ArticlePDF AvailableLiterature Review

Bioactive Functions of Milk Proteins: a Comparative Genomics Approach

Authors:

Abstract

The composition of milk includes factors required to provide appropriate nutrition for the growth of the neonate. However, it is now clear that milk has many functions and comprises bioactive molecules that play a central role in regulating developmental processes in the young while providing a protective function for both the suckled young and the mammary gland during the lactation cycle. Identifying these bioactives and their physiological function in eutherians can be difficult and requires extensive screening of milk components that may function to improve well-being and options for prevention and treatment of disease. New animal models with unique reproductive strategies are now becoming increasingly relevant to search for these factors.
Bioactive Functions of Milk Proteins: a Comparative
Genomics Approach
Julie A. Sharp
1,3
&Vengama Modepalli
2
&Ashwanth Kumar Enjapoori
2
&
Swathi Bisana
1
&Helen E. Abud
3
&Christophe Lefevre
2,4,5,6
&Kevin R. Nicholas
3
Received: 20 March 2015 /Accepted: 19 June 2015 / Published online: 27 June 2015
#Springer Science+Business Media New York 2015
Abstract The composition of milk includes factors required
to provide appropriate nutrition for the growth of the neonate.
However, it is now clear that milk has many functions and
comprises bioactive molecules that play a central role in reg-
ulating developmental processes in the young while providing
a protective function for both the suckled young and the mam-
mary gland during the lactation cycle. Identifying these bio-
actives and their physiological function in eutherians can be
difficult and requires extensive screening of milk components
that may function to improve well-being and options for pre-
vention and treatment of disease. New animal models with
unique reproductive strategies are now becoming increasingly
relevant to search for these factors.
Keywords Mammary gland .Milk .miRNA .Marsupial .
Monotreme .Bioactive
Abbreviations
WFDC2WAP Four-disulfide Core Domain 2
MLP Monotreme Lactation Protein
AMP Antimicrobial Protein
LLP Late Lactation Protein
WAP Whey Acid Protein
ELP Early Lactation Protein
JEOL JEM 2100 Transmission Electron Microscopy
NEC Necrotizing Enterocolitis
Introduction
Understanding the composition of milk has been a fundamen-
tal topic in lactation biology. The molecular basis of mammary
gland functionhas been studied from an evolutionary perspec-
tive to understand the evolution of mammals and adaption of
the lactation system to support the survival of the newborns by
providing nutrients [1,2]. Milk is known as a well-balanced
source of nutrition, providing essential carbohydrates, lipids
and proteins to support the neonate. Milk is well established
nutritional source that also comprises various bioactive com-
ponents supporting early development of the neonate [35]
and also regulates mammary gland development and function
[6,7]. Recent studies have recognized the biofunctional po-
tential of milk in supporting the development of the newborn
by delivering bioactives [8]. Biologically active components
within milk include growth factors, proteins, peptides, fatty
acids, hormones and miRNA [4,9]. The presence of various
sources of bioactivity in milk raises questions about their role
in supporting neonatal growth and development. Studies on
the role of milk in neonates have shown that factors in milk
have a potential role in maturation of the immune system of
the neonate [10]. Milk is a source of different growth factors
*Julie A. Sharp
Julie.Sharp@deakin.edu.au
1
Institute for Frontier Materials, Deakin University, Geelong 3216,
Australia
2
School of Medicine, Deakin University, Geelong 3216, Australia
3
Department of Anatomy and Developmental Biology, Monash
University, Clayton 3800, Australia
4
Division of Bioinformatics, Walter and Eliza Hall Medical Research
Institute, Melbourne 3000, Australia
5
Peter MacCallum Cancer Research Institute, East Melbourne 3002,
Australia
6
Faculty of Science, University of Melbourne, Parkville 3010,
Australia
J Mammary Gland Biol Neoplasia (2014) 19:289302
DOI 10.1007/s10911-015-9331-6
like epidermal growth factor, transforming growth factor,
nerve growth factor, insulin, and insulin like growth factors
supporting development [4]. Growth factors and hormones
have been observed to have a crucial role in regulating organ
development like maturation of the intestinal epithelium for
the preparation of solid food intake following birth [11]. Tra-
ditionally, it was believed that the development of the gut was
not influenced by factors in milk, but recent observations have
shown that bioactive components in colostrum and milk can
influence the development of the gastrointestinal tract [12].
The secretion of milk is a defining feature of all mammali-
an species [13]. The evolution of unique anatomical secretory
mammary glands and development of the lactation process
has seen the evolutionary success of mammals over million
years [14,15]. The term mammalia comes from the latin word
mamma (breast or mammary gland) coined by Carl Linnaeus
in 1758 [16]. A dependency on milk is key to the life history
of all mammals and is evolutionarily ancient in origin [13].
Pond (1984) summarized the process of lactation as a means
by which lactating females can convert their nutrient reserves
for milk production and offer substantial selective advantages
to feed dependent young when food is unavailable, and is a
universal feature of mammalian reproduction. Lactation pro-
vides the essential nutrients required by the smallest and most
altricial neonate to the largest and most precocial offspring
[13](Fig.1). The ability to provide easily digestible, balanced
nutritional components supported evolution of different devel-
opmental and reproductive strategies [13]. In the ancestral
history of mammalian evolution, milk has evolved not only
for nutritional value but also the earliest mechanism through
which mothers signal biochemically to their offspring by pro-
viding factors in milk that play a significant part in improving
health of the young [1721].
Although the lactation cycle is a common feature of all
mammals, the monotremes and marsupials have evolved a
reproductive strategy that is very different from that of most
eutherians. Eutherians have a long gestation relative to their
lactation period, and the composition of milk doesntchange
substantially. This contrasts with reproduction and lactation in
monotremes, such as platypus and echidna, and the marsu-
pials, such as the tammar wallaby (Fig. 2). Deciphering the
functions of milk and discovery of new functional compo-
nents involved in the development of various organs requires
smart, innovative and strategic approaches. One such ap-
proach is the use of comparative models with extreme adap-
tations to lactation such as the monotreme and marsupial
whose young are born highly underdeveloped. When com-
bined with technology platforms that include genomics, pro-
teomics and bioinformatics these powerful models can be
exploited to identify milk proteins (and potentially
microRNA) with bioactivity and subsequent application as
nutraceuticals and pharmaceuticals.
Fig. 1 Developmental differences in newborn offspring of mammalian
lineages. Monotreme and marsupial young begin life as altricial young,
while most eutherian species are born comparatively well developed. a
From left: echidna adult, hatchling emerging from egg and young bFrom
left: tammar wallaby adult, newborn and pouch young cFrom left: human
adult, fetus and newborn
Fig. 2 Schematic representation of differences in length of mammalian
gestation and lactation between lineages. Monotreme and marsupials
have a short gestation and young are highly altricial at birth. This is
followed by a prolonged period of lactation. In contrast, eutherians
evolved longer gestation periods to allow greater in utero development
for the birth of a more highly developed young. The following lactation
period is comparatively short
290 J Mammary Gland Biol Neoplasia (2014) 19:289302
Evolutionary Diversity in Mammalian Lactation
Prototherian, metatherian and eutherian mammals differ prin-
cipally in their strategy of reproduction such as gestation pe-
riod, composition of milk and length of lactation period.
Prototherian mammals comprise the extant species platypus
(Ornithorhynchus anatinus) and 2 genera of echidnas, short-
beaked echidna (Tachyglossus aculeatus)andthelong-beaked
echidna (Zaglossus spp.), which are confined to Australia and
New Guinea [22]. Monotremes have a unique position in
mammalian evolution with a highly specialized reproductive
strategy which facilitates both egg-laying and lactation char-
acterized by the secretion of complex milk [23]. Monotremes
lay eggs after a short gestation period and the egg is incubated
in a burrow (platypus) or pouch (echidna), an immature young
hatches from the egg, and therefore these highly altricial
hatchlings depend entirely on milk secreted by the mother
for development and immune protection [24]. The mother
has a long lactation period extending to 114145 days in
platypus [25] and 160210 days in echinda [26,27], depend-
ing on geographical location. In comparison, marsupials com-
prise over 300 diverse species distributed into two groups
called Ameridelphia and Australidelphia. Marsupials survive
mainly in Australia (~200 species, including tammar wallaby,
possums, kangaroos and koalas). The Virginia opossum is the
only extant species that can be found in North America but
several other species are located in South America (including
more than 65 species of opossums and shrew opossums) [28].
All marsupials have a brief intrauterine life (for example
28 days in the tammar wallaby) supported by a short-lived,
less developed placenta and give birth to an altricial young
which relies on an elaborate and sophisticated lactation period
for developmental cues [10,29]. The tammar wallaby is one
of the most studied marsupials, and its lactation period is di-
vided into three phases (phase 2A, phase 2B and phase 3)
based on the composition of the milk, and growth and behav-
ior of the young. Tammar young are born after 28 days of
gestation and weigh approximately 440 milligrams [30]. Dur-
ing the early phase of lactation (phase 2A), the young is per-
manently attached to the teat and the mother secretes dilute
milk with a low concentration of protein and lipids but a high
concentration of carbohydrates [31]. In the tammar, the ex-
pression of phase specific proteins include early lactation pro-
tein (ELP), whey acid protein (WAP) and late lactation
protein-A &B (LLPs) (Fig. 3)[32]. During the first 100 days,
the development of the neonate is similar to a late stage eu-
therian fetus. Therefore, the signalling factors involved in the
development of the eutherian fetus may be delivered in the
milk secreted by marsupials [33].
In general, pouch young are born with immature organs
due to their short gestation period and during early lactation,
the organs within the pouch young develop according to the
necessity for their survival. This includes developing organs
such as lungs [34], lymphoid tissues [35], nervous system
including brain and spinal cord [36,37] that all develop rap-
idly during this period of milk consumption. In mid-lactation
the young remains in the pouch without being permanently
attached to the teat and milk composition remains similar al-
though changes in milk protein composition have been ob-
served [38]. At this stage, the eyes of the pouch young are
opened, fur is visible and by end of phase 2B, the kidney is
functional toproduce concentrated urine and the thyroid gland
is fully developed [39]. By the end of lactation in phase 3, the
pouch young begins feeding on vegetation but intermittently
consumes milk from the mother until 300350 days of age. In
this phase, the milk becomes rich in protein and lipids with a
reduction in the concentration of carbohydrates [40].
The eutherian mammals are represented by almost 4700
extinct and extant genera [41]. Eutherian mammals evolved
with a deeply invasive placenta to support in utero develop-
ment after which the mother gives birth to a well-developed
offspring compared to monotremes and marsupials [42]. Eu-
therian milk remains relatively unchanged throughout lacta-
tion apart from the initial colostrum [43]. Thus, there are sig-
nificant differences in reproductive strategies among three
groups of mammals but the common feature that unites these
divergent reproduction strategies is that all mammals secrete
milk to supply nutrition and bioactive components to the new-
born until they become independent at weaning [44]. Oftedal
(2002) proposed that mammary gland secretions first originat-
ed from glandular skin secretions in synapsids which laid
parchment-shelled eggs which occurred well before the
Fig. 3 Lactation cycle of the tammar wallaby showing changes in milk
composition during different phases of lactation (phase 2A, phase 2B and
phase 3) and growth of the young. The expression of ELP (early lactation
protein),WAP (whey acidic protein) and LLP (Late lactation protein) was
observed only at specific phases. Modified from [10]
J Mammary Gland Biol Neoplasia (2014) 19:289302 291
common ancestor of living mammals appeared on the earth
[15,23]. The early amniotes (sauropsids and synapsids) in
general laid ancestral parchment-shelled eggs; however,
sauropsids evolved to develop calcified eggshells resistant to
moisture loss when exposed to vapor pressure gradients [13].
In contrast, synapsids did not evolve calcified eggshells but
instead produced glandular skin secretions to prevent eggs
from drying under ambient conditions during the course of
incubation [15,23]. These glandular skin secretions are sug-
gested to have evolved from the primary function of providing
moisture and antimicrobials to parchment-shelled eggs to a
new function associated with the supply of nutrients and other
components like antimicrobials for the young [15]. The egg-
laying ability of synapsids was retained in early mammals
which include the monotremes that lay parchment-shelled
eggs and produce mammary gland secretions to provide nu-
trients for the young [45,46].
Identifying the Potential of Milk as a Source
of Bioactives to Support Development of the Suckled
Yo u n g
Bioactive Milk Peptides
Peptides derived from milk proteins constitute a major family
of signalling molecules [47].Recent studies have revealed that
different fractions of milk protein provide groups of specific
peptides including, casein phosphopeptides, alpha lactorphins
and beta lactorphins, antihypertensive peptides, casokinins,
glycomacropeptide and opioid peptides [48,49]. These
milk-derived peptides have nutraceutical, antimicrobial, anti-
oxidative, antiviral and antihypertensive functions [48,50,51]
and are beneficial to the function of a variety of organs includ-
ing the gastrointestinal, immune, cardiovascular and nervous
systems [52].
Milk proteins are broken down to fragments by the action of
enzymes derived from secretions within the stomach and small
intestine during digestion [4749,53]. However, in the case of
tammar pouch young the digestive system is very underdevel-
oped at the time of birth and only matures during mid-lactation
phase2B (100200 days) [54]. As an alternative, tammar milk
may contain enzymes that support digestion and recent studies
have demonstrated that endogenous enzymatic activities in-
cluding amylase, catalase, lactase, lactoperoxidase, lipase and
phosphatase act on milk proteins to release bioactive peptides
[55,56]. Hence, it is conceivable that tammar milk may include
specific enzymes to hydrolyse milk proteins to produce bioac-
tive peptides at specific times during the lactation cycle as an
additional mechanism to support neonatal development. Inter-
estingly, endogenous human milk peptides have been found to
be greater in milk produced by mothers of preterm infants
compared to term infants [57]. Further studies are needed to
focus on understanding tammar milk derived enzymes and the
potential bioactive peptides derived from fragmentation.
Milk MiRNAs: Potential New Regulators of Newborn
Development
Recent studies have shown milk also contains other compo-
nents such as miRNAs [9,11] indicating that in addition to
proteins and peptides, miRNA may be a new class of signal-
ling molecules that contribute to the development of the neo-
nate. Milk miRNAs may have a crucial role in development
due to their key role in gene regulation [58]. Together with
proteins and peptides, the mammary gland also packages and
secretes miRNAs [59]. MicroRNAs are small untranslated
RNAs involved in post-transcriptional modification of
mRNAs, which in turn regulates protein levels [60]. miRNAs
play a crucial role in regulating a wide range of cellular func-
tions, such as cell differentiation, proliferation and cell death
[61,62]. Recent studies have shown that secretory miRNAs
are found in body fluids including breast milk [59], saliva
[63], plasma [64] and urine [65]. The presence of miRNAs
in these body fluids suggests they can act as paracrine signals
to regulate gene expression in adjacent cells/tissues by
allowing cell to cell signalling and subsequently regulating
cellular function. Discovery of miRNA in the milk of various
species potentially has profound implications in understand-
ing their role in neonate development and immune regulation.
However, the precise mechanism of secretion and the physio-
logical role of these miRNAs is not yet clear. Studies on the
miRNA expression profiles of human breast milk have shown
the presence of functional miRNAs, indicating that milk me-
diates the supply of maternal miRNA to infants, potentially
further regulating the gene expression within infant tissues
during lactation [11]. The existence of exogenous miRNAs
in the blood serum and other body fluids may suggest that
miRNAs can travel throughout he circulation and play a sig-
nificant role in other sites of the body [66,67]. Recent studies
on transportation of milk miRNAs in different eutherian spe-
cies have shown that miRNAs are packed into the exosomes
and passed on to neonates along with other components [11,
68,69]. Comparative studies on milk miRNA from different
eutherian species includes human [11], porcine [68] and goat
[69]. However, their functional role within the neonate has not
been clearly defined but a potential immunological role in the
newborn has been suggested [59].
The miRNA knockout studies in both laboratory models,
zebrafish and mouse have shown miRNAs are involved in
many biological process, including organogenesis and mor-
phogenesis during fetal development including liver [7072],
brain [70,73,74], kidney [75] and retina [76,77]byregulat-
ing mRNA expression. The latest reports on expression pro-
files of miRNAs in the developing lung suggest a role in
292 J Mammary Gland Biol Neoplasia (2014) 19:289302
regulating lung development [78]; for example miR-127 has
been implicated in regulating terminal bud development [79].
Work to date on milk miRNAs has identified their presence
in milk but their functional role in neonatal development still
needs to be comprehensively analyzed. The existence of
miRNAs in milk supports the concept that milk miRNAs could
represent new signalling agents in the molecular relationship
between mother and child that has evolved in the mammal
kingdom through lactation. The monotreme and marsupial are
ideal models to investigate the role of milk miRNAs in neonatal
development. Recently we showed that tammar milk contained
a substantial amount of miRNAs involved in developmental
processes (Table 1) along with other RNA molecules [9]. These
miRNAs were specifically transported in exosomes (Fig. 4a),
as shown in milk from eutherian species [11,68,69,91].
Exosomes (Fig. 4b, c) and miRNA have also been identified
in platypus and echidna milk (Enjapoori unpublished) suggest-
ing these evolved early in the evolution of milk production.
Further analysis of tammar exosomal miRNAs showed they
were stable under harsh conditions, indicating that milk
miRNAs may be successfully transported to the pouch young
without degradation and may survive longer in the intestinal
tract [9] . Therefore, the presence of exosome-like vesicles in
milk suggests that these secretory vesicles may provide protec-
tive packaging and transport of miRNA to signals the neonate.
In addition, the immature intestinal epithelial lining of pouch
young allows nonspecific uptake of large molecules from milk
such as immunoglobulins for a major period of lactation
[9294], a process which could facilitate transfer of exosomes
across the gut. This indicates that tammar wallaby milk
miRNAs are most likely transported into the pouch young
blood circulation. The high levels of miRNA secreted in milk
has allowed the identification of differentially expressed milk
miRNAs during the wallaby lactation cycle and demonstrates
the dynamics of milk miRNA profiles across the tammar wal-
laby lactation cycle [9]. This analysis identified putative
markers of mammary gland activity and functional candidate
miRNAs which may assist growth and timed development of
the young. This approach highlights the value of comparative
quantitative transcriptomics to improve understanding of milk
composition and, more importantly, has paved the way forward
to address the true potential of milk miRNA functionality and
the full impact of milk consumption on health and wellbeing.
Tabl e 1 Differentially expressed
tammar milk miRNAs and their
function during development
miRNA Function Reference
miR-148 Growth and development of normal tissues [80]
miR-184 Development of central nervous system and regulates the
balance between neural stem cell proliferation and differentiation
[70,73,74]
let-7b Neural stem cell differentiation and proliferation [81]
let-7 s Regulates the timing of terminal differentiation during embryogenesis [82,83]
miR-122 Liver-specific and key role in liver development [84][71,72]
miR-22 Neural system and erythroid development and maturation [85,86]
miR-375 Highly expressed in hormone secreting organs
(pancreas and pituitary gland) and regulates pancreas organogenesis
[8789]
miR-204 Lens and retinal development [77,90]
miR-30 Kidney development by regulating Xenopus pronephros development [75]
Fig. 4 Exosomes in monotreme and marsupial milk. Exosomes (30
100 nm) were prepared from atammar wallaby, b, platypus and cand
short-beaked echidna milk by differential centrifugations and visualized
by JEM 2100 transmission electron microscopy (JEOL) with an acceler-
ating voltage of 200 kV
J Mammary Gland Biol Neoplasia (2014) 19:289302 293
Bioactivity of Milk for Immune Defence; Evidence
from the Monotreme and Marsupial
The best known antibacterial proteins family, the cathelicidins
are secreted in milk as well as epithelial surfaces and in neu-
trophils and have been proposed to provide a first line of
defence against infection by acting as natural antibiotics [95,
96]. Tammar mammary cathelicidins are secreted abundantly
in milk during early neonatal life (mid lactation) and weaning
(involution) and display antibacterial activity against the com-
mon Staphylococcus aureus,Pseudomonas aeruginosa,En-
terococcus faecalis and Salmonella enterica species [96].This
correlated with stages of lactation associated with higher risks
of infection in pouch young and mammary gland. Similarly, in
the tammar, WFDC2 (WAP four-disulfide core domain 2) ex-
pression has been identified in the mammary gland during
lactation [97]. Tammar WFDC2 is comprised of two 4-DSC
domains: domain III on the amino terminal end and domain II
at the carboxyl terminal end. The domain II of the protein
exhibited significant antibacterial activity against S. aureus, S.
enterica and P. aeruginosa while no activity was seen against
gut commensal E. faecalis [98]. The timing of WFDC2 expres-
sion during pregnancy, early lactation and involution correlates
with stages of the lactation cycle that have increased risk of
infection in the mammary gland. Lactoferrin and lysozyme
are other examples of milk bioactives present in milk. These
proteins are capable of degrading gram negative and positive
cell walls, respectively [17,99101]. In newborn mammals,
immature intestinal immune function and the intestinal epithe-
lial lining may be susceptible to systemic infections. Secretion
of antimicrobials and functional nutrients into mothersmilkis
a potential strategy to reduce the young early-life infections,
and protect the integrity of the epithelial monolayer.
Recently two novel milk proteins, AMP (antimicrobial pro-
tein) and MLP (monotreme lactation protein) have been iden-
tified only in both platypus and echinda milk [102]. AMP and
MLP are abundantly expressed in milk at similar levels to
caseins [103,104]. Recombinant AMP protein was found to
have significant bacteriostatic activity against Escherichia
coli,S. enterica and S. aureus, and showed significant inhibi-
tion of growth of Staphylococcus epidermidis and
P. aeruginosa [103].However, AMP showed no inhibition
of growth of the bacterial species, E. faecalis demonstrating
that AMP exhibited strain-specific antibacterial activity. Sim-
ilarly, the study of recombinant MLP also showed significant
antibacterial activity against two gram-positive bacterial spe-
cies S. aureus and E. faecalis but had no effect on the third
gram-positive species, S. epidermidis, nor the gram-negative
E. coli 2348/69, P. aeruginosa,E. coli O157:H7 and
S. enterica [104]. Antimicrobial activity was observed but
levels of activity varied between S. aureus and E. faecalis.
Both AMP and MLP protein sequence are highly con-
served between the two monotreme species studied. However
homologues have not been identified in other mammals sug-
gesting that these proteins are specific to this lineage and
therefore highly specialized in their role in monotreme lacta-
tion. Monotreme mammary glands are unique because they
have no nipples. Milk produced by the monotreme emerges
from a cluster of openings of mammary gland ducts (areola or
milk patch) onto the females abdomen and the young suck up
the milk directly from the abdominal surface [105,106]. It is
therefore predicted that AMP and MLP may be necessary to
facilitate nippleless delivery of milk which is more prone to
bacterial exposure. A major rationale behind this hypothesis is
that highly altricial hatchlings are immunologically naïve lack-
ing differentiated lymphoid tissue and mature immune effector
cells [107]. It has been reported that monotreme do not possess
CD4 T cell memory immune response and lymph nodes, which
are characteristic of mammalian immunity [108]. The hatchling
without immunological tissues will have lower resistance to
infection from a wide range of pathogenic organisms in the
non-sterile environment of the burrow or pouch, and therefore
may require high concentrations of proteins like AMP and MLP
in the milk for anti-bacterial protection.
Mother-to-hatchling transfer of pathogens during milk in-
gestion as the young lick the abdominal fur on the milk patch
is very problematic and may present one of the most signifi-
cant challenges to the survival of the young. In marsupials
such as the tammar wallaby this disadvantage appears to have
been minimised by the development of a teat to which the
altricial young permanently attaches for the first 100125 days
of lactation [38,109,110]. Marsupial pouch secretions also
contain antimicrobial proteins which provide immune protec-
tion to the immune deficient pouch young along with antimi-
crobial proteins in milk [10,96,98,111,112]. Interestingly,
some of the antibacterials are delivered in the milk only when
the young is immunocompromised or when the mammary
gland is particularly susceptible to infection. In eutherians,
the developing young are protected by the sterile confines of
the mothers uterus and young are born at a more advanced
stage of development. During intra-uterine development the
immune system of the fetus becomes more developed (al-
though not to full maturity) and a level of protection is pro-
vided postpartum via colostrum and milk bioactives [113].
Therefore the evolution of a sophisticated delivery system of
signalling molecules necessary for continued development of
the young whilst maintaining the necessary protection against
threats such as bacterial infection appear to have evolved dif-
ferently in each of the mammalian lineages.
Eutherian Placenta vs Marsupial Milk: Delivery
of Bioactives to Support Development of the Young
Factors responsible for development of the eutherian fetus are
generally supplied through the placenta via the umbilical cord.
In eutherians, the placenta mediates intrauterine fetal
294 J Mammary Gland Biol Neoplasia (2014) 19:289302
development and also plays a key role in fetal growth by
supplying various bioactives [114,115]. During fetal devel-
opment the placenta performs a wide range of physiological
functions which are subsequently performed by the neonate
after birth including respiration, endocrine and digestive pro-
cesses [116,117]. Blood from the fetus enters the umbilical
arteries of the umbilical cord and blood enters the placenta
with the required amount of oxygen and nutrition [118]. Re-
cent studies have demonstrated that the placenta plays an in-
tegral role in programing both brain and lung development of
the fetus [119122].
In contrast the majority of marsupial young development
occurs postnatally, the associated changes are comparable to
the developmental changes in the fetus in eutherians [123].
Hence, bioactives involved in regulating eutherian fetal devel-
opment delivered to the fetus via the placenta are proposed to
be delivered through milk by the marsupial mammary gland
[54,110,124]. Early attempts to understand the role of milk in
development of tammar pouch young have shown that factors
in milk have a potential role in maturation of the immune
system of the neonate [10]. Recent studies on understanding
the changes in tammar milk protein secreted during lactation
have revealed several factors that may involve maturation of
internal organs [54,110,124].Theseincludethegrowth
hormone-like and growth factors like epidermal growth factor,
transforming growth factor, nerve growth factor, insulin, and
insulin-like growth factors which support development
[125127]. Fostering experiments performed in tammars to
understand regulatory effects of milk composition on the rate
of pouch young development have demonstrated that transfer-
ring early pouch young to late lactating tammar mothers can
accelerate the growth and development of pouch young [54,
128]. This is presumably due to disruption of the timing of
delivery of essential bioactives necessary for growth and
development.
Milk Bioactives in Gut Development
Studies attempting to identify the role of milk factors in gut
development of tammar pouch young have shown that every
phase of milk has specific factors triggering phenotypic
changes in the neonate stomach, where the late phase milk
can drive stomach maturation following cross fostering of
the early phase neonate to late phase lactating mother [54].
Milk is a vital immune modulator aiding in regulation of the
microbial flora of neonates gut [129]. As previously
discussed, studies of gene expression profiles of tammar wal-
laby milk protein genes have shown differential expression
profiles of genes encoding antibacterial proteins WFDC2
[97] and cathelicidin [130]. Further analysis of the WFDC2
protein has shown specific antibacterial activity protecting the
mammary gland of the mother, whilst avoiding the disruption
of neonatal gut microbial flora [98].
Growth factors and hormones have a crucial role in regu-
lating organ development like maturation of neonate gut for
preparation of food intake [11]. Human preterm infants (30
37 weeks) can reduce the risk of necrotizing enterocolitis
(NEC), a result of a poorly developed gastrointestinal tract,
by breast feeding [131,132]. Studies attempting to identify the
role of milk factors in gut development in the marsupial
tammar wallaby pouch young have shown that every phase
of milk has specific factors that trigger phenotypic changes in
the neonate stomach [54]. The physiological delay in gut mat-
uration is considered advantageous in mammals like marsu-
pials, as their gastro-intestinal system is not mature until the
late phase [133]. This allows the pouch young to have the
capacity to absorb macromolecules such as maternal immu-
noglobulins in milk from the gut and this capacity for en-
hanced absorption persists through a majority of pouch life
[133]. This significantly contrasts to gut closure observed in
eutherians at a much earlier stage of development [92,93,
133]. The milk proteins and peptides transmitted from the
gut lumen into the peripheral circulation may play a regulatory
role in pouch young maturation in other organs.
Milk Bioactives in Lung Development
The lungs are a primary organ that undergoes considerable
morphological change to perform adequate levels of respira-
tion in the newborn. At birth, the respiratory system of the
neonate must be mature enough to function with efficient
gas exchange. Respiratory complications are frequently seen
in premature infants and preterm death rates are high due to
incomplete development of the respiratory system. Identifying
the components responsible for lung development and accel-
erated maturation of the lung may provide new intervention
therapies to reduce preterm deaths. During early development
of the respiratory system in eutherians, several factors in-
volved in regulating specific stages of lung development are
signalled by factors secreted by the placenta which drive lung
development for postnatal function. In contrast, marsupial
young are born with immature lungs and the majority of lung
development occurs postnatally and is dependent on factors
provided in milk.
The process of lung development in all mammals is similar
[134], but differs in the perinatal and postnatal development
period. In eutherians, the majority of lung morphogenesis oc-
curs during gestation, where the placenta performs gaseous
exchange between the fetus and the mother [135], and during
this period the lung develops completely to perform gaseous
exchange after birth. In contrast, studies of lung development
in several marsupial species including bandicoot (Isoodon
macrounus)[136], Julia Creek dunnart (Sminthopsis douglasi)
[137], tammar wallaby (Macropus eugenii) and gray short
tailed opossum (Monodelphis domestica)[138]havedemon-
strated that the lungs of marsupial newborns are still under the
J Mammary Gland Biol Neoplasia (2014) 19:289302 295
process of development and major developmental changes in
the respiratory system occur during their early postnatal life
[34].
Newborn marsupials are similar in development to an early
stage eutherian fetus and the immature lung is required to
develop rapidly to become fully functional. Unlike eutherians,
marsupials have the ability to allow the transmission of mac-
romolecules across the gut wall due to their immature gut
[133] and therefore the milk proteins and peptides transmitted
from the gut lumen into the peripheral circulation may play a
regulatory role in lung maturation. For example, in marsupial
species like the tammar wallaby and brushtail possum, the
thyroid gland is non- functional until mid-lactation [139,
140], but analysis of thyroid hormone levels in the neonate
have shown the presence of T4 & T3 thyroid hormones [39]
and the presence of thyroid hormone in the milk throughout
lactation [109]. These findings suggest that the thyroid hor-
mones in the neonate are acquired from the mothers milk
[39]. This theory was later supported by observing the trans-
mission of radiolabelled thyroxin from mother to neonate in
brushtail possums (Trichosurus vulpecular) and bandicoots
(Isoodon macrourus) during early lactation [141].
Recently we performed the most detailed investigation of
gray short tailed opossum and tammar wallaby lung devel-
opment to date, to understand the morphological changes
during the course of early postnatal life [142]. These studies
showed lungs in the newborns were immature, transiting
from the late canalicular to saccular stage of development
(Fig. 5). As the marsupial neonate developed the lung
underwent morphological changes by forming sacs and
gradually increased the respiratory surface area, simulta-
neously the volume of lung increased and by end of lacta-
tion the lungs were well developed and similar to that ob-
served in a eutherian newborn [135,137,138]. During early
postnatal life the lungs comprised a handful of sacs and
connected with primitive air ducts and were too immature
to provide the required amount of oxygen for the young
[143145]. However, studies undertaken to understand the
respiratory mechanism of newborn tammar and dunnarts
[146] have demonstrated respiration occurs through the skin
for a period of early postnatal life to meet oxygen demand.
This unique mechanism diminishes gradually during early
development as the neonate lungs mature to perform effi-
cient respiration [137,147]. It appears that this mechanism
was exclusively developed by these primitive mammals to
survive during their early postnatal life and this mechanism
has been lost during evolution of sophisticated respiration in
mammals. This is also consistent with respiration through
theskininprimitiveanimalslikeamphibians[148].
Based on our observation the majority of tammar pouch
young lung development is carried out during early lactation
phase 2A. In order to test the role of tammar milk in lung
development a study was performed using mouse embryonic
lungs (E-12) cultured in media with tammar skim milk col-
lected at key time points of lactation [142]. Remarkably the
embryonic lungs showed increased branching morphogenesis
when incubated with milk collected at specific time points
between approximately day 40 to 100 lactation (phase 2A)
and reduced lung development when incubated in milk from
day of 20 lactation (Fig. 6). This suggests that day 20 milk
either lacks the necessary factors to stimulate lung develop-
ment or includes inhibitors of this process at a time when
respiration occurs through the skin and is an example of timed
developmental cues delivered through milk. This concept of
the timed presentation of milk bioactives to the young for lung
development is consistent with the timed appearance of milk
bioactives that regulate gut development in the young and
protection of the young from infection [38,54,110,124]. A
cross fostering experiment was performed whereby 25 day old
wallaby pouch young were fostered onto a series of mothers at
day 1525 of lactation (Fig. 7) until the pouch young was
45 days old (Modepalli unpublished). Extraordinarily, the fos-
tered young showed marked reduction in lung volume and
lung development compared to the control group. Notably
total body weight and head length was also reduced in fostered
young when compared with controls.
Overall, these studies have demonstrated the importance of
milk in marsupial postnatal development and showed that
milk has evolved to support the development of immature
pouch young, in addition to providing a source of nutrition.
Fig. 5 Lung development in the tammar wallaby. The majority of lung
development in the tammar wallaby occurs postnatally and progresses
through late canalicular to alveolar stages
296 J Mammary Gland Biol Neoplasia (2014) 19:289302
Importance of These Models for Future Study to Improve
the Health Outcomes of Preterm Neonates
Preterm neonates are highly vulnerable to death due to
interruption of their normal fetal development. Human
neonates born before 37 weeks of gestation are considered
as preterm births [149,150] and are afflicted with various
health issues including respiratory complication, limited
central nervous system and also have decreased risk of
survival [151154]. Even though the initial survival out-
comes are positive, the preterm baby may develop long
term issues that include lifelong mental retardation, poor
health, chronic diseases like hypertension, cardiovascular
and diabetes [155].
In all newborn eutherian mammals, the ability to survive in
the external environment depends on the degree of maturity at
birth. The rate of maturation and the period of gestation are
inversely proportional. A better understanding of the process
of fetal development and the molecular mechanism of organ
maturation may provide new therapeutic approaches for re-
ducing the number of preterm deaths. During fetal develop-
ment in utero, factors involved in regulating specific morpho-
logical changes are signaled by placental secretions and am-
niotic fluid in readiness for postnatal function. In contrast,
Fig. 6 Tammar milk accelerates
lung development. aMouse
E12.5 embryonic lung cultures
with tammar milk protein. bAfter
3 days (T72) lung development is
accelerated when cultured in day
60 milk and abrogated when
cultured in day 20 milk compared
to other time points. cNumber of
terminal endbuds of differentiated
lungs grown in day 20, day 60,
and day 120 milk
Fig. 7 Foster experiment. Pouch
young of the foster group only
receives milk from day 125 of
lactation, while the control group
receives milk from days 145 of
lactation
J Mammary Gland Biol Neoplasia (2014) 19:289302 297
marsupial pouch young are born with immature organs due to
their short gestation and during early lactation the pouch
young organs necessary for survival, such as respiratory sys-
tem [34], lymphoid tissues [35], nervous system including
brain and spinal-cord [36,37] are rapidly developed and are
entirely dependent on factors provided through milk [33,38].
Therefore, the marsupial and monotreme provide exceptional
models to identify and understand the role of these milk fac-
tors in programming and stimulating fetal development.
Future studies are needed to investigate the gene expres-
sion profile in the tammar mammary gland during the lacta-
tion cycle and proteomics to identify those factors in milk
regulating lung development. Identification of bioactives
which progress or halt the development of lung may provide
therapeutic modalities, or targets for therapeutic modalities,
for the treatment of preterm babies at risk of respiratory fail-
ure. In addition, future studies understanding the importance
of tammar milk in driving the maturation of other organs in-
cluding gut, nervous system and kidney may provide signifi-
cant evidence to support preterm neonatal maturation. The
potential approaches to produce intervention therapy will not
only assist in saving lives but may also shorten the duration of
preterm infant hospitalization leading to substantially reduced
costs for hospital-based health care.
Summary
Advances in next-generation sequencing transcriptomics and
mass spectrometry proteome studies have enabled precise de-
termination of the timing of mammary gland gene expression
and protein production during the lactation cycle from diverse
mammalian lineages. RNA profiling of both mammary gland
tissue and mammary epithelial cells secreted in milk have
been used to catalogue the complete lactome and mass spec-
trometry has been used to explore mammary secreted
proteomes. The depth of proteogenomic analysis of milk from
platypus, echidna, tammar wallaby, mouse, fur seal and hu-
man lactation have enabled the discovery of a number of novel
milk protein-coding genes and proteins which are tailored to
the particular needs of each mammalian lineage. The contin-
uous change in the composition of milk in marsupials and
monotremes presents an excellent opportunity to identify
and investigate the role of milk derived factors in neonatal
maturation. Further studies in understanding monotreme and
marsupial milk may reveal those necessary signalling factors
delivered through milk which are involved in regulating de-
velopment of various organs of the young during lactation,
such as lung and gut. These unique models may offer new
opportunities for the identification of signalling molecules that
are presented to the young at a time that correlates with pre-
natal presentation of signalling factors by the placenta and
amniotic fluid for the development of a range of tissues during
eutherian foetal development.
References
1. Lefevre CM, Sharp JA, Nicholas KR. Evolution of lactation: an-
cient origin and extreme adaptations of the lactation system. Annu
Rev Genomics Hum Genet. 2010;11:21938.
2. Blackburn DG. Lactation: historical patterns and potential for ma-
nipulation. J Dairy Sci. 1993;76(10):3195212.
3. Liao Y, Alvarado R, Phinney B, Lönnerdal B. Proteomic charac-
terization of human milk whey proteins during a twelve-month
lactation period. J Proteome Res. 2011;10(4):174654.
4. Donovan SM, Odle J. Growth-factors in milk as mediators of
infant development. Annu Rev Nutr. 1994;14:14767.
5. Oftedal OT, Iverson SJ. CHAPTER 10comparative analysis of
nonhuman milks: a. Phylogenetic variation in the gross composi-
tion of milks. In: Jensen RG, editor. Handbook of milk composi-
tion. San Diego: Academic Press; 1995. p. 74989.
6. Blum JW, Baumrucker CR. Colostral and milk insulin-like growth
factors and related substances: Mammary gland and neonatal (in-
testinal and systemic) targets. Domest Anim Endocrinol.
2002;23(12):10110.
7. Wakao H, Gouilleux F, Groner B. Mammary-gland factor (Mgf) is
a novel member of the cytokine regulated transcription factor gene
family and confers the prolactin response (Vol 13, Pg 2182, 1994).
Embo J. 1995;14(4):8545.
8. Fitzgerald RJ, Meisel H. Milk Protein Hydrolysates and Bioactive
Peptides. In: Fox PF, McSweeney PLH, editors. Advanced Dairy
Chemistry1 Proteins. Springer US; 2003. p. 67598.
9. Modepalli V, Kumar A, Hinds L, Sharp J, Nicholas K, Lefevre C.
Differential temporal expression of milk miRNA during the lacta-
tion cycle of the marsupial tammar wallaby (Macropus eugenii).
BMC Genomics. 2014;15(1):1012.
10. Nicholas K, Sharp J, Watt A, Wanyonyi S, Crowley T, Gillespie
M, et al. The tammar wallaby: a model system to examine domain-
specific delivery of milk protein bioactives. Semin Cell Dev Biol.
2012;23(5):54756.
11. Kosaka N, Izumi H, Sekine K, Ochiya T. microRNA as a new
immune-regulatory agent in breast milk. Silence. 2010;1(1):7.
12. Bösze Z. Bioactive Components of Milk. SpringerVerlag; 2008.
p. 395422.
13. Oftedal O. The evolution of milk secretion and its ancient origins.
Animal. 2012;6(3):35568.
14. Peaker M. The mammary gland in mammalian evolution: a brief
commentary on some of the concepts. J Mammary Gland Biol
Neoplasia. 2002;7(3):34753.
15. Oftedal O. Origin and Evolution of the Major Constituents of
Milk. In: McSweeney PLH, Fox PF, editors. Advanced Dairy
Chemistry. Springer US; 2013. p. 142.
16. Schiebinger L. Why mammals are called mammals: gender poli-
tics in eighteenth-century natural history. Am Hist Rev.
1993;98(2):382411.
17. Séverin S, Wenshui X. Milk biologically active components
as nutraceuticals: review. Crit Rev Food Sci Nutr.
2005;45(78):64556.
18. Zimecki M, Kruzel ML. Milk-derived proteins and peptides of
potential therapeutic and nutritive value. J Exp Ther Oncol.
2007;6(2):89106.
19. Dallas DC, Guerrero A, Khaldi N, Castillo PA, Martin WF,
Smilowitz JT, et al. Extensive in vivo human milk peptidomics
298 J Mammary Gland Biol Neoplasia (2014) 19:289302
reveals specific proteolysis yielding protective antimicrobial pep-
tides. J Proteome Res. 2013;12(5):2295304.
20. Dallas DC, Guerrero A, Parker EA, Garay LA, Bhandari A,
Lebrilla CB, et al. Peptidomic Profile of Milk of Holstein Cows
at Peak Lactation. J Agric Food Chem. 2013.
21. Hettinga K, van Valenberg H, de Vries S, Boeren S, van
Hooijdonk T, et al. The host defense proteome of human and
bovine milk. PLoS ONE. 2011;6(4):e19433.
22. Springer M, Krajewski C. Monotremes (Prototheria). The
Timetree of life. Eds. (Oxford University Press, 2009), UK; 2009.
23. Oftedal O.The origin of lactation as a water source forparchment-
shelled eggs. J Mammary Gland Biol Neoplasia. 2002;7(3):253
66.
24. Griffiths M. The biology of the monotremes. New York:
Academic Press; 1978.
25. Hawkins M, Battaglia A. Breeding behaviour of the platypus
(Ornithorhynchus anatinus) in captivity. Aust J Zool.
2009;57(4):28393.
26. Morrow G, Andersen NA, Nicol SC. Reproductive strategies of
the short-beaked echidnaa review with new data from a long-
term study on the Tasmanian subspecies (Tachyglossus aculeatus
setosus). Aust J Zool. 2009;57(4):27582.
27. Morrow GE, Nicol SC. Maternal care in the Tasmanian echidna
(Tachyglossus aculeatus setosus). Aust J Zool. 2013;60(5):289
98.
28. Tyndale-Biscoe CH. Life ofMarsupials. CSIRO Publishing; 2005.
29. Tyndale-Biscoe CCH, Renfree MB, Renfree D. Reproductive
Physiology of Marsupials. Cambridge University Press; 1987.
30. Ward KL, Renfree MB. Effects of progesterone on parturition in
the tammar, Macropus eugenii. J Reprod Fertil. 1984;72(1):218.
31. Messer M, Nicholas KR. Biosynthesis of marsupial milk oligo-
saccharidescharacterization and developmental-changes of 2
galactosyltransferases in lactating mammary-glands of the tammar
wallaby, macropus-eugenii. Biochim Biophys Acta.
1991;1077(1):7985.
32. Sharp JA, Digby M, Lefevre C, Mailer S, Khalil E, Topcic D, etal.
The comparative genomics of tammar wallaby and fur seal lacta-
tion; models to examine function of milk proteins. In: Thompson
A, Boland M, Singh H, editors. Milk proteins: from expression to
food. USA: Academic; 2009. p. 5580.
33. Brennan AJ, Sharp JA, Digby MR, Nicholas KR. The tammar
wallaby: a model to examine endocrine and local control of lacta-
tion. IUBMB Life. 2007;59(3):14650.
34. Runciman SI, Baudinette RV, Gannon BJ. Postnatal development
of the lung parenchyma in a marsupial: the tammar wallaby. Anat
Rec. 1996;244(2):193206.
35. Basden K, Cooper DW, Deane EM. Development of the lymphoid
tissues of the tammar wallaby Macropus eugenii. Reprod Fertil
Dev. 1997;9(2):24354.
36. HarrisonPH, Porter M. Development of the brachial spinal cord in
the marsupial Macropus eugenii (tammar wallaby). Dev Brain
Res. 1992;70(1):13944.
37. Saunders NR, Adam E, Reader M, Møllgård K. Monodelphis
domestica (grey short-tailed opossum): an accessible model for
studies of early neocortical development. Anat Embryol.
1989;180(3):22736.
38. Nicholas K, Simpson K, Wilson M, Trott J, Shaw D. The
tammar wallaby: a model to study putative autocrine-
induced changes in milk composition. J Mammary Gland
Biol Neoplasia. 1997;2(3):299310.
39. Janssens PA, Grigg JA, Dove H, Hulbert AJ. Thyroid hormones
during development of a marsupial, the tammar wallaby,
Macropus eugenii. J Endocrinol. 1990;127(3):42736.
40. Green B, Griffiths M, Leckie RM. Qualitative and quantitative
changes in milk fat during lactation in the tammar wallaby
(Macropus eugenii). Aust J Biol Sci. 1983;36(56):45561.
41. Archibald JD. Eutheria (Placental Mammals). eLS. John Wiley &
Sons, Ltd; 2001.
42. Wildman DE, Chen C, Erez O, Grossman LI, Goodman M,
Romero R. Evolution of the mammalian placenta revealed by
phylogenetic analysis. Proc Natl Acad Sci U S A. 2006;103(9):
32038.
43. Vaughan T, Ryan J, Czaplewski N. Mammalogy. Jones & Bartlett
Learning; 2011.
44. Hamilton M, Davidson A, Sibly R, Brown J. Universal scaling of
production rates across mammalian lineages. Proc Biol Sci.
2011;278(1705):5606.
45. Blackburn DG, Hayssen V, Murphy CJ. The origins of lactation
and the evolution of milk: a review with new hypotheses.
Mammal Rev. 1989;19(1):126.
46. McClellan HL, Miller SJ, Hartmann PE. Evolution of lactation:
nutrition v. protection with special reference to five mammalian
species. Nutr Res Rev. 2008;21(2):97116.
47. Wada Y, Lonnerdal B. Bioactive peptides derived from human
milk proteinsmechanisms of action. J Nutr Biochem.
2014;25(5):50314.
48. Shah NP. Effects of milk-derived bioactives: an overview. Brit J
Nutr. 2000;84:S310.
49. Livney YD. Milk proteins as vehicles for bioactives. Curr Opin
Colloid Interface Sci. 2010;15(12):7383.
50. Nagpal R, Behare P, Rana R, Kumar A, Kumar M, Arora S, et al.
Bioactive peptides derived from milk proteins and their health
beneficial potentials: an update. Food Funct. 2011;2(1):1827.
51. Pihlanto A. Antioxidative peptides derived from milk proteins. Int
Dairy J. 2006;16(11):130614.
52. Murray BA, FitzGerald RJ. Angiotensin converting enzyme in-
hibitory peptides derived from food proteins: Biochemistry, bio-
activity and production. Curr Pharm Design. 2007;13(8):77391.
53. Dallas DC, Guerrero A, Khaldi N, Borghese R, Bhandari A,
Underwood MA, et al. A peptidomic analysis of human milk
digestion in the infant stomach reveals protein-specific degrada-
tion patterns. J Nutr. 2014;144(6):81520.
54. Kwek JH, Iongh RD, Digby MR, Renfree MB, Nicholas KR,
Familari M. Cross-fostering of the tammar wallaby (Macropus
eugenii) pouch young accelerates fore-stomach maturation.
Mech Dev. 2009;126(56):44963.
55. Fox PF, McSweeney PLH. Advanced dairy chemistry. Volume
1A, Proteins basic aspects. 4th ed. New York: Springer; 2013.
56. Guerrero A, Dallas DC, Contreras S, Chee S, Parker EA, Sun X,
et al. Mechanistic peptidomics: factors that dictate specificity in
the formation of endogenous peptides in human milk. Mol Cell
Proteomics MCP. 2014;13(12):334351.
57. Dallas DC, Smink CJ, Robinson RC, Tian T, Guerrero A, Parker
EA, et al. Endogenous human milk Peptide release is greater after
preterm birth than term birth. J Nutr. 2015;145(3):42533.
58. Ambros V. microRNAs: tiny regulators with great potential. Cell.
2001;107(7):8236.
59. Zhou Q, Li MZ, Wang XY, Li QZ, Wang T, Zhu Q, et al. Immune-
related MicroRNAs are abundant in breast milk exosomes. Int J
Biol Sci. 2012;8(1):11823.
60. Zhu M, Yi M, Kim CH, Deng C, Li Y, Medina D, et al. Integrated
miRNA and mRNA expression profiling of mouse mammary tu-
mor models identifies miRNA signatures associated with mam-
mary tumor lineage. Genome Biol. 2011;12(8):R77.
61. Hwang HW, Mendell JT. MicroRNAs in cell proliferation, cell
death, and tumorigenesis. Brit J Cancer. 2006;94(6):77680.
62. Song L, Tuan RS. MicroRNAs and cell differentiation in
mammalian development. Birth Defects Res C Embryo
Today Rev. 2006;78(2):1409.
63. Michael A, Bajracharya SD, Yuen PST, Zhou H, Star RA, Illei
GG, et al. Exosomes from human saliva as a source of microRNA
biomarkers. Oral Dis. 2010;16(1):348.
J Mammary Gland Biol Neoplasia (2014) 19:289302 299
64. Caby MP, Lankar D, Vincendeau-Scherrer C, Raposo G, Bonnerot
C. Exosomal-like vesicles are present in human blood plasma. Int
Immunol. 2005;17(7):87987.
65. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic
profiling of exosomes in human urine. Proc Natl Acad Sci U S A.
2004;101(36):1336873.
66. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK,
Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable
blood-based markers for cancer detection. Proc Natl Acad Sci.
2008;105(30):105138.
67. Chen X, Ba Y, Ma L, Cai X, Yin Y, WangK, et al. Characterization
of microRNAs in serum: a novel class of biomarkers for diagnosis
of cancer and other diseases. Cell Res. 2008;18(10):9971006.
68. Gu Y, Li M, Wang T, Liang Y, Zhong Z, Wang X, et al. Lactation-
related microRNA expression profiles of porcine breast milk
exosomes. PLoS One. 2012;7(8):e43691.
69. Ji ZB, Wang GZ, Xie ZJ, Zhang CL, Wang JM. Identification and
characterization of microRNA in the dairy goat (Capra hircus)
mammary gland by Solexa deep-sequencing technology. Mol
Biol Rep. 2012;39(10):936171.
70. Filipowicz W, Großhans H. The Liver-Specific MicroRNA miR-
122: Biology and Therapeutic Potential. In: Gasser SM, Li E,
editors. Epigenetics and Disease. Progress in Drug Research:
Springer Basel; 2011. p. 22138.
71. Wienholds E, Kloosterman WP, Miska E, Alvarez-Saavedra E,
Berezikov E, de Bruijn E, et al. MicroRNA expression in zebrafish
embryonic development. Science. 2005;309(5732):3101.
72. Wu X, Wu SQ, Tong L, Luan TA, Lin LX, Lu SL, et al. miR-122
affects the viability and apoptosis of hepatocellular carcinoma
cells. Scand J Gastroenterol. 2009;44(11):13329.
73. Liu C, Teng ZQ, Santistevan NJ, Szulwach KE, Guo W, Jin P,
et al. Epigenetic regulation of miR-184 by MBD1 governs neural
stem cell proliferation and differentiation. Cell Stem Cell.
2010;6(5):43344.
74. McKiernan RC, Jimenez-Mateos EM, Sano T, Bray I, Stallings
RL, Simon RP, et al. Expression profiling the microRNA response
to epileptic preconditioning identifies miR-184 as a modulator of
seizure-induced neuronal death. Exp Neurol. 2012;237(2):346
54.
75. Agrawal R, Tran U, Wessely O. The miR-30 miRNA family reg-
ulates Xenopus pronephros development and targets the transcrip-
tion factor Xlim1/Lhx1. Development. 2009;136(23):392736.
76. Conte I, Carrella S, Avellino R, Karali M, Marco-Ferreres R,
Bovolenta P, et al. miR-204 is required for lens and retinal devel-
opment via Meis2 targeting. Proc Natl Acad Sci. 2010.
77. Avellino R, Carrella S, Pirozzi M, Risolino M, Salierno FG,
Franco P, et al. miR-204 targeting of <italic>Ankrd13A</italic>
controls both mesenchymal neural crest and lens cell migration.
PLoS ONE. 2013;8(4):e61099.
78. Khoshgoo N, Kholdebarin R,IwasiowBM,KeijzerR.
MicroRNAs and lung development. Pediatr Pulmonol.
2013;48(4):31723.
79. Bhaskaran M, Wang Y, Zhang H, Weng T, Baviskar P, Guo Y,
et al. MicroRNA-127 modulates fetal lung development. Physiol
Genomics. 2009;37(3):26878.
80. Chen Y, Song YX, Wang ZN. The microRNA-148/152 family:
multi-faceted players. Mol Cancer. 2013;12:43.
81. Zhao C, Sun G, Li S, Lang MF, Yang S, Li W, et al. MicroRNA
let-7b regulates neural stem cell proliferation and differentiation
by targeting nuclear receptor TLX signaling. Proc Natl Acad Sci U
S A. 2010;107(5):187681.
82. Grosshans H, Johnson T, Reinert KL, Gerstein M, Slack FJ. The
temporal patterning microRNA let-7 regulates several transcrip-
tion factors at the larval to adult transition in C. elegans. Dev Cell.
2005;8(3):32130.
83. Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC,
Rougvie AE, et al. The 21-nucleotide let-7 RNA regulates devel-
opmental timing in Caenorhabditis elegans. Nature.
2000;403(6772):9016.
84. Filipowicz W, Grosshans H. The liver-specific microRNA miR-
122: biology and therapeutic potential. Prog Drug Res Fortschritte
der Arzneimittelforschung Progres des recherches
pharmaceutiques. 2011;67:22138.
85. Choong ML, Yang HH, McNiece I. MicroRNA expression profil-
ing during human cord blood-derived CD34 cell erythropoiesis.
Exp Hematol. 2007;35(4):55164.
86. Jovicic A, Zaldivar Jolissaint JF, Moser R, Silva Santos Mde F,
Luthi-Carter R. MicroRNA-22 (miR-22) overexpression is neuro-
protective via general anti-apoptotic effects and may also target
specific Huntingtons disease-related mechanisms. PLoS One.
2013;8(1):e54222.
87. Avnit-Sagi T, Kantorovich L, Kredo-Russo S, Hornstein E,
Walker MD. The promoter of the pri-miR-375 gene directs ex-
pression selectively to the endocrine pancreas. PLoS One.
2009;4(4):e5033.
88. Kloosterman WP, Lagendijk AK, Ketting RF, Moulton JD,
Plasterk RH. Targeted inhibition of miRNA maturation with
morpholinos reveals a role for miR-375 in pancreatic islet devel-
opment. PLoS Biol. 2007;5(8):e203.
89. Poy MN, Hausser J, Trajkovski M, Braun M, Collins S, Rorsman
P, et al. miR-375 maintains normal pancreatic alpha- and beta-cell
mass. Proc Natl Acad Sci U S A. 2009;106(14):58138.
90. Conte I, Carrella S, Avellino R, Karali M, Marco-Ferreres R,
Bovolenta P, et al. miR-204 is required for lens and retinal devel-
opment via Meis2 targeting. Proc Natl Acad Sci U S A.
2010;107(35):154916.
91. Hata T, Murakami K, Nakatani H, Yamamoto Y, Matsuda T, Aoki
N. Isolation of bovine milk-derived microvesicles carrying
mRNAs and microRNAs. Biochem Bioph Res Co. 2010;396(2):
52833.
92. Old JM, Deane EM. The lymphoid and immunohaematopoietic
tissues of the embryonic brushtail possum (Trichosurus
vulpecula). Anat Embryol. 2003;206(3):1937.
93. Yadav M. Characteristics of blood in the pouch young of a mar-
supial, Setonix brachyurus. Aust J Zool. 1972;20(3):24963.
94. Daly KA, Digby M, Lefevre C, Mailer S, Thomson P, Nicholas K,
et al. Analysis of the expression of immunoglobulins throughout
lactation suggests two periods of immune transfer in the tammar
wallaby (Macropus eugenii). Vet Immunol Immunopathol.
2007;120(34):187200.
95. Bals R, Wilson JM. Cathelicidins-a family of multifunctional an-
timicrobial peptides. Cell Mol Life Sci CMLS. 2003;60(4):711
20.
96. Wanyonyi SS, Sharp JA, Khalil E, Lefevre C, Nicholas KR.
Tammar wallaby mammary cathelicidins are differentially
expressed during lactation and exhibit antimicrobial and cell pro-
liferative activity. Comp Biochem Physiol A Mol Integr Physiol.
2011;160(3):4319.
97. Sharp JA, Lefèvre C, Nicholas KR. Molecular evolution of mono-
treme and marsupial whey acidic protein genes. Evol Dev.
2007;9(4):37892.
98. Watt AP, Sharp JA, Lefevre C, Nicholas KR. WFDC2 is differen-
tially expressed in the mammary gland of the tammar wallaby and
provides immune protection to the mammary gland and the devel-
oping pouch young. Dev Comp Immunol. 2012;36(3):58490.
99. Batish VK, Chander H, Zumdegeni KC, Bhatta KL, Singh RS.
Antibacterial activity of lactoferrin against some common food-
borne pathogenic organisms. Aust J Dairy Technol. 1988;5:168.
100. Lönnerdal B. Nutritional and physiologic significance of human
milk proteins. Am J Clin Nutr. 2003;77(6):1537S43.
300 J Mammary Gland Biol Neoplasia (2014) 19:289302
101. Saito H, Miyakawa H, Tamura Y, Shimamura S, Tomita M. Potent
bactericidal activity of bovine lactoferrin hydrolysate produced by
heat treatment at acidic pH. J Dairy Sci. 1991;74(11):372430.
102. Lefevre CMSJ, Nicholas KR. Characterisation of monotreme ca-
seins reveals lineage-specific expansion of an ancestral casein lo-
cus in mammals. Reprod Fertil Dev. 2009;21(8):101527.
103. Bisana S, Kumar S, Rismiller P, Nicol SC, Lefèvre C, Nicholas
KR, et al. Identification and functional characterization of a novel
monotreme- specific antibacterial protein expressed during lacta-
tion. PLoS ONE. 2013;8(1):e53686.
104. Enjapoori AK, Grant TR, Nicol SC, Lefevre CM, Nicholas KR,
Sharp JA. Monotreme lactation protein is highly expressed in
monotreme milk and provides antimicrobial protection. Genome
Biol Evol. 2014;6(10):275473.
105. Griffiths M, Elliott M, Leckie RMC, Schoefl G. Observations of
the comparative anatomy and ultrastructure of mammary glands
and on the fatty acids of the triglycerides in platypus and echidna
milk fats. J Zool. 1973;169(2):25579.
106. Griffiths M, McIntosh D, Coles REA. The mammary gland of the
echidna, Tachyglossus aculeatuswith observations on the incu-
bation of the egg and on the newly-hatched young. J Zool.
1969;158(3):37186.
107. Connolly JH, Canfield PJ, McClure SJ, Whittington RJ.
Histological and immunohistological investigation of lymphoid
tissue in the platypus (Ornithorhynchus anatinus). J Anat.
1999;195(2):16171.
108. Withers DR, Gaspal FM, Mackley EC, Marriott CL, Ross EA,
Desanti GE, et al. Cutting edge: lymphoid tissue inducer cells
maintain memory CD4 T cells within secondary lymphoid tissue.
J Immunol. 2012;189(5):20948.
109. Nicholas KR. Control of milk protein synthesis in the marsupial
Macropus eugenii: a model system to study prolactin-dependent
development. In: Tyndale-Biscoe CHJ, P.A., editor. The develop-
ing marsupial. Models for biomedical research; 1988. p. 6885.
110. Trott JF, Simpson KJ, Moyle RL, Hearn CM, Shaw G, Nicholas
KR, et al. Maternal regulation of milk composition, milk produc-
tion, and pouch young development during lactation in the tammar
wallaby (Macropus eugenii). Biol Reprod. 2003;68(3):92936.
111. Edwards M, Deakin J. The marsupial pouch: implications for re-
productive success and mammalian evolution. Aust J Zool. 2012.
112. Edwards M, Hinds L, Deane E, Deakin J. A review of comple-
mentary mechanisms which protect the developing marsupial
pouch young. Dev Comp Immunol. 2012;37(2):21320.
113. Power ML, Schulkin J. Maternal regulation of offspring develop-
ment in mammals is an ancient adaptation tied to lactation. Appl
Translat Genomics. 2013;2(1):5563.
114. Garnica AD, Chan WY. The role of the placenta in fetal nutrition
and growth. J Am Coll Nutr. 1996;15(3):20622.
115. Bell AW, Hay WW, Ehrhardt RA. Placental transport of nutrients
and its implications for fetal growth. J Reprod Fertil. 1999;54:
40110.
116. Jansson T, Powell TL. Human placental transport in altered fetal
growth: does the placenta function as a nutrient sensor?areview.
Placenta. 2006;27(Supplement(0)):917.
117. Jansson T, Powell TL. Role of the placenta in fetal programming:
underlying mechanisms and potential interventional approaches.
Clin Sci. 2007;113(1):113.
118. Burton GJ, Barker DJP, MoffettA, Thornburg K. The placenta and
human developmental programming. Cambridge: Cambridge
University Press; 2010.
119. Zeltser LM, Leibel RL. Roles of the placenta in fetal brain devel-
opment. Proc Natl Acad Sci. 2011;108(38):156678.
120. Bonnin A, Goeden N, Chen K, Wilson ML, King J, Shih JC, et al.
A transient placental source of serotonin for the fetal forebrain.
Nature. 2011;472(7343):34750.
121. Bonnin A, Levitt P. Fetal, maternal, and placental sources of sero-
tonin and new implications for developmental programmingof the
brain. Neuroscience. 2011;197:17.
122. Myatt L. Placental adaptive responses and fetal programming. J
Physiol. 2006;572(1):2530.
123. Tyndale-Biscoe C, Renfree M. Reproductive physiology of
Marsupials. Cambridge: Cambridge University Press; 1987.
124. Joss JL, Molloy MP, Hinds L, Deane E. A longitudinal study of
the protein components of marsupialmilk from birth to weaning in
the tammar wallaby (Macropus eugenii). Dev Comp Immunol.
2009;33(2):15261.
125. Xu R-J. Bioactive Peptides in Milk and their Biological and
Health Implications. Food for Health in the Pacific Rim. Food &
Nutrition Press, Inc.; 2008. p. 291301.
126. Grosvenor CE, Picciano MF, Baumrucker CR. Hormones and
growth-factors in milk. Endocr Rev. 1993;14(6):71028.
127. Pearlman WH. Hormones and tissue-growth factors in milkevo-
lutionary implications. Endocr Regul. 1991;25(12):49.
128. Waite R, Giraud A, Old J, Howlett M, Shaw G, Nicholas K, et al.
Cross-fostering in Macropus eugenii leads to increased weight but
not accelerated gastrointestinal maturation. J Exp Zool A Comp
Exp Biol. 2005;303(5):33144.
129. Schanbacher FL, Talhouk RS, Murray FA, Gherman LI, Willett
LB. Milk-borne bioactive peptides. Int Dairy J. 1998;8(56):393
403.
130. Daly KA, Digby MR, Lefévre C, Nicholas KR, Deane EM,
Williamson P. Identification, characterization and expression of
cathelicidin in the pouch young of tammar wallaby (Macropus
eugenii). Comp Biochem Physiol B Biochem Mol Biol.
2008;149(3):52433.
131. Rodriguez NA, Caplan MS. Oropharyngeal administration of
Mothers milk to prevent necrotizing enterocolitis in extremely
low-birth-weight infants: theoretical perspectives. J Perinat
Neonatal Nurs. 2015;29(1):8190.
132. Sullivan S, Schanler R, Kim J, Patel A, Trawöger R, Kiechl-
Kohlendorfer U, et al. An Exclusively Human Milk-Based Diet
Is Associated with a Lower Rate of Necrotizing Enterocolitis than
a Diet of Human Milk and Bovine Milk-Based Products. J Pediatr.
2009;In Press.
133. Yadav M. The transmissions of antibodies across the gut of pouch-
young marsupials. 1971;21(5).
134. Tschanz SA. Strukturelle Aspekte der prä- und postnatalen
Lungenentwicklung. Pneumologie. 2007;61(7):47889.
135. Mess AM, Ferner KJ. Evolution and development of gas exchange
structures in Mammalia: the placenta and the lung. Respir Physiol
Neurobiol. 2010;173(Suppl):S7482.
136. Gemmell RT, Little GJ. The structure of the lung of the newborn
marsupial bandicoot, Isoodon macrounus. Cell Tissue Res.
1982;223(2):44553.
137. Frappell PB, Mortola JP. Respiratory function in a newborn mar-
supial with skin gas exchange. Respir Physiol. 2000;120(1):35
45.
138. Szdzuy K, Zeller U, Renfree M, Tzschentke B, Janke O. Postnatal
lung and metabolic development in two marsupial and four euthe-
rian species. J Anat. 2008;212(2):16479.
139. Setchell PJ. The development of thermoregulation and thyroid
function in the marsupial Macropus eugenii (Desmarest). Comp
Biochem Physiol A Physiol. 1974;47(3):111521.
140. Buaboocha W, Gemmell RT. Thyroid-gland development in the
brushtail possum, trichosurus-vulpecula. Anatomical Record.
1995;243(2):25460.
141. Gemmell RT, Sernia C. The transfer of thyroxine from the mother
to the young of the marsupials, the bandicoot (Isoodon macrourus)
and the possum (Trichosurus vulpecula). Comp Biochem Physiol
A Physiol. 1992;103(3):5413.
J Mammary Gland Biol Neoplasia (2014) 19:289302 301
142. Modepalli V, Hinds L, Sharp J, Lefevre C, Nicholas K. Role of
marsupial tammar wallaby milk in lung maturation of pouch
young. BMC Dev Biol. 2015;15(1):16.
143. Weibel ER. What makes a good lung? The morphometric basis of
lung function. Swiss Med Wkly. 2009;139(2728):37586.
144. Weibel ER. Gas exchange: large surface and thin barrier determine
pulmonary diffusing capacity. Minerva Anestesiol. 1999;65(6):
37782.
145. Fraser J, Walls M, McGuire W. Respiratory complications of pre-
term birth. BMJ. 2004;329(7472):9625.
146. Simpson SJ, Flecknoe SJ, Clugston RD, Greer JJ, Hooper SB,
Frappell PB. Structural and functional development of the respi-
ratory system in a newborn marsupial with cutaneous gas ex-
change. Physiol Biochem Zool PBZ. 2011;84(6):63449.
147. Mortola JP, Frappell PB, Woolley PA. Breathing through skin in a
newborn mammal. Nature. 1999;397(6721):660.
148. Cloudsley-Thompson JL. The significance of cutaneous respira-
tion in Bufo regularis Reuss. Int J Biometeorol. 1970;14(4):3614.
149. Ucar A, Vafaizadeh V, Jarry H, Fiedler J, Klemmt PAB, Thum T,
et al. miR-212 and miR-132 are required for epithelial stromal
interactions necessary for mouse mammary gland development.
Nat Genet. 2010;42(12):1101U100.
150. Goldenberg RL, Rouse DJ. Medical progressPrevention of pre-
mature birth. New Engl J Med. 1998;339(5):31320.
151. Wang ML, Dorer DJ, Fleming MP, Catlin EA. Clinical outcomes
of near-term infants. Pediatrics. 2004;114(2):3726.
152. Petrou S. The economic consequences of preterm birth during the
first 10 years of life. Bjog-Int J Obstet Gy. 2005;112:105.
153. Beck S, Wojdyla D, Say L, Betran AP, Merialdi M, Requejo JH,
et al. The worldwide incidence of preterm birth: a systematic re-
view of maternal mortality and morbidity. B World Health Organ.
2010;88(1):318.
154. Escobar GJ, Clark RH, Greene JD. Short-term outcomes of infants
born at 35 and 36 weeks gestation: We need to ask more questions.
Semin Perinatol. 2006;30(1):2833.
155. Svedenkrans J, Henckel E, Kowalski J, Norman M, Bohlin K.
Long-term impact of preterm birth on exercise capacity in healthy
young Men: a national population-based cohort study. PLoS ONE.
2013;8(12):e80869.
302 J Mammary Gland Biol Neoplasia (2014) 19:289302
... This phase is followed by one of intermittent nursing, where infants are no longer attached to their mothers' nipples. Marsupial milk composition varies dramatically throughout lactation, with changes tailored according to infants' needs (Sharp et al. 2014). Twelve per cent (range: 5-24%) of the time between conception and weaning occurs between conception and birth, and 88% (76-95%) Fig. 1. ...
... Twelve per cent (range: 5-24%) of the time between conception and weaning occurs between conception and birth, and 88% (76-95%) Fig. 1. Duration of gestation, incubation, and lactation in three species that exhibit reproductive strategies representative of their respective mammalian lineages (modified from Sharp et al. 2014). of the time occurs between birth and weaning (data from 31 species, Hayssen 1985; Table 1, Fig. 1). Hence, lactation dominates early growth and development in marsupials. ...
... While some studies have investigated the effects of milk GCs on offspring in eutherian species, none has done so in monotremes or marsupials. The lactation period encompasses the majority of growth and development in these two mammalian lineages and, if Impacts of maternal stress on nursing offspring S. M. Stead, I. Bădescu and R. Boonstra maternal programming occurs, it is likely to occur then (Sharp et al. 2014). Monotremes and marsupials offer a more complete picture of the effects of milk GCs on offspring than eutherians, because milk supports a wider range of developmental processes in these lineages (Fanson & Parrott 2015). ...
Article
• A large body of research shows that maternal stress during an offspring’s early life can impact its phenotype in both the short and long term. In the Vertebrata, most research has been focused on maternal stress during the prenatal period. However, the postnatal period is particularly important in mammals because maternal milk provides a conduit by which maternal hormones secreted in response to stressors (glucocorticoids, GCs) can reach offspring. Moreover, lactation outlasts gestation in many species. • Though GCs were first detected in milk over 40 years ago, few studies have explored how they affect nursing offspring, and no reviews have been written on how maternal stress affects nursing offspring in the natural world. • We discuss the evolution of milk and highlight its importance in each of the three mammalian lineages: monotremes (subclass Monotremata), marsupials (infraclass Marsupialia), and eutherians (infraclass Placentalia). Most research on the effects of milk GCs on offspring has been focused on eutherians, but monotremes and marsupials rely on their mothers’ milk for a proportionally longer period of time, and so research on these taxa may yield more insight. • We show that GCs are important for milk production, both during an individual nursing bout and over the entire lactation period, and review evidence of GCs moving from maternal blood to milk, and eventually to nursing offspring. We examine evidence from rodents and primates of associations between GC levels in lactating females (either blood or milk) and offspring behaviour and growth rates. We discuss ways that maternal stress may impact these offspring phenotypes outside of milk GCs, such as changes to: (1) milk output, (2) other milk constituents (e.g. macronutrients, growth factors, cytokines), and (3) maternal care behaviour. • Critical to understanding the fitness impacts of elevated maternal GC levels during lactation is to place this within the context of the natural environment. Species-specific traits and natural histories will help us to understand why such maternal stress produces different offspring phenotypes that equip them to cope with and succeed in the environment they are about to enter.
... Breast milk contains essential macronutrients including lipids, carbohydrates, and proteins (Boquien, 2018;Pietrzak-Fiećko and Kamelska-Sadowska, 2020;Sharp et al., 2015). Lipids account for approximately 50% of the total energy in breast milk with a majority of lipids being triglycerides (Pietrzak-Fiećko and Kamelska-Sadowska, 2020;Yi and Kim, 2021). ...
... In addition to a complex mix of nutritive components, breast milk contains non-nutritive bioactive compounds that modulate offspring development, including hormones, growth factors, a complex microbiota, immune factors, HMOs, MEVs, and small and long non-coding RNAs (Eisha et al., 2022;Sharp et al., 2015;Vass et al., 2019). Important hormones found in breast milk include the anorexigenic hormone leptin that acts on the arcuate nucleus of the hypothalamus to decrease appetite and regulate energy expenditure, thus playing a significant role in hypothalamic pathway development in neonates (Savino et al., 2009;Vásquez-Garibay et al., 2019); the orexigenic hormone ghrelin, which acts as an antagonist to leptin, stimulates the arcuate nucleus to increase food intake and is implicated in long-term weight regulation (Vásquez-Garibay et al., 2019;Yiş et al., 2010); and adiponectin, which is involved in the metabolic regulation of fatty acids and conveys anti-inflammatory properties (Martin et al., 2006;Mohamad et al., 2018). ...
Article
The Developmental Origins of Health and Disease (DOHaD) hypothesis describes how maternal stress exposures experienced during critical periods of perinatal life are linked to altered developmental trajectories in offspring. Perinatal stress also induces changes in lactogenesis, milk volume, maternal care, and the nutritive and non-nutritive components of milk, affecting short and long-term developmental outcomes in offspring. For instance, selective early life stressors shape the contents of milk, including macro/micronutrients, immune components, microbiota, enzymes, hormones, milk-derived extracellular vesicles, and milk microRNAs. In this review, we highlight the contributions of parental lactation to offspring development by examining changes in the composition of breast milk in response to three well-characterized maternal stressors: nutritive stress, immune stress, and psychological stress. We discuss recent findings in human, animal, and in vitro models, their clinical relevance, study limitations, and potential therapeutic significance to improving human health and infant survival. We also discuss the benefits of enrichment methods and support tools that can be used to improve milk quality and volume as well as related developmental outcomes in offspring. Lastly, we use evidence-based primary literature to convey that even though select maternal stressors may modulate lactation biology (by influencing milk composition) depending on the severity and length of exposure, exclusive and/or prolonged milk feeding may attenuate the negative in utero effects of early life stressors and promote healthy developmental trajectories. Overall, scientific evidence supports lactation to be protective against nutritive and immune stressors, but the benefits of lactation in response to psychological stressors need further investigation.
... In this regard, marsupials may be especially valuable. In contrast to the situation in eutherian mammals (including humans), the majority of development in marsupial young occurs postnatally and is supported exclusively by milk bioactives (Nicholas et al. 2019;Sharp et al. 2014;Sharp et al. 2017). Further, in contrast to eutherian milk, which remains relatively unchanged in composition throughout lactation (Langer 2009), milk composition in both marsupial and monotreme mammals changes systematically throughout lactation. ...
... This provides a natural model system for identification of unique and conserved roles of milk bioactives in lactocrine programming of organ development, including the central nervous system. In marsupial pouch young both the brain and spinal cord develop rapidly in offspring during the first 100 days of milk consumption (Harrison and Porter 1992;Saunders et al. 1989;Sharp et al. 2014). Insights into the role of lactocrine-active milk bioactives in regulation of central nervous system development will benefit markedly by studies designed to exploit comparative methodologies. ...
Article
Full-text available
Human milk is a highly complex liquid food tailor-made to match an infant's needs. Beyond documented positive effects of breastfeeding on infant and maternal health, there is increasing evidence that milk constituents also impact child neurodevelopment. Non-nutrient milk bioactives would contribute to the (long-term) development of child cognition and behavior, a process termed 'Lactocrine Programming'. In this review we discuss the current state of the field on human milk composition and its links with child cognitive and behavioral development. To promote state-of-the-art methodologies and designs that facilitate data pooling and meta-analytic endeavors, we present detailed recommendations and best practices for future studies. Finally, we determine important scientific gaps that need to be filled to advance the field, and discuss innovative directions for future research. Unveiling the mechanisms underlying the links between human milk and child cognition and behavior will deepen our understanding of the broad functions of this complex liquid food, as well as provide necessary information for designing future interventions.
... These bacteria are transferred to the mammary gland and eventually into the milk for microbiome establishment in the neonate [3,12,13]. From an evolutionary perspective, the EMP and the composition of human milk (HM) are important due to certain bacterial species that undergo strong selection to initiate the colonization of the intestinal microbiota of the newborn for subsequent maturation [14]. ...
Article
Full-text available
The entero–mammary pathway is a specialized route that selectively translocates bacteria to the newborn’s gut, playing a crucial role in neonatal development. Previous studies report shared bacterial and archaeal taxa between human milk and neonatal intestine. However, the functional implications for neonatal development are not fully understood due to limited evidence. This study aimed to identify and characterize the microbiota and metabolome of human milk, mother, and infant stool samples using high-throughput DNA sequencing and FT-ICR MS methodology at delivery and 4 months post-partum. Twenty-one mothers and twenty-five infants were included in this study. Our results on bacterial composition suggest vertical transmission of bacteria through breastfeeding, with major changes occurring during the first 4 months of life. Metabolite chemical characterization sheds light on the growing complexity of the metabolites. Further data integration and network analysis disclosed the interactions between different bacteria and metabolites in the biological system as well as possible unknown pathways. Our findings suggest a shared bacteriome in breastfed mother–neonate pairs, influenced by maternal lifestyle and delivery conditions, serving as probiotic agents in infants for their healthy development. Also, the presence of food biomarkers in infants suggests their origin from breast milk, implying selective vertical transmission of these features.
... Leading health organizations such as the American Academy of Family Physicians, and the World Health Organization (WHO) recommend that infants be exclusively breastfed for about the first 6 months, and then combine continued breastfeeding with solid foods. Artificial baby milk (infant formula) should only be given to the baby if they have not been breastfed before one year of age [8][9][10][11][12]. ...
Article
Full-text available
Mothers’ awareness regarding the risk factors for the development of early childhood caries is crucial. The current study aims to examine the knowledge of breastfeeding mothers about their baby’s dental health and prevention of ECC while comparing primiparous mothers to multiparous mothers. A total of 165 mothers aged 20–49 y/o participated in the study by completing questionnaires that assessed the knowledge and attitudes of mothers toward their infants’ oral health. Results showed that (1) mothers were found to be highly knowledgeable regarding the oral hygiene of their infants and the recommended breastfeeding best practices (71%); (2) mothers with lower education showed poor knowledge regarding the recommended practices of infant oral health; (3) a large proportion of the mothers in the sample (62%) reported that they usually tasted the food before giving it to their baby, in a way they could transmit bacteria to infants; (4) most of the mothers (68%) were not aware that their dental health during pregnancy affects the infants’ dental health; and (5) multiparous mothers were more knowledgeable regarding artificial baby milk composition (96%) in comparison with mothers with only a single child (60%). According to the results, there is a need to improve the knowledge of breastfeeding mothers, especially mothers who have one child and mothers with a lower education about their baby’s dental health. The results of this study shed light on the knowledge of breastfeeding mothers on this important topic and could serve policymakers to improve practices toward advancing better oral health for infants, without sacrificing the benefits of breastfeeding, which are so crucial for infant health and development.
... They allow resources acquired by one individual in one time and place to be used by another individual in another time and place. Their components (Table 1) range from structural and nutritional building blocks such as fatty acids, amino acids and carbohydrates, to complex molecules like proteins, RNA or antibodies functioning as allohormones [15], all the way to fully functional cells [16][17][18][19][20][21][22]. Socially transferred materials can have long-lasting effects on the receiver, for example, affecting an individual's development or long-term health [23][24][25][26]. ...
Preprint
Full-text available
Since the dawn of life, transfers of metabolized material between individuals have led to great innovations of evolution. When metabolized material is transferred from one individual’s body to another (as with sperm, eggs, milk, symbionts), secondary manipulative molecules that induce a physiological response in the receiver are often transferred along with the primary cargo. The bioactive and transfer-supporting components in these socially transferred materials have evolved convergently to the point where they can be used in applications across taxa and type of transfer. Because these materials’ composition is typically highly dynamic and context-dependent, their focused study will allow deeper understanding of their transformative evolutionary and physiological role. We synthesize a conceptual framework for their study, and discuss future directions.
... One of the possible consequences for reduced human milk feeding among infants with CLP may be the negative impact on nutrition. Human milk has powerful health advantages which are important for protection against infections while the neonatal immune system completes its development (Sharp et al., 2014). According to the CDCP (2021), nutrition provided by human milk reduces the risk of comorbid conditions such as diabetes, lower respiratory infection, asthma, gastrointestinal infection, ear infection, and sudden infant death syndrome. ...
Article
Objective The objective of this study was to examine differences in human milk feeding outcomes among infants admitted to a neonatal intensive care unit (NICU) with and without cleft lip and palate (CLP). Method Data were used from a sample of infants admitted to the NICU with and without CLP from the 2018 National Vital Statistics System. For baseline comparisons, Chi-square tests of independence were used to compare categorical variables, and independent samples t tests were used for continuous variables. Logistic regression models were performed to determine the odds of human milk feeding at discharge in infants admitted to the NICU with CLP. Results The total sample included 345,429 infants admitted to the NICU, of which 660 had CLP. Significant differences were found among the following variables when baseline comparisons were made between infants admitted to the NICU with and without CLP: mother's race, mother's education, maternal smoking record, childbirth delivery method, presence of maternal pre-pregnancy diabetes, five-minute APGAR score, multiparity record (having more than one baby at birth), gestational age, and gestational weight. After controlling for baseline differences, results indicated reduced odds of human milk feeding at discharge in infants admitted to the NICU with CLP compared to those without CLP (OR = .543; 95% CI.455,.648). Conclusion Results suggest reduced odds of human milk feeding at discharge among infants admitted to the NICU with CLP compared to those without CLP. These findings emphasize the necessity of awareness and facilitation of human milk feeding in this population.
... Human milk is well-recognised as the optimal food for the newborn due to its comprehensive nutrient composition, immune components, anti-infective factors and metabolic enzymes, which are required for growth and development during early life [1]. The significant nutrients available in human milk include vitamins, minerals, amino acids and essential proteins. ...
Article
Full-text available
Human milk is recommended by the World Health Organization (WHO) for the general well-being of infants. However, many mothers face an insufficient milk supply to breastfeed their children. Galactagogue, in particular, plant galactagogue, serves as a method to promote lactation. This in-depth review examines the evidence supporting different plants' galactagogic activity through clinical studies around the globe. A scoping review approach was adopted to establish the research questions, and define the findings, selection and analysis of the study. This scoping review highlights and compiles the clinical research performed globally involving plant galactagogue to better inform the medical practitioners, lactation consultants, nursing mothers, communities and relevant personnel on practicing, guidelines, policymaking and research. In general, a total of 1041 research publications were retrieved from different global bibliographic databases, of which only 13 articles were retained for analysis after applying the exclusion criteria. A total of 14 types of plants have undergone clinical studies in the past decade to verify their galactagogic activity. All but two showed a positive effect on promoting milk production. There were 42 articles categorised as excluded studies. The category includes review articles, surveys, case reports, introductory articles of regional plant galactagogue and preclinical studies, which involves animal testing and the studies exploring other issues related to plant galactagogue. The findings demonstrate that there is a significant research gap on the plant galactagogue using clinical studies. More clinical research is necessary to identify and verify the efficacy of various types of plant galactagogue for the benefit of humankind.
Article
Objective The purpose of this study was to identify infant and maternal factors associated with reduced human milk feeding among infants in a neonatal intensive care unit (NICU) with cleft lip with or without cleft palate (CL ± P). Method Data collected on mothers and infants admitted to the NICU with CL ± P from the 2018 National Vital Statistics System were used for this study. Chi-square tests of independence and independent-samples t tests were used to compare categorical variables and continuous variables, respectively, among two groups of infants admitted to the NICU with CL ± P—those who did and did not receive human milk feeding at discharge. Results The sample included 660 infants admitted to the NICU with CL ± P, of which 353 received human milk at discharge. Significant differences were found between the two groups for marital status, mother's education, maternal smoking record, total number of prenatal visits, multiparity record, gestational age, birth weight, and use of assisted ventilation. Conclusions Results indicated that, as a function of human milk feeding at discharge, mothers and their infants admitted to the NICU with CL ± P exhibited differences across infant and maternal factors. These findings further our understanding of this sample of mothers and infants with CL ± P while potentially identifying determinants to human milk feeding. This study provides insight into infant and maternal characteristics that may be associated with barriers to human milk feeding.
Book
Marsupials differ from most other mammals in their method of reproduction, in that they have chosen, in an evolutionary sense, to develop lactation rather than placentation for the nurture of their young. The neonate is therefore born with a mixture of advanced and embryonic characters, and yet is readily accessible within the pouch, providing a unique system for the study of the ontogeny of various physiological and endocrinological parameters. Marsupials are therefore ideal animals for research into mammalian reproductive physiology. The results of this exciting new research are summarized in this book by two of the foremost workers in the field. Individual chapters analyse the genetic and hormonal control of sexual differentiation, male and female reproductive structures and their functions, the role of the corpus luteum in the oestrous cycle and pregnancy, the hormonal control of embryonic diapause and the role of the marsupial placenta in the development of the embryo. This book is more than just a straightforward review of marsupial reproduction for its detailed analyses and broad comparative coverage will attract mammalogists and reproductive physiologists with a wide range of research interests.
Book
Over the past half a century research has revealed that marsupials – far from being ‘second class’ mammals – have adaptations for particular ways of life quite equal to their placental counterparts. Despite long separate evolution, there are extraordinary similarities in which marsupials have solved the challenges of living in such environments as deserts, alpine snowfields or tropical rainforests. Some can live on grass, some on pollen and others on leaves; some can glide, some can swim and others hop with extraordinary efficiency. In Life of Marsupials, one of the world’s leading experts explores the biology and evolution of this unusual group – with their extraordinary diversity of forms around the world – in Australia, New Guinea and South America. Joint winner of the 2005 Whitley Medal. Included in Choice Magazine's 2006 Outstanding Academic Titles list.
Article
The evolutionary origin of the mammary gland has been difficult to establish because little knowledge can be gained on the origin of soft tissue organs from fossil evidence. One approach to resolve the origin of lactation has compared the anatomy of existing primitive mammals to skin glands, whilst another has examined the metabolic and molecular synergy between mammary gland development and the innate immune system. We have reviewed the physiology of lactation in five mammalian species with special reference to these theories. In all species, milk fulfils dual functions of providing protection and nutrition to the young and, furthermore, within species the quality and quantity of milk are highly conserved despite maternal malnutrition or illness. There are vast differences in birth weight, milk production, feeding frequency, macronutrient concentration, growth rate and length of lactation between rabbits, quokkas (Setonix brachyurus), pigs, cattle and humans. The components that protect the neonate against infection do so without causing inflammation. Many protective components are not unique to the mammary gland and are shared with the innate immune system. In contrast, many of the macronutrients in milk are unique to the mammary gland, have evolved from components of the innate immune system, and have either retained or developed multiple functions including the provision of nourishment and protection of the hatchling/neonate. Thus, there is a strong argument to suggest that the mammary gland evolved from the inflammatory response; however, the extensive protection that has developed in milk to actively avoid triggering inflammation seems to be a contradiction.
Book
Of all food products dairy foods have the most potential concerning functional foods. Therefore, there is a tremendous amount of interest in value-added milk products and the identification of components in food which have health benefits. Bioactive Components of Milk provides an overview of these derived components and their diverse activities including: the stimulation of beneficial microflora, alerting the immune system to the presence of potential pathogens and allergens, binding and eliminating toxins, etc. The book is divided into four parts. The first part focuses on bioactive milk lipid components, which very widely among mammalian species. The second part describes different aspects of biological active colostrums and milk proteins and their derivatives, with special concern on species specific effects. The third part reviews the production of recombinant human proteins in the milk of livestock animals - including ethical issues - and the aims of altering milk composition for the benefit of both the animals themselves and the consumers. The final part focuses on the influence of ruminants nutrition on the biological activity of milk.
Article
OBJECTIVE: To analyse preterm birth rates worldwide to assess the incidence of this public health problem, map the regional distribution of preterm births and gain insight into existing assessment strategies. METHODS: Data on preterm birth rates worldwide were extracted during a previous systematic review of published and unpublished data on maternal mortality and morbidity reported between 1997 and 2002. Those data were supplemented through a complementary search covering the period 2003-2007. Region-specific multiple regression models were used to estimate the preterm birth rates for countries with no data. FINDINGS: We estimated that in 2005, 12.9 million births, or 9.6% of all births worldwide, were preterm. Approximately 11 million (85%) of these preterm births were concentrated in Africa and Asia, while about 0.5 million occurred in each of Europe and North America (excluding Mexico) and 0.9 million in Latin America and the Caribbean. The highest rates of preterm birth were in Africa and North America (11.9% and 10.6% of all births, respectively), and the lowest were in Europe (6.2%). CONCLUSION: Preterm birth is an important perinatal health problem across the globe. Developing countries, especially those in Africa and southern Asia, incur the highest burden in terms of absolute numbers, although a high rate is also observed in North America. A better understanding of the causes of preterm birth and improved estimates of the incidence of preterm birth at the country level are needed to improve access to effective obstetric and neonatal care.
Chapter
Abstract Introduction Natural Bioactive Peptides in Milk EGF Bioactive Peptides Derived from Milk Proteins Conclusions Acknowlegments
Article
Human milk contains a wide variety of proteins that contribute to its unique qualities. Many of these proteins are digested and provide a well-balanced source of amino acids to rapidly growing infants. Some proteins, such as bile salt–stimulated lipase, amylase, β-casein, lactoferrin, haptocorrin, and α1-antitrypsin, assist in the digestion and utilization of micronutrients and macronutrients from the milk. Several proteins with antimicrobial activity, such as immunoglobulins, κ-casein, lysozyme, lactoferrin, haptocorrin, α-lactalbumin, and lactoperoxidase, are relatively resistant against proteolysis in the gastrointestinal tract and may, in intact or partially digested form, contribute to the defense of breastfed infants against pathogenic bacteria and viruses. Prebiotic activity, such as the promotion of the growth of beneficial bacteria such as Lactobacilli and Bifidobacteria, may also be provided by human milk proteins. This type of activity can limit the growth of several pathogens by decreasing intestinal pH. Some proteins and peptides have immunomodulatory activities (eg, cytokines and lactoferrin), whereas others (eg, insulin-like growth factor, epidermal growth factor, and lactoferrin) are likely to be involved in the development of the intestinal mucosa and other organs of newborns. In combination, breast-milk proteins assist in providing adequate nutrition to breastfed infants while simultaneously aiding in the defense against infection and facilitating optimal development of important physiologic functions in newborns.