Gut Microbiota: No Longer The Forgotten Organ

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 52

Vol. 78, Suppl.

2, 20-21

Gut Microbiota: No Longer


the Forgotten Organ

Editor
Hania Szajewska, Warsaw

Editorial Board
Andrew Prentice, Banjul/London
Raanan Shamir, Tel Aviv
Hania Szajewska, Warsaw

Supported by

Basel Freiburg Hartford Oxford Bangkok Dubai Kuala Lumpur


• • • • • • •

Melbourne Mexico City Moscow New Delhi Paris Shanghai Tokyo


• • • • • •

https://www.nestlenutrition-institute.org
Reprint of Annals of Nutrition and Metabolism Vol. 77, Suppl. 3, 2021

Sponsor Note
This publication was supported by an unrestricted educational grant by the
Nestlé Nutrition Institute. The institute is a not-for-profit association which was
created to provide latest medical and scientific information to health profes-
sionals in the field of pediatric and adult nutrition and nutrition-related disorders
(available at www.nestlenutrition-institute.org).
Any liability of the sponsors for the content of the papers is hereby expressly
excluded.

Disclosure Statement Editor


Hania Szajewska has participated during the last 5 years as a clinical investigator,
and/or advisory board member and/or consultant, and/or speaker for Arla, Aus-
nutria, BioGaia, Biocodex, Cargill, Chr. Hansen, Danone, Nestlé, Nestlé Nutrition
Institute, and Merck.

S. Karger Disclaimer All rights reserved.


Medical and Scientific Publishers The statements, opinions and data contained in this publica- No part of this publication may be translated into other
tion are solely those of the individual authors and contributors languages, reproduced or utilized in any form or by any means,
Basel Freiburg Hartford Oxford
• • • •
and not of the publisher and the editor(s). The appearance of electronic or mechanical, including photocopying, recording,
Bangkok Dubai Kuala Lumpur
• • •
advertisements in the journal is not a warranty, endorsement, microcopying, or by any information storage and retrieval
Melbourne Mexico City
• •
or approval of the products or services advertised or of their system, without permission in writing from the publisher or,
Moscow New Delhi Paris
• • • effectiveness, quality or safety. The publisher and the editor(s) in the case of photocopying, direct payment of a specified fee to
Shanghai Tokyo
• disclaim responsibility for any injury to persons or property the Copyright Clearance Center (see “General Information”).
resulting from any ideas, methods, instructions or products
referred to in the content or advertisements. © 2021 Nestlé Nutrition Institute, Switzerland/
S. Karger AG, Basel
Drug Dosage P.O. Box, CH–4009 Basel (Switzerland)
The authors and the publisher have exerted every effort to en- e-ISBN 978–3–318–07025–5
sure that drug selection and dosage set forth in this text are in
accord with current recommendations and practice at the time
of publication. However, in view of ongoing research, changes
in government regulations, and the constant flow of informa-
tion relating to drug therapy and drug reactions, the reader is
urged to check the package insert for each drug for any change
in indications and dosage and for added warnings and precau-
tions. This is particularly important when the recommended
agent is a new and/or infrequently employed drug.

karger@karger.com
Vol. 78, Suppl. 2, 20-21

Contents

11 Editorial
Szajewska, H. (Warsaw)

Gut Microbiota: No Longer the Forgotten Organ

13 Focus on: Microbiome and Infection: A Case for “Selective


Depletion”

14 Microbiome and Infection: A Case for “Selective Depletion”


Hill, C. (Cork)

10 Focus on: Impact of Delivery Mode on Infant Gut Microbiota

11 Impact of Delivery Mode on Infant Gut Microbiota


Korpela, K. (Helsinki)

20 Focus on: Gut Microbiota Development: Influence of Diet


from Infancy to Toddlerhood

21 Gut Microbiota Development: Influence of Diet from Infancy


to Toddlerhood
Laursen, M.F. (Kongens Lyngby)

35 Focus on: Microbiomes and Childhood Malnutrition: What Is


the Evidence?

36 Microbiomes and Childhood Malnutrition: What Is the


Evidence?
McGuire, M.K.; McGuire, M.A. (Moscow, ID)

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/


S. Karger AG, Basel
Policy Statement

The Nestlé Nutrition Institute was created to provide health


professionals with up-to-date information on nutrition and nutrition-
related disorders in order to enable them to continuously improve
patient care based on the latest medical and scientific developments.
One of the key pillars of the Nestlé Nutrition Institute is Annales Nestlé,
a pediatric journal that has been published on a regular basis since 1942.
It contains review articles on clinical practice and research in all fields
of pediatrics with focus on nutrition.
Annales Nestlé appears three times a year. Each article is supported
by a Focus Page, and each issue by an Infographic illustrating the core
topic. Published on www.nestlenutrition-institute.org as well as in print,
Annales Nestlé is one of the most widely read pediatric journals in the
world.
Annales Nestlé is edited by an independent editorial board of opinion
leaders in pediatric research, thus guaranteeing the medical and scientific
impartiality of the journal, and hence the high regard it enjoys in medical
and scientific circles. The editorial board sets the editorial policy,
identifies topics to be addressed, selects authors, and oversees the
review process for each issue.
Every issue of Annales Nestlé initially appears as a supplement to
Annals of Nutrition and Metabolism – a journal from Karger Publishers,
Basel, Switzerland – and is listed in all major bibliographic services, such
as Medline, PubMed, and Web of Science. This has been our practice
since 2011.
We are pleased to offer you our innovative product, which results
from a creative and effective cooperation with Karger Publishers,
Switzerland.

Josephine Yuson-Sunga, MD
Global Head
Nestlé Nutrition Institute
Vevey (Switzerland)

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/


S. Karger AG, Basel
Editorial

Reprint with permission from:


Ann Nutr Metab 2021;77(suppl 3):1–2
DOI: 10.1159/000519223

Gut Microbiota: No Longer the


Forgotten Organ
Hania Szajewska
The Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland

While the gut microbiota was once called “the forgotten the concept of “selective depletion,” where pathogens can be
organ” [1], it is no longer forgotten. On the contrary, it is a hot reduced by narrow-spectrum inhibitors, such as bacteriocins
topic for research, as documented by the rapidly increasing or bacteriophages (phages). The microbiome is protected and
number of scientific papers on this subject as well as coverage can play a role in reducing the spread of infection by sup-
in the lay press, TV/radio programs, and social media. pressing the growth of pathogens.
The aim of this new series is to highlight recent develop- The mode of delivery affects the gut microbiome. The
ments and hot topics in gut microbiome/microbiota research. World Health Organization estimates the cesarean section
These two terms are often used interchangeably. However, rate as 10–15% of all births if only used when medically nec-
there is a subtle difference between them [2]. The term mi- essary. However, cesarean delivery is increasing in prevalence
crobiome refers to all the microorganisms (and their genes) worldwide. Based on data from 169 countries that included
living in a specific environment such as the gut. These micro- over 98% of births worldwide, cesarean section almost dou-
organisms include bacteria, archaea, eukaryotes, and viruses. bled in 2015 compared with 2000 (21.1% vs. 12.1%, respec-
The term microbiota, in principle, refers to the microbes tively) [5]. The second paper by Katri Korpela [6] from the Uni-
themselves. If only the genes are of interest, the term metage- versity of Helsinki, Finland, discusses the impact of delivery
nome is used. mode on the infant gut microbiota, its consequences, and the
Regardless of the terminology, a balanced gut microbiota interventions to restore the microbial dysbiosis created by ce-
is crucial for health. Conversely, dysbiosis, which refers to al- sarean section.
tered gut microbiota diversity and composition (i.e., changes Diet is a key factor that has a substantial impact on the
at the level of phylum, genus, or species) [3], contributes to composition and function of the gut microbiota. The third
the development of gastrointestinal and extraintestinal dis- paper, written by Martin Laursen [7] from the National Food
eases. Among others, the microbiota mediates colonization Institute, Technical University of Denmark, covers the most
resistance and influences susceptibility to infectious diseases. important factors controlling the succession and establish-
The first paper, by Colin Hill [4] from the APC Microbiome Ire- ment of the gut microbiota in infants after birth. Considering
land and School of Microbiology, University College Cork, Ire- how the early-life gut microbiota impacts our immunity, a
land, reviews advances in the research on infections and the better understanding of the process could potentially lead to
microbiome, which provides a barrier to infection but can also strategies modifying the risk of diseases later in life.
serve as a source of novel antimicrobials. He also introduces

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/ Correspondence to:


S. Karger AG, Basel Hania Szajewska, hszajewska @ wum.edu.pl
Some children have too little to eat, while others consume position of the gut microbiota and the regulation of body
too many calories via junk food and beverages. Consequent- weight.
ly, malnutrition consisting of undernutrition, overweight, and In summary, gut microbiome/microbiota research is a hot
obesity is a major public health concern affecting all countries topic and is booming. This series also documents that the gut
– low-, middle-, and high-income – although in different microbiota is indeed no longer forgotten and is increasingly
proportions [8, 9]. In the fourth paper written by Michelle Mc- recognized as a potential key player in the pathogenesis of
Guire and Mark McGuire [10] from the University of Idaho, several diseases.
Moscow, ID, USA, the authors discuss data linking the com-

References
1 Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold 6 Korpela K. Impact of delivery mode on infant gut microbiota. Ann
G, et al. The gut microbiota and host health: a new clinical frontier. Nutr Metab. doi: 10.1159/000518498.
Gut. 2016 Feb;65(2):330–9.
7 Laursen MF. Gut Microbiota development: influence of diet from
2 Quigley EM, Ghoshal UC, editors. WGO Handbook on the Gut Mi- infancy to toddlerhood. Ann Nutr Metab. doi: 10.1159/000517912.
crobiome. World Gastroenterology Organisation; 2020 [cited Aug
8 Hawkes C, Ruel MT, Salm L, Sinclair B, Branca F. Double-duty ac-
22, 2021]. Available from: https://www.worldgastroenterology.
tions: seizing programme and policy opportunities to address
org/wgo-foundation/wdhd/wdhd-2020/tools-and-
malnutrition in all its forms. Lancet. 2020 Jan;395(10218):142–55.
resources#Handbook.
Erratum in: Lancet. 2020 Feb 1;395.
3 Hooks KB, O’Malley MA. Dysbiosis and Its Discontents. MBio. 2017
9 The double burden of malnutrition. Lancet. Dec 16, 2019 [cited
Oct;8(5):e01492–17.
Aug 22, 2021]. Available from: https://www.thelancet.com/series/
4 Hill C. Microbiome and infection: a case for “selective depletion”. double-burden-malnutrition.
Ann Nutr Metab. doi: 10.1159/000516399.
10 McGuire MK, McGuire MA. Microbiomes and childhood mal-
5 Boerma T, Ronsmans C, Melesse DY, Barros AJD, Barros FC, Juan nutrition: what is the evidence? Ann Nutr Metab. doi: 10.1159/
L, et al. Global epidemiology of use of and disparities in caesarean 000519001.
sections. Lancet. 2018 Oct;392(10155):1341–48.

2 Reprint with permission from: Szajewska


Ann Nutr Metab 2021;77(suppl 3):1–2
DOI: 10.1159/000519223
Focus
The future of smart antimicrobial therapies may not involve pathogen
elimination with collateral damage to the microbiome (broad-spectrum
approaches), but may well lie in restricting the growth of pathogens to a
point where they cannot cause disease while harnessing the power of the
microbiome to ensure a return to a healthy state

Reprinted with permission from: Ann Nutr Metab 2021;77(suppl 3):4–9

Microbiome and Infection: A Case for “Selective Depletion”


Colin Hill

Key insights Fecal microbiota


transplantation Selective microbial
Microbial infections can be recognized as a disruption of the consortiums

normal homeostatic relationship between a particular


microbe and the host. There are many instances where the
pathogen is not acquired but is already a member of the Healthy
Probiotics microbiome
microbiome, e.g., Clostridioides difficile, Staphylococcus Purified
aureus, or certain strains of Escherichia coli. Often, these bacteriocins
organisms only cause disease when the surrounding
microbiome is compromised, enabling the pathogen to grow
Bacteriophages
and result in disease symptoms in the host. Thus, the Bacteriophage lysins
microbiome itself should be treated as a virtual organ and
protected during therapeutic interventions. A targeted Mining the microbiome has yielded a variety of therapeutic strategies
approach towards eliminating unwanted microbial pathogens for the treatment of infections and chronic diseases.
is termed “selective depletion,” whereby pathogen numbers
are curtailed by a narrow-spectrum inhibitor, but the
microbiome is protected and can thus play a role in restoring Practical implications
health and suppressing pathogen outgrowth in the infected
patient. What is it about the microbiome that keeps pathogens in check,
and can we harness the microbiome itself for selective therapies
against specific pathogens? Microbiome-derived anti-infection
strategies have ranged from using an entire intact microbiome,
a selected microbial consortium, or single biological entities.
Current knowledge Recent advances in mining the microbiome have taken this one
step further. A targeted approach involves the isolation and pu-
From birth to adulthood, all our exposed bodily surfaces be- rification of bacteriocins, toxins produced by bacteria that in-
come colonized by thousands of different microbial strains. hibit the growth of related bacterial strains. For example, thuri-
The human body consists of hundreds of micro-environ- cin CD is produced by a strain of Bacillus thuringiensis and is a
ments that facilitate the growth of various microbes (the mi- very narrow-spectrum bacteriocin targeting C. difficile. This ap-
crobiome). In turn, the microbiome fulfills many functions proach paves the way for selective depletion of pathogens with-
that are critical for human health. Under a normal state of out collateral damage to the microbiome.
homeostasis, the microbiome acts as a “virtual organ,” con-
stantly signaling to the digestive, immune, nervous, and en-
docrine systems in a process that maintains health and pre- Recommended reading
vents disease. Infection can be viewed as any interaction be-
tween a microbe and the host that results in damage severe De Maesschalck V, Gutiérrez D, Paeshuyse J, Lavigne R, Briers
enough to be manifested in the form of disease symptoms. Y. Advanced engineering of third-generation lysins and for-
mulation strategies for clinical applications. Crit Rev Micro-
biol. 2020;46:548–64.

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/


S. Karger AG, Basel
Review Article

Reprint with permission from:


Ann Nutr Metab 2021;77(suppl 3):4–9
DOI: 10.1159/000516399

Microbiome and Infection:


A Case for “Selective Depletion”
Colin Hill  

APC Microbiome Ireland and School of Microbiology, University College Cork, Cork, Ireland

Key Messages virtual organ within the human body, and we would surely
hesitate to advance any therapeutic approach that would
• The microbiome is a virtual organ that should be protected in cause substantial damage to one of our organs. This is one
therapeutic interventions.
consequence of many broad-spectrum antimicrobial thera-
• Narrow-spectrum inhibitors can “selectively deplete” the
pathogen while protecting the microbiome. pies. There may be instances where a more precise approach
• The microbiome itself can be a valuable source of antimicro- would be useful. I have termed this “selective depletion”; a
bials. concept where pathogen numbers are curtailed by a narrow-
spectrum inhibitor but the microbiome is protected and can
play a role in restoring health and suppressing the outgrowth
Keywords of the pathogen in the infected patient. It may well be that the
Bacteriocin · Bacteriophage · Probiotics · Infection · best reservoir of microbiome-friendly antimicrobial agents is
Microbiome the microbiome itself, and I provide examples of where the
microbiome has been mined for novel precision antimicrobi-
als. © 2021 Nestlé Nutrition Institute, Switzerland/
Abstract S. Karger AG, Basel

In most instances where a pathogen has initiated an infection,


the primary goal of the treating physician or pharmacist is to
eliminate the pathogen. In the absence of knowledge of the Microbiome, Pathogens, and Host
precise identity of the problem-causing microbe, a broad-
spectrum antimicrobial gives the best chance of success. This As a rule, microbes are good for us. We interact with trillions
approach has saved many lives and is an invaluable tool in of microbes every day, and almost all these interactions are
fighting infections. However, perhaps our current apprecia- mutually beneficial, or at the very least are benign. Our most
tion of the importance of the microbiome in human health important microbial interaction is that between us and our
should give us pause. We can regard the microbiome as a microbiome, our fellow travellers on our journey through life.

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/ Correspondence to:


S. Karger AG, Basel Colin Hill, c.hill @ ucc.ie
Colonisation resistance Infection

Pathogen

Microbiome

Host
epithelium

Immune
system

Access to
other organs

Fig. 1. A diverse abundant microbiome (left) can act as a barrier to infection through a variety of mechanisms, includ-
ing direct inhibition, colonisation resistance, or educating the immune system. A damaged microbiome (right) may
be more susceptible to infectious bacteria causing damage.

We are born into a microbial world, and as adults, all our ex- between the pathogen and the host in terms of survival, colo-
posed surfaces are colonised with thousands of microbial nisation, growth, and ultimately damage mediated by viru-
strains selected from the millions we have encountered since lence factors such as invasive mechanisms and/or the pro-
birth [1]. We provide our microbes with a somewhat protect- duction of toxins. The damage caused can result in mild
ed, albeit highly competitive, environment. It is more correct symptoms or death depending on a myriad of factors. These
to state that we provide hundreds of micro-environments or factors include the identity of the pathogen, including the ge-
niches. We as hosts provide sustenance and permissive envi- nus, species, and even the genetic complement of a given
ronmental conditions, while our microbial partners fulfil many strain; the possession and expression of virulence factors; the
functions important to human health. We share the same dose; the circumstances surrounding the exposure in terms
chemical language, and so there is constant communication of food carriers or breach of barriers (skin or mucosal surface);
between our microbes and our nervous system, our immune the genetics of the host; the hosts immune status; and of
systems, and our mucosal surfaces. In a healthy human, these course, the commensal microbes present at the site of infec-
interactions with our microbial “virtual organ” [2] is important tion (the microbiome).
for maintaining health and preventing disease. The microbi- There are many instances where the pathogen is not ac-
ome is a valuable partner and, as with any other organ, should quired but is already a member of our microbiome, for exam-
be protected insofar as possible. ple, Clostridioides difficile, Staphylococcus aureus, or certain
In this context, we can see microbial infections as an aber- strains of Escherichia coli. These pathogens often only cause
ration where a specific microbe has broken with the normal- disease when the surrounding microbiome is compromised in
ly benign or beneficial relationship between microbes and some way, and the pathogen manages to grow and use its
hosts. We call these microbes pathogens or opportunistic virulence factors to cause damage to the host. We can regard
pathogens. Infection describes any interaction between a mi- the microbiome as a barrier to infection (Fig.  1). Antibiotic
crobe and a host that results in damage that is severe enough treatment can be an effective solution to reduce or even elim-
to be manifested in the form of symptoms. Infection usually inate a pathogen but can also lead to further damage to the
follows a very prescribed path, initiated by acquisition or ex- microbiome. For C. difficile, this can result in recurring bouts
posure to the infectious microbe (pathogen), engagement of diarrhoea and even fatal outcomes. Most microbiomes will

A Case for Selective Depletion in Treating Reprint with permission from: 5


Infectious Disease Ann Nutr Metab 2021;77(suppl 3):4–9
DOI: 10.1159/000516399
Faecal Microbiota Transplantation
Faecal microbiota transplantation (FMT) is perhaps the most
FMT
(Microbial consortia) dramatic example of mining the microbiome for solutions to
infections. A recent systematic review and meta-analysis con-
Probiotics cluded that there is high-quality evidence that FMT is effective
(Postbiotics) in breaking the vicious cycle of recurring C. difficile infections
[4], although a similar success rate has not been reported for
Phage non-infectious diseases such as irritable bowel syndrome [5].
(Phage lysins) Essentially, FMT involves collecting the entire microbiome
from one individual in the form of a faecal donation and trans-
Microbiome Bacteriocins planting it with minimal processing to the colon of a patient
RWKHU£DQWLPLFURELDOV
with recurring C. difficile infections. The generally accepted
opinion is that FMT “repairs” a microbiome that has been dam-
aged by the antibiotic therapies used to/try to control the ini-
Fig. 2. Mining the microbiome for selective depletion strategies.
FMT, faecal microbiota transplantation.
tial infection(s) and allows the recipient to use this more di-
verse microbiome to suppress subsequent outgrowth of the
pathogen. Another strategy linked to FMT is using an unde-
fined microbial consortium composed of spores purified from
recover from antibiotic exposure and will revert to a level of a faecal sample and transferred to a patient with a similar ob-
complexity and community structure that approximates the jective of restoring diversity and suppressing pathogen-in-
pre-treatment microbiome, but of course, there will also be an duced damage. This intervention had less successful results
undesirable selection pressure for the emergence of antibi- in published phase 2 clinical trials [6] but has the potential to
otic resistance in both target and nontarget species. A link be- be the platform for a range of microbiome-based intervention
tween microbiome composition and sensitivity to viral infec- strategies.
tion in humans has also been reported recently [3].
What is it about the microbiome that keeps these patho- Probiotics
gens in check, and can we develop effective therapies aimed Probiotics are live microorganisms that, when administered in
at selectively depleting the pathogen during an infection? I adequate amounts, confer a health benefit on the host [7].
use the term “selective depletion” to describe any treatment There is evidence that probiotics can be used to prevent and
that causes minimal damage to the microbiome but reduces treat infection, and some of these data have been generated
the pathogen to levels that do not lead to symptoms or cause in humans. The strength of the data is not entirely consistent,
long-term damage to the host. What kind of strategies would since most meta-analyses and systematic reviews are per-
meet this objective of selective depletion? Perhaps the best formed across a range of different probiotic interventions,
place to start is the microbiome itself. Can we explore the mi- dose ranges, and disease outcomes. Nonetheless, there have
crobiome for strategies that can prevent and even treat infec- been positive outcomes. A recent meta-analysis on the abil-
tions? I will draw upon some of our own work in this area to ity of probiotics to prevent necrotizing enterocolitis in pre-
illustrate some of the possibilities of “mining the microbiome” term neonates concluded that there are “significant benefits
for novel anti-infectives. of probiotic supplements in reducing death and disease in
preterm neonates” [8]. A similar analysis for the use of probi-
Mining the Microbiome otics in upper respiratory tract infections concluded that
Microbiome-derived anti-infection strategies have ranged “low-quality evidence provides support that probiotics have
from using an entire intact microbiome or a microbial con- potential efficacy for preventing upper respiratory tract infec-
sortium to selecting single biological entities (bacteria, fungi, tion episodes in adults” [9]. Yet another meta-analysis con-
bacteriophage, or bacterial metabolites) (Fig. 2). In most in- ducted on the use of probiotics in children with acute diar-
stances, the objective is one of selective depletion, rather than rhoea “supports the potential beneficial roles of probiotics
the broad-spectrum approach typical of many antibiotic or and synbiotics for acute diarrhoea in children” [10]. There are
antiviral strategies. Much of the data generated to date have also single clinical trials with impressive evidence, for exam-
been acquired from animal models since it can be difficult to ple, a randomised clinical trial in India involving over 45,000
design and perform experiments on humans with respect to children using a symbiotic composed of Lactiplantibacillus
infection. plantarum together with fructooligosaccharides reported a

6 Reprint with permission from: Hill


Ann Nutr Metab 2021;77(suppl 3):4–9
DOI: 10.1159/000516399
significant reduction in sepsis and death in the treatment co- Bacteriophage and Bacteriophage Lysins
hort [11]. Bacteriophages (phages) are bacterial viruses that often have
Mechanistic insights have also been generated in animal a very narrow-spectrum of inhibition. There are several ad-
models. We were able to demonstrate that bacteriocin pro- vantages and disadvantages to using bacteriocins to treat in-
duction is responsible for the protective effect of Ligilactoba- fections, a concept often referred to as phage therapy [25].
cillus salivarius UCC118 against deliberate infection of mice Disadvantages include the narrow host ranges, although, of
with Listeria monocytogenes [12]. We showed that the bacte- course, this could be regarded as an advantage in selective
riocin was highly active against L. monocytogenes and that a depletion strategies. This disadvantage can be addressed by
non-bacteriocin-producing genetic knockout had no protec- using cocktails of phages to cover a wider spectrum of target
tive effect against the pathogen in the same model. Other bacteria, or by carefully selecting the correct phage from an
anti-infective mechanisms could include interacting with the existing phage bank to use against a specific pathogenic strain
immune system or improving barrier function [13]. Bacterio- infecting a patient. Another disadvantage is the ease with
cins represent another example of the “selective depletion” which bacteria can develop resistance to the phage attack,
concept in that they are usually narrow spectrum and should again an issue that may be overcome by using multiple phag-
be rapidly broken down by proteases and peptidases in the es in cocktails. Advantages include their widespread distribu-
gut and therefore unlikely to select for resistance. tion in nature and subsequent ease of isolation in many in-
stances, and, of course, their ability to multiply at the site of
Bacteriocins infection. However, the clinical evidence is scarce. Phages
Concentrated or purified bacteriocins have also been ex- have been used for decades in eastern Europe, and it is logical
plored for their potential in preventing or treating infection to assume they must have benefits in that they have survived
(reviewed by [14–16]). Bacteriocins have been largely tested as frontline treatments for so many years [26]. In the West,
in laboratory animal models, although a broad-spectrum lan- phages have been used in some high-profile instances in sin-
tibiotic, nisin, has been used commercially to prevent mastitis gle patients with infections that were recalcitrant to antibi-
in dairy cattle [17, 18]. Some bacteriocins such as nisin are otic therapies [27, 28]. Phages can potentially be applied on
relatively broad spectrum, but an attempt has also been made the skin, by inhalation, or by ingestion [29].
to use narrow-spectrum bacteriocins for “selective depletion” Lysins are enzymes produced by bacteriophages that are
of specific pathogens. One example is the discovery of thuri- responsible for lysing the target bacterium following multipli-
cin CD, a very narrow-spectrum bacteriocin targeting C. dif- cation of phage particles. Phage lysins target the cell wall and
ficile [19, 20]. Thuricin CD is produced by a strain of Bacillus can also act from outside the cell. Lysins can be broader spec-
thuringiensis that was mined from the human gut microbiome trum than their carrying phages, but are still limited to the spe-
in an extensive screening program. It was subsequently dem- cies or genus level, making them good candidates for selec-
onstrated in improvised models of the human colon (faecal tive depletion strategies. There have been many attempts to
fermentations) that thuricin CD could dramatically reduce C. use phage lysins to target specific pathogens (reviewed by [30,
difficile levels in a highly complex microbiome without caus- 31], particularly on the skin [32] and in the respiratory tract [33].
ing substantial collateral damage to the other members of the As is the case for bacteriocins, these gene-encoded antimi-
community. Antibiotics or broad-spectrum bacteriocins de- crobials can readily be engineered for greater efficacy [34].
ployed in the same model also reduced levels of C. difficile
but also caused significant shifts in microbiome composition
[20]. While this approach has not been validated in humans or Conclusions
animals, it provides a glimpse of future developments in this
exciting area. The microbiome provides both a barrier to infection and a
Bacteriocins also have a potential as antimicrobials for source that can be mined for novel antimicrobials. While
treating skin infections, some as broad-spectrum inhibitors these antimicrobials, including FMT, microbial consortia, pro-
such as garvicin KS and micrococcin P1 [21, 22], but others as biotics, bacteriocins, and bacteriophages (and their lysins),
potentially narrow-spectrum inhibitors that could be used in have often been analysed for their solo impact on infection,
selective depletion strategies. Because bacteriocins are gene there is no reason why these could not be used as combina-
encoded, they can be engineered to improve their physico- torial treatments. Perhaps the most attractive feature of these
chemical characteristics and/or inhibition spectrum [23]. Bac- microbiome-based inhibitors is their potential narrow spec-
teriocins can also be used in combination with other antimi- trum of inhibition that would act to selectively deplete the
crobials for greater effect [24]. pathogen, while preserving the complexity of the surrounding

A Case for Selective Depletion in Treating Reprint with permission from: 7


Infectious Disease Ann Nutr Metab 2021;77(suppl 3):4–9
DOI: 10.1159/000516399
microbiome. This might require precise identification of the Statements of Ethics
pathogen before initiating treatment, but this is not a signifi-
No ethical approval is required as there is no original data in this ar-
cant problem with chronic infections and is becoming less of ticle.
a problem with speedy high-throughput bacterial identifica-
tion becoming more accessible for acute infections [35]. The
future of smart antimicrobial therapies may not involve patho- Conflict of Interest Statement
gen elimination with collateral damage to the microbiome
(broad-spectrum approaches) but may well lie in restricting The writing of this article was supported by Nestlé Nutrition Institute
and the author declares no other conflicts of interest.
the growth of pathogens to a point where they cannot cause
disease while harnessing the power of the microbiome to en-
sure a return to a healthy state – a strategy I designate as se-
Funding Sources
lective depletion.
The author is an investigator at APC Microbiome Ireland that receives
funding from the Science Foundation Ireland (SFI) (Grant No. SFI/12/
RC/2273).

References
1 Hill C. You have the microbiome you deserve. Gut Microb. 2020; 11 Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L, Chandel
1: E3. DS, et al. A randomized synbiotic trial to prevent sepsis among
infants in rural India. Nature. 2017; 548(7668): 407–12.
2 O’Hara AM, Shanahan F. The gut flora as a forgotten organ. EMBO
Rep. 2006; 7: 688–93. 12 Corr SC, Li Y, Riedel CU, O’Toole PW, Hill C, Gahan CGM. Bacte-
riocin production as a mechanism for the antiinfective activity of
3 Patin NV, Peña-Gonzalez A, Hatt JK, Moe C, Kirby A, Konstantini-
Lactobacillus salivarius UCC118. Proc Natl Acad Sci U S A. 2007;
dis KT. The role of the gut microbiome in resisting norovirus infec-
104(18): 7617–21.
tion as revealed by a human challenge study. mBio. 2020; 11:
e02634–20. 13 Preidis GA, Hill C, Guerrant RL, Ramakrishna BS, Tannock GW, Ver-
salovic J. Probiotics, enteric and diarrheal diseases, and global
4 Baunwall SMD, Lee MM, Eriksen MK, Mullish BH, Marchesi JR, Dah-
health. Gastroenterology. 2011; 140(1): 8–14.
lerup JF, et al. Faecal microbiota transplantation for recurrent
Clostridioides difficile infection: an updated systematic review and 14 Lopetuso LR, Giorgio ME, Saviano A, Scaldaferri F, Gasbarrini A,
meta-analysis. EClinicalMedicine. 2020; 29–30: 100642. Cammarota G. Bacteriocins and bacteriophages: therapeutic
weapons for gastrointestinal diseases? Int J Mol Sci. 2019; 20(1):
5 Xu D, Chen VL, Steiner CA, Berinstein JA, Eswaran S, Waljee AK, et
183.
al. Efficacy of fecal microbiota transplantation in irritable bowel
syndrome: a systematic review and meta-analysis. Am J Gastro- 15 Meade E, Slattery MA, Garvey M. Bacteriocins, potent antimicro-
enterol. 2019; 114: 1043–50. bial peptides and the fight against multi drug resistant species:
resistance is futile? Antibiotics. 2020; 9(1): 32.
6 McGovern BH, Ford CB, Henn MR, Pardi DS, Khanna S, Hohmann
EL, et al. SER-109, an investigational microbiome drug to reduce 16 Soltani S, Hammami R, Cotter PD, Rebuffat S, Said LB, Gaudreau
recurrence after Clostridioides difficile infection: lessons learned H, et al. Bacteriocins as a new generation of antimicrobials: toxic-
from a phase 2 trial. Clin Infect Dis. 2020; ciaa387. ity aspects and regulations. FEMS Microbiol Rev. 2021; 45(1):
fuaa039.
7 Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al.
Expert consensus document. The international scientific associa- 17 Cao LT, Wu JQ, Xie F, Hu SH, Mo Y. Efficacy of nisin in treatment
tion for probiotics and prebiotics consensus statement on the of clinical mastitis in lactating dairy cows. J Dairy Sci. 2007; 90(8):
scope and appropriate use of the term probiotic. Nat Rev Gastro- 3980–5.
enterol Hepatol. 2014; 11(8): 506–14.
18 Halasa T, Huijps K, Østerås O, Hogeveen H. Economic effects of
8 Deshpande G, Rao S, Patole S, Bulsara M. Updated meta-analysis bovine mastitis and mastitis management: a review. Vet Q. 2007;
of probiotics for preventing necrotizing enterocolitis in preterm 29(1): 18–31.
neonates. Pediatrics. 2010; 125: 921–30.
19 Rea MC, Sit CS, Clayton E, O’Connor PM, Whittal RM, Zheng J, et
9 Li L, Hong K, Sun Q, Xiao H, Lai L, Ming M, et al. Probiotics for pre- al. Thuricin CD, a posttranslationally modified bacteriocin with a
venting upper respiratory tract infections in adults: a systematic narrow spectrum of activity against Clostridium difficile. Proc Natl
review and meta-analysis of randomized controlled trials. Evid Acad Sci U S A. 2010; 107(20): 9352–7.
Based Complement Alternat Med. 2020; 2020: 8734140.
20 Rea MC, Dobson A, O’Sullivan O, Crispie F, Fouhy F, Cotter PD, et
10 Yang B, Lu P, Li M-X, Cai X-L, Xiong W-Y, Hou H-J, et al. A meta- al. Effect of broad- and narrow-spectrum antimicrobials on Clos-
analysis of the effects of probiotics and synbiotics in children with tridium difficile and microbial diversity in a model of the distal co-
acute diarrhea. Medicine. 2019; 98: e16618. lon. Proc Natl Acad Sci U S A. 2011; 108(Suppl 1): 4639–44.

8 Reprint with permission from: Hill


Ann Nutr Metab 2021;77(suppl 3):4–9
DOI: 10.1159/000516399
21 Kranjec C, Ovchinnikov KV, Grønseth T, Ebineshan K, Srikantam A, 28 Strathdee SA, Patterson TL, Barker T. The perfect predator: a sci-
Diep DB. A bacteriocin-based antimicrobial formulation to effec- entist’s race to save her husband from a deadly superbug. 1st ed.
tively disrupt the cell viability of methicillin-resistant Staphylococ- New York ; Boston: Hachette Books; 2019.
cus aureus (MRSA) biofilms. NPJ Biofilms Microbiomes. 2020;6(1):
29 Loh B, Gondil VS, Manohar P, Khan FM, Yang H, Leptihn S. Encap-
58.
sulation and delivery of therapeutic phages. Appl Environ Micro-
22 Ovchinnikov KV, Kranjec C, Thorstensen T, Carlsen H, Diep DB. biol AEM. Forthcoming 2020.
Successful development of bacteriocins into therapeutic formula-
30 Vázquez R, García E, García P. Phage lysins for fighting bacterial
tion for treatment of MRSA skin infection in a murine model. An-
respiratory infections: a new generation of antimicrobials. Front
timicrob Agents Chemother. 2020; 64(12): e00829–20.
Immunol. 2018; 9: 2252.
23 Field D, Cotter PD, Hill C, Ross RP. Bioengineering lantibiotics for
31 Fischetti V. Development of phage lysins as novel therapeutics: a
therapeutic success. Front Microbiol. 2015; 6: 1363.
historical perspective. Viruses. 2018; 10: 310.
24 Todorov SD, de Paula OAL, Camargo AC, Lopes DA, Nero LA.
32 Wang Z, Kong L, Liu Y, Fu Q, Cui Z, Wang J, et al. A phage lysin
Combined effect of bacteriocin produced by Lactobacillus plan-
fused to a cell-penetrating peptide kills intracellular methicillin-
tarum ST8SH and vancomycin, propolis or EDTA for controlling
resistant Staphylococcus aureus in keratinocytes and has poten-
biofilm development by Listeria monocytogenes. Rev Argent Mi-
tial as a treatment for skin infections in mice. Appl Environ Micro-
crobiol. 2018; 50(1): 48–55.
biol. 2018; 84(12): e00380.
25 Principi N, Silvestri E, Esposito S. Advantages and limitations of
33 Wienhold S-M, Lienau J, Witzenrath M. Towards inhaled phage
bacteriophages for the treatment of bacterial infections. Front
therapy in Western Europe. Viruses. 2019; 11: 295.
Pharmacol. 2019; 10: 513.
34 De Maesschalck V, Gutiérrez D, Paeshuyse J, Lavigne R, Briers Y.
26 Kutateladze M, Adamia R. Phage therapy experience at the Eliava
Advanced engineering of third-generation lysins and formulation
Institute. Med Mal Infect. 2008; 38(8): 426–30.
strategies for clinical applications. Crit Rev Microbiol. 2020; 46:
27 Dedrick RM, Guerrero-Bustamante CA, Garlena RA, Russell DA, 548–64.
Ford K, Harris K, et al. Engineered bacteriophages for treatment of
35 Özenci V, Rossolini GM. Rapid microbial identification and antimi-
a patient with a disseminated drug-resistant Mycobacterium ab-
crobial susceptibility testing to drive better patient care: an evolv-
scessus. Nat Med. 2019; 25(5): 730–3.
ing scenario. J Antimicrob Chemother. 2019; 74: i2–5.

A Case for Selective Depletion in Treating Reprint with permission from: 9


Infectious Disease Ann Nutr Metab 2021;77(suppl 3):4–9
DOI: 10.1159/000516399
Focus
Emerging evidence suggests that antibiotic exposure during
birth can be likened to a chemical C-section from the
microbial perspective, eliminating the natural transmission of
gut microbiota, especially bifidobacteria, from mother to infant

Reprinted with permission from: Ann Nutr Metab 2021;77(suppl 3):11–19

Impact of Delivery Mode on Infant Gut Microbiota


Katri Korpela

Key Insight
Exposure to maternal gut microbes
Infant gut is colonized
C-section birth or exposure to intrapartum antibiotics by specific maternal
Infant Vaginal birth microbes
eliminates the normal mother-to-infant transmission of (bifidobacterial and
beneficial microbes. This alters the development of the infant Bacteroides species)
gut microbiota, with negative consequences on immune and Exposure to skin microbes, microbes
metabolic development. C-section deliveries are increasing from the hospital environment
Lower abundance of
worldwide, affecting over 50% of infants in certain regions. In bifidobacterial and
Infant C-section birth
developed countries, over 30% of infants are also exposed to almost total lack of
Bacteroides species
antibiotics during vaginal birth. This suggests that natural
microbial colonization at birth is currently disrupted in up to
half of infants, even in regions with low C-section rates and
C-section birth eliminates the normal mother-to-infant transmission
good antimicrobial stewardship.
of beneficial microbes.

Current knowledge
During vaginal birth, the infant is exposed to diverse maternal addressing the microbiota imbalance in infants born via C-
microbes, of which specific fecal microbes colonize the in- section and in infants exposed to antibiotics before or during
fant’s gut. However, not all maternal gut bacteria have the birth. The impact of antibiotic exposure can be modified by
ability to colonize infants. It is important to note that the ma- feeding practices: evidence shows that formula-fed infants
ternal gut microbiota is not transmitted as an entire commu- who are on antibiotics are more severely affected by antibi-
nity: only specific members are able to establish persistent otic exposure. Breastfeeding and probiotics are particularly
populations in the infant. The dominant maternally-derived important for infants with disrupted gut colonization, as these
colonizers of the infant gut are bifidobacteria and Bacteroi- measures enhance the growth of beneficial bifidobacteria. Fe-
des. Infants born by C-section are colonized by bacteria from cal microbiota transplant from mother to neonate has also
the environment, including potential pathogens from the been shown to restore normal gut microbiota in C-section-
hospital environment. Therefore, there is a pressing need to born infants but requires careful screening of the mother.
restore normal gut microbiota composition in C-section-
born and antibiotic-exposed infants.
Recommended reading

Practical implications Korpela K, Helve O, Kolho K, Saisto T, Skogberg K, Dikareva E,


et al. Maternal fecal microbiota transplantation in cesarean-
Gut microbiota development is an important part of the phys- born infants rapidly restores normal gut microbial develop-
iological development of infants and should be considered in ment: a Proof-of-Concept Study. Cell. 2020;183:324–34.e5.
clinical practice. Attention should therefore be paid towards

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/


S. Karger AG, Basel
Review Article

Reprint with permission from:


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498

Impact of Delivery Mode on Infant


Gut Microbiota
Katri Korpela  

Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland

Key Messages with maternal microbes, preventing vertical transmission of


gut microbes. Consequently, infants are colonized by bacte-
• Gut microbiota development is part of the physiological ria from the environment, including potential pathogens from
development of infants and should be considered in clinical
the hospital environment. Recent studies have shown that
practice.
• A large proportion of infants are exposed to C-section birth intrapartum antibiotic exposure has a C-section-like effect
or intrapartum antibiotics that eliminate normal mother-to- on the infant gut microbiota. While the composition of the
infant transmission of beneficial microbes, altering the gut microbiota largely normalizes during the first year of life,
development of their gut microbiota and consequently epidemiological studies suggest that the aberrant early mi-
immune and metabolic development.
crobial exposures have long-term immunological and meta-
• Breastfeeding and probiotics are especially important for
infants with disrupted gut microbiota colonization. bolic consequences. Because of the high prevalence of pro-
• Faecal microbiota transplant from mother to neonate has cedures that prevent normal gut microbiota development,
been shown to restore normal gut microbiota in C-section- effective methods to normalize the gut microbiota of neo-
born infants but requires careful screening of the mother. nates are urgently needed. Even more importantly, attention
should be paid to the microbiota imbalance in C-section-
Keywords born and antibiotic-exposed infants in clinical practice.
Antibiotics · Bifidobacteria · Breastfeeding · C-section · Breastfeeding and probiotics are particularly important for in-
Infancy and Childhood · Microbiota · Pediatrics · Probiotics fants with disrupted gut colonization.
© 2021 Nestlé Nutrition Institute, Switzerland/
S. Karger AG, Basel

Abstract
Microbial colonization of the neonate is an important feature Colonization and Early Development of Infant
of normal birth. The gut microbiota has a central role in the Gut Microbiota
programming of the host’s metabolism and immune func-
tion, with both immediate and long-term health conse- Although the notion of infants being born sterile has been
quences. During vaginal birth, the infant is exposed to diverse contested by the detection of both bacteria and bacterial DNA
maternal microbes, of which specific faecal microbes colo- in the placenta, amniotic fluid, and meconium passed within
nize the infant’s gut. C-section eliminates the infant’s contact hours of birth, the expert consensus is currently that micro-

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/ Correspondence to:


S. Karger AG, Basel Katri Korpela, katri.korpela @ helsinki.fi
Microbial signals to the host Microbial signals to the host

Clostridia Enterobacteria

Relative abundances

Relative abundances
Bifidobacteria Clostridia
Enterobacteria

Bifidobacteria
Bacteroides
Bacteroides

Age Age
a Weaning b Weaning

Fig. 1. Temporal development of the relative abundances of the dominant classes of bacteria in the infant gut in
vaginally born, breastfed infants (a) and CS-born infants (b). CS, C-section.

bial colonization of the infant, that is, the establishment of broad range of HMOs [13]. In contrast, Firmicutes and Proteo-
replicating microbial cells, does not commonly occur in bacteria generally do not degrade HMOs, with the exception
healthy pregnancies [1, 2]. In any case, all infants are colonized of certain Lactobacillus, Streptococcus, and Enterococcus
by microbes during birth. The microbes that have been ob- strains with narrow HMO utilization spectra [11].
served in utero do not include the normal infant gut microbes, The importance of early maternal colonization is exempli-
which must be acquired during birth. fied by the finding that maternally derived microbes colonize
During birth, infants are exposed to maternal vaginal, fae- the infant essentially permanently, while non-maternal mi-
cal, and skin bacteria, which colonize the infant body sites that crobes are more transient [6]. This is suggestive of special
provide a suitable habitat and nutrients. Each bacterial species compatibility between infant and maternal microbes, poten-
has specific habitat requirements and cannot permanently tially related to partly genetically determined factors such as
colonize inhospitable body sites. The infant gut is colonized breast milk oligosaccharide composition, gut mucus compo-
at birth by maternal gut microbes [3–8], while maternal vagi- sition, and immune system.
nal microbes do not colonize the infant gut [7, 9, 10]. Not all While the gut microbiota is generally characterized by tre-
maternal gut bacteria have the ability to colonize infants, and mendous interindividual variation, the microbiota develop-
it is important to note that the gut microbiota is not transmit- ment during infancy follows a fairly clear and uniform pattern,
ted as a whole community, but only specific members are dependent on birth mode (Fig. 1). During the first year of life,
able to establish persistent populations. The dominant mater- ecological succession takes place in the infant gut with a con-
nally derived colonizers of the infant gut are bifidobacteria sistent temporal pattern across cultures [14]. The first stools
and Bacteroides [6, 7]. Several species of Bifidobacterium and harbour a highly diverse microbiota of mostly transient pass-
Bacteroides have the capacity to ferment human milk oligo- ers-by [7]. After the first few days, the diversity decreases as
saccharides (HMOs) [11], which very likely explains their suc- specific microbes bloom while others disappear [7]. Initially
cess in colonizing the infant gut [5]. While certain infant gut- dominant species include aerotolerant taxa such as staphylo-
adapted Bifidobacterium strains, such as B. longum subsp. in- cocci, enterococci, and streptococci as well as enterobacte-
fantis, B. bifidum, and B. breve, are specialized in HMO ria [12, 14]. Within weeks, the composition changes as breast
utilization [12], Bacteroides spp. are substrate generalists. milk oligosaccharide-degrading bifidobacteria outgrow. Bifi-
Bacteroides fragilis, thetaiotaomicron, and vulgatus are effi- dobacteria are usually the dominant group until weaning after
cient HMO utilizers [11]. They exploit the same pathways for which taxa that utilize plant-based carbohydrates, mainly
host mucus and HMO degradation and are able to degrade a members of the clostridial families Lachnospiraceae and Ru-

12 Reprint with permission from: Korpela


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498
Table 1. Consistent gut microbiota patterns observed in CS-born infants compared to vaginally born

Increased in CS Decreased in CS Reference (large birth cohort


studies or meta-analyses)

Bacteroides x [12, 14, 16, 17]


Bifidobacteria x [12, 14, 16]
Clostridia x [12]
Gram-negative pathogens x [14, 18]
Gram-positive pathogens x [14, 16, 18]

minococcaceae, begin to gradually increase in relative abun- It has been suggested that in terms of microbial contact,
dance [14]. The abundance of Bacteroides can be high already CS delivery after rupture of membranes would more closely
very early and remains fairly stable through the early develop- resemble vaginal delivery than elective CS in which the infant
ment [14]. This likely reflects the generalist nature of Bacte- has no contact with maternal birth canal microbiota. While
roides species in terms of substrate utilization potential, in- the exposure of mother and infant to the labour process un-
cluding utilization of both diverse dietary polysaccharides and doubtedly has physiological effects, there is no reason to ex-
host-derived glycans [13]. There are some geographical dif- pect much benefit for the microbial colonization from labour,
ferences in the average gut microbiota composition of in- even with rupture of membranes, as maternal faecal microbes
fants. African infants show an increased relative abundance of are unlikely to be present in the birth canal of healthy moth-
Bacilli, while North American infants have more Bacteroidetes ers. Indeed, the hypothesis has not received much support in
and less Actinobacteria than infants in other continents [14]. the scientific literature. Only a few studies have compared the
Whether these differences are caused by technical or bio- effect of different CS types on infant gut microbiota. Emer-
logical reasons is unknown. gency CS seems to result in more drastic and long-lasting
changes in infant gut microbiota [18, 20], possibly due to
health differences. To conclude, there is currently no evi-
Impact of C-Section Birth on Gut Microbiota dence suggesting a beneficial effect of rupture of membranes
or CS during labour versus pre-labour CS on infant gut micro-
The natural microbiota transmission from mother to infant is biota.
disrupted by C-section (CS) birth [6], resulting in a deviation Thus far, only a few studies have looked into the viral com-
of microbiota development, most notably in the first 6–12 ponent of the gut microbiota, the so-called gut virome, in
months of life [14]. In fact, CS birth is perhaps the factor with early infancy. Recent results show that most of the viruses in
the most uniform effects on the gut microbiota across indi- the neonate gut are phages, that is, virus-infecting bacteria
viduals and studies. A large study on nearly 600 infants found [21]. There is evidence that phages are transmitted from the
delivery mode to be the most important factor influencing mother at birth together with their host bacteria [5, 22]. Con-
infant gut microbiota composition, with a stronger effect than sequently, total virus and phage diversity has been shown to
postnatal antibiotic treatments [15]. A particularly prominent be reduced in CS-born infants at the age of 1 year, with par-
and ubiquitous feature of the CS-born infant’s gut microbiota ticularly low abundances of Anelloviridae and Caudovirales in
is the low relative abundance of bifidobacteria and almost to- CS-born infants [23]. In fact, the effect of birth mode on the
tal lack of Bacteroides [14], that is, the taxa that would nor- gut virome was much more prominent at 1 year than the ef-
mally be obtained from the mother at birth and that are large- fect on the bacteriome [23]. This indicates that birth is crucial
ly responsible for breast milk oligosaccharide fermentation in for the acquisition of the gut phageome, and the potential for
the infant gut. These patterns are observed in practically all postnatal colonization, for example, via breast milk or the en-
studies investigating the impact of CS on infant gut microbi- vironment, is limited.
ota (Table 1), including several large cohort studies of 600– A meta-analysis of the dominant bacterial classes revealed
1,000 infants each [15–18]. In addition, the relative abundance that the differences between birth modes gradually even out
of pathogenic bacteria is often increased in CS-born infants. and mostly disappear by the age of 12 months [14]. However,
These taxa are common in the hospital and are transmitted only a few studies have investigated the associations between
from hospital surfaces to new-born infants [19]. birth mode and gut microbiota development after infancy.
Reduced diversity of Bacteroides spp. has been reported up

Impact of Delivery Mode Reprint with permission from: 13


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498
to the age of 2 years [24]. Another study found reduced rela- Table 2. Consistent gut microbiota patterns observed in vaginally
born infants exposed to intrapartum antibiotics compared to those
tive abundances of several members of Lachnospiraceae and
not exposed to antibiotics
Ruminococcaceae in CS-born children at the age of 4 years
[25]. Comparably, a study using fluorescent in situ hybridiza- Increased Decreased Reference
tion found a reduced abundance of clostridia in CS-born in exposed in exposed
compared to vaginally born 7 year olds [26]. A study using
Bifidobacteria x [30–34]
quantitative PCR found significant differences in gut micro-
Clostridia x [20, 30, 32]
biota composition between CS-born and vaginally born Enterobacteria x [20, 30, 32]
young adults [27]. In particular, relatives of Bacteroides fragilis Streptococci x [30, 32]
and Lactobacillus sakei were reduced in CS-born children. Gram-negative pathogens x [18]
The results suggest that birth mode effects on the gut micro- Gram-positive pathogens x [18]

biota may in some cases persist even into adulthood. Notably,


the latter 2 studies employed quantitative measures (FISH and
qPCR, respectively) that can estimate the absolute abundance
of the bacteria, in contrast to sequencing which only reveals teria from mother to infant. Antibiotics are not selective be-
relative abundances. Clearly, future studies need to employ tween pathogens and non-pathogens, so if the transmission
methods that allow the absolute abundances to be estimated of pathogens is prevented, so must be the transmission of
[28]. non-pathogenic bacteria with a similar antibiotic sensitivity
profile.
In studies investigating the impact of intrapartum antibiot-
Impact of Intrapartum Antibiotics on Infant Gut ics in vaginally born infants, bifidobacteria appear to be par-
Microbiota ticularly strongly negatively affected by the antibiotic expo-
sure (Table 2). There is discrepancy regarding the impact of
In most hospitals, antibiotic treatment given to the mother is intrapartum antibiotic exposure on Bacteroides, with effects
an integral part of CS delivery. Therefore, there has been some varying from negative [17, 20, 30] to neutral [20, 31–33] to
unclarity as to whether the effect of CS birth is exclusively due positive [30]. Importantly, the impact of antibiotic exposure is
to the lack of exposure to maternal faecal microbes or partly modified by feeding practice, with formula-fed infants more
due to the antibiotic treatment. Considering that regardless of severely affected [34–36]. Overall, the emerging evidence
maternal microbiota composition, CS eliminates the contact suggests that antibiotic exposure during birth can be likened
between maternal faecal microbes and infant, thus preventing to a chemical CS from the microbial perspective, eliminating
colonization, and any additional microbiota-affecting treat- the natural transmission of gut microbiota, especially bifido-
ments seem trivial. If a microbe has no contact with the infant, bacteria, from mother to infant.
it cannot colonize the infant regardless of exposure to antibi-
otics. This view was recently confirmed in a study comparing
the effects of pre- versus post-CS antibiotic prophylaxis on Consequences of Disrupted Vertical Microbial
the infants’ gut microbiota composition [29]. No significant Transfer
differences in gut microbiota composition were found be-
tween infants exposed and not exposed to maternal antibi- Due to the age-associated microbiota succession, the host
otic prophylaxis, demonstrating that antibiotic exposure is not receives specific microbial signals at specific age windows
a driver of the CS-induced microbiota imbalance. Along the (Fig. 1). If these signals are altered or missing due to abnormal
same lines, in an analysis of 700 infants, stratification by intra- microbiota development, the host’s immune and metabolic
partum antibiotic exposure did not alter the results regarding development may be affected. Deviations from normal mi-
the effect of CS birth on infant gut microbiota [18]. crobiota succession in infants and young children usually oc-
The effect of antibiotic treatment during vaginal birth is an cur without overt symptoms, but can increase the susceptibil-
important issue in itself, since a large proportion of infants are ity to later health problems, such as allergic disease [37–39],
exposed to intrapartum antibiotics either due to prophylaxis autoimmune diseases [40], and overweight [41].
or treatment of maternal infection. In some countries, all Bifidobacteria normally form the dominant taxon in healthy
mothers who carry group B Streptococcus are given antibi- vaginally born breastfed infants, representing up to 90% of the
otic prophylaxis during labour. The aim of intrapartum antibi- total gut microbiota. Due to their abundance and metabolic
otic prophylaxis is to prevent transmission of pathogenic bac- capacities, they have important effects on gut microbiota

14 Reprint with permission from: Korpela


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498
Increased risk of allergic disease and overweight in later childhood

Altered immune and metabolic programming

Pain, gut symptoms

Altered microbial Reduced colonisation


signals to the host resistance against
pathogens Inflammation

Altered SCFA and


bile acid compositions Increased pH Weakened gut barrier

Low abundance of Bifidobacteria


Fig. 2. Effects of low abundance of bifido-
bacteria in infants.

composition and function and host health (Fig. 2). Unfortu- Not only are bifidobacteria important for early-life gut
nately, bifidobacteria are perhaps the most vulnerable bacte- health but also have widespread immunological effects,
rial group in infants, affected negatively by all unnatural prac- which is particularly important during the time of immune
tices such as CS birth, antibiotics, formula feeding, early system development. Indeed, numerous studies have shown
weaning, and even maternal stress. Bifidobacteria are the that a low abundance of bifidobacteria, especially B. longum,
dominant HMO-utilizing group in the infant gut. and a high abundance of Proteobacteria in infancy is associ-
As a consequence of the reduced abundance of dominant ated with an increased risk of allergic diseases in later child-
HMO fermenters and lower fermentation activity, an increased hood [48–51]. Several studies have shown that low early
intestinal pH and decreased levels of the short-chain fatty acid abundance of bifidobacteria is predictive of later adiposity, as
propionate has been observed in CS-born infants [42]. As well [41, 52, 53].
HMOs constitute approximately 20% of all carbohydrates in As a demonstration of the clinical consequences of the
breast milk [43], and the majority of them are degraded by gut disrupted microbial colonization, several large cohort studies
bacteria into short-chain fatty acids [44], HMO fermentation and meta-analyses have shown that birth by CS is associated
represents a significant energy source for the infant. This indi- with long-term health effects, including increased risk of
cates that CS-born infants do not receive the full nutritional chronic immune diseases and obesity [54–56], with some of
value of breast milk. Furthermore, the altered gut pH has been the negative health associations reaching into adulthood [54].
shown to be permissive for pathogen colonization [42], and While some associations may be caused by confounding ef-
indeed several studies have observed increased abundances fects, the clear alterations in gut microbiota and the impor-
of pathogens in CS-born infants (Table 1). Bifidobacteria have tance of the gut microbiota for host physiological develop-
anti-streptococcal activity, indicating that a low abundance of ment support the causal attribution.
bifidobacteria in the gut may increase the growth of patho-
genic streptococci [31]. In addition to the increase in patho-
genic microbes, there is evidence that a microbiota with re- Correction of Infant Microbiota after Disrupted
duced capacity for oligosaccharide utilization degrades the Colonization
intestinal mucus, potentially leading to weakened intestinal
barrier [45]. This together with the increase in pathogenic mi- CS deliveries are increasing worldwide, affecting over 50% of
crobes is likely to stimulate inflammation both systemically infants in certain regions [57]. Additionally, over 30% of infants
and locally. Indeed, signs of pain and distress in infants are cor- are exposed to antibiotics during vaginal birth in developed
related with low abundance of bifidobacteria [46, 47]. countries [58, 59]. These figures are alarmingly high, suggest-

Impact of Delivery Mode Reprint with permission from: 15


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498
Benefits Disadvantages

• The cheapest and least invasive • Efficacy dependent on


method maternal genotype
• Promotes normal microbiota • Ineffective at full microbiota
composition and function restoration
• Additional health benefits for
Breastfeeding both infant and mother

• Can be effective at normalising • Ineffective in formula-fed


bifidobacterial abundance infants without added prebiotics
when combined with • Effect is strain-specific
breastfeeding or prebiotics • Ineffective at full microbiota
• Safe and widely available restoration
Probiotics

• The most effective method • Requires careful screening of


• Full microbiota restoration the mother and monitoring of
• Restores vertical transmission the infant
of microbes from mother • Only in hospital
to infant • Not suitable if mother carries
Fig. 3. Studied methods of microbiota • One-time application potential pathogens
normalization in CS-born infants. CS, C-
FMT
section; FMT, faecal microbiota trans-
plantation.

ing that natural microbial colonization at birth is currently dis- low abundance of lactobacilli, but those are gut adapted or
rupted in up to half of infants even in regions with low CS rates food derived, rather than of vaginal origin [61]. Whether the
and good antibiotic stewardship. Therefore, there is a pressing transient exposure to vaginal microbes during birth plays a
need to restore normal gut microbiota composition in CS- role in, for example, immune stimulation, is currently un-
born and antibiotic-exposed infants. Only a few different known.
methods have been studied in terms of their efficacy in restor- A few studies have tested the efficacy of probiotics as a
ing normal gut microbiota in CS-born infants (Fig. 3). means of microbiota restoration in CS-born infants. A
A study comparing the microbiota found on different in- 6-month daily supplementation with a probiotic product
fant body sites within minutes after birth and meconium sam- containing several Lactobacillus and Bifidobacterium strains
ples taken within the first 24 h to maternal skin and vaginal has been shown to alleviate the CS- and antibiotic-associated
samples showed, unsurprisingly, that vaginally born infants, microbiota disturbance in breastfed infants [62]. Importantly,
still covered with vaginal fluids at the time of sampling, har- the treatment reduced the incidence of allergic diseases in
boured vaginal microbes, and CS-born infants harboured skin CS-born infants, demonstrating that gut microbiota-targeting
microbes [60]. These results were interpreted to mean that interventions have beneficial health effects [63]. However, the
the infant gut is colonized by maternal vaginal microbes at effect of the probiotic intervention on the gut microbiota was
birth, although faecal microbiota was not sampled in the dependent on breastfeeding and failed to increase bifidobac-
study. However, the samples were taken too early to distin- teria in the exclusively formula-fed infants [62]. In another tri-
guish colonizers from transient passers-by. In fact, it was al, Lactobacillus reuteri-containing formula was fed to vagi-
shown already in 1990 that vaginal lactobacilli do not colonize nally born and CS-born infants for 6 months, with beneficial
the infant gut [9], and later research has confirmed this [7, 10]. effects on the abundances of bifidobacteria and lactobacilli in
The adult vagina is a very different environment compared to the CS-born group [64]. These results are very promising,
infant body sites, and consequently microbes adapted to the demonstrating the effectiveness of such simple, safe, and
vaginal environment, predominantly specific Lactobacillus cost-effective interventions. However, probiotics represent a
species, are unlikely to find suitable habitats on the infant small fraction of the total microbial diversity that normally
body. Human vaginal lactobacilli are highly adapted to that colonize infants and cannot fully restore normal gut micro-
environment and rarely observed in other body sites [61]. biota. Importantly, probiotics are not maternally derived. It is
Therefore, vaginal microbes are not useful in terms of infant likely that maternal strains are adapted to the specific mother-
gut microbiota restoration [8]. The infant gut does harbour a infant dyad and have been preselected by maternal immune

16 Reprint with permission from: Korpela


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498
system and gut secretions to suit her infant and her breast milk Conclusion
composition.
Breast milk is highly bifidogenic, so breastfeeding could be Because CS eliminates the contact between infant and ma-
expected to restore the low abundance of bifidobacteria in ternal faecal microbes, natural vertical transfer of gut micro-
CS-born infants. Exclusive breastfeeding has been found to biota from mother to infant is prevented. This results in aber-
have a beneficial impact on the gut microbiota of CS-born rant gut microbiota composition with reduced abundance of
infants, especially regarding bifidobacteria [65, 66]. However, breast milk fermenters and increased abundance of patho-
the effect of breastfeeding is dependent on breast milk oligo- gens during the first year of life, possibly longer. Intrapartum
saccharide composition [67]. The oligosaccharide 2′fucosyl- antibiotic exposure during vaginal birth has a similar but
lactose in breast milk alleviates the effect of CS birth on the somewhat weaker impact on the infant gut microbiota as CS.
gut microbiota, but not all mothers secrete 2′fucosyllactose Accumulating evidence indicates that the disrupted gut colo-
[67]. The presence of fucosylated oligosaccharides in breast nization in early life carries long-term health risks, including
milk, the so-called secretor status, is determined by the moth- increased risk of chronic immune diseases and overweight.
er’s FUT2 genotype. The results indicate that infants of non- Due to the high prevalence of both CS and intrapartum anti-
secretor mothers and formula-fed infants are at a particular biotic use, methods to correct the infant gut microbiota are
risk of gut dysbiosis if born by CS. Overall, the results show needed. Breastfeeding together with probiotics, especially
that breastfeeding should be considered essential for infants those containing bifidobacteria, can be used to alleviate the
with disrupted gut colonization. microbiota imbalance and are highly recommendable for all
Due to the fact that infants are normally colonized by ma- CS-born and antibiotic-exposed infants. The efficacy of
ternal faecal microbes during birth, birth can be seen as a fae- mother-to-infant FMT in correcting the infant gut microbiota
cal microbiota transplantation (FMT) event from mother to has been recently demonstrated.
infant. In a recent pilot study, CS-born infants were given an
oral FMT, a few grams of faecal matter mixed with breast milk,
from their own mothers within hours after delivery [8]. The Conflict of Interest Statement
treatment fully restored normal gut microbiota composition,
The writing of this article was supported by Nestlé Nutrition Institute
validating that maternal faecal microbes are the natural colo-
and the author declares no other conflicts of interest.
nizers of the infant gut. While all procedures transferring a
community of microbes from 1 person to another are inher-
ently risky due to the potential of transferring pathogens, FMT Funding Sources
from carefully screened healthy mothers was shown to be
safe [8]. The writing of this article was supported by funding from the Univer-
sity of Helsinki.

References
1 Blaser MJ, Devkota S, McCoy KD, Relman DA, Yassour M, Young 6 Korpela K, Costea P, Coelho LP, Kandels-Lewis S, Willemsen G,
VB. Lessons learned from the prenatal microbiome controversy. Boomsma DI, et al. Selective maternal seeding and environment
Microbiome. 2021; 9(1): 8–7. shape the human gut microbiome. Genome Res. 2018;28:561–8.
2 Walter J, Hornef MW. A philosophical perspective on the prenatal 7 Ferretti P, Pasolli E, Tett A, Asnicar F, Gorfer V, Fedi S, et al. Moth-
in utero microbiome debate. Microbiome. 2021; 9: 1–9. er-to-infant microbial transmission from different body sites
shapes the developing infant gut microbiome. Cell Host Microbe.
3 Makino H, Kushiro A, Ishikawa E, Kubota H, Gawad A, Sakai T, et
2018; 24(1): 133–45.
al. Mother-to-infant transmission of intestinal bifidobacterial
strains has an impact on the early development of vaginally deliv- 8 Korpela K, Helve O, Kolho KL, Saisto T, Skogberg K, Dikareva E, et
ered infant’s microbiota. PLoS One. 2013; 8: e78331. al. Maternal fecal microbiota transplantation in cesarean-born in-
fants rapidly restores normal gut microbial development: a Proof-
4 Asnicar F, Manara S, Zolfo M, Truong DT, Scholz M, Armanini F, et
of-Concept Study. Cell. 2020; 183: 324–34.e5.
al. Studying vertical microbiome transmission from mothers to
infants by strain-level metagenomic profiling. bioRxiv. 2016. Epub 9 Tannock GW, Fuller R, Smith SL, Hall MA. Plasmid profiling of
ahead of print. members of the family Enterobacteriaceae, lactobacilli, and bifi-
dobacteria to study the transmission of bacteria from mother to
5 Duranti S, Lugli GA, Mancabelli L, Armanini F, Turroni F, James K,
infant. J Clin Microbiol. 1990; 28: 1225–8.
et al. Maternal inheritance of bifidobacterial communities and bi-
fidophages in infants through vertical transmission. Microbiome.
2017; 5: 66.

Impact of Delivery Mode Reprint with permission from: 17


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498
10 Sakwinska O, Foata F, Berger B, Brüssow H, Combremont S, Mer- 26 Salminen S, Gibson GR, McCartney AL, Isolauri E. Influence of
cenier A, et al. Does the maternal vaginal microbiota play a role in mode of delivery on gut microbiota composition in seven year old
seeding the microbiota of neonatal gut and nose? Benef Microbes. children. Gut. 2004; 53: 1388–9.
2017; 8: 763–78.
27 Nagpal R, Yamashiro Y. Gut microbiota composition in healthy
11 Yu ZT, Chen C, Newburg DS. Utilization of major fucosylated and Japanese infants and young adults born by C-section. Ann Nutr
sialylated human milk oligosaccharides by isolated human gut mi- Metab. 2018; 73 Suppl 3: 4–11.
crobes. Glycobiology. 2013; 23: 1281–92.
28 Jian C, Luukkonen P, Yki-Järvinen H, Salonen A, Korpela K. Quan-
12 Thomson P, Medina DA, Garrido D. Human milk oligosaccharides titative PCR provides a simple and accessible method for quantita-
and infant gut bifidobacteria: molecular strategies for their utiliza- tive microbiota profiling. PLoS One. 2020; 15: e0227285.
tion. Food Microbiol. 2018; 75: 37–46.
29 Kamal SS, Hyldig N, Krych Ł, Greisen G, Krogfelt KA, Zachariassen
13 Marcobal A, Barboza M, Sonnenburg ED, Pudlo N, Martens EC, G, et al. Impact of early exposure to cefuroxime on the composi-
Desai P, et al. Bacteroides in the infant gut consume milk oligosac- tion of the gut microbiota in infants following cesarean delivery. J
charides via mucus-utilization pathways. Cell Host Microbe. 2011; Pediatr. 2019; 210: 99–105.e2.
10: 507–14.
30 Stearns JC, Simioni J, Gunn E, McDonald H, Holloway AC, Tha-
14 Korpela K, de Vos WM. Early life colonization of the human gut: bane L, et al. Intrapartum antibiotics for GBS prophylaxis alter col-
microbes matter everywhere. Curr Opin Microbiol. 2018;44:70–8. onization patterns in the early infant gut microbiome of low risk
infants. Sci Rep. 2017; 7: 16527–9.
15 Shao Y, Forster SC, Tsaliki E, Vervier K, Strang A, Simpson N, et al.
Stunted microbiota and opportunistic pathogen colonization in 31 Aloisio I, Mazzola G, Corvaglia LT, Tonti G, Faldella G, Biavati B, et
caesarean-section birth. Nature. 2019; 574: 117–21. al. Influence of intrapartum antibiotic prophylaxis against group B
Streptococcus on the early newborn gut composition and evalu-
16 Penders J, Thijs C, Vink C, Stelma FF, Snijders B, Kummeling I, et
ation of the anti-Streptococcus activity of Bifidobacterium strains.
al. Factors influencing the composition of the intestinal microbi-
Appl Microbiol Biotechnol. 2014; 98: 6051–60.
ota in early infancy. Pediatrics. 2006; 118: 511–21.
32 Bugmann H. Functional types of trees in temperate and boreal
17 Fallani M, Young D, Scott J, Norin E, Amarri S, Adam R, et al. Intes-
forests: classification and testing. J Veg Sci. 1996; 7(3): 359–70.
tinal microbiota of 6-week-old infants across Europe: geograph-
ic influence beyond delivery mode, breast-feeding, and antibiot- 33 Imoto N, Morita H, Amanuma F, Maruyama H, Watanabe S, Hashi-
ics. J Pediatr Gastroenterol Nutr. 2010; 51: 77–84. guchi N. Maternal antimicrobial use at delivery has a stronger im-
pact than mode of delivery on bifidobacterial colonization in in-
18 Stokholm J, Thorsen J, Chawes BL, Schjørring S, Krogfelt KA,
fants: a pilot study. J Perinatol. 2018; 38: 1174–81.
Bønnelykke K, et al. Cesarean section changes neonatal gut colo-
nization. J Allergy Clin Immunol. 2016; 138: 881–9.e2. 34 Nogacka A, Salazar N, Suárez M, Milani C, Arboleya S, Solís G, et
al. Impact of intrapartum antimicrobial prophylaxis upon the in-
19 Brooks B, Olm MR, Firek BA, Baker R, Thomas BC, Morowitz MJ,
testinal microbiota and the prevalence of antibiotic resistance
et al. Strain-resolved analysis of hospital rooms and infants reveals
genes in vaginally delivered full-term neonates. Microbiome.
overlap between the human and room microbiome. Nat Com-
2017; 5: 93.
mun. 2017; 8: 1814–7.
35 Corvaglia L, Tonti G, Martini S, Aceti A, Mazzola G, Aloisio I, et al.
20 Azad MB, Konya T, Persaud RR, Guttman DS, Chari RS, Field CJ, et
Influence of intrapartum antibiotic prophylaxis for group B strep-
al. Impact of maternal intrapartum antibiotics, method of birth and
tococcus on gut microbiota in the first month of life. J Pediatr
breastfeeding on gut microbiota during the first year of life: a pro-
Gastroenterol Nutr. 2016; 62(2): 304–8.
spective cohort study. BJOG. 2016; 123: 983–93.
36 Mazzola G, Murphy K, Ross RP, Di Gioia D, Biavati B, Corvaglia LT,
21 Liang G, Zhao C, Zhang H, Mattei L, Sherrill-Mix S, Bittinger K, et
et al. Early gut microbiota perturbations following intrapartum an-
al. The stepwise assembly of the neonatal virome is modulated by
tibiotic prophylaxis to prevent group B streptococcal disease.
breastfeeding. Nature. 2020; 581: 470–4.
PLoS One. 2016; 11(6): e0157527.
22 Siranosian BA, Tamburini FB, Sherlock G, Bhatt AS. Acquisition,
37 Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Eng-
transmission and strain diversity of human gut-colonizing crass-
strand L, Jenmalm MC. Low diversity of the gut microbiota in in-
like phages. Nat Commun. 2020; 11: 280–11.
fants with atopic eczema. J Allergy Clin Immunol. 2012; 129: 434.
23 McCann A, Ryan FJ, Stockdale SR, Dalmasso M, Blake T, Ryan CA,
38 Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Eng-
et al. Viromes of one year old infants reveal the impact of birth
strand L, Jenmalm MC. Low gut microbiota diversity in early in-
mode on microbiome diversity. PeerJ. 2018; 6: e4694.
fancy precedes asthma at school age. Clin Exp Allergy. 2014; 44:
24 Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince 842–50.
C, Jernberg C, et al. Decreased gut microbiota diversity, delayed
39 Azad MB, Konya T, Guttman DS, Field CJ, Sears MR, HayGlass KT,
Bacteroidetes colonisation and reduced Th1 responses in infants
et al. Infant gut microbiota and food sensitization: associations in
delivered by caesarean section. Gut. 2014; 63: 559–66.
the first year of life. Clin Exp Allergy. 2015; 45: 632–43.
25 Fouhy F, Watkins C, Hill CJ, O'Shea CA, Nagle B, Dempsey EM, et
40 Kostic AD, Gevers D, Siljander H, Vatanen T, Hyötyläinen T,
al. Perinatal factors affect the gut microbiota up to four years after
Hämäläinen AM, et al. The dynamics of the human infant gut mi-
birth. Nat Commun. 2019; 10: 1517.
crobiome in development and in progression toward type 1 dia-
betes. Cell Host Microbe. 2015; 17: 260–73.

18 Reprint with permission from: Korpela


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498
41 Korpela K, Zijlmans MA, Kuitunen M, Kukkonen K, Savilahti E, Sa- 55 Keag OE, Norman JE, Stock SJ. Long-term risks and benefits as-
lonen A, et al. Childhood BMI in relation to microbiota in infancy sociated with cesarean delivery for mother, baby, and subsequent
and lifetime antibiotic use. Microbiome. 2017; 5: 26. pregnancies: systematic review and meta-analysis. PLoS Med.
2018; 15: e1002494.
42 Nagpal R, Tsuji H, Takahashi T, Nomoto K, Kawashima K, Nagata
S, et al. Gut dysbiosis following C-section instigates higher colo- 56 Andersen V, Möller S, Jensen PB, Møller FT, Green A. Caesarean
nisation of toxigenic Clostridium perfringens in infants. Benef Mi- delivery and risk of chronic inflammatory diseases (inflammatory
crobes. 2017; 8: 353–65. bowel disease, rheumatoid arthritis, coeliac disease, and diabetes
mellitus): a population based registry study of 2,699,479 births in
43 Thurl S, Munzert M, Boehm G, Matthews C, Stahl B. Systematic
Denmark during 1973–2016. Clin Epidemiol. 2020; 12: 287.
review of the concentrations of oligosaccharides in human milk.
Nutr Rev. 2017; 75: 920–33. 57 Births by caesarean section. WHO; 2018. Available from: https: //
apps.who.int/gho/data/node.main.BIRTHSBYCAESAREAN?
44 Coppa GV, Pierani P, Zampini L, Bruni S, Carloni I, Gabrielli O.
lang=en. Accessed 2020 Sep 22.
Characterization of oligosaccharides in milk and feces of breast-
fed infants by high-performance anion-exchange chromatogra- 58 Stokholm J, Schjørring S, Pedersen L, Bischoff AL, Følsgaard N,
phy. In: Anonymous bioactive components of human milk. Carson CG, et al. Prevalence and predictors of antibiotic admin-
Springer; 2001. p. 307–14. istration during pregnancy and birth. PLoS One. 2013; 8: e82932.
45 Karav S, Casaburi G, Frese SA. Reduced colonic mucin degradation 59 Persaud RR, Azad MB, Chari RS, Sears MR, Becker AB, Kozyrskyj AL.
in breastfed infants colonized by Bifidobacterium longum subsp. Perinatal antibiotic exposure of neonates in Canada and associ-
infantis EVC001. FEBS Open Bio. 2018; 8: 1649–57. ated risk factors: a population-based study. J Matern Fetal Neo-
natal Med. 2015; 28: 1190–5.
46 Pärtty A, Kalliomäki M, Endo A, Salminen S, Isolauri E. Composi-
tional development of Bifidobacterium and Lactobacillus micro- 60 Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hi-
biota is linked with crying and fussing in early infancy. PLoS One. dalgo G, Fierer N, et al. Delivery mode shapes the acquisition and
2012; 7: e32495. structure of the initial microbiota across multiple body habitats in
newborns. Proc Natl Acad Sci U S A. 2010; 107: 11971–5.
47 de Weerth C, Fuentes S, Puylaert P, de Vos WM. Intestinal micro-
biota of infants with colic: development and specific signatures. 61 Mendes-Soares H, Suzuki H, Hickey RJ, Forney LJ. Comparative
Pediatrics. 2013; 131: e550–8. functional genomics of Lactobacillus spp. reveals possible mech-
anisms for specialization of vaginal lactobacilli to their environ-
48 Sjogren YM, Jenmalm MC, Bottcher MF, Bjorksten B, Sverremark-
ment. J Bacteriol. 2014; 196: 1458–70. 7
Ekstrom E. Altered early infant gut microbiota in children develop-
ing allergy up to 5 years of age. Clin Exp Allergy. 2009;39:518–26. 62 Korpela K, Salonen A, Vepsäläinen O, Suomalainen M, Kolmeder
C, Varjosalo M, et al. Probiotic supplementation restores normal
49 Akay HK, Tokman HB, Hatipoglu N, Hatipoglu H, Siraneci R, Demir-
microbiota composition and function in antibiotic-treated and in
ci M, et al. The relationship between bifidobacteria and allergic
caesarean-born infants. Microbiome. 2018; 6: 182.
asthma and/or allergic dermatitis: a prospective study of 0–3
years-old children in Turkey. Anaerobe. 2014; 28: 98–103. 63 Kuitunen M, Kukkonen K, Juntunen-Backman K, Korpela R, Pous-
sa T, Tuure T, et al. Probiotics prevent IgE-associated allergy until
50 Enomoto T, Sowa M, Nishimori K, Shimazu S, Yoshida A, Yamada
age 5 years in cesarean-delivered children but not in the total co-
K, et al. Effects of bifidobacterial supplementation to pregnant
hort. J Allergy Clin Immunol. 2009; 123: 335–41.
women and infants in the prevention of allergy development in
infants and on fecal microbiota. Allergol Int. 2014; 63: 575–85. 64 Garcia Rodenas CL, Lepage M, Ngom-Bru C, Fotiou A, Papaga-
roufalis K, Berger B. Effect of formula containing Lactobacillus re-
51 Low JSY, Soh SE, Lee YK, Kwek KYC, Holbrook JD, Van der Beek
uteri DSM 17938 on fecal microbiota of infants born by cesarean-
EM, et al. Ratio of Klebsiella/Bifidobacterium in early life correlates
section. J Pediatr Gastroenterol Nutr. 2016; 63: 681–7.
with later development of paediatric allergy. Benef Microbes.
2017; 8: 681–95. 65 Yasmin F, Tun HM, Konya TB, Guttman DS, Chari RS, Field CJ, et
al. Cesarean section, formula feeding, and infant antibiotic expo-
52 Kalliomaki M, Collado MC, Salminen S, Isolauri E. Early differences
sure: separate and combined impacts on gut microbial changes
in fecal microbiota composition in children may predict over-
in later infancy. Front Pediatr. 2017; 5: 200.
weight. Am J Clin Nutr. 2008; 87: 534–8.
66 Liu Y, Qin S, Song Y, Feng Y, Lv N, Xue Y, et al. The perturbation of
53 Dogra S, Sakwinska O, Soh SE, Ngom-Bru C, Brück WM, Berger B,
infant gut microbiota caused by cesarean delivery is partially re-
et al. Dynamics of infant gut microbiota are influenced by delivery
stored by exclusive breastfeeding. Front Microbiol. 2019; 10: 598.
mode and gestational duration and are associated with subse-
quent adiposity. mBio. 2015; 6: e02419–14. 67 Korpela K, Salonen A, Hickman B, Kunz C, Sprenger N, Kukkonen
K, et al. Fucosylated oligosaccharides in mother’s milk alleviate the
54 Hansen S, Halldorsson TI, Olsen SF, Rytter D, Bech BH, Granström
effects of caesarean birth on infant gut microbiota. Sci Rep. 2018;
C, et al. Birth by cesarean section in relation to adult offspring
8: 13757.
overweight and biomarkers of cardiometabolic risk. Int J Obes.
2018; 42: 15–9.

Impact of Delivery Mode Reprint with permission from: 19


Ann Nutr Metab 2021;77(suppl 3):11–19
DOI: 10.1159/000518498
Focus
Early childhood development of a mature and
diverse adult-like gut microbiota, which is
stable and resilient towards perturbation,
appears to be an important asset of health

Reprinted with permission from: Ann Nutr Metab 2021;77(suppl 3):21–34

Gut Microbiota Development: Influence of Diet from Infancy to


Toddlerhood
Martin Frederik Laursen

Key Insight
Birth mode
Furred
Upon birth, the infant gastrointestinal tract becomes colonized animals
Maternal

Gut microbiota
with a diversity of microbes, which gradually develop into the diet Antibiotics
complex microbial community known as the gut microbiota. Infant feeding
Contact with and diet
The gut microbiota interacts with the gut epithelium, immune siblings
and nervous systems and influences metabolism. Aberrations Geographic
Rural/urban location
in the gut microbiota have been associated with a range of environment
inflammatory, infectious, autoimmune, neurological, and
metabolic conditions. Thus, understanding the processes that
govern the initial colonization and establishment of microbes Infant feeding and diet are key forces that drive the development of the
in our gastrointestinal tract is of great importance. gut microbiota from infancy and throughout childhood.

species in the infant gut. The metabolites of Bifidobacterium


Current knowledge species lower intestinal pH, creating an environment hostile to
pathogenic bacteria. In addition, they exert anti-inflammatory
A key factor influencing gut microbiota development in early life effects, protect the gut epithelium, and regulate the immune
is diet. The succession of microbes from infancy to early tod- system. These properties may result in reduced risk of infec-
dlerhood is driven by the type of milk-feeding and complemen- tious and immune-related diseases for breastfed infants har-
tary feeding. Breastfeeding maintains the gut microbiota in a boring these species. The addition of human milk oligosac-
state of low diversity, enabling Bifidobacterium species to pre- charides to infant formulas may constitute an avenue for im-
dominate and favoring saccharolytic fermentation. Formula-fed proving the gut microbiota in infants that cannot be breastfed.
infants have a more diverse gut microbiota, characterized by Lack of diet-promoted maturation of the gut microbiota dur-
higher prevalence of potentially pathogenic taxa, such as Clos- ing complementary feeding is associated with poor growth
tridium and Enterobacteriaceae, and a higher degree of proteo- and neuro-, bone, and immune development. Further studies
lytic fermentation. The gradual progression towards comple- disentangling the complex interactions between infant diet,
mentary feeding results in further diversification and maturation gut microbiota and health are warranted in order to approach
of the gut microbial community, as well as expansion of bacte- clinical implication.
rial products from saccharolytic and proteolytic metabolism.

Recommended reading
Practical implications
Laursen MF, Bahl MI, Licht TR. Settlers of our inner surface:
Due to the abundance of human milk oligosaccharides, breast- factors shaping the gut microbiota from birth to toddlerhood.
feeding promotes the growth of beneficial Bifidobacterium FEMS Microbiol Rev. 2021;1:1–14.

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/


S. Karger AG, Basel
Review Article

Reprint with permission from:


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912

Gut Microbiota Development:


Influence of Diet from Infancy to
Toddlerhood
Martin Frederik Laursen  

National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark

Key Messages Abstract


Early life is a critical period as our gut microbiota establishes
• The gut microbiota develops drastically during the first year of here and may impact both current and future health. Thus, it
life and is influenced by a range of external factors, with diet
is of importance to understand how different factors govern
being a major player.
• Breastfeeding versus formula feeding strongly affects gut mi- the complex microbial colonization patterns in this period.
crobiota composition and metabolism during early infancy. The gut microbiota changes substantially during infancy and
Breastfeeding promotes the dominance of specific human milk toddlerhood in terms of both taxonomic composition and
oligosaccharide-degrading Bifidobacterium species in the gut, diversity. This developmental trajectory differs by a variety of
which may contribute to protection against infectious and im-
factors, including term of birth, mode of birth, intake of anti-
mune-related diseases. Formula feeding results in a more di-
verse gut microbiota with a higher prevalence of opportunistic biotics, presence of furred pets, siblings and family members,
pathogenic bacterial taxa and proteolytic metabolism in the gut. host genetics, local environment, geographical location, and
• Complementary feeding and the increasing dietary fiber and maternal and infant/toddler diet. The type of milk feeding and
protein intake induces a shift from the milk-adapted gut micro- complementary feeding is particularly important in early and
biota and metabolism toward one with vastly increased diver-
late infancy/toddlerhood, respectively. Breastfeeding, due to
sity and proportions of the fiber-degrading bacterial families
Lachnospiraceae, Ruminococcaceae, and Bacteroidaceae, and the supply of human milk oligosaccharide into the gut, pro-
their major metabolic end products short chain fatty acids. In- motes the growth of specific human milk oligosaccharide
adequate maturation of the gut microbiota during comple- (HMO)-utilizing Bifidobacterium species that dominate the
mentary feeding is associated with poor growth and develop- ecosystem as long as the infant is primarily breastfed. These
ment in early life.
species perform saccharolytic fermentation in the gut and
produce metabolites with physiological effects that may con-
tribute to protection against infectious and immune-related
Keywords diseases. Formula feeding, due to its lack of HMOs and high-
Gut microbiota · Diet · Breastfeeding · Complementary er protein content, give rise to a more diverse gut microbiota
feeding that contains more opportunistic pathogens and results in a

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/ Correspondence to:


S. Karger AG, Basel Martin Frederik Laursen, mfrla @ food.dtu.dk
more proteolytic metabolism in the gut. Complementary complementary feeding, characterized by increased con-
feeding, due to the introduction of dietary fibers and new sumption of dietary fibers and proteins, results in a further
protein sources, induces a shift in the gut microbiota and me- diversification and maturation of the microbial community in
tabolism away from the milk-adapted and toward a more ma- the gut with increased abundance of the bacterial families
ture and diverse adult-like community with increased abun- Lachnospiraceae, Ruminococcaceae, and Bacteroidaceae, as
dances of short chain fatty acid-producing bacterial taxa. well as expansion of bacterial products from saccharolytic
While the physiological implication of these complementary and proteolytic metabolism.
diet-induced changes remains to be established, a few recent
studies indicate that an inadequately matured gut microbiota
may be causally related to poor growth and development. Practical Implications
Further studies are required to expand our knowledge on in-
teractions between diet, gut microbiota, and health in the The breastfeeding promoted Bifidobacterium species pro-
early life setting. © 2021 Nestlé Nutrition Institute, Switzerland/ duce metabolites that lower intestinal pH and thus creates an
S. Karger AG, Basel environment hostile to pathogenic bacteria, they exert anti-
inflammatory properties, protect the gut epithelium and reg-
ulate the immune system. These properties may result in re-
Key Insight duced the risk of infectious and immune-related diseases for
breastfed infants harboring these species. Formula fed infant
The healthy fetus is devoid of microbial organisms, but upon are more frequently colonized with opportunistic pathogenic
birth our gastrointestinal (GI) tract becomes colonized with and toxin-carrying bacterial species and exhibit a mostly pro-
a multitude of microbes, gradually developing into a com- teolytic metabolism in the gut, which may influence the risk
plex microbial community during the first year of life. This of infectious, but also metabolic and immune-related diseas-
community, termed the gut microbiota, interacts with our es. Lack of diet-promoted maturation of the gut microbiota
gut epithelium, immune, and nervous systems and influenc- during complementary feeding is associated with poor growth
es our metabolism. During the last decades, the gut micro- and neuro, bone and immune development, although the ev-
biota has been linked with a range of gut inflammatory, in- idence base for this is extremely limited at present. Further
fectious, allergic, autoimmune, neurological, and metabolic studies disentangling the complex interactions between in-
diseases. Thus, understanding the processes that govern ini- fant diet, gut microbiota and health are warranted in order to
tial colonization and establishment of microbes in our GI approach clinical implication.
tract is of great importance. A key factor influencing the gut
microbiota development in early life is diet. This article re-
view our current knowledge of how the type of milk feeding Recommended Reading
and complementary feeding affects the microbial ecosys-
tem in the gut and how this may impact current and future Laursen MF, Bahl MI, Licht TR. Settlers of our inner surface:
health. factors shaping the gut microbiota from birth to toddlerhood.
FEMS Microbiol Rev. 2021;1:1–14 [1].

Current Knowledge
Gut Microbiota Development in Early Life
Type of milk-feeding and complementary feeding dictates the
microbial succession throughout infancy and into early tod- Despite recent controversies, the general consensus is that
dlerhood. Breastfeeding keeps the gut microbiota in a state of the healthy fetus is devoid of microbial organisms [2]. During
low diversity dominated by human milk oligosaccharide-uti- and following birth the neonatal external body surfaces and
lizing Bifidobacterium species that produce various metabo- gastrointestinal (GI) tract rapidly becomes colonized with a
lites characteristic of saccharolytic fermentation. Formula multitude of microorganisms, reaching >108 microorganisms
feeding allows for a somewhat more diverse gut microbiota per gram feces in a matter of hours to days [3] and over weeks
to colonize and is characterized by higher prevalence of po- to months increase up to 1011–1012 microorganisms per gram
tential pathogenic taxa such as Clostridium and Enterobacte- feces [4, 5], comparable to the microbial densities found in the
riaceae and a higher degree of proteolytic fermentation. The adult gut [6, 7]. The infant gut microbiota undergoes drastic
gradual cessation of milk-based feeding and progression in changes during the first years of life in terms of taxonomic

22 Reprint with permission from: Laursen


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
Within individuals
(alpha diversity)
Bacteroidaceae
Microbiota diversity

Relative abundance
Lachnospiraceae

Ruminococcaceae

Between individuals
(beta diversity) Bifidobacteriaceae
Lactobacillales Enterobacteriaceae
1 2 3 4+ 1 2 3 4+
a Age, years b Age, years

Fig. 1. Development of the gut microbiota in early life. a Develop- average gut microbiota composition with age, showing the relative
ment of gut microbiota diversity with age [97]. b Development of the abundance of the major microbial families/orders [11, 13].

composition and diversity (Fig. 1). As a function of age, the in- siblings and family members, geographical location/popula-
fant becomes exposed to and colonized by a range of new tion, growing up environment (i.e., urban vs. rural), maternal
microbes, increasing diversity within the individual (termed al- diet (through breast milk), and infant diet (Fig. 2). While all of
pha diversity). However, differences in gut microbiota com- these individually have shown impact, infant diet has been
position between individuals (termed beta diversity), is initial- identified as a major contributor to gut microbiota develop-
ly high, resulting from individual exposures to various different ment in early life [11, 12]. In this regard, early infancy diet, such
environmental sources of microbes, but these differences as formula versus breast milk has been relatively well studied,
gradually decrease as selective forces, such as diet, converge whereas much less is known about the effects complemen-
microbiota compositions (Fig. 1a). Thus, the individual infant tary feeding on infant gut microbiota composition [13]. Given
continuously acquires new microbial taxa with age, but when the dominating influence of breastfeeding on gut microbial
comparing across infants the gut microbiota gradually be- composition as well as its protective effects against infectious
comes taxonomically more similar. Aerotolerant and faculta- and some metabolic diseases [14], and the involvement of gut
tive anaerobes, such as lactic acid bacteria (Lactobacillales bacteria in regulation of host immune system and metabolism
genera, such as Streptococcus, Enterococcus and Lactoba- [15], it is of great importance to obtain a deep understanding
cillus) and Enterobacteriaceae are among the major initial of the potential microbe-mediated host effects of feeding
colonizers of the neonatal gut, but their proportions rapidly mode in early infancy. Also, the complementary feeding pe-
decrease within days to weeks and the gut microbiota in riod (6–24 months of life) coincides with a critical period in
breastfed (BF) infants becomes dominated by anaerobic gut microbiota development, transitioning away from the in-
breast milk-promoted Bifidobacteriaceae. As the gut micro- fluence of milk-based diet. As early childhood development
biota develops during infancy and early childhood, abun- of a mature and diverse adult-like gut microbiota, which is
dance of other anaerobic bacterial families, such as Bacteroi- stable and resilient toward perturbation, appears to be an im-
daceae, Lachnospiraceae, and Ruminococcaceae increase portant asset of health [16], assessing how dietary changes in
(Fig. 1b). In early life, the gut microbiota is unstable and less early life affect gut microbiota trajectory is of great value.
resilient to perturbation than the adult gut microbiota [8], but
the ecosystem develops in alpha diversity until around 3–5
years of age, where a stable adult-like microbiota has estab- Early Infancy Feeding and Gut Microbiota
lished [9]. However, before this state is reached, the gut mi-
crobiota is more susceptible to modulation by external fac- Breast milk is the recommended first nutrition for the infant,
tors. Factors influencing the infant gut microbiota develop- providing all necessary nutrients to support growth and de-
ment include (as reviewed in [10]); term of birth, mode of birth, velopment, as well as passive immunity to protect against in-
intake of antibiotics, presence of furred pets, host genetics, fectious diseases during infancy [17]. After lactose and lipids,

Diet and Infant Gut Microbiota Reprint with permission from: 23


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
Mode of feeding
(early infancy diet)

Antibiotics Maternal diet

Term of birth
Mode of birth Furret pets

Host genetics

Geographical location/population Gut microbiota

Siblings and family members

Urban vs. rural

Complementary diet
(late infancy/toddler diet)

Fig. 2. Factors influencing the gut microbiota in early life. Gut micro- environment, such as urban versus rural and geographical location/
biota vary by term of birth, mode of birth, oral antibiotics, mode of population [10]. Mode of milk feeding and complementary diet has
feeding, maternal diet, presence of furred pets, host genetics, pres- been identified as the strongest determinants of infant gut micro-
ence of siblings and family members, complementary diet, and local biota composition [11, 12, 89] as indicated by the arrow sizes.

human milk oligosaccharides (HMOs) represent the third Throughout the GI tract, they can exert direct biological func-
most abundant component of breast milk, reaching 20–25 tions, such as antiadhesion of pathogens and modulation of
g/L in colostrum and ranging between 5 and 15 g/L in mature epithelial cell responses [18], but in the colon they also serve
milk [18]. HMOs are short saccharides composed of 5 mono- as metabolic substrates for gut bifidobacteria. Some of these
meric building blocks (Glucose, Galactose, N-acetylglycos- bifidobacteria harbor an arsenal of membrane transporters
amine, Fucose, and Sialic acid). From these 5 building blocks, and saccharolytic enzymes which cleave and internalize
>200 different HMOs structures have been identified [19]. HMOs [20] to implement the monomers into the central cat-
Whereas lactose, protein, and lipid serve as important macro- abolic pathways, leading to the main end products lactate,
nutrients for the BF infant, HMOs are virtually ingestible by the acetate, formate, and 1,2-propandiol [21–24] (Fig.  3a). Al-
infant’s own GI saccharolytic enzymes. Therefore, HMOs pass though other bacteria (e.g., Bacteroides, Lactobacillus, and
through the upper GI tract and reach the colon largely intact. recently the Roseburia/Eubacterium group) exhibit some de-

24 Reprint with permission from: Laursen


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
Breastfed Formulafed

HMOs Breast milk Formula milk Proteins Peptides Amino acids


5–15 g/L 100–150 g/L
>200 different structures
Some proteins (and peptides) escape digestion and
HMOs are not digested in upper GI tract and reach the absorption in upper GI tract and reach colon were they are
colon were they are degraded into monosaccharides degraded into amino acids and fermented by gut microbes
and fermented by gut microbes

C. perfringens C. difficile

B. breve
B. bifidum
B. longum subsp. infantis
E. coli
HMO degrading Bifidobacterium species Protein degrading species
(e.g., B. bifidum and B. breve or B. longum subsp. infantis) (e.g., Clostridium, Enterobacteriaceae)

HMO catabolites
Acetate BCAA catabolites
Lactate lsovalerate
Formate Isobutyrate
1,2-Propanediol AAA catabolites
AAA-derived Co-HMO metabolism products p-cresol
ILA Phenylacatate
PLA Tryptamine
4-OH-PLA
Potential benefical health effects
Fewer opportunistic pathogens Potential detrimental health effects
Fewer antibiotic resistance genes More opportunistic pathogens
Lower gut inflammation More antibiotics resistance genes
Protect gut epithelium Higher gut inflammation
Immune system regulation Increased risk of metabolic and
Decreased risk of immune-related immune-related diseases?
a diseases? b

Fig. 3. Mode of feeding influences early infancy microbiota and sac- ed protein, peptides, and individual amino acids are metabolized by
charolytic versus proteolytic fermentation in the gut with potential gut microbes, including opportunistic pathogenic bacteria, such as
implications for health. a Breast milk contains HMOs, which upon Clostridium and Enterobacteriaceae species, yielding various amino
ingestion by the infant pass undigested through the upper GI tract acid catabolites, some of which have potential detrimental health
until they reach the colon. Here, they are digested by specific HMO- effects. HMO, human milk oligosaccharide; AAA, aromatic amino
degrading Bifidobacterium species into various metabolites with po- acid; BCAA, branched chain amino acid; GI tract, gastrointestinal
tential beneficial health effects. b Formula milk contain excess pro- tract; ILA, indolelactate; PLA, phenyllactate; 4-OH-PLA, 4-hy-
teins, some of which, upon ingestion, is incompletely digested and droxypheyllactate.
absorbed in the upper GI tract and reaches the colon. Partly digest-

gree of HMO utilization [25–27], the extensive arsenal of en- lescentis, B. catenulatum, and B. angulatum) cannot utilize
zymes and transporters is restricted to specific species within HMOs and therefore are less abundant and/or infrequent
the Bifidobacterium genus. Whereas some species, such as B. members of the gut microbiota of BF infants [20]. Thus, due
bifidum, employ extracellular saccharolytic enzymes to de- to the HMO content, breast milk represents a strong selective
grade HMOs and internalize mono- and disaccharides, other factor for shaping the early infancy microbiota, dominated by
species such and B. breve and especially B. longum subsp. specific Bifidobacterium species (Fig. 3a). Through HMO ca-
infantis transport the intact HMO structures and degrades tabolism, these Bifidobacterium species produce metabolites
them inside the cell [20]. The ability of these species to effi- with potential host-health effects. Acetate and lactate are 2
ciently utilize HMOs makes them dominant members of the main end products of bifidobacterial HMO metabolism and
gut of BF infants [11, 28, 29]. Importantly, other Bifidobacte- are responsible for the low pH found in feces from BF infants
rium species commonly found in the adult gut (e.g., B. ado- [24, 30, 31], which is likely to suppress the growth of oppor-

Diet and Infant Gut Microbiota Reprint with permission from: 25


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
tunistic pathogenic species within Clostridiaceae, Enterobac- in the FF gut [52, 53]. Whereas remnants of HMO metabolism
teriaceae, and Staphylococcaceae [30, 32]. Further, Bifido- (e.g., fucose), their direct catabolic end products (e.g., lactate
bacterium-produced acetate was found to inhibit entero- and 1,2 propanediol) and aromatic amino acid derived co-
pathogenic E. coli infection in a mouse model, by maintaining HMO metabolism products [29, 35] (e.g., indolelactate and
the epithelial barrier function upon insult [33]. HMO-utilizing 4-hydroxypheyllactate) dominate in BF infants, various differ-
Bifidobacterium species are also main producers of a limited ent protein fermentation products such as isovalerate, isobu-
set of the aromatic amino acid catabolites, namely indolelac- tyrate, phenylacetate, p-cresol and tryptamine dominate in
tate (ILA), phenyllactate, and 4-hydroxypheyllactate (4-OH- FF infants [52, 53] (Fig.  3). Some of these metabolites (e.g.,
PLA), in the gut of BF infants [29]. These metabolites are pro- p-cresol and phenylacetate) are converted in the liver to me-
duced when the bacteria grow on HMOs (and have access to tabolites with detrimental effects, such as p-cresol-sulfate,
the aromatic amino acids tryptophan, tyrosine, and phenyl- which is a uremic toxin [54] and phenylacetateglutamine
alanine, also contained in breast milk), and interact with vari- which may contribute to development cardiovascular diseas-
ous receptors expressed in immune cells, exhibit immuno- es [55]. Phenylacetate has also been shown to promote ac-
modulatory potential and protect the gut epithelium [29, 34– cumulation of fat in the liver [56]. Further, bacterially pro-
36]. This may contribute to the anti-inflammatory status duced tryptamine in FF infant gut may shift tryptophan me-
observed in the GI tract of BF infants with Bifidobacterium- tabolism away from serotonin and influence immune system
rich communities [37]. However, despite being BF, some indi- development [57]. Biogenic amines produced by bacterial
viduals (e.g., infants born preterm, by C-section or in popula- amino acid metabolism (such as tryptamine and histamine)
tion with high consumption of antibiotics which disrupts ver- can cause GI symptoms and allergic reactions [58, 59]. In ad-
tical transmission from the maternal gut) lack Bifidobacterium dition C. perfringens and C. difficile that often colonize the
species, such as B. longum, B. breve, and B. bifidum [38, 39]. gut of FF infants are potential toxin-carrying opportunistic
Lack of Bifidobacterium is associated with immune dysregu- pathogens [60, 61] and the gut microbiota of FF infants has a
lation [40] and development of asthma [41] and autoimmune higher abundance of antimicrobial resistance genes [62, 63].
diseases [42], but early life intervention with a HMO-degrad- Last, FF infant have higher levels of inflammation markers
ing B. longum subsp. infantis strain in BF infants modulates than BF infants [64].
immune responses away from the allergy and autoimmunity In summary, breastfeeding, due to the presence of HMOs,
associated immune-phenotypes [40]. In both full term [31] promotes growth of beneficial bifidobacteria in the infant gut,
and preterm [43, 44] BF infants lacking bifidobacteria, oral ad- which produces metabolites that may contribute to preven-
ministration of HMO-utilizing Bifidobacterium spp. were tion of GI infections and support immune system develop-
found to decrease the abundance of opportunistic patho- ment. By contrast, formula feeding leads to a gut microbiota
gens, antibiotic resistance genes, and reduce intestinal in- with higher abundances of opportunistic pathogens and a
flammation, demonstrating the importance of this group of mostly proteolytic gut metabolism, leading to potentially det-
bacteria for infant health (Fig. 3a). rimental health effects (Fig. 3). However, it has recently be-
On the contrary, exclusively formula-fed (FF) infants har- come technically and legally possible to add synthetically
bor a more diverse microbiota with lower abundances of produced HMOs (e.g., 2′FL) to infant formula in order to stim-
HMO-utilizing Bifidobacterium species, often with increased ulate a BF-like gut microbiota and metabolism [65]. A recent
abundances of Clostridium species (C. difficile and C. perfrin- double-blinded randomized controlled trial investigated how
gens) and Enterobacteriaceae species (e.g., E. coli) [45–47] addition of 2 HMOs, namely 2′FL and LNnT, to infant formula
(Fig. 3b). The lack of HMOs and higher protein content in for- influence the gut microbiota, and compared HMO-supple-
mula is likely to explain these observations. Although many mented and unsupplemented formula groups with a refer-
infant formula products are supplemented with fructo-oligo- ence group of BF infants [66]. The addition of HMOs to the
saccharides and/or galacto-oligosaccharides, these are not formula resulted in a microbiota configuration that was more
as selective since they can be utilized by most Bifidobacte- similar to that of the BF infants. Infants receiving HMO-sup-
rium species (including the adult-associated B. adolescentis plemented formula, similar to the BF infants, had higher rela-
and B. catenulatum) [48, 49] and additional can stimulate tive abundances of Bifidobacterium and lower relative abun-
growth of various Lactobacillus, Streptococcus, and Bacte- dances of Enterobacteriaceae and Peptostreptococcaceae
roides species [50, 51]. Studies of the fecal metabolome in (the bacterial family that includes C. difficile) compared to in-
exclusively FF (even when formula is containing galacto-oli- fants receiving unsupplemented formula. Although promis-
gosaccharides) versus exclusively BF infants, suggest that ing, this study is the first of its kind in investigating the influ-
proteolytic rather than saccharolytic metabolism dominates ence on the gut microbiota and confirmatory studies are re-

26 Reprint with permission from: Laursen


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
quired. Nonetheless, this strategy may constitute an avenue diet but will also vary with passage of luminal content through
to improve the gut microbiota in infants that for some reason the colon, as dietary fibers will gradually become depleted [76].
cannot be BF. The main catabolic end products of dietary fiber metabolism
are the short chain fatty acids (SCFAs) acetate, butyrate, and
propionate [77]. Whereas the former (together with lactate,
Complementary Feeding and Gut Microbiota formate, and succinate) is produced in high quantities in early
infancy (e.g., by Bifidobacterium, Lactobacillus, and Entero-
At some point during infancy milk-based feeding is no longer bacteriaceae spp.), butyrate and propionate concentrations
adequate to cover the nutritional requirements of the infant. are initially very low but increase with infant age [24]. In addi-
Therefore, supplementation with additional foods, alongside tion, products of protein fermentation, such as branched chain
milk-feeding, is needed. The period of transition from exclu- fatty acids (BCFAs) are almost undetectable during breastfeed-
sive milk-feeding toward eating family foods is termed com- ing, but follow a similar pattern of increase with age [24]. These
plementary feeding and usually covers age 6–24 months [67, changes in SCFAs and BCFAs are coinciding with the introduc-
68]. It is recommended by WHO to start complementary tion of solid foods and cessation of breastfeeding [24]. Con-
feeding around 6 months of age [67]; however, in some coun- sistent with the typical gut microbiota developmental pattern,
tries/populations is it not uncommon to introduce other some key Lachnospiraceae (Anaerostipes, Roseburia, and Eu-
foods as early as 2–3 months of age [69]. Early complemen- bacterium) and Ruminococcaceae (Faecalibacterium, Gem-
tary feeding (before 3 months of age) has been linked to in- minger, and Subdoligranulum) species are butyrate producers
creased risk of GI and respiratory infections, obesity, and al- [24, 78], whereas Bacteroides are common propionate pro-
lergies, but this may rather be attributed to shorter duration of ducers [79]. These species harbor an extensive catalog of en-
breastfeeding [70]. However, late introduction to comple- zymes (glycosyl hydrolases) for degradation of dietary fibers
mentary feeding can also be problematic as it may result in [80] into these SCFAs [79]. In addition, some Bacteroides and
feeding problems, inadequate nutrition and growth [70], and Clostridium species may utilize various amino acids from di-
failure to induce oral tolerance [71]. Recent evidence suggests etary proteins to form BCFAs [81]. Thus, complementary feed-
that the infant gut microbiota development may be causally ing is very likely to causally affect microbiota composition and
linked to healthy growth [72, 73] and protection against food metabolism. As SCFAs have a range of physiological effects,
allergies [74, 75]. including influence on intestinal barrier function, host metab-
Despite extensive progression, during the last decade, in olism, immune system, and nervous system [82], these are
our understanding of gut microbial succession in early life, and plausible mediators of microbiota-host interactions during
our continuously expanding knowledge about microbial ca- complementary feeding affecting host health (Fig.  4). Much
pacity to consume various dietary compounds, we know sur- less is known about the physiological effects of BCFAs [83].
prisingly little about causal effects of diet on gut microbiota Longitudinal studies, designed with multiple samplings
during complementary feeding. As outlined above, the com- around the period of first introduction to solid foods, have
plementary feeding period coincides with a phase of drastic demonstrated increased alpha diversity and abundance of
changes in the gut microbiota (Fig. 1), including a rapid decline Lachnospiraceae genera (such as Blautia) after introduction of
in HMO-degrading Bifidobacterium species. The prominent solid foods [84, 85]. Supporting this, earlier introduction to
increase in alpha diversity and appearance/bloom in Bacteroi- complementary foods is associated with higher microbial al-
daceae, Lachnospiraceae, and Ruminococcaceae species, pha diversity throughout infancy, including higher abundance
mirror the increased complexity of the diet with the introduc- of the butyrate producing Lachnospiraceae genera Roseburia
tion of fibers from various fruits, vegetables, cereals/porridge, [86]. In a randomized controlled trial [87] the authors com-
and breads as well as new protein sources that is, in the form pared the gut microbiota in infants that were weaned tradi-
of meats, dairy products, and legumes/lentils. As dietary fibers tionally (with spoon feeding) to infants weaned with a baby-
and secondly (incompletely digested) proteins/peptides are led approach (no spoon feeding, but only complementary
the main sources of energy for gut microbes [76] these mac- “finger foods” consumed), with complementary diet assessed
ronutrients would be expected to have most impact on the by 3-day dietary records. The baby-led weaned infants were
microbial composition. Carbohydrates (dietary fibers) are pre- introduced to solid foods roughly 3 weeks later (6 months of
ferred energy-sources for microbes, but in shortage of these age) and consumed significantly less fruits and vegetables and
a higher degree of proteolytic fermentation occurs in the gut total dietary fiber at 7 months of age, which was associated
(as illustrated in the example of metabolism in the gut of FF with a reduced alpha diversity and lower abundance of spe-
infants mentioned above). This balance not only depends on cific Lachnospiraceae (Roseburia facies and Eubacterium rec-

Diet and Infant Gut Microbiota Reprint with permission from: 27


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
Meats and dairy
Fruits and
vegetables Breads and cereals

Complementary diet

Fig. 4. Complementary feeding increases Protein


gut microbial diversity and production of Dietary fiber
BCFAs and SCFAs with potential implica-
BCFAs (protein)
tion for growth, neuro, bone, and immune
Isovalerate
development. As complementary feeding Aplha diversity Isobutyrate
progresses, the milk-based component is Lachnospiraceae
gradually replaced by other foods such as Ruminococcaceae
SCFAs (dietary fiber)
Bacteroidaceae Butyrate
meats and dairy, fruits and vegetables,
Propionate
and bread and cereals, which are directly Acetate
and possibly indirectly (through the gut Gut microbiota
microbiota) impacting growth and devel-
opment. These foods contain protein and
dietary fibers that modulate the infant gut ?
microbiota, increasing alpha diversity and
?
the abundance of key bacterial families
that produce SCFAs (note that acetate is
also produced in high amount during ear-
ly infancy) and BCFAs during comple-
Neuro
mentary feeding. This “natural” develop- Growth
development
ment of the gut microbiota and its me-
tabolites is associated with healthy
growth, neuro, bone development, and
Bone
appropriate immune system regulation. Immune system regulation
development
BCFAs, branched chain fatty acids; SCFAs,
short chain fatty acids.

tale) and Ruminococcaceae (Faecalibacterium prausnitzii) ucts were main food groups driving these associations [89],
species during complementary feeding [87]. In addition, inde- suggesting that these complementary foods are contributing
pendent of the feeding groups, consumption of breads and to the diversification of the infant gut microbiota, at least in
cereals, as well as meat products at 7 months of age were this cohort. Importantly, these associations were not solely
positively associated with alpha diversity at 12 months of age mediated by cessation of breastfeeding, as they were found
[87]. in both partially BF and partially FF infants [13]. Progression in
A study including 9 months old Danish infants assessed the complementary feeding also correlated positively to the
complementary feeding diet by 7-days dietary records and a abundance of several Lachnospiraceae and Ruminococcace-
used multivariate statistical analysis to generate the dietary ae spp., but negatively to Bifidobacterium [13], thus marking
pattern “family foods” [88, 89], describing the infant’s progres- the transition of the breast milk promoted Bifidobacterium-
sion in complementary feeding (from milk-based toward rich gut community toward the fiber and protein promoted
family foods). Progression in complementary feeding, char- (more diverse) gut microbial community, characterized by
acterized by higher dietary fiber and protein intake, correlated butyrate, propionate, and BCFA-producing bacteria (Fig. 4).
significantly with gut microbial alpha diversity [89]. Specifi- Indeed, interventions with meat as a main complemen-
cally, consumption of rye bread and cheese and meat prod- tary food compared to dairy [90] or cereals [91] has revealed

28 Reprint with permission from: Laursen


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
significantly increased abundance of butyrate-producing onto an antigen-containing diet [75]. In addition, colonization
Lachnospiraceae genera. A recent 7-week intervention of previously germ-free mice with gut commensal microbes
study comparing a refined grain cereal product to a whole inhibit the increase in food-specific IgE and disruption of the
grain cereal product as first complementary food, demon- gut microbiota by oral administration of antibiotics to con-
strated a significant increase in Bacteroides and Lachno- ventional mice increases food-specific IgE concentrations in
clostridium (Lachnospiraceae family) and a decrease in blood [75]. Thus, gut microbes play a causal role in regulation
Escherichia (Enterobacteriaceae family) over time only in of IgE levels during the food antigen exposure and may affect
the whole grain cereal group. However, other intervention the risk of developing food allergy. Another study performed
with specific complementary foods such as legumes versus gut microbiota transplantation from healthy or cow’s milk al-
corn-soy flour [92] or meat versus cereals [93] show limited lergic 6-month old infants into germ-free mice and chal-
differential impact on the gut microbiota, possibly due to lenged them with the cow’s milk allergen β-lactoglobulin [74].
these infants still receiving a high proportion of breast milk In contrast to the mice colonized with the cow’s milk allergic
in their diet or other study participant characteristics. Ulti- infant gut microbiota, mice colonized with the healthy infant
mately, more randomized controlled trials are needed to gut microbiota were protected against allergic responses. In
establish direct causal proof of links between specific com- the healthy infant gut microbiota, Anaerostipes coccae, a
plementary foods and specific microbial taxa. Nonetheless, Lachnospiraceae species that increases during complemen-
as outlined below, a few recent studies indicate that spe- tary feeding and associates with consumption of vegetables
cific Lachnospiraceae species affected by complementary and fruits [96], was identified as a bacterial taxon that medi-
foods may influence growth and development and protect ated this protection. By comparing mice colonized with cow’s
against food allergies (Fig. 4). milk allergic microbiota to mice mono-colonized with A. cac-
Stunted undernourished Bangladeshi infants/toddlers cae it was demonstrated that this species protected the mice
have an immature gut microbiota compared to same age against allergic food response [74]. While results from these
healthy well-nourished counterparts [94], likely as a result of studies are encouraging, it must be emphasized that our cur-
inadequate and/or delayed complementary feeding. Trans- rent knowledge on the effects of different complementary
plantation of the gut microbiota, using fecal samples from foods on gut microbiota and thereof potential health impact
stunted, malnourished versus healthy infants, into germ-free is still extremely limited.
mice (devoid of microbes) has revealed differential growth
patterns, recapitulating the growth phenotypes observed in
these infants [73]. Oral introduction of some of the Lachno- Future Directions
spiraceae species (such as Ruminococcus gnavus and Clos-
tridium symbiosum) that appear during complementary feed- Undoubtedly, diet plays a major role for the succession of the
ing in healthy infants was shown to promote growth and bone infant gut microbiota, yet we still have a very incomplete un-
development in mice previously transplanted with the mal- derstanding of the details. There is a need for well-designed
nourished gut microbiota [73]. Furthermore, complementary and well-powered interventions in various different popula-
food intervention in stunted malnourished Bangladeshi tod- tions in order to assess the impact of formula supplemented
dlers rationally designed to repair the immature gut microbi- with different HMOs on gut microbiota and metabolome in
ota suggest that a combination of banana, chickpea, soy, and early infancy, including relevant measures of host impact,
peanut matures the gut microbiota (increase abundance of such as prevalence of infectious and immune-related diseas-
some Lachnospiraceae and Ruminococcaceae species) and es, during and after intervention. There is a lack of studies with
influence blood markers of healthy growth, bone, immune, detailed dietary recordings coupled to measurements of the
and neurodevelopment [72, 95]. However, despite having a gut microbiota at a finer resolution scale. In example, to im-
similar energy, macronutrient and fiber content, other ratio- prove our knowledge on dietary factors shaping the infant gut
nally designed microbiota-directed complementary food microbiota, we need prospective cohort studies with detailed
combinations were not effective, illustrating the need for a information about infant diet, obtained from multiple con-
deeper understanding of the effects of specific food combi- secutive days of dietary recordings throughout periods of
nations. complementary feeding, combined with dense fecal sampling
Recent studies have demonstrated that blood concentra- and state of the art microbiome (obtaining species and strain
tions of food specific immunoglobulin E (IgE), an important level resolution) and metabolome profiling, as well as deep
mediator in food allergy, is elevated in germ-free mice com- genomic and phenotypic characterization of fecal microbial
pared to conventional (colonized) counterparts after weaning isolates to decipher molecular mechanisms. These studies

Diet and Infant Gut Microbiota Reprint with permission from: 29


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
should be accompanied by intervention studies, in relevant butyrate and propionate as well as the BCFAs. While the phys-
study populations, designed to introduce specific comple- iological relevance of these bacterial taxa and metabolites
mentary foods using frequent longitudinal sampling and the remains to be established in this early life context, recent re-
state-of-the-art methods of microbiome and metabolome search suggests that they may be important for healthy
profiling. Hopefully, this will lead to identification of key diet- growth and development and protection against food aller-
microbe-metabolite interactions that beneficially impact as- gies.
pects of host physiology, such as metabolism and immunol- Clearly, the question of when to start introducing various
ogy. Ultimately, if evidence is adequate, specific dietary inter- complementary foods in order to achieve optimal health and
vention should be tested in disposed/susceptible study gut microbiota progression is central. Early introduction,
populations (e.g., disposed to allergic or autoimmune diseas- combined with reduced or lack of breastfeeding is presum-
es or individuals at risk of malnourishment and stunting) to ably not beneficial, as it will disrupt the Bifidobacterium dom-
provide proof of applicability. inated gut community, associated with protection against in-
fectious and immune-related diseases. On the other hand,
very late introduction as observed in populations where food
Summary and Conclusions is scare and breastfeeding of the infant is prolonged without
adequate complementary foods would also be expected to
The infant gut microbiota undergoes significant temporal de- be adverse, given the inability of the gut microbiota to mature
velopment during the first years of life. A multitude of factors when deprived of key nutrients and the obvious negative im-
such term of birth, mode of birth, antibiotics, contact with plications for growth and development. Undoubtedly, further
furred animals and siblings/family members, local environ- studies are required to improve our understanding of the in-
ment, geographical location, and maternal and infant diet af- triguing links between complementary feeding, gut microbi-
fect the trajectory of this development. Of these, infant/tod- ota, and health.
dler diet has been identified as one of the most influential
factors. The neonatal gut microbiota is initially characterized
by random colonization of environmental microbes, but in a Conflict of Interest Statement
matter of days to weeks the gut ecosystem and the type milk-
feeding selects for microbes adapted to the GI environment The writing of this article was supported by Nestlé Nutrition
and equipped to consume the indigestible (or incompletely Institute and the author declares no other conflicts of interest.
digested) constituents of breast or formula milk. Breast milk
feeding selects for specific HMO-degrading Bifidobacterium
species that vastly dominate the gut ecosystem and produce Funding Sources
metabolites with beneficial effects likely contributing to the
protection against GI infections and supporting immune sys- The writing of the manuscript was supported by Nestlé Nutri-
tem development. On the contrary, formula-milk feeding is tion Institute.
less selective and the gut microbiota of FF infants is more di-
verse with a higher frequency and proportion of protein de-
grading opportunistic pathogenic species. This may increase References
risk of GI infections and the apparent mostly proteolytic me- 1 Laursen MF, Bahl MI, Licht TR. Settlers of our inner surface: factors
tabolism is possibly detrimental to health as some of the me- shaping the gut microbiota from birth to toddlerhood. FEMS Mi-
tabolites are associated with kidney, liver, and cardiovascular crobiol Rev. 2021; 1: 1–14.
diseases in adults. 2 Blaser MJ, Devkota S, McCoy KD, Relman DA, Yassour M, Young
As the infant transitions into complementary feeding the VB. Lessons learned from the prenatal microbiome controversy.
Microbiome. 2021 Dec; 9(1): 8.
gut microbiota diversifies along with the increasing amounts
of dietary fiber and proteins in the diet. Specifically, members 3 Bittinger K, Zhao C, Li Y, Ford E, Friedman ES, Ni J, et al. Bacterial
colonization reprograms the neonatal gut metabolome. Nat Mi-
of the bacterial families Lachnospiraceae, Ruminococcace-
crobiol. 2020.
ae, and Bacteroidaceae capable of consuming various dietary
4 Pham VT, Lacroix C, Braegger CP, Chassard C. Early colonization
fibers from fruits, vegetables, breads and cereals, and possibly
of functional groups of microbes in the infant gut. Environ Micro-
proteins from meat and dairy sources, increase in abundance biol. 2016 Jul; 18(7): 2246–58.
during complementary feeding. This leads to a shift in gut
metabolism with the appearance or increase of the SCFAs

30 Reprint with permission from: Laursen


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
5 Obermajer T, Grabnar I, Benedik E, Tušar T, Robič Pikel T, Fidler 21 Bunesova V, Lacroix C, Schwab C. Fucosyllactose and L-fucose
Mis N, et al. Microbes in infant gut development: placing abun- utilization of infant Bifidobacterium longum and Bifidobacterium
dance within environmental, clinical and growth parameters. Sci kashiwanohense. BMC Microbiol. 2016 Oct; 16(1): 248–12.
Rep. 2017 Dec; 7(1): 11230–14.
22 Özcan E, Sela DA. Inefficient metabolism of the human milk oli-
6 Jian C, Luukkonen P, Yki-Järvinen H, Salonen A, Korpela K. Quan- gosaccharides lacto-N-tetraose and lacto-N-neotetraose shifts
titative PCR provides a simple and accessible method for quantita- Bifidobacterium longum subsp. infantis physiology. Front Nutr.
tive microbiota profiling. PLoS One. 2020 Jan; 15(1): e0227285. 2018 May; 5: 46.
7 Tsuji H, Matsuda K, Nomoto K. Counting the countless: bacterial 23 Dedon LR, Özcan E, Rani A, Sela DA. Bifidobacterium infantis me-
quantification by targeting rRNA molecules to explore the human tabolizes 2′Fucosyllactose-derived and free fucose through a
gut microbiota in health and disease. Front Microbiol. 2018 Jun; common catabolic pathway resulting in 1,2-propanediol secre-
9: 1417. tion. Front Nutr. 2020 Nov; 7: 583397.
8 Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. 24 Tsukuda N, Yahagi K, Hara T, Watanabe Y, Matsumoto H, Mori H,
Diversity, stability and resilience of the human gut microbiota. Na- et al. Key bacterial taxa and metabolic pathways affecting gut
ture. 2012 Sep; 489(7415): 220–30. short-chain fatty acid profiles in early life. ISME J. 2021 Mar: 1–17.
9 Rodríguez JM, Murphy K, Stanton C, Ross RP, Kober OI, Juge N, 25 Pichler MJ, Yamada C, Shuoker B, Alvarez-Silva C, Gotoh A, Leth
et al. The composition of the gut microbiota throughout life, with ML, et al. Butyrate producing colonic Clostridiales metabolise hu-
an emphasis on early life. Microb Ecol Health Dis. 2015 Jan; 26: man milk oligosaccharides and cross feed on mucin via conserved
26050. pathways. Nat Commun. 2020; 11(1): 3285.
10 Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, et al. 26 Marcobal A, Barboza M, Sonnenburg ED, Pudlo N, Martens EC,
The first microbial colonizers of the human gut: composition, ac- Desai P, et al. Bacteroides in the infant gut consume milk oligosac-
tivities, and health implications of the infant gut microbiota. Mi- charides via mucus-utilization pathways. Cell Host Microbe. 2011
crobiol Mol Biol Rev. 2017 Dec; 81(4): e00036–17. Nov; 10(5): 507–14.
11 Stewart CJ, Ajami NJ, O’Brien JL, Hutchinson DS, Smith DP, Wong 27 Rodríguez-Díaz J, Monedero V, Yebra MJ. Utilization of natural
MC, et al. Temporal development of the gut microbiome in early fucosylated oligosaccharides by three novel α-L-fucosidases
childhood from the TEDDY study. Nature. 2018 Oct; 562(7728): from a probiotic lactobacillus casei strain. Appl Environ Microbiol.
583–8. 2011 Jan; 77(2): 703–5.
12 Galazzo G, van Best N, Bervoets L, Dapaah IO, Savelkoul PH, Hor- 28 Turroni F, Peano C, Pass DA, Foroni E, Severgnini M, Claesson MJ,
nef MW, et al. Development of the microbiota and associations et al. Diversity of bifidobacteria within the infant gut microbiota.
with birth mode, diet, and atopic disorders in a longitudinal analy- PLoS One. 2012 Jan; 7(5): e36957.
sis of stool samples, collected from infancy through early child-
29 Laursen MF, Sakanaka M, von Burg N, Andersen D, Mörbe U, Rivol-
hood. Gastroenterology. 2020 May; 158(6): 1584–96.
lier A, et al. Breastmilk-promoted bifidobacteria produce aromat-
13 Laursen MF, Bahl MI, Michaelsen KF, Licht TR. First foods and gut ic lactic acids in the infant gut. bioRxiv. 2020 Jan.
microbes. Front Microbiol. 2017; 8: 356.
30 Matsuki T, Yahagi K, Mori H, Matsumoto H, Hara T, Tajima S, et al.
14 Victora CG, Bahl R, Barros AJ, França GV, Horton S, Krasevec J, et A key genetic factor for fucosyllactose utilization affects infant gut
al. Breastfeeding in the 21st century: epidemiology, mechanisms, microbiota development. Nat Commun. 2016 Sep; 7(1): 11939.
and lifelong effect. Lancet. 2016 Jan; 387(10017): 475–90.
31 Frese SA, Hutton AA, Contreras LN, Shaw CA, Palumbo MC, Casa-
15 Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold buri G, et al. Persistence of supplemented Bifidobacterium long-
G, et al. The gut microbiota and host health: a new clinical frontier. um subsp. infantis EVC001 in breastfed infants. mSphere. 2017
Gut. 2016 Feb; 65(2): 330–9. Dec; 2(6): e00501–17.
16 Mosca A, Leclerc M, Hugot JP. Gut microbiota diversity and hu- 32 Henrick BM, Hutton AA, Palumbo MC, Casaburi G, Mitchell RD,
man diseases: Should we reintroduce key predators in our eco- Underwood MA, et al. Elevated fecal pH indicates a profound
system? Front Microbiol. 2016 Mar; 7: 455. change in the breastfed infant gut microbiome due to reduction
of Bifidobacterium over the past century. mSphere. 2018 Mar;3(2):
17 WHO. Breastfeeding [Internet]. [cited 2021 Mar 18]. Available from:
e00041–18.
https: //www.who.int/health-topics/breastfeeding#tab=tab_2.
33 Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, et
18 Bode L. Human milk oligosaccharides: every baby needs a sugar
al. Bifidobacteria can protect from enteropathogenic infection
mama. Glycobiology. 2012 Sep; 22(9): 1147–62.
through production of acetate. Nature. 2011; 469(7331): 543–7.
19 Davis JC, Totten SM, Huang JO, Nagshbandi S, Kirmiz N, Garrido
34 Peters A, Krumbholz P, Jäger E, Heintz-Buschart A, Çakir MV,
DA, et al. Identification of oligosaccharides in feces of breast-fed
Rothemund S, et al. Metabolites of lactic acid bacteria present in
infants and their correlation with the gut microbial community.
fermented foods are highly potent agonists of human hydroxycar-
Mol Cell Proteomics. 2016; 15(9): 2987–3002.
boxylic acid receptor 3. PLoS Genet. 2019 May; 15(5): e1008145.
20 Sakanaka M, Gotoh A, Yoshida K, Odamaki T, Koguchi H, Xiao JZ,
35 Ehrlich AM, Pacheco AR, Henrick BM, Taft D, Xu G, Huda MN, et
et al. Varied pathways of infant gut-associated Bifidobacterium to
al. Indole-3-lactic acid associated with Bifidobacterium-dominat-
assimilate human milk oligosaccharides: prevalence of the gene
ed microbiota significantly decreases inflammation in intestinal
set and its correlation with bifidobacteria-rich microbiota forma-
epithelial cells. BMC Microbiol. 2020 Dec; 20(1): 357.
tion. Nutrients. 2020 Dec; 12(1): 71.

Diet and Infant Gut Microbiota Reprint with permission from: 31


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
36 Meng D, Sommella E, Salviati E, Campiglia P, Ganguli K, Djebali K, 50 Schwab C, Gänzle M. Lactic acid bacteria fermentation of human
et al. Indole-3-lactic acid, a metabolite of tryptophan, secreted by milk oligosaccharide components, human milk oligosaccharides
Bifidobacterium longum subspecies infantis is anti-inflammatory and galactooligosaccharides. FEMS Microbiol Lett. 2011 Feb;
in the immature intestine. Pediatr Res. 2020 Aug; 88(2): 209–17. 315(2): 141–8.
37 Henrick BM, Chew S, Casaburi G, Brown HK, Frese SA, Zhou Y, et 51 Ma C, Wasti S, Huang S, Zhang Z, Mishra R, Jiang S, et al. The gut
al. Colonization by B. infantis EVC001 modulates enteric inflam- microbiome stability is altered by probiotic ingestion and im-
mation in exclusively BF infants. Pediatr Res. 2019 Dec;86(6):749– proved by the continuous supplementation of galactooligosac-
57. charide. Gut Microbes. 2020 Nov; 12(1): 1–13.
38 Casaburi G, Duar RM, Brown H, Mitchell RD, Kazi S, Chew S, et al. 52 Chow J, Panasevich MR, Alexander D, Vester Boler BM, Rossoni
Metagenomic insights of the infant microbiome community Serao MC, Faber TA, et al. Fecal metabolomics of healthy breast-
structure and function across multiple sites in the United States. fed versus formula-fed infants before and during in vitro batch
Sci Rep. 2021 Dec; 11(1): 1472. culture fermentation. J Proteome Res. 2014 May; 13(5): 2534–42.
39 Forsgren M, Isolauri E, Salminen S, Rautava S. Late preterm birth 53 He X, Parenti M, Grip T, Lönnerdal B, Timby N, Domellöf M, et al.
has direct and indirect effects on infant gut microbiota develop- Fecal microbiome and metabolome of infants fed bovine MFGM
ment during the first six months of life. Acta Paediatr. 2017 Jul; supplemented formula or standard formula with breast-fed in-
106(7): 1103–9. fants as reference: a randomized controlled trial. Sci Rep. 2019
Dec; 9(1): 1–14.
40 Henrick BM, Rodriguez L, Lakshmikanth T, Pou C, Henckel E, Olin
A, et al. Bifidobacteria-mediated immune system imprinting early 54 Gryp T, Vanholder R, Vaneechoutte M, Glorieux G. p-Cresyl Sul-
in life. bioRxiv. 2020 Oct. fate. Toxins. 2017 Jan; 9(2): 52.
41 Stokholm J, Blaser MJ, Thorsen J, Rasmussen MA, Waage J, Vind- 55 Nemet I, Saha PP, Gupta N, Zhu W, Romano KA, Skye SM, et al. A
ing RK, et al. Maturation of the gut microbiome and risk of asthma cardiovascular disease-linked gut microbial metabolite acts via
in childhood. Nat Commun. 2018 Dec; 9(1): 141. adrenergic receptors. Cell. 2020 Mar; 180(5): 862–e22.
42 Vatanen T, Kostic AD, d’Hennezel E, Siljander H, Franzosa EA, Yas- 56 Delzenne NM, Bindels LB. Microbiome metabolomics reveals new
sour M, et al. Variation in microbiome LPS immunogenicity con- drivers of human liver steatosis. Nat Med. 2018 Jul; 24(7): 906–7.
tributes to autoimmunity in humans. Cell. 2016 May; 165(4): 842–
57 Saraf MK, Piccolo BD, Bowlin AK, Mercer KE, LeRoith T, Chintapal-
53.
li SV, et al. Formula diet driven microbiota shifts tryptophan me-
43 Alcon-Giner C, Dalby MJ, Caim S, Ketskemety J, Shaw A, Sim K, et tabolism from serotonin to tryptamine in neonatal porcine colon.
al. Microbiota supplementation with bifidobacterium and lactoba- Microbiome. 2017 Jul; 5(1): 77.
cillus modifies the preterm infant gut microbiota and metabo-
58 Fernández-Reina A, Urdiales J, Sánchez-Jiménez F. What we
lome: an Observational Study. Cell Rep Med. 2020 Aug; 1(5):
know and what we need to know about aromatic and cationic
100077.
biogenic amines in the gastrointestinal tract. Foods. 2018 Sep;7(9):
44 Nguyen M, Holdbrooks H, Mishra P, Abrantes MA, Eskew S, Garma 145.
M, et al. Impact of probiotic B. infantis EVC001 feeding in prema-
59 Daniel Collins J, Noerrung B, Budka H, Andreoletti O, Buncic S,
ture infants on the gut microbiome, nosocomially acquired anti-
Griffin J, et al. Scientific Opinion on risk based control of biogen-
biotic resistance, and enteric inflammation. Front Pediatr. 2021
ic amine formation in fermented foods. EFSA J. 2011 Oct; 9(10):
Feb; 9: 618009.
2393.
45 Penders J, Thijs C, van den Brandt PA, Kummeling I, Snijders B,
60 Shaw AG, Cornwell E, Sim K, Thrower H, Scott H, Brown JCS, et
Stelma F, et al. Gut microbiota composition and development of
al. Dynamics of toxigenic Clostridium perfringens colonisation in
atopic manifestations in infancy: the KOALA Birth Cohort Study.
a cohort of prematurely born neonatal infants. BMC Pediatr. 2020
Gut. 2007 May; 56(5): 661–7.
Feb; 20(1): 75.
46 Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary
61 Kuiper GA, van Prehn J, Ang W, Kneepkens F, van der Schoor S, de
P, et al. Dynamics and stabilization of the human gut microbiome
Meij T. Clostridium difficile infections in young infants: case pre-
during the first year of life. Cell Host Microbe. 2015 May; 17(5):
sentations and literature review. IDCases. 2017 Jan; 10: 7–11.
690–703.
62 Rahman SF, Olm MR, Morowitz MJ, Banfield JF. Machine learning
47 Benno Y, Sawada K, Mitsuoka T. The intestinal microflora of in-
leveraging genomes from metagenomes identifies influential an-
fants: composition of fecal flora in breast-fed and bottle-fed in-
tibiotic resistance genes in the infant gut microbiome. mSystems.
fants. Microbiol Immunol. 1984 Sep; 28(9): 975–86.
2018 Jan; 3(1): e00123–17.
48 Rossi M, Corradini C, Amaretti A, Nicolini M, Pompei A, Zanoni S,
63 Pärnänen K, Hultman J, Satokari R, Rautava S, Lamendella R,
et al. Fermentation of fructooligosaccharides and inulin by bifido-
Wright J, et al. Formula alters preterm infant gut microbiota and
bacteria: a comparative study of pure and fecal cultures. Appl En-
increases its antibiotic resistance load. bioRxiv. 2019 Sep: 782441.
viron Microbiol. 2005 Oct; 71(10): 6150–8.
64 Ossa JC, Yáñez D, Valenzuela R, Gallardo P, Lucero Y, Farfán MJ.
49 Akiyama T, Kimura K, Hatano H. Diverse galactooligosaccharides
Intestinal inflammation in Chilean infants fed with bovine formula
consumption by bifidobacteria: Implications of β-galactosidase-
vs. breast milk and its association with their gut microbiota. Front
LacS operon. Biosci Biotechnol Biochem. 2015 Apr;79(4):664–72.
Cell Infect Microbiol. 2018 Jun; 8: 190.

32 Reprint with permission from: Laursen


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
65 Salminen S, Stahl B, Vinderola G, Szajewska H. Infant formula sup- 82 Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From di-
plemented with biotics: current knowledge and future perspec- etary fiber to host physiology: short-chain fatty acids as key bac-
tives. Nutrients. 2020 Jul; 12(7): 1–20. terial metabolites. Cell. 2016 Jun; 165(6): 1332–45.
66 Berger B, Porta N, Foata F, Grathwohl D, Delley M, Moine D, et al. 83 Rios-Covian D, González S, Nogacka AM, Arboleya S, Salazar N,
Linking human milk oligosaccharides, infant fecal community Gueimonde M, et al. An overview on fecal branched short-chain
types, and later risk to require antibiotics. MBio. 2020 Apr;11(2):17. fatty acids along human life and as related with body mass index:
associated dietary and anthropometric factors. Front Microbiol.
67 WHO. Complementary feeding: global [Internet]. 2020 [cited 2021
2020 May; 11: 973.
Mar 16]. Available from: https://www.who.int/health-topics/com-
plementary-feeding#tab=tab_2. 84 Roger LC, McCartney AL. Longitudinal investigation of the faecal
microbiota of healthy full-term infants using fluorescence in situ
68 Castenmiller J, De Henauw S, Hirsch-Ernst K-I, Kearney J, Maciuk
hybridization and denaturing gradient gel electrophoresis. Micro-
A, Mangelsdorf I, et al. Scientific Opinion appropriate age range
biology. 2010; 156(Pt 11): 3317–28.
for introduction of complementary feeding into an infant’s diet
EFSA panel on nutrition, novel foods and food allergens (NDA). 85 Thompson AL, Monteagudo-Mera A, Cadenas MB, Lampl ML,
2019. Azcarate-Peril MA. Milk- and solid-feeding practices and daycare
attendance are associated with differences in bacterial diversity,
69 Schiess S, Grote V, Scaglioni S, Luque V, Martin F, Stolarczyk A, et
predominant communities, and metabolic and immune function
al. Introduction of complementary feeding in 5 european coun-
of the infant gut microbiome. Front Cell Infect Microbiol. 2015; 5:
tries. J Pediatr Gastroenterol Nutr. 2010 Jan; 50(1): 92–8.
3.
70 Przyrembel H. Timing of introduction of complementary food:
86 Differding MK, Benjamin-Neelon SE, Hoyo C, Østbye T, Mueller
Short- and long-term health consequences. Ann Nutr Metab.
NT. Timing of complementary feeding is associated with gut mi-
2012; 60(Suppl 2): 8–20.
crobiota diversity and composition and short chain fatty acid con-
71 Krawiec M, Fisher HR, Du Toit G, Bahnson HT, Lack G. Overview centrations over the first year of life. BMC Microbiol. 2020; 20(1):
of oral tolerance induction for prevention of food allergy – Where 56–13.
are we now? Allergy. 2021 Mar.
87 Leong C, Haszard JJ, Lawley B, Otal A, Taylor RW, Szymlek-Gay
72 Gehrig JL, Venkatesh S, Chang HW, Hibberd MC, Kung VL, Cheng EA, et al. Mediation analysis as a means of identifying dietary com-
J, et al. Effects of microbiota-directed foods in gnotobiotic ani- ponents that differentially affect the fecal microbiota of infants
mals and undernourished children. Science. 2019 Jul(6449): 365. weaned by modified baby-led and traditional approaches. Appl
Environ Microbiol. 2018 Sep; 84(18): e00914–18.
73 Blanton LV, Charbonneau MR, Salih T, Barratt MJ, Venkatesh S,
Ilkaveya O, et al. Gut bacteria that prevent growth impairments 88 Andersen LB, Pipper CB, Trolle E, Bro R, Larnkjær A, Carlsen EM,
transmitted by microbiota from malnourished children. Science. et al. Maternal obesity and offspring dietary patterns at 9 months
2016; 351(6275): aad3311. of age. Eur J Clin Nutr. 2015 Jun; 69(6): 668–75.

74 Feehley T, Plunkett CH, Bao R, Choi Hong SM, Culleen E, Belda- 89 Laursen MF, Andersen LB, Michaelsen KF, Mølgaard C, Trolle E,
Ferre P, et al. Healthy infants harbor intestinal bacteria that protect Bahl MI, et al. Infant gut microbiota development is driven by tran-
against food allergy. Nat Med. 2019 Mar; 25(3): 448–53. sition to family foods independent of maternal obesity. mSphere.
2016 Feb; 1(1): e00069–15.
75 Hong SW, O E, Lee JY, Lee M, Han D, Ko HJ, et al. Food antigens
drive spontaneous IgE elevation in the absence of commensal 90 Tang M, Frank D, Hendricks A, Ir D, Krebs N. Protein intake during
microbiota. Sci Adv. 2019 May; 5(5): eaaw1507. early complementary feeding affects the gut microbiota in U.S.
formula-fed infants (FS04-03-19). Curr Dev Nutr. 2019;3(Suppl 1):
76 Cummings JH, Macfarlane GT. The control and consequences of 930.
bacterial fermentation in the human colon. J Appl Bacteriol. 1991
Jun; 70(6): 443–59. 91 Krebs NF, Sherlock LG, Westcott J, Culbertson D, Hambidge KM,
Feazel LM, et al. Effects of different complementary feeding regi-
77 Scott KP, Duncan SH, Flint HJ. Dietary fibre and the gut microbi- mens on iron status and enteric microbiota in BF infants. J Pediatr.
ota. Nutr Bull. 2008; 33(3): 201–11. 2013; 163(2): 416–23.
78 Appert O, Garcia AR, Frei R, Roduit C, Constancias F, Neuzil-Bune- 92 Ordiz MI, Janssen S, Humphrey G, Ackermann G, Stephenson K,
sova V, et al. Initial butyrate producers during infant gut microbi- Agapova S, et al. The effect of legume supplementation on the gut
ota development are endospore formers. Environ Microbiol. 2020 microbiota in rural Malawian infants aged 6 to 12 months. Am J
Sep; 22(9): 3909–21. Clin Nutr. 2020; 111(4): 884–92.
79 Louis P, Flint HJ. Formation of propionate and butyrate by the hu- 93 Qasem W, Azad MB, Hossain Z, Azad E, Jorgensen S, Castillo San
man colonic microbiota. Environ Microbiol. 2017 Jan;19(1):29–41. Juan S, et al. Assessment of complementary feeding of Canadian
80 Flint HJ, Scott KP, Duncan SH, Louis P, Forano E. Microbial degra- infants: effects on microbiome & oxidative stress, a randomized
dation of complex carbohydrates in the gut. Gut Microbes. 2012 controlled trial. BMC Pediatr. 2017; 17(1): 54.
Jul-Aug; 3(4): 289–306. 94 Subramanian S, Huq S, Yatsunenko T, Haque R, Mahfuz M, Alam
81 Smith EA, Macfarlane GT. Enumeration of amino acid fermenting MA, et al. Persistent gut microbiota immaturity in malnourished
bacteria in the human large intestine: effects of pH and starch on Bangladeshi children. Nature. 2014; 510(7505): 417–21.
peptide metabolism and dissimilation of amino acids. FEMS Mi-
crobiol Ecol. 1998 Apr; 25(4): 355–68.

Diet and Infant Gut Microbiota Reprint with permission from: 33


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
95 Raman AS, Gehrig JL, Venkatesh S, Chang HW, Hibberd MC, Sub- 97 Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG,
ramanian S, et al. A sparse covarying unit that describes healthy Contreras M, et al. Human gut microbiome viewed across age and
and impaired human gut microbiota development. Science. 2019; geography. Nature. 2012 Jun; 486(7402): 222–7.
365(6449): eaau4735.
96 Planer JD, Peng Y, Kau AL, Blanton LV, Ndao IM, Tarr PI, et al. De-
velopment of the gut microbiota and mucosal IgA responses in
twins and gnotobiotic mice. Nature. 2016 May; 534(7606): 263–6.

34 Reprint with permission from: Laursen


Ann Nutr Metab 2021;77(suppl 3):21–34
DOI: 10.1159/000517912
Focus
When possible, studies should be conducted in more than one
location so that we can better understand microbe-health
relationships that are specific to one culture/region versus those
that are common to all

Reprinted with permission from: Ann Nutr Metab 2021;77(suppl 3):36–48

Microbiomes and Childhood Malnutrition: What Is the Evidence?


Michelle K. McGuire and Mark A. McGuire

Key insights
Feeding practices,
genetics, gender,
Malnutrition is a complex state that goes beyond the nutrient status, lifestyle,
deprivation or excess of nutrients. There is growing evidence endemic microbial
exposure, location
that dysbiosis of the enteric microbial community may play
an important role in predisposing an individual to both
extremes of malnutrition. Healthy and malnourished infants Healthy children Malnourished children
(Microbiome) (Microbiome)
have different fecal microbiomes, and these differences may
precede the onset of physiological symptoms. This highlights
the importance of the gut microbial community as a
modulator of risk or resilience towards the effects of
ࣼ Higher diversity ࣼ Lower diversity
malnutrition. However, other factors are also involved in ࣼ Prevalence of certain strains ࣼ Different microbial profile
predisposing an individual towards nutrition-related disorders, (Bacteroidetes, Bifidobacteria) from that of healthy children
ࣼ Steadily evolves over time ࣼ Age-delayed
including genetic background, gender, infant feeding
practices, and nutritional status.
Many factors influence the development of the gastrointestinal micro-
biome, resulting in key differences in the microbiomes of healthy and
malnourished children.
Current knowledge
The greatest opportunity for altering the enteric microbiome have a higher diversity of bacterial species, as well as a greater
to optimize lifelong health lies within the first 1,000 days of life. prevalence of certain strains, such as Bacteroidetes and bifido-
Underpinning this is the concept that variations in fetal and bacteria. Healthy fecal microbiomes also evolve with age. An-
infant programming through environmental exposures can other relevant finding is that microbial communities vary along
impact health in later life. This also fits with the hypotheses that the gastrointestinal tract, suggesting that the small intestinal mi-
(1) early life exposure to microorganisms protects against al- crobiome should also be taken into account when performing
lergic disease (hygiene hypothesis) and (2) early life exposure studies. Finally, there is no one-size-fits-all definition of a
to those microorganisms with which we have co-evolved is “healthy” microbiome. Future studies should account for these
needed to develop tolerance and avoid unhealthy inflamma- and other confounding variables in the study design and data
tory responses (“related old friends” hypothesis). However, the analyses. Going forwards, we should aim to advance beyond the
current state of knowledge is not sufficient to enable us to cataloging of microbes towards understanding their functions
conclude how variations in the early life microbiome contrib- within the organism as a whole.
ute towards the systemic manifestations of health or disease.

Recommended reading
Practical implications Robertson RC, Manges AR, Finlay BB, Prendergast AJ. The hu-
Studies comparing the gastrointestinal microbiomes of healthy man microbiome and child growth – first 1000 days and be-
and malnourished children have revealed that healthy children yond. Trends Microbiol. 2019;27(2):131–47.

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/


S. Karger AG, Basel
Review Article

Reprint with permission from:


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001

Microbiomes and Childhood


Malnutrition: What Is the Evidence?
Michelle K. McGuire a Mark A. McGuire b    

aMargaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, USA;
bDepartment of Animal, Veterinary, and Food Sciences, University of Idaho, Moscow, ID, USA

Key Messages in individuals and populations (referred to as the double bur-


den of malnutrition), understanding their biological and envi-
• Healthy and malnourished infants have different fecal ronmental causes is a primary research and humanitarian ne-
microbiomes, and this difference can precede onset of
cessity. There is growing evidence of a bidirectional associa-
malnutrition suggesting that microbiome community
structure might impart resilience and/or risk for malnutrition. tion between variation in the gastrointestinal (GI) microbiome
• Early differences in fecal microbiomes can be associated with and risk of/resilience to malnutrition during early life. For ex-
later risk for overweight and obesity, but these differences ample, studies of siblings who discordantly do or do not de-
vary by population and sometimes by sex. velop severe malnutrition show clear differences in the diver-
• Longitudinal studies designed to prospectively assess
sity and composition of fecal microbiomes. These differenc-
variation among infant gastrointestinal microbiomes (and
their likely sources, such as the human milk microbiome), es are transiently lessened during refeeding but re-emerge
infant feeding practices, nutritional status (including thereafter. These findings have been somewhat recapitulated
micronutrients), and systemic and intestinal inflammation are using animal models, but small sample sizes and limited range
needed to tease apart these potentially important complicate interpretation of results and applicability to hu-
relationships.
mans. Mechanisms driving these differences are currently un-
known but likely involve a combination of inflammatory path-
ways (and perhaps antioxidant status of the host) and effects
Keywords on nutrient availability, requirements, and utilization by both
Growth · Health · Human milk · Infant · Malnutrition · host and microbe. A less robust literature also suggests that
Microbiome · Human nutrition · Obesity variation in GI microbiome is associated with risk for obesity
during childhood. The putative impact of GI microbiomes on
malnutrition is likely modified by a variety of important vari-
Abstract ables such as genetics (likely driven, in part, by evolution),
Both undernutrition and overnutrition continue to represent environmental pathogen exposure and its timing, dietary fac-
enduring global health crises, and with the growing implica- tors, and cultural/societal pattern (e.g., use of antibiotics).
tions of both forms of malnutrition occurring simultaneously Given the growing double burden of malnutrition, this topic

karger@karger.com © 2021 Nestlé Nutrition Institute, Switzerland/ Correspondence to:


S. Karger AG, Basel Michelle K. McGuire, smcguire @ uidaho.edu
demands a focused interdisciplinary approach that expands sembly of an “optimal” GI microbiome likely differs depending
from merely characterizing differences and longitudinal on a multitude of factors such as genetics, endemic microbes,
changes in fecal microbes to examining their functionality chronic food availability, and cultural practices. Relationships
during early life. Understanding the complex composition of among these concepts, nutrition, the enteric microbiome,
human milk and how its components impact establishment and short- and long-term health are illustrated in Figure 1.
and maintenance of the recipient infant’s GI microbiome will The focus of this study is to, largely through the lenses of
also undoubtedly shed important light on this topic. these various overlapping concepts and hypotheses, review
© 2021 Nestlé Nutrition Institute, Switzerland/ the complex interactions among early life nutrition, establish-
S. Karger AG, Basel ment and variation in GI microbiomes, and overall health and
nutritional status during childhood. We mainly discuss human
Introduction studies of general malnutrition, but also briefly consider indi-
vidual nutrients (particularly iron) and selected animal studies.
Preventing and treating malnutrition, particularly in children,
remain a global health priority [1]. Preventing undernutrition is
important because inadequate food intake increases risks for Factors Impacting Early Establishment of GI
nutritional deficiencies, impaired immune function, and de- Microbiome
layed cognitive and physical development. Similarly, being
overweight increases one’s risk for a variety of diet-related Whereas it has long been held that the healthy infant is born
noncommunicable diseases such as heart disease, stroke, di- with a sterile GI tract, some evidence suggests that initial mi-
abetes, and cancer. Undernutrition and overnutrition have, in crobial colonization may occur in utero [11, 12], although
the past, been considered somewhat separate health chal- skepticism remains (e.g., [13]). Nonetheless, the origin of a
lenges affecting different populations. However, with indus- large proportion (if not all) of the pioneering bacteria is re-
trialization has come the simultaneous double burden of mal- lated to exposure during birth. Azad and colleagues estimated
nutrition (DBM), even within the same household or popula- that ∼70% of colonizing GI bacteria originate from maternal
tion [2]. Indeed, the prevalence of obesity continues to rise on feces in vaginally delivered infants, whereas a smaller propor-
a global basis while nutrient deficiencies remain common [2, tion (∼40%) originate from this source in infants born via Cae-
3]. sarean section [14]; whether the Caesarean section is planned
Whereas the drivers of this somewhat new nutrition reality or emergency as well as antibiotic use also appear to impact
of DBM are clearly complex, malnutrition is often not as sim- early colonization [15, 16]. However, not all studies have re-
ple as nutrient deprivation or excess [4, 5]. For example, there ported an enduring impact of delivery mode on GI microbi-
is mounting evidence that a dysbiosis of enteric microbes ome [17].
might play a role in predisposing an individual to both ex- After initial colonization, feeding mode directs establish-
tremes of the nutrient status continuum (see Table  1 for a ment and stabilization of the GI microbiome – likely driven by
summary of common measures of malnutrition in children). differences between the components found in human milk
The opportunity to alter the enteric microbiome for optimiz- (HM) and formula. The fecal microbiota of breastfed infants is
ing lifelong health is likely greatest in the first 1,000 days of life not only different in composition than that of formula-fed in-
[6], dovetailing nicely the concept of the developmental ori- fants, but it is also more stable and less diverse [15, 18]. There
gins of health and disease (DOHaD), which proposes that vari- are likely many reasons for these differences including HM
ation in fetal and infant programming through environmental oligosaccharides (HMOs) indigestible by human enzymes but
exposures impact lifelong health [7], and the hygiene hypoth- selectively metabolized by microbes [19]. Even within exclu-
esis and related old friends hypothesis which posit that (1) sively breastfed infants, different HMO profiles have been as-
early life exposure to microorganisms protects against allergic sociated with variation in infant fecal microbiomes [20, 21].
disease [8] and (2) early life exposure to microorganisms with Because HMO profiles are largely driven by maternal genetics
which we have co-evolved is needed to develop tolerance so [22], it is possible that variation in HMOs and the resulting in-
they do not evoke unhealthy inflammatory responses [9], re- fant GI microbiome represent eco-homeorhetic adjustments
spectively. Another important concept is eco-homeorhesis to “normal” to support optimal health in a particular environ-
which posits that what is “normal” and “healthy” in 1 ecosys- ment and may also support the “old friends” hypothesis in that
tem may not confer optimal fitness in another [10]. Indeed, it these microbial profiles nurture endemic, nonpathogenic
is possible that there exists no 1-size-fits-all construct when bacterial taxa in such a way that they are tolerated by and per-
it comes to a healthy GI microbiome in early life. Rather, as- haps even benefit the host. HMO concentrations and profiles

Infant Nutrition, Microbiomes, and Health Reprint with permission from: 37


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
Table 1. Common measures of malnutrition in children

Condition Commonly used metric and/or sign

Low birth weight, g <2,500


MAM Weight-for-height Z score between −2 and −3 without bilateral pitting edema
SAM Weight-for-height Z score <−3, or mid-upper arm circumference <115 mm, or bilateral pitting edema
Kwashiorkor SAM with bilateral pitting edema
Marasmus SAM without bilateral pitting edema
Underweight Weight-for-age Z score <−2
Stunting Height-for-age Z score <−2
Wasting Weight-for-height Z score <−2
Overweight Body mass index Z score >2 or weight-for-length Z score >97th percentile
At risk of overweight Weight-for-length Z score >85th percentile

MAM, moderate acute malnutrition; SAM, severe acute malnutrition.

Hygiene and Old Friends Hypotheses: Safe exposure to endemic, environmental


microbes in early life is important to development of tolerance.

Feeding choices

Eco-homeorhesis: Genetic, Genetics


behavioral, and Emerging
environmental factors Environmental evidence
likely customize microbes Overweight suggests that
microbiome for optimal microbial
fitness for a given location dysbiosis can
and culture. The thrifty Bacterial dysbiosis increase risk of
gene hypothesis fits within and/or resilience
this eco-homeorhetic to malnutrition.
construct. A B1 B2 B3

Well nourished
B5 B6 B7 C

B9 B12 D Fe

E Protein Se K

Nutrient intake

Undernourished

Developmental Origins of Health and Disease: Establishment of the


gastrointestinal microbiome in the “1st 1,000 days” impacts lifelong health.

Fig. 1. Putative relationships among early-life microbial exposures, nutrition GI microbiomes, and malnutrition. GI, gastrointestinal.

can also vary by season, suggesting that environment (e.g., of milk’s own inherent microbiome is likely one of the most
food availability, pathogens) can impact their synthesis [23]. important [24]. Like HMO profiles [25], those of the HM mi-
Many additional components (e.g., lysozyme and lactofer- crobiome (HMM) vary greatly among women and globally
rin) in milk are known to affect GI microbes, but the presence [26]. Although milk and infant fecal microbiomes differ sub-

38 Reprint with permission from: McGuire/McGuire


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
stantially in composition, variation in one is highly correlated amount of food (example of the thrifty gene concept). None-
with variation in the other [27, 28]. The origins of the HMM are theless, it is important to remember that only a small propor-
thought to be a combination of maternal skin and GI tract, tion (∼5–10%) of energy obtained from the diet comes
also fitting nicely into the old friend hypothesis and highlight- through microbial action in the large intestine. Rather, most is
ing the likely importance of a tempered introduction of envi- obtained via digestion by host enzymes and absorption in the
ronmental microbes via breastfeeding/HM to develop toler- stomach and small intestine. Some taxa can also synthesize
ance in the infant. It is also noteworthy that this microbial essential vitamins (e.g., vitamin B12), therefore supplying these
communication between mother and infant is likely bidirec- nutrients to the host and possibly shielding him/her from de-
tional, with continuous inoculation of the mammary gland via ficiencies. While the absorption of vitamin B12 synthesized in
the infant’s mouth [29]. the colon or cecum is unlikely [37], some bacteria may ben-
Introduction of complementary foods also impacts the in- efit from increased levels. As such, variation in early-life GI
fant’s GI microbiome [30, 31] although cessation of breast- microbiome might also predispose an infant/child to malnu-
feeding [32] might be more important suggesting that HM trition and, conversely, variation in early life nutritional status
helps stabilize the infant’s GI microbiome. Changes that occur may have acute and/or enduring impacts on the GI microbi-
around weaning are associated with shifts in the microbial ome. It is also possible that variation in the GI microbiome
community’s capacity to metabolize typically consumed plays a role in environmental enteric dysfunction, a subclinical
foods [32]. In addition, there are shifts in species that produce and chronic disorder characterized by poor growth, blunting
short-chain fatty acids and B vitamins (e.g., folate and vitamin of the small intestine villi, nutrient malabsorption, and chron-
B12), as well as taxa involved in amino acid metabolism [30, ic inflammation [38].
33–35]. To our knowledge, there have been no studies de- To our knowledge, the first study comparing GI microbi-
signed to understand whether these longitudinal changes in ome between healthy and malnourished children was con-
GI microbiomes and their functional capacities are custom- ducted by Monira et al. [39] and included 7 Bangladeshi chil-
ized to best suit the future dietary landscape, but this has been dren with low weight-for-height and 7 healthy children of
hypothesized [34]. study staff. There were substantial differences in both diver-
In summary, many factors impact the diversity and com- sity and composition of the GI microbiome: compared to
position of the GI microbiome during early life, and there ex- the malnourished children, samples from healthy children
ists substantial variation in GI microbiomes (and their func- had higher bacterial diversity and greater relative abun-
tional capacities) among infants. Central to the aim of this dance of Bacteroidetes, whereas Klebsiella and Escherichia
manuscript, we ask whether this variability predisposes an in- were 174- and 9-fold lower. Interpretation, however, was
fant to secondary malnutrition – be it undernutrition or over- severely limited by the fact that the study was small, cross-
nutrition – and if so, is this a one-size-fits-all construct or is sectional, and the 2 cohorts were from different socioeco-
this effect modified by environment? Related, can nutritional nomic groups.
status (e.g., iron) of an infant impact GI microbiome to predis- This study was followed by another [40] which compared
pose the infant to acute and/or long-term health outcomes, GI microbiome of Malawian twins discordant for kwashiorkor
and if so, is this relationship context specific? – a form of severe acute malnutrition (SAM) characterized by
nutritional edema. GI microbiomes of 9 healthy, same-gender
twins were compared with those of 13 same-gender twin
GI Microbiomes and Childhood Undernutrition pairs who were discordant for kwashiorkor. Principal coordi-
nates analysis was used to visualize composite variation in
It is well established that fecal microbiomes differ between inferred fecal microbiome functionality with age. Overall met-
lean and obese adults and that – although these differences abolic functions of the healthy twin pairs’ fecal microbiomes
are clearly due to a complex constellation of genetics, diet, were found to steadily change over time as would be expect-
and other biological and behavioral patterns – there might be ed; the same was found for the healthy co-twins from discor-
particular GI microbiomes that impart resilience to or risk of dant twin pairs. Conversely, metabolic functions of feces col-
both under- and overnutrition. For instance, some taxa are lected from children with kwashiorkor were age-delayed, and
genetically equipped to harvest a greater proportion of the although they transiently became more like healthy children
energy in our foods than others, supplying volatile short- of similar ages during feeding this regressed when feeding
chained fatty acids that can serve as ATP sources to entero- was ceased. To assess causality, this research group then
cytes and colonocytes [36]. Individuals hosting a greater per- treated gnotobiotic mice with feces collected from twins in
centage of these taxa likely garner more calories from a given this study [41]. Results from mice (5 per single stool inoculum)

Infant Nutrition, Microbiomes, and Health Reprint with permission from: 39


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
in which transplantation efficiency was >50% demonstrated subsequent growth in the Malawian cohort [43]. They found
reduced growth and lean mass in mice receiving stool from 5 that MAZ at 12 months predicted future ponderal (but not lin-
stunted/underweight donors compared to mice receiving ear) growth at 18 months, but again myriad confounding and
stool from 3 healthy donors. It is unclear if this reduction in possibly interacting factors (e.g., maternal nutritional status,
replicates from the total donor population allowed for com- variation in milk composition, and infant feeding practices)
parison between the mice inoculated with feces from twin cannot be ruled out. In fact, lack of controlling for these and
cohorts or from a similar age cohort. A direct comparison other confounding factors either in experimental design or
within twin pairs would strengthen the confidence in the re- statistical analysis is a major weakness of most of the research
sults. Further, transplant efficiency was defined based upon conducted on this topic and the reader will notice that this
operational taxonomic unit (OTU) diversity relative to the un- theme represents a common thread running through this
cultured donor sample, which suggests that relative abun- study. A subsequent study [44] also suggested an expected
dance of bacterial taxa is not as important as mere presence maturation in the GI virome among the Malawian twin cohort;
in the community. Finally, clarity on food intake of the mice this trajectory was impaired in SAM and not repaired with
would be helpful in understanding the changes in growth. The refeeding.
authors [41] report that food intake was not different for 4 days The researchers then used a defined 25-bacterium com-
during the 35-day period, but it is not clear what days intake munity isolated from a single fecal sample collected from 1
was measured. Random forest-based modeling identified Bi- stunted Malawian infant transplanted into germ-free mice fed
fidobacterium longum in discriminating weight and lean mass a diet representative of that which is consumed in Malawi to
gain when utilizing feces from children at 6 months of age test whether the addition of bovine milk oligosaccharides
while Faecalibacterium prausnitzii was more important when (BMOs) could rescue growth [43]. Mice (4–5 per group) fed
utilizing feces of children at 18 months of age. The potential the BMO-fortified diet had increased body weight compared
impact of age-related GI microbiome could be critical for po- to mice fed the control diet with or without inulin. Unfortu-
tential success of interventions impacting malnutrition and nately, comparison to mice transplanted with microbiota
emphasizing the importance of clear comparisons within an present in healthy Malawi infants was absent in the experi-
age group. mental design. Mice not fed BMO had sustained body weight
The same group then characterized what it considered losses for 30 days after colonization differing from the work
to be healthy GI microbiome projections during the first 2 of Blanton et al. [41] where only temporary body weight (<8
years of life in 12 healthy Bangladeshi infants and estab- days) losses were noted in any group. Further, while food con-
lished a “microbiome-for-age Z score” (MAZ) to assess GI sumption was not statistically different, the mice fed the
microbiome “maturity” [42]. This metric was applied to 64 BMO-fortified diet consumed 25% more than controls. Similar
Bangladeshi infants with SAM before and after nutritional effects were found with gnotobiotic piglets. Additional studies
treatment; SAM was associated with microbiome immatu- utilizing larger sample sizes and appropriate controls are
rity, and although MAZ score improved upon treatment, needed to further understand these findings.
there was regression to immaturity afterward. In a subset of It is interesting to speculate what causes or prevents “mat-
malnourished infants who provided samples before antibi- uration” of the fecal microbiota, but Vonaesch and colleagues
otic administration (common to SAM treatment), lower mi- [45] provide strong evidence that stunted children may have
crobial diversity prior to feeding was not attributable to an- small intestine bacterial overgrowth dominated by taxa typi-
tibiotics. The authors concluded that microbiome immatu- cally found in the oropharyngeal cavity. Their data suggest a
rity may play a role in predisposing young children to “decompartmentalization” of the GI tract and are not only in-
malnutrition. However, it should be noted that feeding teresting from a mechanistic point of view but also remind us
mode and duration of exclusive breastfeeding were not that microbial communities vary along the GI tract, and that
controlled for, even though the authors provided some ev- the fecal microbiome likely provides a poor representation of
idence that intake of infant formula was associated with what is likely more important for nutrition, that is, the small
higher maturity values. This weakness in experimental de- intestine microbiome.
sign makes its results difficult to interpret. In another related study utilizing data from the Bangladeshi
This study was followed by another that (1) assessed GI and Malawi cohorts described above, Gough and colleagues
microbiome in 27 twin pairs and 2 sets of triplets (from the [46] used a case-control approach to investigate whether
same population) with healthy growth patterns, (2) compared particular fecal microbial community structures or individual
findings with those published previously by Subramanian et al. taxa are associated with linear growth faltering. A total of 10
[42] in Bangladesh (they were similar), (3) and related MAZ to cases and 8 controls from Malawi, and 6 cases and 5 controls

40 Reprint with permission from: McGuire/McGuire


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
from Bangladesh, where cases were defined as HAZ ≤−3 and In a subsequent study [49], the same group compared GI
controls as −3 < HAZ ≤−2. They found that reduced fecal mi- microbiomes of 5 healthy infants and 10 infants with kwashi-
crobiome diversity and increased relative abundance of Acid- orkor; they also found a depletion in oxygen-sensitive pro-
aminococcus sp. predicted more severe later growth deficits karyotes including M. smithii. Diversity of anaerobic taxa was
and hypothesized that microbial fermentation of glutamate lower in the malnourished infants, and much of this difference
might play a role in this association. To test this, they used was due to species not previously identified in human stool
KEGG enzyme abundance data for the Malawi cohort and samples. The authors posited that there is a “loss” of anaerobic
found that the abundance of genes encoding key glutamate- species in patients with kwashiorkor. It is worth noting, how-
metabolizing enzymes was negatively associated with future ever, that although ages were not statistically different be-
height-for-age Z score. However, cases in both cohorts were tween the groups the malnourished cohort was substantially
younger and relatively shorter (HAZ) than controls making it younger than the controls (13.4 vs. 25.1 months). As such, dif-
impossible to disentangle whether the differences in fecal mi- ferences could be explained by myriad other factors such as
crobiome are related to age-related factors (e.g., exclusive breastfeeding status. In addition, the small sample sizes com-
breastfeeding status) or health status. In addition, because the bined with the fact that subjects were recruited from 2 distinct
controls were also stunted, the data cannot be used to infer locations (Senegal and Niger) make the results difficult to in-
differences between well growing and growth-stunted in- terpret. Future studies should endeavor to recruit healthy and
fants. Future studies should better account for these impor- malnourished subjects at similar ages living in the same loca-
tant variables. tion.
In a set of studies carried out in Senegal and Niger, Million In the only study to our knowledge to compare infants with
and colleagues [47] studied 69 children with varying nutri- kwashiorkor (n = 54) to those with marasmus (n = 33), Kris-
tional status. Location was found to be the major driver of tensen and colleagues [50] studied Ugandan children aged
overall fecal microbial community structure when all children 6–24 months. After adjusting for sex and diarrhea at admis-
were considered together, whereas age, sex, and nutritional sion, they found higher diversity in samples collected from
status did not explain much of the overall taxonomic variation. infants with kwashiorkor. When severity of wasting, mid-up-
However, the researchers found lower levels of obligate an- per arm circumference, and stunting were considered sepa-
aerobes associated with malnutrition. This approach was fol- rate from type of SAM, no relationships were found with over-
lowed up with an individual patient data meta-analysis which all GI microbiome. There were also no differences in individ-
included 108 children with SAM and 77 well-nourished chil- ual taxa. Unfortunately, markers of inflammation and
dren from 5 African and Asian countries. Results indicate that differences in microbial community function were not as-
members of the Bacteroidaceae, Eubacteriaceae, Lachnospi- sessed. In addition, children with kwashiorkor were older than
raceae, and Ruminococceae families were depleted while En- those with marasmus (17 vs. 15 months, respectively; p =
terococcus faecalis, Escherichia coli, and Staphylococcus au- 0.04), and age and infant feeding patterns were not controlled
reus were consistently more abundant in feces collected from for in the analyses. This is particularly unfortunate given the
children with SAM. Depletion of obligate anaerobes, de- fact that these authors had previously published findings [51]
creased total bacterial number, and lower relative abundance that children with kwashiorkor in this population were less
of Methanobrevibacter smithii (a member of the Archaea do- likely to be breastfed and that household dietary diversity
main) were also associated with malnutrition. M. smithii is scores were also lower in these children.
thought to enhance GI bacterial polysaccharide digestion by In contrast, in a longitudinal study of 78 Peruvian infants
optimizing hydrogen levels; its potential role in increasing between 5 and 12 months of age [52], those who became
metabolizable energy has been discussed as has its use as a stunted during the study had higher bacterial diversity and al-
therapeutic for modulating energy balance in humans [48]. tered GI microbiome coupled with elevated biomarkers of in-
The authors suggest that maternal antioxidant intake (and flammation that preceded growth faltering. Differences in fe-
thus, milk antioxidant composition) might modify the impor- cal microbial diversity also preceded growth faltering. Impor-
tance of fecal redox capacity to infant nutritional status. If cor- tantly, the researchers took care to document infant feeding
rect, then it is likely that the presence of GI microbiota with practices and reported no differences in duration of exclusive
redox capacity might be particularly important in cases of ma- breastfeeding and age at introduction of solid foods; this rep-
ternal antioxidant deficiency and/or nonexclusive breastfeed- resents an important strength of this study compared to al-
ing and inadequate infant antioxidant consumption – again, most all others on the topic. The authors concluded that the
suggesting that what would be considered “optimal” microbi- origin of immune activation in stunted children may be sus-
omes in various contexts differs. tained, low-grade microbial translocation across the GI mu-

Infant Nutrition, Microbiomes, and Health Reprint with permission from: 41


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
cosa. However, because differences in GI microbiome were amino acids to the infant and lead to decreased growth and
also present prior to growth stunting it is also possible that synthesis of related compounds such as serotonin? Answer-
microbially derived inflammatory metabolites might drive ing these questions will require studies coupling longitudinal
chronic local irritation. Metagenomic analysis to examine mi- assessment of infant growth, GI microbiomes and metabo-
crobial function would be helpful to tease apart these inter- lites, possibly stably labelled amino acids, and circulating me-
twining possibilities. tabolomics.
In another study [53] characterizing longitudinal relation- To help in this regard, Giallourou et al. [56] evaluated lon-
ships among GI microbiome, growth, and inflammation in 691 gitudinally collected urine samples and growth in malnour-
Malawian infants, MAZ and GI microbial diversity were associ- ished Peruvian, Bangladeshi, and Tanzanian infants (n = 281,
ated with weight gain from 6 to 12 months, but associations 249, and 249, respectively) during the first 24 months of life.
with systemic inflammation were inconsistent. Although in- Infants from each location (n = 22, 28, and 13 for Peru, Ban-
fant feeding status was not reported, the analyses were ad- gladesh, and Tanzania, respectively) with LAZ ≥−0.75 at birth
justed for infant age on day of fecal collection. Other studies and ≥−1.25 at 24 months of age were classified as a “healthy”
(e.g., [54]), have also provided weak evidence for a link be- reference group. Both plasma and urinary metabolomes were
tween an “inflammatory” GI microbiome and growth, but assessed, with the latter (in healthy infants) being used to ex-
these studies lack evaluation of inflammatory status in the in- plore the existence of age-discriminatory metabolites com-
fant. mon across cohorts (phenome-for-age Z score, PAZ). PAZ
scores in malnourished children were lower than those of
their healthy counterparts, and low PAZ predicted later linear
GI Microbiome Functionality and Childhood and ponderal growth faltering. After controlling for cofound-
Undernutrition ers such as breastfeeding and diarrhea (none of which were
found to be important), several urinary metabolites arising
Highlighting the potential importance of GI microbiome from microbial-host co-metabolism were positively correlat-
function rather than composition, Mayneris-Perxachs et al. ed with age and/or nutritional status – many related to protein
[55] utilized a case-control study in malnourished versus well- and amino acid metabolism. Importantly, these generalized
nourished Brazilian infants (n = 326) 6–24 months of age. findings were consistent among locations suggesting that
Importantly, cases (n = 158) and controls (n = 168) were they might represent relationships that might be applicable to
matched for age, although matching was within 6 months – a other populations. Whether these associations between PAZ
relatively long period of time in this context – and cases were and nutritional status are causal in nature is unknown, as is
older than controls (14.8 vs. 12.3 months, p < 0.001). In addi- whether differences in GI microbiomes represent mediating
tion, birth weight was lower in cases than controls (−1.7 vs. factors.
−0.52 WAZ, p < 0.001). Whether the infant was breastfed was
apparently similar between groups, although it is unclear from
the report if this refers to ever having been breastfed or breast- Micronutrients (Particularly Iron), Microbiomes,
fed at the time of sample/data collection. Whereas they did and Malnutrition
not directly assess GI microbiome, they evaluated urinary
metabolic phenotypes and indirectly inferred source (host vs. Although most research regarding GI microbiome and early-
microbe). They found that both stunting and wasting were as- life malnutrition deals with generalized undernutrition (e.g.,
sociated with increased proteolytic activity of the infants’ GI kwashiorkor), micronutrient intake might also be important.
microbiomes as well as dysregulated choline metabolism and Although many microbes synthesize vitamins that can be
metabolic adaptations to reduce energy expenditure. Accel- used both by themselves and their hosts, they also rely on the
erated catch-up growth was seen in children with the most presence of some nutrients (e.g., some vitamins, essential
pronounced energy-saving metabolic adaptations. The au- minerals) for survival. It is plausible, therefore, that endemic
thors posit that alterations in methyl donor capacity might nutrient deficiencies might evolutionarily select for GI mi-
trigger early-life epigenetic programming toward a “thrifty” crobes that produce those nutrients – another possible ex-
phenotype. Whereas a favoring of GI microbiota that is able ample of eco-homeorhesis. Yet, aside from iron, very little
to utilize protein as an energy source seems like a reasonable research has been conducted on this topic resulting in a weak
and complementary response to inadequate nutrient avail- knowledge base in this regard. As such, here we focus mainly
ability, it remains unclear how this would occur. Also, will in- on studies relating infant iron consumption and status on the
creased proteolytic activity reduce availability of essential GI microbiome.

42 Reprint with permission from: McGuire/McGuire


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
In a double-blind, controlled intervention trial carried GI Microbiomes and Childhood Obesity
out in Côte d’Ivoire, iron supplementation of 6- to 14-year-
old children (n = 139) increased fecal enterobacteria and There is also substantial interest in the complex interactions
decreased lactobacilli and bifidobacteria [57]. This shift was among GI microbiome, obesity risk, and inflammation in ear-
accompanied by increased calprotectin, a marker of GI in- ly life. One of the first studies to examine this was a case-
flammation. Additional negative outcomes of iron supple- control design in Finland; 24 normal-weight children were
mentation were reported in 6-month-old Kenyan infants matched for gestational age and BMI at birth, delivery mode,
(n = 115) [58] and 1-year-old Peruvian infants (n = 95) [59], probiotic intervention, duration of breastfeeding, and antibi-
with the latter study revealing sex-specific responses. These otic use [65]. GI microbiome was assessed at 6 and 12 months
findings are bolstered by an extensive literature relating iron and relationship to risk for obesity during the first 7 years of
status (and supplementation) to risk of infection, particu- life assessed. Bifidobacterial numbers in stool samples during
larly by pathogens with substantial requirements for this infancy were higher in children who remained healthy weight
mineral – the most studied being those that cause malaria than those who became overweight; S. aureus was higher in
[60]. Indeed, Plasmodium falciparum has evolved elaborate those who became overweight. The researchers hypothe-
mechanisms to sequester iron, and supplementing the host sized that S. aureus may trigger low-grade inflammation, con-
with iron can enhance this pathogen’s survival and repro- tributing to the development of obesity and link their findings
duction. This finding has led the WHO to recommend that to an extended concept of the hygiene hypothesis. In a com-
iron interventions only be administered in areas of endemic panion case-control study involving 15 overweight children at
malaria when diagnosis, prevention, and treatment schemes 10 years of age and 15 healthy-weight children matched for
are employed [61]. sex, gestational age and BMI at birth, mode of delivery, and
Because most microbes have a requirement for iron, hu- duration of breastfeeding, Luoto et al. [66] found that bifido-
mans have evolved a constellation of mechanisms whereby bacterial numbers tended to be lower in fecal samples col-
iron is withheld from pathogens during infection, ultimately lected at 3 months of age in the overweight children. The
impairing bacterial growth while increasing risk for host iron same research team later reported that Staphylococcus was
deficiency. Interestingly, HM is a poor source of iron, and its elevated in feces of infants’ overweight mothers compared to
iron content is relatively refractory to maternal iron status – healthy-weight mothers during pregnancy [67], supporting
suggesting an adaptive advantage to low levels of iron con- the possibility that the infants’ fecal microbiomes were shaped,
sumption in early life. Several studies have provided evidence at least in part, either during delivery, via their mothers’ HMM,
that, in addition to iron supplementation increasing risk for or through other physical contacts in the shared environment.
certain infections including those causing diarrhea, it can also Evidence of intergenerational transmission of the overweight
impact the GI microbiome of children. For instance, several phenotype from mother to offspring via microbiome transfer
have found that iron interventions increase Enterobacteria- has also been provided by Tun and colleagues [68], who uti-
ceae and decrease Lactobacilli and Bifidobacteria [57, 58, 62, lized multiple mediator path modeling to show that infant fe-
63]. This finding is particularly interesting because members cal microbiome (particularly Lachnospiraceae) mediated the
of the Lactobacillaceae family, common in milk and dairy association between maternal prepregnancy overweight and
products and considered to be health-promoting probiotics, risk of overweight in children at 1 and 3 years of age.
have an extremely low requirement for iron. As such, when In a larger study of 330 healthy Danish infants, Bergstrom
iron is abundant in the environment, these taxa are likely out- and colleagues [69] found that increasing abundance of
competed by iron-requiring taxa. It is noteworthy that the im- several taxa (e.g., Firmicutes) and decreasing levels of M.
pact of iron supplementation on GI microbiome appears to smithii in feces of infants between 9 and 18 months of age
be context specific, likely impacted by typical core GI micro- were associated with greater BMI gains – a finding some-
bial community structures, chronic dietary intake, and en- what in conflict with that reported in malnourished children
demic pathogen exposure. For example, Dostal and col- and described previously suggesting that M. smithii might
leagues [64] found no effect of high-dose iron supplements facilitate microbial fermentation of polysaccharides in the
on fecal microbiomes in iron-deficient children with low en- colon. The reason for this discrepancy is unknown, but
teropathogen burden living in South Africa. This discrepancy again highlights the importance of not assuming similar
in findings across locations highlights the need to study these findings in disparate locations. Results from the CHILD study
interactions in each location of interest and the critical impor- in Canada suggested that higher microbial richness and en-
tance of not extending findings and conclusions drawn from richment of Lachnospiraceae at 3–4 months increased risk
one locale to another. of obesity at 12 months [70]. In a companion study [68],

Infant Nutrition, Microbiomes, and Health Reprint with permission from: 43


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
abundance of Lachnospiraceae at 3–4 months was associ- absorption of nutrients by the host and production and use of
ated with more frequent use of household disinfectants, and nutrients by bacteria can create competitive scenarios that
was found to statistically mediate an apparent association are typically not considered in any of these studies. Impor-
between disinfectant use and increased odds of becoming tantly, what is a “healthy” GI microbiome might be modified
overweight or obese at 3 years. However, exclusive breast- by genetics, nutrient availability, endemic microbe exposures,
feeding was associated with lower use of disinfectant, com- feeding patterns, and sex (eco-homeorhesis), but these inter-
plicating interpretation. Dogra et al. [71] reported that in- actions have not been studied using adequate prospective,
fants who acquire higher levels of Bifidobacterium relative- longitudinal experimental designs in at-risk populations. Be-
ly later had lower adiposity at 18 months, but differences in cause GI microbiomes are clearly different across locations
feeding practices were not accounted for. and these customized differences might be important for
Several studies have also considered this topic in prema- acute and chronic health, researchers and public health ex-
ture infants. Scheepers et al. [72] studied 909 preterm infants perts should apply a healthy dose of restraint when consider-
and found that higher levels of fecal B. fragilis at 1 month was ing extending findings from 1 location to another. In sum-
associated with higher BMI between 1 and 10 years, but only mary, the current state of the science is not adequate to con-
when the infants consumed a low-fiber diet. This association clude that variation in the early-life microbiome can lead to
was modified by maternal lifestyle factors, making the results either undernutrition or obesity, and additional rigorous stud-
challenging to interpret. Importantly, feeding practices in the ies are needed in this regard.
first month (exclusive breastfeeding, formula feeding, or a Research in this area must carefully take into account the
combination) and duration of breastfeeding were controlled age and feeding history of study participants, or these impor-
for in the analysis. White and colleagues [73] found that early tant factors might completely confound research results and
detection of Bacteroides in stool samples from 218 preterm substantially complicate their interpretation. When possible,
and term infants at 1 month was associated with reduced studies should be conducted in more than one location so
growth in the first 6 months in male infants; although the au- that we can better understand microbe-health relationships
thors controlled for whether or not the infant had received that are specific to one culture/region versus those that are
formula they did not control for duration of breastfeeding. common to all. Scientific focus should expand well beyond
Additional evidence exists that the association between GI mi- characterizing the association between generalized protein-
crobiome and growth might be sex-specific [69, 74, 75]. energy malnutrition, impaired growth, and energy imbalance
It should be noted that, similar to the literature on antibiot- (e.g., kwashiorkor and marasmus) and GI microbiome. Rather,
ics and growth faltering, there is a relatively large literature and as highlighted by evidence that iron supplementation
examining the association between antibiotic use in infancy during infancy and early childhood might negatively impact
and risk of developing obesity. Findings from these studies, early-life GI microbiome, micronutrients should also be con-
however, are notably inconsistent [75–77]. Again, it is likely sidered. And, as always, studies should be powered sufficient-
that confounders (e.g., infant GI microbiome community ly to address the major hypotheses being tested.
structure and feeding patterns) modify any effect that might Elegant and complex animal studies have examined the
exist and that any impacts are geographically/culturally spe- effect of GI microbiome through inoculation from healthy or
cific. stunted infants. These studies have examined an array of
-omics to identify potential markers or causative factors for
the phenotypes observed in the afflicted children. However,
Conclusions and Major Research Gaps relatively poor establishment of the bacterial community into
the germ-free models and limited numbers of replicates, both
Risk of and resilience to malnutrition as they relate to variation from donors and recipient animals within a donor cohort, re-
in the early-life GIM warrants continued examination because strict our ability to interpret the results – let alone apply them
it is likely that, at least in some contexts, different GI microbi- to the human condition. In addition, a lack of growth or even
omes predispose (or program) individuals to acute and chron- body weight loss with the limited animal numbers is a concern
ic health trajectories (hygiene and old friends hypotheses, for validity of these models. Finally, these models which utilize
DOHaD, 1st 1,000 days emphasis). Most studies to date are weaned animals do not include the potential contribution of
understandably epidemiologic in nature, and the design of HM to the nutrient or health status of children, which would
most interventions has focused on describing members of the be the normal feeding for the children studied. Thus, these
fecal microbiome that could impact health through inflam- models fail to reflect the various food sources consumed by
mation and nutrient utilization. However, consumption and children.

44 Reprint with permission from: McGuire/McGuire


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
Given the growing double burden of malnutrition, under- Conflict of Interest Statement
standing the complex relationships among infant nutrition,
The writing of this article was supported by Nestlé Nutrition Institute
nutritional status, and microbiomes should garner intense re-
and the authors declare no other conflicts of interest.
search activity and funding. Adequately describing this com-
plicated interaction will demand a focused interdisciplinary
approach among nutritionists, microbiologists, immunolo-
Author Contributions
gists, epidemiologists, and clinicians. For these studies to pro-
vide insight, they should expand from simply cataloging mi- Both authors contributed equally to the writing of this manuscript.
crobes to examining their functionality. Because HM contains
a constellation of components (e.g., antimicrobial factors, nu-
trients, and microbes) that can impact establishment of the GI
microbiome, understanding these biological compounds and
microorganisms is likely crucial to this type of research. The
expanded use of well-powered, mechanistic studies using
validated animal models is also clearly needed.

References
1 Black MM, Lutter CK, Trude ACB. All children surviving and thriv- 10 McGuire MK, Meehan CL, Brooker SA, Williams JE, Foster JA, Mc-
ing: re-envisioning UNICEF’s conceptual framework of malnutri- Guire MA. An evolutionary, biosocial perspective on variation in
tion. Lancet Glob Health. 2020; 8(6): e766–7. human milk microbes and oligosaccharides: An example of eco-
homeorhesis? In: McGuire MK, McGuire MA, Bode L, editors. Pre-
2 Popkin BM, Corvalan C, Grummer-Strawn LM. Dynamics of the
biotics and probiotics in human milk. Elsevier Publishing; 2017.
double burden of malnutrition and the changing nutrition reality.
Lancet. 2020; 395(10217): 65–74. 11 Wilczyska P, Skaryska E, Lisowska-Myjak B. Meconium microbi-
ome as a new source of information about long-term health and
3 United Nations Children’s Fund (UNICEF), World Health Organiza-
disease: questions and answers. J Matern Fetal Neonatal Med.
tion, International Bank for Reconstruction and Development/The
2019; 32(4): 681–6.
World Bank. Levels and trends in child malnutrition: key findings
of the 2020 edition of the joint child malnutrition estimates. Ge- 12 Collado MC, Isolauri E, Laitinen K, Salminen S. Distinct composi-
neva: World Health Organization; 2020. tion of gut microbiota during pregnancy in overweight and nor-
mal-weight women. Am J Clin Nutr. 2008; 88(4): 894–9.
4 Prendergast AJ, Humphrey JH. The stunting syndrome in devel-
oping countries. Paediatr Int Child Health. 2014; 34(4): 250–65. 13 Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J. A critical
assessment of the “sterile womb” and “in utero colonization” hy-
5 Martorell R, Zongrone A. Intergenerational influences on child
potheses: implications for research on the pioneer infant micro-
growth and undernutrition. Paediatr Perinat Epidemiol. 2012;
biome. Microbiome. 2017; 5(1): 48.
26(Suppl 1): 302–14.
14 Azad MB, Konya T, Persaud RR, Guttman DS, Chari RS, Field CJ, et
6 Robertson RC, Manges AR, Finlay BB, Prendergast AJ. The human
al. Child study investigators. Impact of maternal intrapartum anti-
microbiome and child growth – first 1000 days and beyond.
biotics, method of birth and breastfeeding on gut microbiota dur-
Trends Microbiol. 2019; 27(2): 131–47.
ing the first year of life: a prospective cohort study. BJOG. 2016;
7 Stiemsma LT, Michels KB. The role of the microbiome in the de- 123(6): 983–93.
velopmental origins of health and disease. Pediatrics. 2018;141(4):
15 Azad MB, Konya T, Maughan H, Guttman DS, Field CJ, Chari RS, et
e20172437.
al. Child study investigators. Gut microbiota of healthy Canadian
8 Sicherer SH, Sampson HA. Food allergy: a review and update on infants: profiles by mode of delivery and infant diet at 4 months.
epidemiology, pathogenesis, diagnosis, prevention, and manage- CMAJ. 2013; 185(5): 385–94.
ment. J Allergy Clin Immunol. 2018; 141: 41–58.
16 Langdon A, Crook N, Dantas G. The effects of antibiotics on the
9 Rook GA, Raison CL, Lowry CA. Microbial “old friends”, immuno- microbiome throughout development and alternative approaches
regulation and socioeconomic status. Clin Exp Immunol. 2014; for therapeutic modulation. Genome Med. 2016; 8(1): 39.
177(1): 1–12.
17 Chu DM, Ma J, Prince AL, Antony KM, Seferovic MD, Aagaard KM.
Maturation of the infant microbiome community structure and
function across multiple body sites and in relation to mode of de-
livery. Nat Med. 2017; 23(3): 314–26.

Infant Nutrition, Microbiomes, and Health Reprint with permission from: 45


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
18 Fan W, Huo G, Li X, Yang L, Duan C, Wang T, et al. Diversity of the 32 Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary
intestinal microbiota in different patterns of feeding infants by Il- P, et al. Dynamics and stabilization of the human gut microbiome
lumina high-throughput sequencing. World J Microbiol Biotech- during the first year of life. Cell Host Microbe. 2015; 17(5): 690–
nol. 2013; 29(12): 2365–72. 703.
19 Vandenplas Y, Carnielli VP, Ksiazyk J, Luna MS, Migacheva N, Mos- 33 Tannock GW, Lawley B, Munro K, Gowri Pathmanathan S, Zhou
selmans JM, et al. Factors affecting early-life intestinal microbiota SJ, Makrides M, et al. Comparison of the compositions of the stool
development. Nutrition. 2020; 78: 110812. microbiotas of infants fed goat milk formula, cow milk-based for-
mula, or breast milk. Appl Environ Microbiol. 2013; 79(9): 3040–8.
20 Lewis ZT, Totten SM, Smilowitz JT, Popovic M, Parker E, Lemay
DG, et al. Maternal fucosyltransferase 2 status affects the gut bifi- 34 Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG,
dobacterial communities of breastfed infants. Microbiome. 2015; Contreras M, et al. Human gut microbiome viewed across age and
3: 13. geography. Nature. 2012; 486(7402): 222–7.
21 Smith-Brown P, Morrison M, Krause L, Davies PS. Mothers secretor 35 Vallès Y, Artacho A, Pascual-García A, Ferrús ML, Gosalbes MJ,
status affects development of children’s microbiota composition Abellán JJ, et al. Microbial succession in the gut: directional trends
and function: a pilot study. PLoS One. 2016; 11(9): e0161211. of taxonomic and functional change in a birth cohort of Spanish
infants. PLoS Genet. 2014; 10(6): e1004406.
22 Williams JE, McGuire MK, Meehan CL, McGuire MA, Brooker SL,
Kamau-Mbuthia EW, et al. Key genetic variants associated with 36 Holmes ZC, Silverman JD, Dressman HK, Wei Z, Dallow EP, Arm-
variation of milk oligosaccharides from diverse human popula- strong SC, et al. Short-chain fatty acid production by gut micro-
tions. Genomics. 2021; 113(4): 1867–75. biota from children with obesity differs according to prebiotic
choice and bacterial community composition. mBio. 2020; 11(4):
23 Davis JC, Lewis ZT, Krishnan S, Bernstein RM, Moore SE, Prentice
e00914–20.
AM, et al. Growth and morbidity of gambian infants are influenced
by maternal milk oligosaccharides and infant gut microbiota. Sci 37 Schjønsby H. Vitamin B12 absorption and malabsorption. Gut.
Rep. 2017; 7: 40466. 1989; 30: 1686–91.
24 Pace RM, Williams JE, Robertson B, Lackey KA, Meehan CL, Price 38 Bartelt LA, Bolick DT, Guerrant RL. Disentangling microbial me-
WJ, et al. Variation in human milk composition is related to differ- diators of malnutrition: modeling environmental enteric dysfunc-
ences in milk and infant fecal microbial communities. Microor- tion. Cell Mol Gastroenterol Hepatol. 2019; 7(3): 692–707.
ganisms. 2021; 9(6): 1153.
39 Monira S, Nakamura S, Gotoh K, Izutsu K, Watanabe H, Alam NH,
25 McGuire MK, Meehan CL, McGuire MA, Williams JE, Foster J, Sell- et al. Gut microbiota of healthy and malnourished children in Ban-
en DW, et al. What’s normal? Oligosaccharide concentrations and gladesh. Front Microbiol. 2011; 2: 228.
profiles in milk produced by healthy women vary geographically.
40 Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, Cheng
Am J Clin Nutr. 2017; 105(5): 1086–100.
J, et al. Gut microbiomes of Malawian twin pairs discordant for
26 Lackey KA, Williams JE, Meehan CL, Zachek JA, Benda ED, Price kwashiorkor. Science. 2013; 339(6119): 548–54.
WJ, et al. What’s normal? Microbiomes in human milk and infant
41 Blanton LV, Charbonneau MR, Salih T, Barratt MJ, Venkatesh S,
feces are related to each other but vary geographically: the inspire
Ilkaveya O, et al. Gut bacteria that prevent growth impairments
study. Front Nutr. 2019; 6: 45.
transmitted by microbiota from malnourished children. Science.
27 Pannaraj PS, Li F, Cerini C, Bender JM, Yang S, Rollie A, et al. As- 2016 Feb 19; 351(6275): 10.
sociation between breast milk bacterial communities and estab-
42 Subramanian S, Huq S, Yatsunenko T, Haque R, Mahfuz M, Alam
lishment and development of the infant gut microbiome. JAMA
MA, et al. Persistent gut microbiota immaturity in malnourished
Pediatr. 2017; 171(7): 647–54.
Bangladeshi children. Nature. 2014; 510(7505): 417–21.
28 Williams JE, Carrothers JM, Lackey KA, Beatty NF, Brooker SL, Pe-
43 Charbonneau MR, Blanton LV, DiGiulio DB, Relman DA, Lebrilla
terson HK, et al. Strong multivariate relations exist among milk,
CB, Mills DA, et al. A microbial perspective of human developmen-
oral, and fecal microbiomes in mother-infant dyads during the
tal biology. Nature. 2016; 535(7610): 48–55.
first six months postpartum. J Nutr. 2019; 149(6): 902–14.
44 Reyes A, Blanton LV, Cao S, Zhao G, Manary M, Trehan I, et al. Gut
29 Ramsay DT, Mitoulas LR, Kent JC, Larsson M, Hartmann PE. The
DNA viromes of Malawian twins discordant for severe acute mal-
use of ultrasound to characterize milk ejection in women using an
nutrition. Proc Natl Acad Sci USA. 2015; 112(38): 11941–6.
electric breast pump. J Hum Lact. 2005; 21(4): 421–8.
45 Vonaesch P, Morien E, Andrianonimiadana L, Sanke H, Mbecko JR,
30 Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R,
Huus KE, et al. Afribiota investigators: stunted childhood growth
et al. Succession of microbial consortia in the developing infant
is associated with decompartmentalization of the gastrointestinal
gut microbiome. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4578–
tract and overgrowth of oropharyngeal taxa. Proc Natl Acad Sci
85.
USA. 2018; 115(36): E8489.
31 Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO. Develop-
46 Gough EK, Stephens DA, Moodie EE, Prendergast AJ, Stoltzfus RJ,
ment of the human infant intestinal microbiota. PLoS Biol. 2007;
Humphrey JH, et al. Linear growth faltering in infants is associated
5(7): e177.
with Acidaminococcus sp. and community-level changes in the
gut microbiota. Microbiome. 2015; 3: 24.

46 Reprint with permission from: McGuire/McGuire


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
47 Million M, Tidjani Alou M, Khelaifia S, Bachar D, Lagier JC, Dione 60 Armitage AE, Moretti D. The importance of iron status for young
N, et al. Increased gut redox and depletion of anaerobic and meth- children in low- and middle-income countries: a narrative review.
anogenic prokaryotes in severe acute malnutrition. Sci Rep. 2016; Pharmaceuticals. 2019; 12(2): 59.
6: 26051.
61 World Health Organization. World malaria report 2016. Geneva:
48 Samuel BS, Hansen EE, Manchester JK, Coutinho PM, Henrissat B, World Health Organization; 2016.
Fulton R, et al. Genomic and metabolic adaptations of Methano-
62 Tang M, Frank DN, Hendricks AE, Ir D, Esamai F, Liechty E, et al.
brevibacter smithii to the human gut. Proc Natl Acad Sci USA.
Iron in micronutrient powder promotes an unfavorable gut micro-
2007; 104(25): 10643–8.
biota in Kenyan infants. Nutrients. 2017; 9(7): 776.
49 Tidjani Alou M, Million M, Traore SI, Mouelhi D, Khelaifia S, Bachar
63 Paganini D, Uyoga MA, Kortman GAM, Cercamondi CI, Moretti D,
D, et al. Gut bacteria missing in severe acute malnutrition, can we
Barth-Jaeggi T, et al. Prebiotic galacto-oligosaccharides mitigate
identify potential probiotics by culturomics? Front Microbiol.
the adverse effects of iron fortification on the gut microbiome: a
2017; 8: 899.
randomised controlled study in Kenyan infants. Gut. 2017; 66(11):
50 Kristensen KH, Wiese M, Rytter MJ, Özçam M, Hansen LH, Namu- 1956–67.
soke H, et al. Gut microbiota in children hospitalized with oede-
64 Dostal A, Baumgartner J, Riesen N, Chassard C, Smuts CM, Zim-
matous and non-oedematous severe acute malnutrition in Ugan-
mermann MB, et al. Effects of iron supplementation on dominant
da. PLoS Negl Trop Dis. 2016; 10(1): e0004369.
bacterial groups in the gut, faecal SCFA and gut inflammation: a
51 Rytter MJ, Namusoke H, Babirekere-Iriso E, Kæstel P, Girma T, randomised, placebo-controlled intervention trial in South African
Christensen VB, et al. Dietary and clinical correlates of oedema in children. Br J Nutr. 2014; 112(4): 547–56.
children with severe acute malnutrition: a cross-sectional study.
65 Kalliomäki M, Collado MC, Salminen S, Isolauri E. Early differences
BMC Pediatr. 2015; 15: 25.
in fecal microbiota composition in children may predict over-
52 Zambruni M, Ochoa TJ, Somasunderam A, Cabada MM, Morales weight. Am J Clin Nutr. 2008; 87(3): 534–8.
ML, Mitreva M, et al. Stunting is preceded by intestinal mucosal
66 Luoto R, Kalliomäki M, Laitinen K, Delzenne NM, Cani PD, Salmin-
damage and microbiome changes and is associated with system-
en S, et al. Initial dietary and microbiological environments deviate
ic inflammation in a cohort of Peruvian infants. Am J Trop Med
in normal-weight compared to overweight children at 10 years of
Hyg. 2019; 101(5): 1009–17.
age. J Pediatr Gastroenterol Nutr. 2011; 52(1): 90–5.
53 Kamng’ona AW, Young R, Arnold CD, Kortekangas E, Patson N,
67 Collado MC, Isolauri E, Laitinen K, Salminen S. Distinct composi-
Jorgensen JM, et al. The association of gut microbiota character-
tion of gut microbiota during pregnancy in overweight and nor-
istics in Malawian infants with growth and inflammation. Sci Rep.
mal-weight women. Am J Clin Nutr. 2008; 88(4): 894–9.
2019; 9(1): 12893.
68 Tun HM, Bridgman SL, Chari R, Field CJ, Guttman DS, Becker AB,
54 Dinh DM, Ramadass B, Kattula D, Sarkar R, Braunstein P, Tai A, et
et al. Canadian healthy infant longitudinal development (CHILD)
al. Longitudinal analysis of the intestinal microbiota in persistent-
study investigators. Roles of birth mode and infant gut microbiota
ly stunted young children in South India. PLoS One. 2016; 11(5):
in intergenerational transmission of overweight and obesity from
e0155405.
mother to offspring. JAMA Pediatr. 2018; 172(4): 368–77.
55 Mayneris-Perxachs J, Lima AA, Guerrant RL, Leite ÁM, Moura AF,
69 Bergström A, Skov TH, Bahl MI, Roager HM, Christensen LB, Ejler-
Lima NL, et al. Urinary N-methylnicotinamide and β-amino-
skov KT, et al. Establishment of intestinal microbiota during early
isobutyric acid predict catch-up growth in undernourished Brazil-
life: a longitudinal, explorative study of a large cohort of Danish
ian children. Sci Rep. 2016; 6: 19780.
infants. Appl Environ Microbiol. 2014; 80(9): 2889–900.
56 Giallourou N, Fardus-Reid F, Panic G, Veselkov K, McCormick BJJ,
70 Azad MB, Bridgman SL, Becker AB, Kozyrskyj AL. Infant antibiotic
Olortegui MP, et al. Metabolic maturation in the first 2 years of life
exposure and the development of childhood overweight and cen-
in resource-constrained settings and its association with postna-
tral adiposity. Int J Obes. 2014; 38(10): 1290–8.
tal growths. Sci Adv. 2020; 6(15): eaay5969.
71 Dogra S, Sakwinska O, Soh SE, Ngom-Bru C, Brück WM, Berger B,
57 Zimmermann MB, Chassard C, Rohner F, N’goran EK, Nindjin C,
et al. Dynamics of infant gut microbiota are influenced by delivery
Dostal A, et al. The effects of iron fortification on the gut micro-
mode and gestational duration and are associated with subse-
biota in African children: a randomized controlled trial in Cote
quent adiposity. mBio. 2015; 6(1): e02419–14.
d’Ivoire. Am J Clin Nutr. 2010; 92(6): 1406–15.
72 Scheepers LE, Penders J, Mbakwa CA, Thijs C, Mommers M, Arts
58 Jaeggi T, Kortman GA, Moretti D, Chassard C, Holding P, Dostal A,
IC. The intestinal microbiota composition and weight develop-
et al. Iron fortification adversely affects the gut microbiome, in-
ment in children: the koala birth cohort study. Int J Obes. 2015;
creases pathogen abundance and induces intestinal inflammation
39(1): 16–25.
in Kenyan infants. Gut. 2015; 64(5): 731–42.
73 White RA, Bjørnholt JV, Baird DD, Midtvedt T, Harris JR, Pagano M,
59 McClorry S, Zavaleta N, Llanos A, Casapía M, Lönnerdal B, Slupsky
et al. Novel developmental analyses identify longitudinal patterns
CM. Anemia in infancy is associated with alterations in systemic
of early gut microbiota that affect infant growth. PLoS Comput
metabolism and microbial structure and function in a sex-specif-
Biol. 2013; 9(5): e1003042.
ic manner: an observational study. Am J Clin Nutr. 2018; 108(6):
1238–48.

Infant Nutrition, Microbiomes, and Health Reprint with permission from: 47


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001
74 Smith-Brown P, Morrison M, Krause L, Davies PSW. Male-specific 76 Trasande L, Blustein J, Liu M, Corwin E, Cox LM, Blaser MJ. Infant
association between fat-free mass index and fecal microbiota in antibiotic exposures and early-life body mass. Int J Obes. 2013;
2–3-year-old Australian children. J Pediatr Gastroenterol Nutr. 37(1): 16–23.
2018; 66(1): 147–51.
77 Gerber JS, Bryan M, Ross RK, Daymont C, Parks EP, Localio AR, et
75 Ajslev TA, Andersen CS, Gamborg M, Sørensen TI, Jess T. Child- al. Antibiotic exposure during the first 6 months of life and weight
hood overweight after establishment of the gut microbiota: the gain during childhood. JAMA. 2016; 315(12): 1258–65.
role of delivery mode, pre-pregnancy weight and early adminis-
tration of antibiotics. Int J Obes. 2011; 35(4): 522–9.

48 Reprint with permission from: McGuire/McGuire


Ann Nutr Metab 2021;77(suppl 3):36–48
DOI: 10.1159/000519001

You might also like