due to low bacterial yield, were more 114

susceptible to sequencing contaminations. This was done by eliminating OTUs in IgA sorted 115

fractions that were absent in corresponding total non-sorted milk samples. Moreover, OTUs 116

were also removed in cases where either IgA positive or IgA negative fractions presented an 117

abundance of less than five reads, compared to its corresponding OTU in non-sorted milk 118

sample. In addition, we filtered out low signal OTUs that were presented in less than five 119

sequences through the total set of samples. 120

α-diversity analyses (presented here as Shannon and Chao1 indices), were utilized to estimate 121

the samples’ diversity and richness at the 97% OTU level using the R-package Vegan.11

122

Constrained correspondence analysis (CCA) was used here to emphasize variation and bring 123

out strong patterns in the dataset. This analysis was performed by R software ade4 package 124

together with permutational multivariate analysis (Adonis) determining the differences in 125

(29)

For analyzing IgA-coating patterns, the threshold used for including the genera was 0.5 % or 127

greater in relative abundance in either the IgA positive or IgA negative fractions. A pseudocount 128

that was equal to 0.001 was added to every genus dedicated in both the IgA positive or IgA 129

negative fractions, thus avoiding the fractions with a value of zero. The abundance proportions 130

of a given genera were log-transformed before calculating the ratio between the IgA positive or 131

IgA negative fractions, resulting in the IgA index.12,13 The IgA index (calculated according the

132

formula log(IgA+/IgA-)) score reflects the degree of mucosal immune responsiveness to the 133

microbiota, where the positive values represent the genera predominantly found IgA-coated 134

while the negative values the bacterial genera predominantly found IgA uncoated. 135

The MaAslin multivariate statistical framework was used in this study in order to evaluate if 136

the confounding factors, including probiotics supplementation during pregnancy and maternal 137

atopy, could influence microbial community abundance.14 Statistical analyses were performed

138

in R version 3.2.2 and GraphPad Prism 6 (GraphPad Software, San Diego, CA, USA, Version 139

6.1f), where p < 0.05 was considered significant. Specific statistical tests (including Mann– 140

Whitney U-test/ Wilcoxon rank-sum test for nonparametric comparisons together with false 141

discovery rate control giving the adjusted p-value) are stated in figure legends. 142

143 144 145

(30)

Table S1. Chao1 and Shannon indices describing the species richness and diversity, respectively, in total milk 148

samples, in the IgA bound and IgA free fractions. Mean and standard error of the mean (SEM) are indicated.

149

HEALTHY non-sorted

(n=20) ALLERGIC non-sorted (n=20) ASTHMA non-sorted (n=10) p-value

Chao1 SEM Chao1 SEM Chao1 SEM H vs Allergic / H vs Asthmatic

542.26 32.96 430.30 40.74 424.88 71.38 0.018*/ 0.065

HEALTHY IgA-bound

(n=40) ALLERGIC IgA-bound (n=40) ASTHMA IgA-bound (n=20) p-value

Chao1 SEM Chao1 SEM Chao1 SEM H vs Allergic / H vs Asthmatic

62.67 6.26 65.88 9.39 70.33 17.05 0.733/0.820

HEALTHY IgA-free

(n=40) ALLERGIC IgA-free (n=40) ASTHMA IgA-free (n=20) p-value

Chao1 SEM Chao1 SEM Chao1 SEM H vs Allergic / H vs Asthmatic

57.09 5.45 58.14 6.75 58.42 11.78 0.898/ 0.820

150

HEALTHY non-sorted

(n=20) ALLERGIC non-sorted (n=20) ASTHMA non-sorted (n=10) p-value

Shannon SEM Shannon SEM Shannon SEM H vs Allergic / H vs Asthmatic

1.96 0.11 1.87 0.19 1.82 0.30 0.351/0.411

HEALTHY IgA-bound

(n=40) ALLERGIC IgA-bound (n=40) ASTHMA IgA-bound (n=20) p-value

Shannon SEM Shannon SEM Shannon SEM H vs Allergic / H vs Asthmatic

2.45 0.10 2.38 0.11 2.42 0.18 0.490/0.893

HEALTHY IgA-free

(n=40) ALLERGIC IgA-free (n=40) ASTHMA IgA-free (n=20) p-value

Shannon SEM Shannon SEM Shannon SEM H vs Allergic / H vs Asthmatic

2.40 0.09 2.32 0.15 2.18 0.28 0.875/0.859

* Mann-Whitney U test. 151

152

153

Fig. S1. IgA-coating levels in the breastmilk of mothers treated, or not, with L. reuteri, as determined with flow 154

cytometry based – cell sorting. (A) Proportions of IgA bound bacteria in breastmilk of mothers treated with L.

155

reuteri probiotics and mothers treated with placebo. p= 0.04; nPlacebo=27, nProbiotics=24 (B) Proportions of IgA

156

HEALTHY ALLERGIC

*

B

A

Probiotics Placebo Probiotics Placebo 0 20 40 60 80 100 IgA bound ba c te ria (% ) p=0,0256

*

Probiotics Placebo 0 20 40 60 80 100 % Ig A b o u n d b act eri a

(31)

nPlacebo=13, nProbiotics=14. Media with interquartile ranges are indicated; * Mann-Whitney U test.

159 160

161

Fig. S2. Bacterial load in breastmilk collected at one month post partum. Quantification of bacterial numbers 162

was obtained by using qPCR detection with universal primers targeting the 16S rRNA bacterial gene. (A)

163

Bacterial load in breastmilk samples fed to children developing allergies or staying healthy. (B) Bacterial load in

164

breastmilk samples fed to children developing asthmatic disease or staying healthy. nHealthy =19; and nAllergic = 19;

165

nAsthmatic =9. Media with interquartile ranges are indicated and no statistically significant differences were

166 observed. 167 168 169 170 171 172 173

Fig. S3. Relative abundance of selected bacterial genera found in non-sorted breastmilk samples of mothers 174

whose children stay healthy or develop allergic and/or asthmatic manifestations. Mean with SEM are presented.

175

* p < 0.05, Mann-Whitney U test, FDR adjusted p-value.

176 177 Healthy Allergic 104 105 106 107 108 109 1010 Lo g (b acter ial cells / 1 ml milk ) Healthy Asthma 104 105 106 107 108 109 1010 Lo g (b acter ial cells / 1 ml milk ) B A Healthy Allergic Asthmatic 0.0 0.2 0.4 10 20 30 40 Pseudomonas Re la tiv e a b u n d a n c e (%) Healthy Allergic Asthmatic 0.0 0.2 0.4 0.6 0.8 1.01 2 3 4 5 6 Veillonella Re la tiv e a b u n d a n ce (%) B A C Healthy Allergic Asthmatic 0.00 0.02 0.04 0.06 0.08 0.10 2 4 6 Enterococcus Re la tiv e a b u n d a n ce (%) *

(32)

180

181

Fig. S4. Breastmilk microbiota composition patterns of sorted IgA fractions, IgA-coated and IgA-free, in allergy 182

development. Constrained correspondence analyses (CCA) based on breastmilk microbiota patterns of the

183

dominant bacterial genera (>0.5% of total) coated with IgA (A) or not coated with IgA (B), from mothers whose

184

children did (n=20) or did not (n=20) develop allergic diseases (p=0.19 and p=0.44 respectively).

185 186 187

REFERENCES 188

1. Abrahamsson TR, Jakobsson T, Böttcher MF, Fredrikson M, Jenmalm MC, Björkstén B, et al. Probiotics

189

in prevention of IgE-associated eczema: A double-blind, randomized, placebo-controlled trial. J Allergy

190

Clin Immunol. 2007 May;119(5):1174–80.

191

2. Abrahamsson TR, Jakobsson T, Björkstén B, Oldaeus G, Jenmalm MC. No effect of probiotics on

192

respiratory allergies: a seven-year follow-up of a randomized controlled trial in infancy. Pediatr Allergy

193

Immunol. 2013 Sep;24(6):556–61.

194

3. Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low gut

195

microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy. 2014

196

Jun;44(6):842–50.

197

4. Böttcher MF, Abrahamsson TR, Fredriksson M, Jakobsson T, Björkstén B. Low breast milk TGF-β2 is

198

induced by Lactobacillus reuteri supplementation and associates with reduced risk of sensitization during

199

infancy. Pediatr Allergy Immunol. 2008 Aug;19(6):497–504.

200

5. Simón-Soro Á, D’Auria G, Collado MC, Džunková M, Culshaw S, Mira A. Revealing microbial

201

recognition by specific antibodies. BMC Microbiol. 2015 Jul;15(1):132.

202

6. Klindworth A, Pruesse E, Schweer T, Peplies J, Quast C, Horn M, et al. Evaluation of general 16S

203

ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies.

204

Nucleic Acids Res. 2013 Jan;41(1):e-publication.

205

7. Dzidic M, Collado MC, Abrahamsson T, Artacho A, Stensson M, Jenmalm MC, et al. Oral microbiome

206

development during childhood: an ecological succession influenced by postnatal factors and associated

207

with tooth decay. ISME J. 2018;12(9):2292–306.

208

8. Schmieder R, Edwards R. Quality control and preprocessing of metagenomic datasets. Bioinformatics.

209 −4 −2 0 2 4 6 −2 0 2 4 6

CCA p−value: 0.099 − ADONIS p−value: 0.19

CCA1 4.051% CA1 26.259% ALLERGIC ● ● ● ● ● ● ● ● ●● ● ● ● ● ● ● ● ● HEALTHY ● ●● ● ● ● ● ● ● ● ● ● ●●● ● ● ● ● ● −4 −2 0 2 4 6 −4 −2 0 2 4 6

CCA p−value: 0.35 − ADONIS p−value: 0.44

CCA1 2.922% CA1 26.665% ALLERGIC ● ● ● ● ● ● ● ● ● ● ●● ● ● ● ● ● ● ● HEALTHY ● ●● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● B A

(33)

10. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. QIIME allows

213

analysis of high-throughput community sequencing data. Nat Methods. 2010 Apr;7:335.

214

11. Oksanen J. Vegan:community ecology. R package version 2.4-6. 2018. p.

<http://cran.r-215

project.org/web/packages/vegan/>.

216

12. Dzidic M, Abrahamsson TRTR, Artacho A, Björkstén B, Collado MCMC, Mira A, et al. Aberrant IgA

217

responses to the gut microbiota during infancy precede asthma and allergy development. J Allergy Clin

218

Immunol. 2017 Mar;139(3):1017–1025.e14.

219

13. Bunker JJ, Flynn TM, Koval JC, Shaw DG, Meisel M, McDonald BD, et al. Innate and Adaptive

220

Humoral Responses Coat Distinct Commensal Bacteria with Immunoglobulin A. Immunity.

221

2015;43(3):541–53.

222

14. Morgan X, Tickle T, Sokol H, Gevers D, Devaney K, Ward D. Dysfunction of the intestinal microbiome

223

in inflammatory bowel disease and treatment. Genome Biol [Internet]. 2012;13(R79).

224 225

References

Related documents

Germ Free Mice Express High IgA Class Switch Recombination Activity But Develop Few IgA Producing Plasma Cells.. Bergqvist, P., Stensson, A., Bemark, M., &amp;

Linköping Studies in Science and Technology, Dissertation No.. 1713 Department of Physics, Chemistry and

By new learning, we do not only mean learning taking place in those areas where the participants lack knowledge (e.g. medical knowledge which requires additional teaching),

Aggregates that were formed from poly-E solution not containing p-FTAA showed the same morphology (see Supporting Figure S3a), indicating that p-FTAA does not affect the

Denna vy är till för att visa en grafisk representation av användarens värden. Vyn finns med i båda versionerna men är väldigt begränsad i iOS då TeeChart inte är designat för

The study shows that (i) working condition challenges were drivers for innovative solution-oriented approaches with potential for rapid decision-making, flexibility, and to attract,

Syftet i föreliggande studie var att undersöka tilltro till de tre olika behandlingsmetoderna psykodynamisk terapi (PDT), kognitiv beteendeterapi (KBT) samt

Forskning visar enligt Ryman (2004) att regelbundna lönesamtal tycks stärka lönesystemets acceptans och legitimitet, vilket stödjer vår slutsats om att försöka skapa intresse