Over the last 2 decades, the study of the microbiome has vastly expanded. What was once an underappreciated contributor to human health—outside of consideration as a source of pathogens and contamination—is now understood as an “organ” in its own right. Dysbiosis, an unhealthy shift in the microbiome, has been associated with disease states throughout the body. Moreover, we have come to realize that the microbiome, like other body systems, undergoes a characteristic developmental trajectory throughout infancy and childhood. Neonates and young infants have a unique microbiome, with bacterial communities that continue to develop until at least 5 years of age [1, 2]. The developing microbiome is also sensitive to dysbioses that have been linked to long-term sequelae, including immunological and metabolic disease [3–6].

The preterm neonatal microbiome is particularly sensitive to microbiome-modulating factors. Preterm infants are anatomically and immunologically underdeveloped [7, 8]. They are also more likely to be born via cesarean delivery, which can disrupt normal colonization of the infant gut [9–11]. Further, hospitalized neonates are at increased risk of colonization with nosocomial pathogens [12, 13] and have substantially increased exposure to antibiotics. The resulting dysbiosis has been shown to contribute to the development of high-mortality infectious and inflammatory conditions, including necrotizing enterocolitis (NEC) [13–16], bloodstream infection (BSI) [14, 17, 18], and as now described by Frerichs and Deianova et al, late-onset meningitis (LOM) [19].

Late-onset meningitis, that is, neonatal meningitis presenting between postnatal days 3 and 28 [20], is associated with both a high mortality rate and poor neurodevelopmental outcomes for survivors [20, 21]. Current diagnostic approaches are invasive: The gold standard consists of analysis and culture of cerebrospinal fluid (CSF). Lumbar punctures can be difficult to obtain and/or contraindicated. Further, culture-based approaches require days of incubation and may be falsely negative due to antibiotic administration prior to CSF collection.

Given such challenges, noninvasive, point-of-care diagnostics would represent a major advance, especially in resource-limited settings. Frerichs and Deianova et al address this compelling need, as their findings suggest that LOM, similar to NEC and BSI, is preceded by a shift in the intestinal bacterial community characterized by intestinal pathogen outgrowth. Importantly, this dysbiosis can be detected prior to clinical symptoms of LOM—suggesting the possibility of identifying at-risk infants in time to intervene. Importantly, this work also suggests that many serious bacterial infections of infancy (NEC, BSI, and LOM) may have a similar mechanism of disease, or alternatively, that they exist on a sort of continuum. In this model (Figure 1), a dysregulation of the microbiome leads to an outgrowth of pathogenic taxa. These unwelcome blooms may directly contribute to the development of NEC, or given the immature nature of the gut epithelium, may translocate across the gut epithelium to the bloodstream, causing BSI. Bacteremia pathogens have been identified in the infant gut prior to the onset of BSI [17, 18, 22]. In a subset of bacteremic neonates, the bacteria further translocate to the meninges, possibly via the highly vascularized choroid plexus, which is incompletely polarized in preterm neonates [23], causing LOM.

Prospective model of the relationship between gut dysbiosis and serious bacterial infections in neonates. Gut dysbiosis, characterized by an outgrowth of pathogenic taxa, contributes to the development of necrotizing enterocolitis, an inflammatory condition. The pathogenic taxa may also translocate the gut epithelium to the bloodstream, initiating a bloodstream infection (BSI). In a subset of neonates, the pathogen further translocates from the bloodstream to the meninges, causing late-onset meningitis with or without BSI. Created with BioRender.com.
Figure 1.

Prospective model of the relationship between gut dysbiosis and serious bacterial infections in neonates. Gut dysbiosis, characterized by an outgrowth of pathogenic taxa, contributes to the development of necrotizing enterocolitis, an inflammatory condition. The pathogenic taxa may also translocate the gut epithelium to the bloodstream, initiating a bloodstream infection (BSI). In a subset of neonates, the pathogen further translocates from the bloodstream to the meninges, causing late-onset meningitis with or without BSI. Created with BioRender.com.

In this issue of The Journal of Infectious Diseases, Frerichs and Deianova et al detail their findings that LOM is preceded by intestinal pathogen outgrowth using a prospective longitudinal study of preterm infants (<30 weeks’ gestational age) across 9 neonatal intensive care units in the Netherlands and Belgium. Fecal samples were collected from neonates during the first 28 days of life; the microbiota was assessed via 16S ribosomal RNA gene sequencing and the fecal volatile metabolome through both gas chromatography–ion mobility spectrometry (GC-IMS) and gas chromatography–time-of-flight mass spectrometry (GC-TOF-MS). Cases and controls were matched by center, gestational age, birth weight, and postnatal age.

Of this 1397-member cohort, 21 neonates were diagnosed with LOM, and 19 of these had preceding bacteremia. Microbiota analysis demonstrated an increased relative abundance of Pseudomonadota (formerly Proteobacteria) concomitant with decreases in the relative abundance of Bacillota (formerly Firmicutes) and Bacteroidota (formerly Bacteroidetes) in subjects who developed LOM relative to controls. Using a binned time-interval approach, the researchers were able to discriminate between cases and controls using both Random Forest and MaAsLin2 approaches. Indeed, the best-performing random forest classification model based on the fecal microbiota was associated with the 1- to 3-day prediagnosis interval (area under the curve [AUC]: 0.88) and was partially driven by a reduced relative abundance of Bacteroides and increased relative abundance of Escherichia/Shigella, as has previously been noted preceding both NEC and BSI [16, 18, 24].

Excitingly, fecal volatile profiles also appear to change prior to LOM diagnosis. GC-IMS analysis identified significant differences between cases and controls 1 day prior to diagnosis (random forest, AUC: 0.74) and GC-TOF-MS distinguished between cases and controls 2 days prior to diagnosis (random forest, AUC: 0.82). This aligns with similar findings from this group and others where multiple analytical techniques identified discriminatory changes in fecal volatile organic compounds within 3 days of diagnosis for both BSI and NEC [25–31].

This study builds on the growing literature linking pathogen outgrowth in the gastrointestinal tract and serious bacterial infections in infancy. As suggested by Frerichs and Deianova et al, the field would benefit from larger, longitudinal multi-omics studies to validate these findings. Optimally, these studies would account for within-patient variability, as study of the preterm gut microbiome is complicated by its plastic nature [17, 32, 33]. However, the current study provides exciting support for the possibility of surveillance and early intervention with microbiota-directed therapeutics—perhaps antibiotics, probiotics, or some other strategy to protect the immature mucosal barrier. Given the suspected role of pathogen outgrowth in serious bacterial infections in neonates, and the negative association between Bifidobacterium and pathogenic taxa in the preterm neonate gut [17, 32, 34], the possibility of probiotic administration presents a particularly intriguing approach that already has shown some promise in prevention of NEC [35].

References

1

Yatsunenko
 
T
,
Rey
 
FE
,
Manary
 
MJ
, et al.   
Human gut microbiome viewed across age and geography
.
Nature
 
2012
;
486
:
222
7
.

2

Roswall
 
J
,
Olsson
 
LM
,
Kovatcheva-Datchary
 
P
, et al.   
Developmental trajectory of the healthy human gut microbiota during the first 5 years of life
.
Cell Host Microbe
 
2021
;
29
:
765
76.e3
.

3

Zimmermann
 
P
,
Messina
 
N
,
Mohn
 
WW
,
Finlay
 
BB
,
Curtis
 
N
.
Association between the intestinal microbiota and allergic sensitization, eczema, and asthma: a systematic review
.
J Allergy Clin Immunol
 
2019
;
143
:
467
85
.

4

Galazzo
 
G
,
van Best
 
N
,
Bervoets
 
L
, et al.   
Development of the microbiota and associations with birth mode, diet, and atopic disorders in a longitudinal analysis of stool samples, collected from infancy through early childhood
.
Gastroenterol
 
2020
;
158
:
1584
96
.

5

Miller
 
S
,
Wu
 
R
,
Oremus
 
M
.
The association between antibiotic use in infancy and childhood overweight or obesity: a systematic review and meta-analysis
.
Obes Rev
 
2018
;
19
:
1463
75
.

6

Stokholm
 
J
,
Blaser
 
MJ
,
Thorsen
 
J
, et al.   
Maturation of the gut microbiome and risk of asthma in childhood
.
Nat Commun
 
2018
;
9
:
141
.

7

Healy
 
DB
,
Ryan
 
CA
,
Ross
 
RP
,
Stanton
 
C
,
Dempsey
 
EM
.
Clinical implications of preterm infant gut microbiome development
.
Nat Microbiol
 
2022
;
7
:
22
33
.

8

Duess
 
JW
,
Sampah
 
ME
,
Lopez
 
CM
, et al.   
Necrotizing enterocolitis, gut microbes, and sepsis
.
Gut Microbes
 
2023
;
15
:
2221470
.

9

Bokulich
 
NA
,
Chung
 
J
,
Battaglia
 
T
, et al.   
Antibiotics, birth mode, and diet shape microbiome maturation during early life
.
Sci Transl Med
 
2016
;
8
:
343ra82
.

10

Al
 
KF
,
Allen
 
L
,
Bedell
 
S
,
Burton
 
JP
,
Vrijer
 
BD
.
Assessing the impact of pregnancy and birth factors on the maternal and infant microbiota
.
Microbiome Res Rep
 
2023
;
2
:
29
.

11

Shao
 
Y
,
Forster
 
SC
,
Tsaliki
 
E
, et al.   
Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth
.
Nature
 
2019
;
574
:
117
21
.

12

Brooks
 
B
,
Firek
 
BA
,
Miller
 
CS
, et al.   
Microbes in the neonatal intensive care unit resemble those found in the gut of premature infants
.
Microbiome
 
2014
;
2
:
1
.

13

Rao
 
C
,
Coyte
 
KZ
,
Bainter
 
W
,
Geha
 
RS
,
Martin
 
CR
,
Rakoff-Nahoum
 
S
.
Multi-kingdom ecological drivers of microbiota assembly in preterm infants
.
Nature
 
2021
;
591
:
633
8
.

14

Masi
 
AC
,
Stewart
 
CJ
.
The role of the preterm intestinal microbiome in sepsis and necrotising enterocolitis
.
Early Hum Dev
 
2019
;
138
:
104854
.

15

Stewart
 
CJ
,
Nelson
 
A
,
Scribbins
 
D
, et al.   
Bacterial and fungal viability in the preterm gut: NEC and sepsis
.
Arch Dis Child Fetal Neonatal Ed
 
2013
;
98
:
F298
303
.

16

Moschino
 
L
,
Verlato
 
G
,
Duci
 
M
, et al.   
The metabolome and the gut microbiota for the prediction of necrotizing enterocolitis and spontaneous intestinal perforation: a systematic review
.
Nutrients
 
2022
;
14
:
3859
.

17

Stewart
 
CJ
,
Embleton
 
ND
,
Marrs
 
ECL
, et al.   
Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls
.
Microbiome
 
2017
;
5
:
75
.

18

Schwartz
 
DJ
,
Shalon
 
N
,
Wardenburg
 
K
, et al.   
Gut pathogen colonization precedes bloodstream infection in the neonatal intensive care unit
.
Sci Transl Med
 
2023
;
15
:
eadg5562
.

19

Frerichs
 
NM
,
Deianova
 
N
,
El Manouni El Hassani
 
S
, et al.   
Fecal microbiota and volatile metabolome pattern alterations precede late-onset meningitis in preterm neonates [manuscript published online ahead of print 23 May 2024]
.
J Infect Dis
 
2024
. doi:

20

Brumbaugh
 
JE
,
Bell
 
EF
,
Do
 
BT
, et al.   
Incidence and neurodevelopmental outcomes after late-onset meningitis among children born extremely preterm
.
JAMA Netw Open
 
2022
;
5
:
e2245826
.

21

Thomas
 
R
,
Bijlsma
 
MW
,
Gonçalves
 
BP
,
Nakwa
 
FL
,
Velaphi
 
S
,
Heath
 
PT
.
Long-term impact of serious neonatal bacterial infections on neurodevelopment
.
Clin Microbiol Infect
 
2024
;
30
:
28
37
.

22

Carl
 
MA
,
Ndao
 
IM
,
Springman
 
AC
, et al.   
Sepsis from the gut: the enteric habitat of bacteria that cause late-onset neonatal blood stream infections
.
Clin Infect Dis
 
2014
;
58
:
1211
8
.

23

Travier
 
L
,
Alonso
 
M
,
Andronico
 
A
, et al.   
Neonatal susceptibility to meningitis results from the immaturity of epithelial barriers and gut microbiota
.
Cell Rep
 
2021
;
35
:
109319
.

24

Graspeuntner
 
S
,
Waschina
 
S
,
Künzel
 
S
, et al.   
Gut dysbiosis with bacilli dominance and accumulation of fermentation products precedes late-onset sepsis in preterm infants
.
Clin Infect Dis
 
2019
;
69
:
268
77
.

25

Berkhout
 
DJC
,
Niemarkt
 
HJ
,
Buijck
 
M
, et al.   
Detection of sepsis in preterm infants by fecal volatile organic compounds analysis: a proof of principle study
.
J Pediatr Gastroenterol Nutr
 
2017
;
65
:
e47
52
.

26

de Meij
 
TGJ
,
van der Schee
 
MPC
,
Berkhout
 
DJC
, et al.   
Early detection of necrotizing enterocolitis by fecal volatile organic compounds analysis
.
J Pediatr
 
2015
;
167
:
562
7.e1
.

27

Berkhout
 
DJC
,
van Keulen
 
BJ
,
Niemarkt
 
HJ
, et al.   
Late-onset sepsis in preterm infants can be detected preclinically by fecal volatile organic compound analysis: a prospective, multicenter cohort study
.
Clin Infect Dis
 
2019
;
68
:
70
7
.

28

Berkhout
 
DJC
,
Niemarkt
 
HJ
,
Andriessen
 
P
, et al.   
Preclinical detection of non-catheter related late-onset sepsis in preterm infants by fecal volatile compounds analysis: a prospective, multi-center cohort study
.
Pediatr Infect Dis J
 
2020
;
39
:
330
5
.

29

Frerichs
 
NM
,
El Manouni El Hassani
 
S
,
Deianova
 
N
, et al.   
Fecal volatile metabolomics predict gram-negative late-onset sepsis in preterm infants: a nationwide case-control study
.
Microorganisms
 
2023
;
11
:
572
.

30

Garner
 
CE
,
Ewer
 
AK
,
Elasouad
 
K
, et al.   
Analysis of faecal volatile organic compounds in preterm infants who develop necrotising enterocolitis: a pilot study
.
J Pediatr Gastroenterol Nutr
 
2009
;
49
:
559
65
.

31

Probert
 
C
,
Greenwood
 
R
,
Mayor
 
A
, et al.   
Faecal volatile organic compounds in preterm babies at risk of necrotising enterocolitis: the DOVE study
.
Arch Dis Child Fetal Neonatal Ed
 
2020
;
105
:
474
9
.

32

Alcon-Giner
 
C
,
Dalby
 
MJ
,
Caim
 
S
, et al.   
Microbiota supplementation with Bifidobacterium and Lactobacillus modifies the preterm infant gut microbiota and metabolome: an observational study
.
Cell Rep Med
 
2020
;
1
:
100077
.

33

Gasparrini
 
AJ
,
Wang
 
B
,
Sun
 
X
, et al.   
Persistent metagenomic signatures of early-life hospitalization and antibiotic treatment in the infant gut microbiota and resistome
.
Nat Microbiol
 
2019
;
4
:
2285
97
.

34

Bajorek
 
S
,
Duar
 
RM
,
Corrigan
 
M
, et al.   
B. infantis EVC001 is well-tolerated and improves human milk oligosaccharide utilization in preterm infants in the neonatal intensive care unit
.
Front Pediatr
 
2022
;
9
:
795970
.

35

Sharif
 
S
,
Meader
 
N
,
Oddie
 
SJ
,
Rojas-Reyes
 
MX
,
McGuire
 
W
.
Probiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants
.
Cochrane Database Syst Rev
 
2023
;
7
:
CD005496
.

Author notes

Potential conflicts of interest. The authors: No reported conflicts.

Both authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/pages/standard-publication-reuse-rights)