Ajpgi 00399 2020

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 36

Dynamics of the Preterm Gut Microbiome in Health and Disease

2  Authors: Alain Cuna1,2, Michael J. Morowitz3, Ishfaq Ahmed 4, Shahid Umar5,

3  Venkatesh Sampath1,2

4  Affiliations: 1Children’s Mercy Kansas City, Kansas City MO; 2University of Missouri

5  Kansas City, Kansas City MO; 3University of Pittsburgh, Pittsburgh PA; 4Kansas City

6  Kansas Community College, Kansas City KS; 5University of Kansas Medical Center,

7  Kansas City KS

9  Corresponding author: Venkatesh Sampath, MD, Department of Pediatrics, Division of

10  Neonatology, Children’s Mercy Kansas City, 2401 Gillham Rd, Kansas City, MO 64108,

11  Tel: (816) 234-3591, Fax: (816) 234-3590, Email: vsampath@cmh.edu

12 

13  Disclosure statement: The authors have no potential or perceived conflicts of interest
14  to disclose.
15 

16  Statement of financial support: This study was supported by institutional funds at

17  Children’s Mercy Hospital (VS, AC), R01DK117296 (VS) and K08DK125735 (AC).

18 

19 
20  ABSTRACT

21  Advances in metagenomics have allowed a detailed study of the gut microbiome, and

22  its role in human health and disease. Infants born prematurely possess a fragile gut

23  microbial ecosystem that is vulnerable to perturbation. Alterations in the developing gut

24  microbiome in preterm infants are linked to life-threatening diseases such as necrotizing

25  enterocolitis (NEC) and late onset sepsis; and may impact future risk of asthma, atopy,

26  obesity, and psychosocial disease. In this mini review, we summarize recent literature

27  on the origins and patterns of development of the preterm gut microbiome in the

28  perinatal period. The host-microbiome-environmental factors that portend development

29  of dysbiotic intestinal microbial patterns associated with NEC and sepsis are reviewed.

30  Strategies to manipulate the microbiome and mitigate dysbiosis, including the use of

31  probiotics and prebiotics will also be discussed. Finally, we explore the challenges and

32  future directions of gut microbiome research in preterm infants.

33 
34  Introduction

35  The human gastrointestinal (GI) tract harbors a complex and diverse microbial

36  community which is dominated by bacteria, but also includes viruses, archaea, fungi,

37  and other eukaryotes. This complex ecosystem with 1012-14 cells from 100-1000

38  species, is considered to be the most dense microbial habitat on earth (74). Compared

39  to approximately 23,000 genes of human genome, the gut microbiome has been

40  referred to as a ‘hidden’ metabolic organ and encodes over 3 million genes producing

41  thousands of metabolites (78). 90% of the GI microbiota is composed of bacteria from

42  two major phyla namely Bacteroidetes and Firmicutes. Other phyla consistently found in

43  human gut include Proteobacteria, Actinobacteria, Fusobacteria, and Verrucomicrobia

44  (4). At lower taxonomic levels however, individuals differ considerably in composition of

45  fecal microbiota and each individual may have his or her own distinctive pattern of

46  microbial profile (25).

47  Studies suggest that the early pattern of infant gut microbial colonization is

48  critical for proper development of the human GI tract. The neonatal gut microbiota plays

49  an essential role in the acquisition of postnatal intestinal endotoxin tolerance (45) and

50  specifically regulates maturation of regulatory T cells (CD4+, Foxp3+), natural killer cell

51  and gamma delta T cells (69). Dysbiosis or imbalance in gut microbial communities

52  during this critical period is linked to several diseases including inflammatory, metabolic,

53  neurologic, cardiovascular, and GI illnesses (73). Compared to term neonates, infants

54  born prematurely are at greater risk for disruptions to the gut microbiota. Herein, we

55  briefly review commonly used metagenomic methods for microbiome analysis, compare

56  the challenges of preterm versus term gut microbiome, delineate antecedents and
57  consequences of preterm gut dysbiosis, and discuss advances in microbiome

58  modulation therapy in preterm infants. We focus predominantly on the bacterial

59  communities as most research has been done in this area.

60  Methods of microbial profiling with 16S and shotgun sequencing

61  Currently available high-throughput sequencing technologies used to profile the

62  genomic composition of a microbial community in a culture-independent manner can be

63  sub-divided into two different approaches, namely 16S rRNA gene sequencing and

64  shotgun metagenomics. In 16S rRNA sequencing, the 16S rRNA gene is used as a

65  genetic fingerprint to decipher bacterial phylogeny and taxonomy. A component of the

66  30S subunit of prokaryotic ribosome, the 16S rRNA gene is 1500 base pair long and is

67  comprised of nine variable regions (V1 – V9) interspersed among the conserved regions

68  of its sequence. These hypervariable regions are unique to each bacterial taxon while

69  the conserved regions allow for the development of universal primers that bind to known

70  sequences shared among most bacteria. During data analysis, amplified 16S rRNA

71  sequences are assigned to Operational Taxonomic Units (OTUs) and OTU clustering

72  methods using a moderately stringent sequence identity threshold allowing for basic

73  taxonomic assignments (family level or higher) (26). Amplicon sequencing targeting the

74  16S rRNA gene is limited by differences arising from the different variable region

75  chosen for PCR amplification, sequencing errors, poor discriminatory power for some

76  genera, and severely limited resolution at the species level due to short-read amplicon

77  sequencing, potential for contamination, and sequencing platform used (64, 76, 87).

78  Unlike 16S sequencing which only targets 16S rRNA sequences, shotgun

79  metagenomics generates whole genome sequencing data that can be compared to
80  existing reference databases for accurate taxonomic identification. By sequencing all

81  genetic information within a sample, shotgun metagenomic studies can extract both

82  taxonomic and functional information from complex microbial communities and therefore

83  guide phenotypic studies to understand their potential roles in health and disease. While

84  shotgun metagenomics can be used to extract species- and even strain-level

85  information through computational approaches, it can still be challenging to differentiate

86  between closely related or co-existing bacterial taxa (65). This has mandated the need

87  for extensive and well-characterized collections of reference genomes including those

88  from the Human Microbiome Project and the Human Gastrointestinal Bacteria Genome

89  Collection (28, 63).

90  Challenges of the preterm infant that shapes the gut microbiome in comparison

91  with the term infant

92  Preterm infants face several unique environmental and host conditions that negatively

93  impact development of their gut microbiome. Even before birth, about 25% to 30% of

94  preterm infants are exposed to microbes in the context of preterm premature rupture of

95  membranes and intra-amniotic infection (32). Cesarean section, with subsequent gut

96  colonization by skin microbiota rather than vaginal and rectal microbiota from vaginal

97  delivery, is also more common in preterm than term infants (67). After delivery, most

98  preterm infants need stabilization and subsequent care in the neonatal intensive care

99  unit which, while necessary, is also invasive and results in exposure to the hospital

100  microbial environment. Consequently, as risk of serious nosocomial infection is high,

101  preterm infants are often exposed to powerful broad-spectrum antibiotics throughout

102  their hospital stay. Preterm nutrition and feeding practices – such as delayed
103  introduction of enteral feedings, frequent withholding of feeds, use of acid-suppressive

104  medications, and provision of exclusive human milk diet or formula – are also major

105  modifiers of gut microbiota composition (13). Together, these environmental factors

106  interact with the intrinsically immature GI tract and immune system of the preterm infant

107  in a highly complex but poorly defined process that significantly impact the developing

108  preterm gut microbiome (36, 85).

109  In general, the intestinal microbiota in the preterm infant differs from the term

110  infant in having delayed colonization, fewer bacterial species, and less diversity and

111  abundance (2, 36, 59). There is also a predilection to being colonized by potentially

112  pathogenic facultative anaerobes (e.g. Enterobacter, Escherichia, and Klebsiella) and

113  decreased levels of commensal strictly anaerobic organisms (e.g. Bifidobacterium,

114  Bacteroides, and Clostridium) (Table 1) (34). A shared patterned progression of

115  microbial communities from Bacilli to Gammaproteobacteria to Clostridia has also been

116  described in preterm infants (41). The pace of this patterned progression is

117  predominantly influenced by gestational age, while mode of delivery, antibiotic

118  exposure, and type of feeding appear to have less impact (41).

119  Whether the differences in gut microbiota of preterm versus term infant

120  constitutes true dysbiosis or merely represents the common gut microbiome signature in

121  very low birth weight infants is debatable. Current studies do not clarify whether a

122  distinct pattern of dysbiosis that portends disease susceptibility exists, apart from a

123  modest increase in Enterobacteriaceae in infants who develop necrotizing enterocolitis

124  (NEC), as further discussed below.

125  Antecedents of dysbiosis in preterm infants


126  Prenatal factors. The development of intestinal microbiome in the preterm neonate and

127  the proclivity to a dysbiotic signature is influenced by several prenatal factors (Fig 1).

128  Maternal gestational diet appears to alter the composition of the first stool after birth

129  (meconium), with depletion of Bacteroides observed with high-fat diet (9). Other studies

130  have described an interactive effect between maternal diet and mode of delivery on the

131  neonatal gut microbiome, with fruit intake increasing intestinal Streptococcus/

132  Clostridium in infants delivered vaginally, while increased dairy intake was associated

133  with higher abundance of Clostridium among infants by Cesarean section (46). In

134  pregnancies complicated by intrauterine amniotic infection or chorioamnionitis, a higher

135  relative abundance of Mycoplasmataceae and phylum Bacteroidetes has been identified

136  in stool of infants postnatally (62). While previous studies had suggested the presence

137  of microbial communities in the placenta and amniotic fluid of healthy pregnancies

138  potentially colonizing the gut prenatally, more vigorous recent studies have disproved

139  the existence of a distinct placental microbiome (14, 20).

140  Intrapartum antibiotics (IAP) administered to prevent Group B streptococcal

141  transmission has also been shown to have significant early effects on the composition

142  of gut microbiome in neonates that persist through the first year of life (12, 15, 56).

143  Nogacka et al. (56) showed a persistent increase in Proteobacteria and Firmicutes with

144  decreases in Acinetobacter and Bacteroides in association with higher occurrence of

145  some bacterial genes that code for β-lactamase resistance. Others have shown that the

146  use of IAP was associated with decreases in Bifidobacteria spp at 7 days of life, while

147  Lactobacillus spp remains unchanged (15). Further, the effects of IAP on the infant

148  microbiome may be selective with penicillin suppressing Bacteroides spp., and
149  cephalosporin delaying the increase in Bifidobacteria (12). These studies suggest that

150  maternal gestational diet, chorioamnionitis and IAP influence the composition of the

151  neonatal microbiome and these effects can last well beyond the neonatal period.

152  Postnatal factors. Several postnatal factors influence the development of gut

153  microbiome and can program intestinal dysbiosis (Fig 1). The mode of birth, i.e., vaginal

154  vs. cesarean, has been shown to influence the neonatal microbiome (10, 23, 67).

155  Infants born by cesarean section have less intestinal microbial diversity, decreased

156  colonization with Bifidobacteria, Bacteroides and Lactobacilli, and tended to have

157  increased skin microbiota such as Streptococcus and Staphylococcus in the first weeks

158  after birth (23, 67). Interestingly, differences related to mode of delivery are less evident

159  with maturity, likely because other factors like postnatal diet impact the gut microbiota

160  (10).

161  Early exposure to parenteral antibiotics is also a risk factor for gut dysbiosis (27,

162  72). Fouhy et al. (29) demonstrated that early (<48hr) antibiotic treatment with ampicillin

163  and gentamicin was associated with increases in Proteobacteria and decreased

164  abundance of Actinobacteria, Bifidobacteria and Lactobacillus. Gibson et al. (31)

165  demonstrated that broad spectrum antibiotics such as meropenem, cephalosporins and

166  ticarcillin-clavulanate decreased species richness whereas vancomycin and gentamicin

167  had non-uniform results. They also noted emergence of antibiotics resistant genes that

168  was antibiotic- and species-specific. Zwittink et al. (90) noted that the use of short- or

169  long-term amoxicillin/ceftazidime treatment was associated with significant higher

170  abundance of Enterococcus during the first two postnatal weeks at the expense of

171  Bifidobacterium and Streptococcus.


172  Nutrition is also a strong modifier of the of the gut microbiome. Preterm infants

173  fed with formula milk exhibit a less diverse gut microbiome with less enrichment of

174  Clostridiales and Bifidobacteria compared to preterm infants fed mothers’ own milk (84).

175  In summary, major postnatal factors linked to a dysbiotic gut signature in preterm infants

176  include mode of delivery, exposure to antibiotics and other medications, and type of milk

177  feeding.

178  Consequences of dysbiosis in preterm infants

179  Necrotizing enterocolitis (NEC). NEC is the most widely recognized consequence of

180  gut dysbiosis in preterm infants (Fig 1). The role of gut bacteria in NEC is demonstrated

181  from mouse models showing absence of disease in a germfree environment, direct

182  studies of human stools showing dysbiotic signatures, and demonstration of bacterial

183  invasion by fluorescent techniques in surgically resected human tissues (39, 66, 79).

184  Clusters of NEC outbreaks related to specific organisms that have varied by institution

185  have also been reported, providing additional evidence of the role of microbes in NEC

186  pathogenesis. With advances in metagenomics, several studies have demonstrated that

187  preterm infants who develop NEC have altered gut microbiota signatures compared to

188  preterm infants who do not develop NEC, though these signatures have varied between

189  studies (50).

190  In a small study, Heida et al. (37) identified enrichment of Clostridrium

191  perfringens in meconium of preterm infants who subsequently developed NEC,

192  suggesting that a pathogenic microbial signature for NEC may be present in the very

193  first days of life. However, meconial or very early stool signatures for NEC have not

194  been demonstrated in subsequent studies (59, 82). In one of largest longitudinal studies
195  in preterm infants, Warner et al. (82) studied >2400 stool samples from 122 infants born

196  with a birthweight <1500g to demonstrate that NEC was associated with

197  Gammaproteobacteria abundance, and relative depletion of Negativicutes, an

198  anaerobe. Interestingly, these patterns emerged typically after the first 4 weeks of life.

199  Other interesting observations were that antibiotic use increased proportions of Bacilli

200  while vaginal birth was associated with decreased proportions of Bacilli, and the volume

201  of human milk feeds did not impact bacterial community architecture.

202  Similar to the landmark study by Warner et al. (82), several investigators have

203  reported an enrichment of potentially pathogenic Gram-negative bacteria in the intestine

204  in addition to a relative decrease in the abundance of Firmicutes and Bacteroides prior

205  to the onset of NEC (51, 57, 59, 81, 82). Furthermore, while preterm infants in general

206  have low gut microbial diversity compared to term infants, several studies have noted

207  further reduced levels of diversity in preterm infants who develop NEC compared to

208  preterm infants without NEC (22, 50, 59, 70). A meta-analysis of published studies

209  confirmed a relative abundance of Gammaproteobacteria, with decreased relative

210  proportions of Firmicutes and Bacteroides preceding NEC, but did not confirm reduced

211  microbial diversity prior to NEC onset (59). Gammaproteobacteria, which include

212  several well-known pathogenic species that have the endotoxin lipopolysaccharide, can

213  induce excessive intestinal and systemic inflammation through stimulation of Toll-like

214  receptors (39); while Firmicutes and Bacteroides can enhance gut development by

215  facilitating intestinal epithelial cell differentiation and preserving mucin and tight junction

216  integrity (88).


217  While these studies have primarily focused on bacterial signatures, a potential

218  role for viruses such as norovirus, rotavirus, and cytomegalovirus (CMV) in NEC

219  causation is also recognized (11, 50). For example, case reports (75) have described

220  infants with CMV-associated NEC, and retrospective studies using surgical specimens

221  (58, 60) have detected CMV in intestinal tissue of preterm infants with NEC or

222  spontaneous intestinal perforation. Infants with CMV-associated NEC tend to have

223  lower gestational age and thrombocytopenia than non-CMV associated cases. The

224  relative impact of viruses on NEC remains poorly characterized as reported rates of

225  detection of CMV and other viruses can vary depending on the sample (blood, stool,

226  intestinal tissue), technique (immunohistochemistry, polymerase chain reaction,

227  enzyme-linked immunosorbent assay), and sample size of NEC cohort (7, 71, 77). The

228  potential role of fungi and the mycobiome in NEC also remain largely unexplored (38).

229  While metagenomic studies provide a snapshot of microbial patterns associated

230  with NEC, more rigorous studies are needed to elucidate their mechanistic role and

231  establish their contribution in NEC. More recently, multi-omic studies combining

232  metagenomics, metatranscriptomics, and metabolomics are beginning to shed new light

233  on bacterial replication rates, quorum sensing patterns, and metabolic profiles

234  associated with intestinal dysbiosis preceding NEC (57, 59, 70). For example, Morrow

235  et al. (51) evaluated the urinary metabolome and gut microbiome of 11 preterm infants

236  with NEC and 21 matched controls. Using this two complementary “-omic” approaches,

237  the authors found that the urinary metabolite alanine was directly correlated with

238  Firmicutes and indirectly correlated with Proteobacteria in the gut, suggesting that

239  specific intestinal bacteria can influence production and utilization of specific
240  metabolites which may be useful as surrogate biomarkers for early diagnosis of NEC.

241  While still at its infancy, these novel multi-omic approaches are anticipated to further

242  advance our understanding and provide a full mechanistic picture of NEC pathogenesis

243  (54).

244  Sepsis and other diseases. The consequences of dysbiosis in the preterm infants

245  extend beyond the predisposition to develop NEC (Fig 1). Stewart et al. (72) noted that

246  the bacteria isolated from a blood culture during sepsis corresponded to the dominant

247  bacterial genera in the gut microbiome, and that Bifidobacteria abundance was

248  observed in infants who did not develop sepsis or NEC. Other studies have also noted

249  decreased diversity and lower abundance of Bifidobacteria among infants who

250  developed sepsis (47). Potential mechanisms by which Bifidobacteria can protect

251  against sepsis include reduced bacterial translocation through enhanced intestinal

252  barrier function and increased production of beneficial short-chain fatty acids like

253  acetate (30, 43). Sepsis related to antibiotic-resistant strains of bacteria originating from

254  the gut are also increased in preterm infants treated with broad spectrum antibiotics that

255  increase dysbiosis (31, 72, 82). While not the focus here, dysbiosis in preterm infants

256  has also been linked to bronchopulmonary dysplasia (a chronic debilitating lung

257  condition of infants) and growth failure (68, 86).

258  Potential long-term consequences. Epidemiological studies suggest a link between

259  early disturbances to the gut microbiome from antibiotics or cesarean delivery with later

260  childhood diseases including allergy (3, 48, 55), obesity (1, 42), and attention deficit

261  hyperactivity disorder (6). A recent population-based cohort study using the Rochester

262  Epidemiological Project found similar associations between early life antibiotic exposure
263  and increased risk for several childhood immunological, metabolic, and neurobehavioral

264  health conditions (5). While these studies did not differentiate term from preterm infants,

265  the aforementioned stressors that preterm infants experience in the hospital

266  environment puts them at greatest risk for early gut microbial dysbiosis (35). Additional

267  studies in mice provide supporting evidence for the hypothesis that early life

268  perturbations during key developmental periods can have long-term consequences. For

269  example, in a mouse model of early gut microbial disruption with low-dose penicillin,

270  long-lasting metabolic effects of increased fat mass were observed even with limited

271  antibiotic administration where microbial communities recovered after cessation of

272  antibiotics (16).

273  Microbiome modulation therapy

274  With accumulating evidence of how gut microbiome perturbations contribute to preterm

275  diseases, novel strategies that reverse or alleviate dysbiosis are being explored.

276  Probiotics. Probiotics are defined as living microorganisms that confer health benefits

277  when administered in adequate amounts. In addition to favorable alteration of gut

278  microbiome, probiotics are now recognized to confer benefit through other mechanisms

279  such as colonization resistance against pathogenic bacteria, enhanced gut barrier

280  function, and immunomodulation (18). Numerous clinical trials have demonstrated the

281  safety and efficacy of prophylactic administration of probiotics in preterm infants for the

282  prevention of NEC. A recent network meta-analysis (63 trials, 15,712 preterm infants)

283  identified that a multi-species combination that contains Lactobacillus and

284  Bifidobacterium provided superior benefits for NEC prevention compared to single
285  species or other multi-species probiotic combinations for reducing mortality and

286  necrotizing enterocolitis in preterm infants (49).

287  Prebiotics. Prebiotics are nutrient substrates that selectively enrich beneficial

288  microbiota in the gut. The most important source of prebiotics for preterm infants is

289  human milk oligosaccharides (HMO) - complex sugars not otherwise digestible by the

290  human gut but utilized by specific microbes such as Bifidobacteria and Bacteroides to

291  promote their growth and activity. In addition to stimulating probiotic growth, prebiotics

292  also help improve intestinal mobility and gut barrier function. Artificial or non-human milk

293  oligosaccharides designed to function in a similar manner to HMOs have also been

294  tested in small clinical trials with promising results (8). Other active areas of

295  investigation include synbiotics which are combination products that contain both

296  prebiotics and probiotics (53).

297  Postbiotics. Postbiotics are metabolic byproducts of probiotic bacteria that can exert

298  positive biologic activity in the host. In a recent study, Zheng et al. (89) demonstrated

299  that short chain fatty acids, such as butyrate and acetate produced by Bifidobacterium

300  infantis from metabolism of complex carbohydrates expressed in breastmilk, have anti-

301  inflammatory effects in immature enterocytes. Other studies have shown how butyrate,

302  can also enhance intestinal barrier function and regulate mucosal immunity (44). By

303  using metabolites instead of bacteria, postbiotics potentially provide an effective,

304  simpler, and safer alternative to ingestion of live microorganisms. A related strategy is

305  the use of para-probiotics, which are inactivated microbial cells or cell fractions typically

306  produced by heat-killing probiotics (21).


307  Fecal microbiota transplantation (FMT). In FMT, fecal material from a healthy donor

308  is transplanted to the recipient’s GI tract to restore microbial homeostasis. Compared to

309  probiotics, FMT allows more robust and longer-lasting community of diverse microbes

310  with a single-dose. Safety concerns include inadvertent introduction or promotion of

311  pathogenic species and transference of antibiotic resistance. FMT in preterm infants

312  remains largely unexplored except in experimental animal research (61). A similar

313  thematic approach is vaginal seeding, wherein bacteria from the mother’s vaginal tract

314  is transferred to the infants face and body when infants are born via cesarean section

315  (52).

316  Conclusion

317  The concept of the holobiont, consisting of the human host and the resident microbial

318  communities, is important in considering perturbations of intestinal microbial assembly

319  in preterm infants (24, 73). The development of a symbiotic intestinal ecosystem is a

320  central event for successful adaptation to postnatal life. Maternal disease, intrauterine

321  infection, and perinatal use of antibiotics can disrupt establishment of co-evolved

322  microbial communities, which are integral for the optimal function and development of

323  the intestine, the immune system, the brain, and other physiological processes (24, 73).

324  Restoration to a normal microbial assembly is further impeded in the preterm neonate

325  fed formula milk, exposed to long term antibiotics and deviant microbial dispersal

326  patterns in a pathogen-rich milieu, and microbial community priority effects. These

327  adverse influences result in the establishment of a dysbiotic microbial signature

328  characterized by a relative abundance in Gammaproteobacteria, decreased diversity,

329  and a modest decrease in Bifidobacteria. Several studies link these dysbiotic signatures
330  to increased vulnerability to life-threatening neonatal illness such as NEC and sepsis,

331  and increasingly to childhood asthma, allergic disease, disorders of mood and obesity

332  (24, 73). Efforts at restoring microbial community structure using probiotics have shown

333  promise in alleviating NEC and sepsis risk. Other approaches based on prebiotics or

334  enteral immune therapy, however, warrant careful trials. Understanding the role of host

335  genetics in programming the neonatal gut microbial assembly, combining metabolomic

336  and transcriptomic signatures concurrent with dysbiotic microbiota signatures, and

337  personalized approaches in reconstituting the deviant microbial assembly in preterm

338  neonates to prevent disease and ensure healthy outcomes remain highly significant

339  areas for translational research (17, 40, 70).

340 
341  ACKNOWLEDGMENTS

342  Fig 1 created using www.biorender.com.

343 

344 

345 

346 

347 
348  REFERENCES

349  1. Ajslev TA, Andersen CS, Gamborg M, Sorensen TI, and Jess T. Childhood

350  overweight after establishment of the gut microbiota: the role of delivery mode, pre-

351  pregnancy weight and early administration of antibiotics. Int J Obes (Lond) 35: 522-529,

352  2011.

353  2. Arboleya S, Sanchez B, Milani C, Duranti S, Solis G, Fernandez N, de los

354  Reyes-Gavilan CG, Ventura M, Margolles A, and Gueimonde M. Intestinal microbiota

355  development in preterm neonates and effect of perinatal antibiotics. The Journal of

356  pediatrics 166: 538-544, 2015.

357  3. Arrieta MC, Stiemsma LT, Dimitriu PA, Thorson L, Russell S, Yurist-

358  Doutsch S, Kuzeljevic B, Gold MJ, Britton HM, Lefebvre DL, Subbarao P,

359  Mandhane P, Becker A, McNagny KM, Sears MR, Kollmann T, Investigators CS,

360  Mohn WW, Turvey SE, and Finlay BB. Early infancy microbial and metabolic

361  alterations affect risk of childhood asthma. Sci Transl Med 7: 307ra152, 2015.

362  4. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR,

363  Fernandes GR, Tap J, Bruls T, Batto JM, Bertalan M, Borruel N, Casellas F,

364  Fernandez L, Gautier L, Hansen T, Hattori M, Hayashi T, Kleerebezem M,

365  Kurokawa K, Leclerc M, Levenez F, Manichanh C, Nielsen HB, Nielsen T, Pons N,

366  Poulain J, Qin J, Sicheritz-Ponten T, Tims S, Torrents D, Ugarte E, Zoetendal EG,

367  Wang J, Guarner F, Pedersen O, de Vos WM, Brunak S, Dore J, Antolin M,

368  Artiguenave F, Blottiere HM, Almeida M, Brechot C, Cara C, Chervaux C, Cultrone

369  A, Delorme C, Denariaz G, Dervyn R, Foerstner KU, Friss C, van de Guchte M,

370  Guedon E, Haimet F, Huber W, van Hylckama-Vlieg J, Jamet A, Juste C, Kaci G,


371  Knol J, Lakhdari O, Layec S, Le Roux K, Maguin E, Merieux A, Melo Minardi R,

372  M'Rini C, Muller J, Oozeer R, Parkhill J, Renault P, Rescigno M, Sanchez N,

373  Sunagawa S, Torrejon A, Turner K, Vandemeulebrouck G, Varela E, Winogradsky

374  Y, Zeller G, Weissenbach J, Ehrlich SD, and Bork P. Enterotypes of the human gut

375  microbiome. Nature 473: 174-180, 2011.

376  5. Aversa Z, Atkinson EJ, Schafer MJ, Theiler RN, Rocca WA, Blaser MJ, and

377  LeBrasseur NK. Association of Infant Antibiotic Exposure With Childhood Health

378  Outcomes. Mayo Clin Proc 2020.

379  6. Axelsson PB, Clausen TD, Petersen AH, Hageman I, Pinborg A, Kessing LV,

380  Bergholt T, Rasmussen SC, Keiding N, and Lokkegaard ECL. Investigating the

381  effects of cesarean delivery and antibiotic use in early childhood on risk of later attention

382  deficit hyperactivity disorder. J Child Psychol Psychiatry 60: 151-159, 2019.

383  7. Cheng C, He Y, Xiao S, Ai Q, and Yu J. The association between enteric

384  viruses and necrotizing enterocolitis. Eur J Pediatr 2020.

385  8. Chi C, Buys N, Li C, Sun J, and Yin C. Effects of prebiotics on sepsis,

386  necrotizing enterocolitis, mortality, feeding intolerance, time to full enteral feeding,

387  length of hospital stay, and stool frequency in preterm infants: a meta-analysis.

388  European journal of clinical nutrition 73: 657-670, 2019.

389  9. Chu DM, Antony KM, Ma J, Prince AL, Showalter L, Moller M, and Aagaard

390  KM. The early infant gut microbiome varies in association with a maternal high-fat diet.

391  Genome medicine 8: 77, 2016.


392  10. Chu DM, Ma J, Prince AL, Antony KM, Seferovic MD, and Aagaard KM.

393  Maturation of the infant microbiome community structure and function across multiple

394  body sites and in relation to mode of delivery. Nature medicine 23: 314-326, 2017.

395  11. Coggins SA, Wynn JL, and Weitkamp JH. Infectious causes of necrotizing

396  enterocolitis. Clinics in perinatology 42: 133-154, ix, 2015.

397  12. Coker MO, Hoen AG, Dade E, Lundgren S, Li Z, Wong AD, Zens MS, Palys

398  TJ, Morrison HG, Sogin ML, Baker ER, Karagas MR, and Madan JC. Specific class

399  of intrapartum antibiotics relates to maturation of the infant gut microbiota: a prospective

400  cohort study. BJOG : an international journal of obstetrics and gynaecology 127: 217-

401  227, 2020.

402  13. Collado MC, Cernada M, Neu J, Perez-Martinez G, Gormaz M, and Vento M.

403  Factors influencing gastrointestinal tract and microbiota immune interaction in preterm

404  infants. Pediatric research 77: 726-731, 2015.

405  14. Collado MC, Rautava S, Aakko J, Isolauri E, and Salminen S. Human gut

406  colonisation may be initiated in utero by distinct microbial communities in the placenta

407  and amniotic fluid. Scientific reports 6: 23129, 2016.

408  15. Corvaglia L, Tonti G, Martini S, Aceti A, Mazzola G, Aloisio I, Di Gioia D, and

409  Faldella G. Influence of Intrapartum Antibiotic Prophylaxis for Group B Streptococcus

410  on Gut Microbiota in the First Month of Life. Journal of pediatric gastroenterology and

411  nutrition 62: 304-308, 2016.

412  16. Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, Kim SG,

413  Li H, Gao Z, Mahana D, Zarate Rodriguez JG, Rogers AB, Robine N, Loke P, and
414  Blaser MJ. Altering the intestinal microbiota during a critical developmental window has

415  lasting metabolic consequences. Cell 158: 705-721, 2014.

416  17. Cuna A, George L, and Sampath V. Genetic predisposition to necrotizing

417  enterocolitis in premature infants: Current knowledge, challenges, and future directions.

418  Semin Fetal Neonatal Med 23: 387-393, 2018.

419  18. Cuna A, Yu W, Menden HL, Feng L, Srinivasan P, Chavez-Bueno S, Ahmed

420  I, Umar S, and Sampath V. NEC-like intestinal injury is ameliorated by Lactobacillus

421  rhamnosus GG in parallel with SIGIRR and A20 induction in neonatal mice. Pediatric

422  research 88: 546-555, 2020.

423  19. Dahl C, Stigum H, Valeur J, Iszatt N, Lenters V, Peddada S, Bjornholt JV,

424  Midtvedt T, Mandal S, and Eggesbo M. Preterm infants have distinct microbiomes not

425  explained by mode of delivery, breastfeeding duration or antibiotic exposure. Int J

426  Epidemiol 47: 1658-1669, 2018.

427  20. de Goffau MC, Lager S, Sovio U, Gaccioli F, Cook E, Peacock SJ, Parkhill J,

428  Charnock-Jones DS, and Smith GCS. Human placenta has no microbiome but can

429  contain potential pathogens. Nature 572: 329-334, 2019.

430  21. Deshpande G, Athalye-Jape G, and Patole S. Para-probiotics for Preterm

431  Neonates-The Next Frontier. Nutrients 10: 2018.

432  22. Dobbler PT, Procianoy RS, Mai V, Silveira RC, Corso AL, Rojas BS, and

433  Roesch LFW. Low Microbial Diversity and Abnormal Microbial Succession Is

434  Associated with Necrotizing Enterocolitis in Preterm Infants. Frontiers in microbiology 8:

435  2243, 2017.


436  23. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G,

437  Fierer N, and Knight R. Delivery mode shapes the acquisition and structure of the

438  initial microbiota across multiple body habitats in newborns. Proceedings of the National

439  Academy of Sciences of the United States of America 107: 11971-11975, 2010.

440  24. Dominguez-Bello MG, Godoy-Vitorino F, Knight R, and Blaser MJ. Role of

441  the microbiome in human development. Gut 68: 1108-1114, 2019.

442  25. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill

443  SR, Nelson KE, and Relman DA. Diversity of the human intestinal microbial flora.

444  Science 308: 1635-1638, 2005.

445  26. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon

446  reads. Nat Methods 10: 996-998, 2013.

447  27. Esaiassen E, Fjalstad JW, Juvet LK, van den Anker JN, and Klingenberg C.

448  Antibiotic exposure in neonates and early adverse outcomes: a systematic review and

449  meta-analysis. The Journal of antimicrobial chemotherapy 72: 1858-1870, 2017.

450  28. Forster SC, Kumar N, Anonye BO, Almeida A, Viciani E, Stares MD, Dunn M,

451  Mkandawire TT, Zhu A, Shao Y, Pike LJ, Louie T, Browne HP, Mitchell AL, Neville

452  BA, Finn RD, and Lawley TD. A human gut bacterial genome and culture collection for

453  improved metagenomic analyses. Nat Biotechnol 37: 186-192, 2019.

454  29. Fouhy F, Guinane CM, Hussey S, Wall R, Ryan CA, Dempsey EM, Murphy B,

455  Ross RP, Fitzgerald GF, Stanton C, and Cotter PD. High-throughput sequencing

456  reveals the incomplete, short-term recovery of infant gut microbiota following parenteral

457  antibiotic treatment with ampicillin and gentamicin. Antimicrobial agents and

458  chemotherapy 56: 5811-5820, 2012.


459  30. Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T,

460  Clarke JM, Topping DL, Suzuki T, Taylor TD, Itoh K, Kikuchi J, Morita H, Hattori M,

461  and Ohno H. Bifidobacteria can protect from enteropathogenic infection through

462  production of acetate. Nature 469: 543-547, 2011.

463  31. Gibson MK, Wang B, Ahmadi S, Burnham CA, Tarr PI, Warner BB, and

464  Dantas G. Developmental dynamics of the preterm infant gut microbiota and antibiotic

465  resistome. Nature microbiology 1: 16024, 2016.

466  32. Goldenberg RL, Culhane JF, Iams JD, and Romero R. Epidemiology and

467  causes of preterm birth. Lancet 371: 75-84, 2008.

468  33. Gorreja F, Rush ST, Kasper DL, Meng D, and Walker WA. The

469  developmentally regulated fetal enterocyte gene, ZP4, mediates anti-inflammation by

470  the symbiotic bacterial surface factor polysaccharide A on Bacteroides fragilis. Am J

471  Physiol Gastrointest Liver Physiol 317: G398-G407, 2019.

472  34. Gritz EC, and Bhandari V. The human neonatal gut microbiome: a brief review.

473  Frontiers in pediatrics 3: 17, 2015.

474  35. Groer MW, Gregory KE, Louis-Jacques A, Thibeau S, and Walker WA. The

475  very low birth weight infant microbiome and childhood health. Birth Defects Res C

476  Embryo Today 105: 252-264, 2015.

477  36. Groer MW, Luciano AA, Dishaw LJ, Ashmeade TL, Miller E, and Gilbert JA.

478  Development of the preterm infant gut microbiome: a research priority. Microbiome 2:

479  38, 2014.

480  37. Heida FH, van Zoonen A, Hulscher JBF, Te Kiefte BJC, Wessels R, Kooi

481  EMW, Bos AF, Harmsen HJM, and de Goffau MC. A Necrotizing Enterocolitis-
482  Associated Gut Microbiota Is Present in the Meconium: Results of a Prospective Study.

483  Clinical infectious diseases : an official publication of the Infectious Diseases Society of

484  America 62: 863-870, 2016.

485  38. James SA, Phillips S, Telatin A, Baker D, Ansorge R, Clarke P, Hall LJ, and

486  Carding SR. Preterm Infants Harbour a Rapidly Changing Mycobiota That Includes

487  Candida Pathobionts. J Fungi (Basel) 6: 2020.

488  39. Jilling T, Simon D, Lu J, Meng FJ, Li D, Schy R, Thomson RB, Soliman A,

489  Arditi M, and Caplan MS. The roles of bacteria and TLR4 in rat and murine models of

490  necrotizing enterocolitis. J Immunol 177: 3273-3282, 2006.

491  40. Kurilshikov A, Wijmenga C, Fu J, and Zhernakova A. Host Genetics and Gut

492  Microbiome: Challenges and Perspectives. Trends in immunology 38: 633-647, 2017.

493  41. La Rosa PS, Warner BB, Zhou Y, Weinstock GM, Sodergren E, Hall-Moore

494  CM, Stevens HJ, Bennett WE, Jr., Shaikh N, Linneman LA, Hoffmann JA, Hamvas

495  A, Deych E, Shands BA, Shannon WD, and Tarr PI. Patterned progression of

496  bacterial populations in the premature infant gut. Proc Natl Acad Sci U S A 111: 12522-

497  12527, 2014.

498  42. Leong KSW, McLay J, Derraik JGB, Gibb S, Shackleton N, Taylor RW,

499  Glover M, Audas R, Taylor B, Milne BJ, and Cutfield WS. Associations of Prenatal

500  and Childhood Antibiotic Exposure With Obesity at Age 4 Years. JAMA Netw Open 3:

501  e1919681, 2020.

502  43. Ling X, Linglong P, Weixia D, and Hong W. Protective Effects of

503  Bifidobacterium on Intestinal Barrier Function in LPS-Induced Enterocyte Barrier Injury

504  of Caco-2 Monolayers and in a Rat NEC Model. PLoS One 11: e0161635, 2016.
505  44. Liu J, Zhu H, Li B, Lee C, Alganabi M, Zheng S, and Pierro A. Beneficial

506  effects of butyrate in intestinal injury. Journal of pediatric surgery 55: 1088-1093, 2020.

507  45. Lotz M, Gutle D, Walther S, Menard S, Bogdan C, and Hornef MW. Postnatal

508  acquisition of endotoxin tolerance in intestinal epithelial cells. J Exp Med 203: 973-984,

509  2006.

510  46. Lundgren SN, Madan JC, Emond JA, Morrison HG, Christensen BC,

511  Karagas MR, and Hoen AG. Maternal diet during pregnancy is related with the infant

512  stool microbiome in a delivery mode-dependent manner. Microbiome 6: 109, 2018.

513  47. Mai V, Torrazza RM, Ukhanova M, Wang X, Sun Y, Li N, Shuster J, Sharma

514  R, Hudak ML, and Neu J. Distortions in development of intestinal microbiota

515  associated with late onset sepsis in preterm infants. PloS one 8: e52876, 2013.

516  48. Mitre E, Susi A, Kropp LE, Schwartz DJ, Gorman GH, and Nylund CM.

517  Association Between Use of Acid-Suppressive Medications and Antibiotics During

518  Infancy and Allergic Diseases in Early Childhood. JAMA Pediatr 172: e180315, 2018.

519  49. Morgan RL, Preidis GA, Kashyap PC, Weizman AV, Sadeghirad B,

520  McMaster Probiotic P, and Synbiotic Work G. Probiotics Reduce Mortality and

521  Morbidity in Preterm, Low-Birth-Weight Infants: A Systematic Review and Network

522  Meta-analysis of Randomized Trials. Gastroenterology 159: 467-480, 2020.

523  50. Morowitz MJ, Poroyko V, Caplan M, Alverdy J, and Liu DC. Redefining the

524  role of intestinal microbes in the pathogenesis of necrotizing enterocolitis. Pediatrics

525  125: 777-785, 2010.

526  51. Morrow AL, Lagomarcino AJ, Schibler KR, Taft DH, Yu Z, Wang B, Altaye M,

527  Wagner M, Gevers D, Ward DV, Kennedy MA, Huttenhower C, and Newburg DS.
528  Early microbial and metabolomic signatures predict later onset of necrotizing

529  enterocolitis in preterm infants. Microbiome 1: 13, 2013.

530  52. Mueller NT, Dominguez-Bello MG, Appel LJ, and Hourigan SK. 'Vaginal

531  seeding' after a caesarean section provides benefits to newborn children: FOR: Does

532  exposing caesarean-delivered newborns to the vaginal microbiome affect their chronic

533  disease risk? The critical need for trials of 'vaginal seeding' during caesarean section.

534  BJOG : an international journal of obstetrics and gynaecology 127: 301, 2020.

535  53. Nandhini LP, Biswal N, Adhisivam B, Mandal J, Bhat BV, and Mathai B.

536  Synbiotics for decreasing incidence of necrotizing enterocolitis among preterm neonates

537  - a randomized controlled trial. The journal of maternal-fetal & neonatal medicine : the

538  official journal of the European Association of Perinatal Medicine, the Federation of Asia

539  and Oceania Perinatal Societies, the International Society of Perinatal Obstet 29: 821-

540  825, 2016.

541  54. Neu J. Necrotizing Enterocolitis: A Multi-omic Approach and the Role of the

542  Microbiome. Dig Dis Sci 65: 789-796, 2020.

543  55. Ni J, Friedman H, Boyd BC, McGurn A, Babinski P, Markossian T, and

544  Dugas LR. Early antibiotic exposure and development of asthma and allergic rhinitis in

545  childhood. BMC Pediatr 19: 225, 2019.

546  56. Nogacka A, Salazar N, Suarez M, Milani C, Arboleya S, Solis G, Fernandez

547  N, Alaez L, Hernandez-Barranco AM, de Los Reyes-Gavilan CG, Ventura M, and

548  Gueimonde M. Impact of intrapartum antimicrobial prophylaxis upon the intestinal

549  microbiota and the prevalence of antibiotic resistance genes in vaginally delivered full-

550  term neonates. Microbiome 5: 93, 2017.


551  57. Olm MR, Bhattacharya N, Crits-Christoph A, Firek BA, Baker R, Song YS,

552  Morowitz MJ, and Banfield JF. Necrotizing enterocolitis is preceded by increased gut

553  bacterial replication, Klebsiella, and fimbriae-encoding bacteria. Science advances 5:

554  eaax5727, 2019.

555  58. Omarsdottir S, Agnarsdottir M, Casper C, Orrego A, Vanpee M, Rahbar A,

556  and Soderberg-Naucler C. High prevalence of cytomegalovirus infection in surgical

557  intestinal specimens from infants with necrotizing enterocolitis and spontaneous

558  intestinal perforation: A retrospective observational study. J Clin Virol 93: 57-64, 2017.

559  59. Pammi M, Cope J, Tarr PI, Warner BB, Morrow AL, Mai V, Gregory KE, Kroll

560  JS, McMurtry V, Ferris MJ, Engstrand L, Lilja HE, Hollister EB, Versalovic J, and

561  Neu J. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: a

562  systematic review and meta-analysis. Microbiome 5: 31, 2017.

563  60. Panesso-Gomez S, Shimamura M, Conces M, Talavera MM, Moallem M,

564  Sanchez PJ, and Malleske DT. Detection of Cytomegalovirus in Intestinal Tissue of

565  Infants with Necrotizing Enterocolitis or Spontaneous Intestinal Perforation. J Pediatr

566  214: 34-40, 2019.

567  61. Prado C, Michels M, Avila P, Burger H, Milioli MVM, and Dal-Pizzol F. The

568  protective effects of fecal microbiota transplantation in an experimental model of

569  necrotizing enterocolitis. Journal of pediatric surgery 54: 1578-1583, 2019.

570  62. Puri K, Taft DH, Ambalavanan N, Schibler KR, Morrow AL, and Kallapur SG.

571  Association of Chorioamnionitis with Aberrant Neonatal Gut Colonization and Adverse

572  Clinical Outcomes. PloS one 11: e0162734, 2016.


573  63. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T,

574  Pons N, Levenez F, Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B,

575  Liang H, Zheng H, Xie Y, Tap J, Lepage P, Bertalan M, Batto JM, Hansen T, Le

576  Paslier D, Linneberg A, Nielsen HB, Pelletier E, Renault P, Sicheritz-Ponten T,

577  Turner K, Zhu H, Yu C, Li S, Jian M, Zhou Y, Li Y, Zhang X, Li S, Qin N, Yang H,

578  Wang J, Brunak S, Dore J, Guarner F, Kristiansen K, Pedersen O, Parkhill J,

579  Weissenbach J, Bork P, Ehrlich SD, and Wang J. A human gut microbial gene

580  catalogue established by metagenomic sequencing. Nature 464: 59-65, 2010.

581  64. Quince C, Lanzen A, Davenport RJ, and Turnbaugh PJ. Removing noise from

582  pyrosequenced amplicons. BMC Bioinformatics 12: 38, 2011.

583  65. Quince C, Walker AW, Simpson JT, Loman NJ, and Segata N. Shotgun

584  metagenomics, from sampling to analysis. Nat Biotechnol 35: 833-844, 2017.

585  66. Remon JI, Amin SC, Mehendale SR, Rao R, Luciano AA, Garzon SA, and

586  Maheshwari A. Depth of bacterial invasion in resected intestinal tissue predicts

587  mortality in surgical necrotizing enterocolitis. Journal of perinatology : official journal of

588  the California Perinatal Association 35: 755-762, 2015.

589  67. Rutayisire E, Huang K, Liu Y, and Tao F. The mode of delivery affects the

590  diversity and colonization pattern of the gut microbiota during the first year of infants'

591  life: a systematic review. BMC gastroenterology 16: 86, 2016.

592  68. Ryan FJ, Drew DP, Douglas C, Leong LEX, Moldovan M, Lynn M, Fink N,

593  Sribnaia A, Penttila I, McPhee AJ, Collins CT, Makrides M, Gibson RA, Rogers GB,

594  and Lynn DJ. Changes in the Composition of the Gut Microbiota and the Blood
595  Transcriptome in Preterm Infants at Less than 29 Weeks Gestation Diagnosed with

596  Bronchopulmonary Dysplasia. mSystems 4: 2019.

597  69. Sanidad KZ, and Zeng MY. Neonatal gut microbiome and immunity. Curr Opin

598  Microbiol 56: 30-37, 2020.

599  70. Sher Y, Olm MR, Raveh-Sadka T, Brown CT, Sher R, Firek B, Baker R,

600  Morowitz MJ, and Banfield JF. Combined analysis of microbial metagenomic and

601  metatranscriptomic sequencing data to assess in situ physiological conditions in the

602  premature infant gut. PloS one 15: e0229537, 2020.

603  71. Skeath T, Stewart C, Waugh S, Embleton N, Cummings S, and Berrington J.

604  Cytomegalovirus and other common enteric viruses are not commonly associated with

605  NEC. Acta Paediatr 105: 50-52, 2016.

606  72. Stewart CJ, Embleton ND, Marrs ECL, Smith DP, Fofanova T, Nelson A,

607  Skeath T, Perry JD, Petrosino JF, Berrington JE, and Cummings SP. Longitudinal

608  development of the gut microbiome and metabolome in preterm neonates with late

609  onset sepsis and healthy controls. Microbiome 5: 75, 2017.

610  73. Tamburini S, Shen N, Wu HC, and Clemente JC. The microbiome in early life:

611  implications for health outcomes. Nat Med 22: 713-722, 2016.

612  74. Thursby E, and Juge N. Introduction to the human gut microbiota. Biochem J

613  474: 1823-1836, 2017.

614  75. Tran L, Ferris M, Norori J, Stark M, Craver R, Dowd S, and Penn D.

615  Necrotizing enterocolitis and cytomegalovirus infection in a premature infant. Pediatrics

616  131: e318-322, 2013.


617  76. Tremblay J, Singh K, Fern A, Kirton ES, He S, Woyke T, Lee J, Chen F,

618  Dangl JL, and Tringe SG. Primer and platform effects on 16S rRNA tag sequencing.

619  Front Microbiol 6: 771, 2015.

620  77. Ullrich T, Tang YW, Correa H, Garzon SA, Maheshwari A, Hill M, Matta P,

621  Krishnan MK, and Weitkamp JH. Absence of gastrointestinal pathogens in ileum

622  tissue resected for necrotizing enterocolitis. Pediatr Infect Dis J 31: 413-414, 2012.

623  78. Valdes AM, Walter J, Segal E, and Spector TD. Role of the gut microbiota in

624  nutrition and health. Bmj 361: k2179, 2018.

625  79. Waligora-Dupriet AJ, Dugay A, Auzeil N, Huerre M, and Butel MJ. Evidence

626  for clostridial implication in necrotizing enterocolitis through bacterial fermentation in a

627  gnotobiotic quail model. Pediatric research 58: 629-635, 2005.

628  80. Wandro S, Osborne S, Enriquez C, Bixby C, Arrieta A, and Whiteson K. The

629  Microbiome and Metabolome of Preterm Infant Stool Are Personalized and Not Driven

630  by Health Outcomes, Including Necrotizing Enterocolitis and Late-Onset Sepsis.

631  mSphere 3: 2018.

632  81. Wang Y, Hoenig JD, Malin KJ, Qamar S, Petrof EO, Sun J, Antonopoulos

633  DA, Chang EB, and Claud EC. 16S rRNA gene-based analysis of fecal microbiota

634  from preterm infants with and without necrotizing enterocolitis. ISME J 3: 944-954,

635  2009.

636  82. Warner BB, Deych E, Zhou Y, Hall-Moore C, Weinstock GM, Sodergren E,

637  Shaikh N, Hoffmann JA, Linneman LA, Hamvas A, Khanna G, Rouggly-Nickless

638  LC, Ndao IM, Shands BA, Escobedo M, Sullivan JE, Radmacher PG, Shannon WD,
639  and Tarr PI. Gut bacteria dysbiosis and necrotising enterocolitis in very low birthweight

640  infants: a prospective case-control study. Lancet 387: 1928-1936, 2016.

641  83. Wong CB, Iwabuchi N, and Xiao JZ. Exploring the Science behind

642  Bifidobacterium breve M-16V in Infant Health. Nutrients 11: 2019.

643  84. Xu W, Judge MP, Maas K, Hussain N, McGrath JM, Henderson WA, and

644  Cong X. Systematic Review of the Effect of Enteral Feeding on Gut Microbiota in

645  Preterm Infants. Journal of obstetric, gynecologic, and neonatal nursing : JOGNN 47:

646  451-463, 2018.

647  85. Yasmin F, Tun HM, Konya TB, Guttman DS, Chari RS, Field CJ, Becker AB,

648  Mandhane PJ, Turvey SE, Subbarao P, Sears MR, Investigators CS, Scott JA, Dinu

649  I, and Kozyrskyj AL. Cesarean Section, Formula Feeding, and Infant Antibiotic

650  Exposure: Separate and Combined Impacts on Gut Microbial Changes in Later Infancy.

651  Frontiers in pediatrics 5: 200, 2017.

652  86. Younge NE, Newgard CB, Cotten CM, Goldberg RN, Muehlbauer MJ, Bain

653  JR, Stevens RD, O'Connell TM, Rawls JF, Seed PC, and Ashley PL. Disrupted

654  Maturation of the Microbiota and Metabolome among Extremely Preterm Infants with

655  Postnatal Growth Failure. Sci Rep 9: 8167, 2019.

656  87. Youssef N, Sheik CS, Krumholz LR, Najar FZ, Roe BA, and Elshahed MS.

657  Comparison of species richness estimates obtained using nearly complete fragments

658  and simulated pyrosequencing-generated fragments in 16S rRNA gene-based

659  environmental surveys. Appl Environ Microbiol 75: 5227-5236, 2009.


660  88. Yu Y, Lu J, Oliphant K, Gupta N, Claud K, and Lu L. Maternal administration

661  of probiotics promotes gut development in mouse offsprings. PLoS One 15: e0237182,

662  2020.

663  89. Zheng N, Gao Y, Zhu W, Meng D, and Walker WA. Short chain fatty acids

664  produced by colonizing intestinal commensal bacterial interaction with expressed breast

665  milk are anti-inflammatory in human immature enterocytes. PLoS One 15: e0229283,

666  2020.

667  90. Zwittink RD, van Zoeren-Grobben D, Renes IB, van Lingen RA, Norbruis OF,

668  Martin R, Groot Jebbink LJ, Knol J, and Belzer C. Dynamics of the bacterial gut

669  microbiota in preterm and term infants after intravenous amoxicillin/ceftazidime

670  treatment. BMC pediatrics 20: 195, 2020.

671 

672 
673  FIGURE LEGENDS

674  Fig 1. Summary of antecedents and consequences of gut dysbiosis in preterm

675  infants. Several prenatal, birth, and postnatal factors negatively impact the developing

676  gut microbiome of the preterm infant. The resulting dysbiotic gut microbial communities

677  during this critical period of development has been linked to both short-term and long-

678  term morbidities. * highlights factors considered to have relatively greater impact than

679  others.

680 
Antecedents Consequences
Birth Short-term
NEC
Mode of delivery
Sepsis
GBS status Long-term
BPD
Antibiotics * Postnatal Asthma
Allergies
NICU environment
Obesity
Formula vs Breastmilk *
Psychiatric illness
Antibiotics *
Anti-acid therapy

Prenatal
Maternal diet
Chorioamnionitis
Antibiotics *
Table 1. Major differences in bacterial gut colonization of preterm infants compared to

term infants.

Bacterial groups Important characteristics and function

Decreased in preterm infants

Bacteroidetes Anti-inflammatory function through surface

(eg Bacteroides fragilis) component polysaccharide A (33)

Actinobacteria Anti-inflammatory function through secretion of

(eg Bifidobacterium infantis, short chain fatty acids (89), and acceleration of

Bifidobacterium breve) maturation of intestinal innate immune response

genes (83)

Increased in preterm infants

Firmicutes Gram positive cocci that can act as opportunistic

(eg Enterococcus faecalis, pathogens, especially in patients with prolonged

Staphylococcus) hospitalization or has received multiple antibiotic

therapy (19)

Proteobacteria Gram negative bacteria commonly associated with

(eg Escherichia coli, Klebsiella opportunistic nosocomial infections; have an outer

pneumoniae) membrane that contains the endotoxin

lipopolysaccharide (80)
Antecedents Consequences
Birth Short-term
NEC
Mode of delivery
Sepsis
GBS status Long-term
BPD
Antibiotics * Postnatal Asthma
Allergies
NICU environment
Obesity
Formula vs Breastmilk *
Psychiatric illness
Antibiotics *
Anti-acid therapy

Prenatal
Maternal diet
Chorioamnionitis
Antibiotics *

You might also like