ArticlePDF Available

Detection and Quantification of Immunoregulatory miRNAs in Human Milk and Infant Milk Formula

Authors:

Abstract and Figures

Mammary gland secretory cells produce miRNA-rich milk. In humans, these miRNAs reach infant/neonate bloodstream, playing diverse roles, like neural system development, metabolism, and immune system maturation. Notwithstanding, still few works explore human milk miRNA content, and there are no reports at the population level. Our hypothesis was that miR-146b-5p, miR148a-3p, miR155-5p, mir181a-5p, and mir200a-3p immunoregulatory miRNAs are expressed in human colostrum/milk at a higher level than infant milk formulae. The aim of this work was to evaluate the expression of the five immunoregulatory miRNAs in human milk and compare it with their expression in infant milk formula. For this purpose, miRNA relative expression was measured by qPCR in cDNA prepared from total RNA extracted from sixty human colostrum/milk samples and six different formulae. The comparative Cт method 2−ΔCт using exogenous cel-miR-39 as internal control was employed, followed by statistical analysis. We found the relative expression levels of miRNAs are comparable among colostrum/milk samples, and these miRNAs are present in infant milk formulae but at very low concentrations. We conclude that the relative expression of the immunomodulatory miRNAs is comparable in all the human colostrum/milk samples and is higher than the expression in formulae.
Content may be subject to copyright.
Citation: Vélez-Ixta, J.M.; Benítez-
Guerrero, T.; Aguilera-Hernández, A.;
Martínez-Corona, H.; Corona-
Cervantes, K.; Juárez-Castelán, C.J.;
Rangel-Calvillo, M.N.; García-Mena,
J. Detection and Quantification of
Immunoregulatory miRNAs in
Human Milk and Infant Milk
Formula. BioTech 2022,11, 11.
https://doi.org/10.3390/biotech
11020011
Academic Editor: Maria Teresa Di
Martino
Received: 16 December 2021
Accepted: 18 March 2022
Published: 20 April 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Article
Detection and Quantification of Immunoregulatory miRNAs in
Human Milk and Infant Milk Formula
Juan Manuel Vélez-Ixta 1, Tizziani Benítez-Guerrero 1, Arlene Aguilera-Hernández 1, Helga Martínez-Corona 1,
Karina Corona-Cervantes 1, Carmen Josefina Juárez-Castelán1, Martín NoéRangel-Calvillo 2
and Jaime García-Mena 1, *
1Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del
Instituto Politécnico Nacional (Cinvestav), Av. Instituto Politécnico Nacional 2508,
Ciudad de México 07360, Mexico;
juan.velez@cinvestav.mx (J.M.V.-I.); tizziani.benitez@cinvestav.mx (T.B.-G.);
arlene.aguilera@cinvestav.mx (A.A.-H.); helga.martinez@cinvestav.mx (H.M.-C.);
karina.corona@cinvestav.mx (K.C.-C.); carmen.juarez@cinvestav.mx (C.J.J.-C.)
2Hospital General “Dr. JoséMaría Rodríguez”, Instituto de Salud del Estado de Mexico,
Ecatepec de Morelos 55200, Estado de México, Mexico; drrangelcalvillo@gmail.com
*Correspondence: jgmena@cinvestav.mx; Tel.: +52-5747-3800 (ext. 5328)
Abstract:
Mammary gland secretory cells produce miRNA-rich milk. In humans, these miRNAs reach
infant/neonate bloodstream, playing diverse roles, like neural system development, metabolism,
and immune system maturation. Notwithstanding, still few works explore human milk miRNA
content, and there are no reports at the population level. Our hypothesis was that miR-146b-5p,
miR148a-3p, miR155-5p, mir181a-5p, and mir200a-3p immunoregulatory miRNAs are expressed
in human colostrum/milk at a higher level than infant milk formulae. The aim of this work was
to evaluate the expression of the five immunoregulatory miRNAs in human milk and compare it
with their expression in infant milk formula. For this purpose, miRNA relative expression was
measured by qPCR in cDNA prepared from total RNA extracted from sixty human colostrum/milk
samples and six different formulae. The comparative C
т
method 2
Cт
using exogenous cel-miR-39
as internal control was employed, followed by statistical analysis. We found the relative expression
levels of miRNAs are comparable among colostrum/milk samples, and these miRNAs are present in
infant milk formulae but at very low concentrations. We conclude that the relative expression of the
immunomodulatory miRNAs is comparable in all the human colostrum/milk samples and is higher
than the expression in formulae.
Keywords: human milk; qPCR; microRNA; RNA; formula milk; breastfeeding
1. Introduction
According to the Mexican national survey ENSANUT [
1
], the panorama of breast-
feeding in Mexican women 12–49 years old with children up to 24 months of age is not
favorable since only 28.3% of children under six months are exclusively breastfed. It is
common that children under 12 months of age are fed infant formula, and just a small
portion of children corresponding to 29% of the studied population was breastfed up to
two years of age. Therefore, these data allow us to conclude that there is still a deficiency in
breastfeeding practices in Mexico that must be tackled and managed from a collaborative
perspective [2].
MicroRNAs (miRNAs) are short 19–24 nucleotides non-coding RNAs that derive from
characteristic hairpin precursors. They constitute one of the more abundant classes of
gene-regulatory molecules, suppressing the expression profiles of protein-coding genes
at the post-transcriptional level [
3
,
4
]. miRNAs have been described in human milk (HM)
as unique bioactive components. During lactogenesis, milk particles are produced by
secretory epithelial cells located in the alveoli of the mammary gland and are secreted in
BioTech 2022,11, 11. https://doi.org/10.3390/biotech11020011 https://www.mdpi.com/journal/biotech
BioTech 2022,11, 11 2 of 9
vesicles into the lumen [
5
]. The vesicles carrying miRNAs are taken up by the infant during
feeding. Later, they are transported and assimilated by the cells to perform their function.
Colostrum has an important immune function due to its high immunoglobulins and
bioactive components content [
6
]. In the human body, miRNAs are present in extracellular
fluids like saliva, urine, plasma, and milk [
7
]. HM contains macronutrients, micronutrients,
bioactive compounds, growth factors, and immunological factors [8].
In recent years, the study of bioactive components, for example, miRNAs, immunoglob-
ulins, maternal cells, among others, has been of great interest. There is solid evidence of the
presence of miRNAs in HM, in addition, they have been found expressed in milk of other
species like goat, cow, and pig [
9
]. In HM, miRNAs remain stable since they are associated
with extracellular vesicles, fat globules, and cellular components. There is evidence that
miRNAs play an important role in establishing the neonate’s immune system and have
other beneficial effects on infant health [9].
Exosome-derived miRNAs, such as miR-148a, induce cell proliferation and protein
expression in normal colon epithelial cells [
10
]. Moreover, miR-148a promotes CD70 and
LFA-1 gene overexpression through hypomethylation. These genes are associated with cell
survival and T-cell activation, respectively [
11
]. In a study done using six-month mature
HM, high content of miR-181 and miR-155 was found. These miRNAs are involved in
the differentiation of B cells [
12
]. Examples of other highly enriched miRNAs in HM are
miR-17 and miR-92, which have functions, such as regulation of monocyte development,
differentiation of B cells and T cells [
13
]. In another report, the presence of miRNAs with
important immunological functions, for example, miR-148a-3p, miR-30b-5p, miR-182-5p,
and miR-200a-3p, was demonstrated in exosomes derived from HM [
14
]. Another group
showed the presence of miR-21, miR-16, and miR-146b-5p in pre-term HM [15].
The presence of miRNAs has been established in cow’s milk based infant milk formula
(IMF) by next-generation sequencing and quantitative real-time PCR. In these products,
the miRNA content is generally lower than HM [
7
]. Since cow’s milk is the most frequently
used component of infant formulas, the same group reported that around 90% of miRNAs
found in human milk are also found in cow and goat’s milk. On the other hand, previous
studies did not detect abundant miRNA content in IMFs [
16
]. The aim of this work was to
explore the expression of five different immunoregulatory miRNAs in Mexican HM and
compare it with the content in IMF.
2. Materials and Methods
2.1. Type of Study
Descriptive cross-sectional study of five selected miRNA expression in human milk
and infant milk formula (IMF).
2.2. Sample
Human colostrum/milk (HCM) samples were selected from a collection of samples
of a previous study made in the public hospital “Dr. JoséMaría Rodríguez” located in
Ecatepec de Morelos, State of Mexico (19_36 03500N, 99_ 303600W), and kept at
70
C
until used. The protocol was approved by the Ethics Committee of the General Hospital
(Project identification code: 217B560002018006) [
17
]. The inclusion criteria were: Healthy
women between 0 to 7 postpartum days, of Mexican origin for at least two generations, who
gave birth by vaginal delivery or non-elective C-section with 37–41 weeks of gestational age.
The exclusion criteria were: Smoking, probiotic or alcohol consumption, being affected by
diabetes, overweight, or obesity, and using antibiotics during the last trimester of pregnancy.
Infant milk formulae (IMF) were selected among the most consumed products in Mexico
City (Table S1). IMF were reconstituted with nuclease-free water (Sigma-Aldrich
®
, St. Louis,
MO, USA, Cat. W4502-1L) following the manufacturer’s instruction.
BioTech 2022,11, 11 3 of 9
2.3. Oligonucleotide Selection and Design
For this research, miR-146b-5p, miR148a-3p, miR155-5p, mir181a-5p, and mir200a-3p
were selected based on literature search in PubMed portal (https://pubmed.ncbi.nlm.nih.
gov/, accessed on 15 August 2020), including original and review articles from years 2000 to
2020, searching for the terms “miRNAs, human milk, milk formula and immunoregulatory
miRNAs”. Raw sequences were obtained from miRBase (www.mirbase.org/, accessed
on 15 August 2020). Stem-loop primers for cDNA synthesis and primers for qPCR were
designed using CLC Workbench v.8.1.3 software (https://www.qiagen.com/, accessed on
15 August 2020) based on a previous report [18] (Table S2).
2.4. RNA Extraction
RNA was extracted from 1 mL HCM or IMF, respectively. Samples were centrifugated
at max speed, for 10 min, at 4
C in Neofuge 13R Centrifuge (HealForce
®
, Shanghai, China).
Cellular pellet and lipid phase were washed with cold PBS pH 7.4. Subsequently, total RNA
was extracted with mirVana
miRNA Isolation Kit (Invitrogen
, Cat. AM1560, Carlsbad,
CA, USA), following manufacturer instructions. 3
µ
L of exogenous miRNA (33 fmol)
cel-miR-39 (microRNA (cel-miR-39) Spike-In Kit, Cat. 59000, Thorold ON, Canada) were
added to each sample before lysis buffer. RNA was eluted in 100
µ
L volume and stored
at
20
C. RNA concentration and purity were assessed with NanoDrop
2000 (Thermo
Fisher Scientific, Cat. ND-2000, Waltham, MA, USA).
2.5. cDNA Synthesis
cDNA synthesis was made using 5–50 ng extracted RNA in 15
µ
L reaction volume,
using MicroRNA Reverse Transcription Kit (Applied Biosystems
, Cat. 4366596, Waltham,
MA, USA), following manufacturer’s instructions. Individual cDNA reactions were pre-
pared for each miRNA evaluated for each sample. Reactions were placed in Thermocycler
2720 (Applied Biosystems, Waltham, MA, USA) for 30 min at 16
C; 30 min, 42
C; 5 min,
85 C; and 10 min, 4 C.
2.6. miRNA Quantification by qPCR
qPCR was made using 1
µ
L of cDNA and Maxima SYBR Green/ROX qPCR Master
Mix (Thermo Scientific
, Cat. K0222, Waltham, MA, USA) reagents in a final volume of
25
µ
L, as described by the manufacturer. Reactions were placed on PCR plates (Applied
Biosystems
, Cat. 4375816, Waltham, MA, USA) and qPCR run in StepOne
Real-Time
PCR System Thermocycler (Applied Biosystems
, Cat. 4376357, Waltham, MA, USA)
according to the two-step cycling protocol: Denaturation of 95
C for 10 min; 40 cycles of
95
C for 15 s, 60
C for 60 s and finally, a melt curve of 95
C for 15 s, 60
C for 60 s, and
95 C for 15 s. Quantitation was made in triplicate.
2.7. Bioinformatics and Statistical Analyses
Data obtained from this work were analyzed using R environment [
19
] and Rstu-
dio 1.4.1717 software [
20
]. The R-libraries employed were “readr” [
21
], “dplyr” [
22
],
“scales” [
23
], and “tidyr” [
24
]. Cтwere calculated by Step One Software v2.2.2 (Applied
Biosystems
, Waltham, Massachusetts, USA) and exported to comma separated values
(.csv) format. Afterwards, the comparative Cтmethod 2
Cт
[
25
] was used for statistical
analysis. Internal control for this study was cel-miR-39. One way Analysis of Variance
(ANOVA) was performed to seek differences in relative miRNA levels in HCM. Kruskal–
Wallis one-way analysis of variance was carried out to seek for differences in miRNA levels
at different postpartum days. Further, Mann–Whitney U test (Wilcoxon rank-sum test)
was used to seek differences in relative miRNA levels between both groups (HCM and
IMF). Tables were elaborated with R and exported to html for inclusion in this work using
‘sjPlot’ library [
26
]. Plots were elaborated with ‘ggplot2’ library [
27
] and exported to Joint
Photographic Experts Group format (.jpg) to 600 dpi/ppi resolution.
BioTech 2022,11, 11 4 of 9
3. Results
3.1. miRNA Relative Expression Is Comparable among the Studied Human Milk Samples
We studied the expression of five different immuno-related miRNAs in human colostrum/
milk samples collected from 60 women in a period of 0 to 7 postpartum days. Women
were healthy, normal weight, around 23 years-old, mostly with vaginal delivery (Table 1).
Samples were collected in a period of three months from 16 October 2017 to 29 January
2018 (Figure S1). Total RNA was extracted, and miR-146b-5p, miR148a-3p, miR155-5p,
mir181a-5p, mir200a-3p were quantified as described in Materials and Methods. Results for
mir181a-5p were discarded due to technical problems with the primer FmiR181 (Table S2),
which amplified a spurious product in the qPCR reaction for the non-template control. The
results for miR-146b-5p, miR148a-3p, miR155-5p, and mir200a-3p indicated that the relative
expression for all of them was comparable, as shown by the one-way ANOVA statistical
test of the relative expression units (2
Cт
) (Pr(>F) = 0.7136) (Figure 1). There is a tendency
of higher miRNAs relative expression at postpartum day-3 (Figure S2). We also evaluated
whether there was an association between miRNA relative expression and mother age but
found no association in our data (Figure S3).
Table 1. Data for 60 participant mothers in the study.
Anthropometric Data Mean SD
Age (years) 22.93 ±4.95
Height (m) α1.57 ±0.07
Weight (Kg) β56.48 ±10.01
BMI γ23.22 ±3.74
Type of delivery n %
Elective caesarean 4 6.67
Non elective caesarean 13 21.67
Vaginal 43 71.66
Parity n %
Primiparous δ16 27.12
Multiparous δ43 72.88
Milk extraction n %
Manual ε34 82.93
Pump ε7 17.07
Socioeconomic data n %
Residence Mexico City (192501000 N 99080440 0 O) 15 25.00
Estado de México (19210150 0 N 99370510 0 O) 36 60.00
Guerrero State (173604700 N 99570000 0 O) 2 3.33
Hidalgo State (20280420 0 N 98510490 0 O) 1 1.67
Oaxaca State (16530530 0 N 96240510 0 O) 3 5.00
Puebla State (19000130 0 N 97530180 0 O) 2 3.33
Veracruz State (192600500 N 96220590 0 O) 1 1.67
Activity Housewife δ52 88.14
Student δ2 3.39
General employee δ5 8.47
Education Primary school (6 years) 20 33.33%
Secondary school (3 years) 36 60.00%
High school (3 years) 1 1.67%
University school (4–5 years) 3 5.00%
α
55 mothers;
β
50 mothers;
γ
48 mothers;
δ
59 mothers;
ε
41 mothers. BMI = weight (kg)/[height (m)]
2
, n,
indicates number of data for the category. Anthropometric data correspond to the start of pregnancy.
BioTech 2022,11, 11 5 of 9
BioTech 2021, 10, x FOR PEER REVIEW 5 of 10
Education Primary school (6 years) 20 33.33%
Secondary school (3 years) 36 60.00%
High school (3 years) 1 1.67%
University school (4–5 years) 3 5.00%
α 55 mothers; β 50 mothers; γ 48 mothers; δ 59 mothers; ε 41 mothers. BMI = weight (Kg)/[height
(m)]2, n, indicates number of data for the category. Anthropometric data correspond to the start of
pregnancy.
Figure 1. Expression of selected miRNAs in human colostrum/milk. hsa-miR146 (miR-146b-5p); hsa-
miR-148 (miR148a-3p), hsa-miR-155 (miR155-5p), hsa-miR-200 (mir200a-3p). The Y-axis shows the
relative expression of each miRNA with respect to the internal control cel-miR-39, and the X-axis
shows the corresponding miRNA. Each dot in the plot represents a sample. The double horizontal
lines indicate the standard error of the mean. One-way ANOVA statistical test shows no significant
differences in miRNA levels, Pr(>F) = 0.7136.
3.2. Selected miRNAs Were Identified in Infant Milk Formulae
Next, we studied the relative expression of miR-146b-5p, miR-148a-3p, miR-155-5p,
mir-181a-5p, mir-200a-3p in six different infant milk formulae (IMF) available in the local
market (Table S1). Total RNA was extracted after reconstituting the powder, as instructed
by the manufacturer in triplicate. miR-146b-5p, miR-148a-3p, miR-155-5p, mir-181a-5p,
and miR-200a-3p miRNAs were quantified as described in Materials and Methods. As
mentioned before, the results for miR-181a-5p were discarded due to technical problems
with the primer FmiR181 (Table S2). The results for miR-146b-5p, miR-148a-3p, miR-155-
5p, and mir-200a-3p indicated that the relative expression of miR-146b-5p and miR-155-
5p was higher in IMF than the expression of hsa-miR-148 and hsa-miR-200. In addition,
the Mann-Whitney U (Wilcoxon rank-sum) test indicated that the relative expression of
miRNAs was higher in the human milk samples in comparison to the IMF (p = 9.0 × 107
for miR-146; p = 1.1 × 107 for miR-148; p = 9.3 × 103 for miR-155, and p = 8.0 × 106 for mir-
200) (Figure 2).
Figure 1.
Expression of selected miRNAs in human colostrum/milk. hsa-miR146 (miR-146b-5p);
hsa-miR-148 (miR148a-3p), hsa-miR-155 (miR155-5p), hsa-miR-200 (mir200a-3p). The Y-axis shows
the relative expression of each miRNA with respect to the internal control cel-miR-39, and the X-axis
shows the corresponding miRNA. Each dot in the plot represents a sample. The double horizontal
lines indicate the standard error of the mean. One-way ANOVA statistical test shows no significant
differences in miRNA levels, Pr(>F) = 0.7136.
3.2. Selected miRNAs Were Identified in Infant Milk Formulae
Next, we studied the relative expression of miR-146b-5p, miR-148a-3p, miR-155-5p,
mir-181a-5p, mir-200a-3p in six different infant milk formulae (IMF) available in the local
market (Table S1). Total RNA was extracted after reconstituting the powder, as instructed
by the manufacturer in triplicate. miR-146b-5p, miR-148a-3p, miR-155-5p, mir-181a-5p,
and miR-200a-3p miRNAs were quantified as described in Materials and Methods. As
mentioned before, the results for miR-181a-5p were discarded due to technical problems
with the primer FmiR181 (Table S2). The results for miR-146b-5p, miR-148a-3p, miR-155-5p,
and mir-200a-3p indicated that the relative expression of miR-146b-5p and miR-155-5p was
higher in IMF than the expression of hsa-miR-148 and hsa-miR-200. In addition, the Mann-
Whitney U (Wilcoxon rank-sum) test indicated that the relative expression of miRNAs was
higher in the human milk samples in comparison to the IMF (p= 9.0
×
10
7
for miR-146;
p= 1.1
×
10
7
for miR-148; p= 9.3
×
10
3
for miR-155, and p= 8.0
×
10
6
for mir-200)
(Figure 2).
BioTech 2022,11, 11 6 of 9
BioTech 2021, 10, x FOR PEER REVIEW 6 of 10
Figure 2. Comparative expression of selected hsa-miRs in colostrum/milk (n = 60) with respect to
milk formula (n = 18). The graphics show the relative expression of each miRNA with respect to the
internal control cel-miR-39. (A) hsa-miR-146, (B) hsa-miR-148, (C) hsa-miR-155, (D) hsa-miR-200.
The Y-axis shows the relative expression of each miRNA, and the X-axis shows the type of sample.
Each dot in the plot represents a sample. The double horizontal lines indicate the standard error of
the mean. Mann-Whitney U (Wilcoxon rank-sum) test was made. ** indicates p < 0.01 and ***
indicates p < 0.001.
4. Discussion
miRNAs play an important role in the regulation of gene expression and contribute
to the pathogenesis of complex diseases, such as IBD [28]. Several studies have revealed
that levels of certain miRNAs are altered in IBD patients in comparison to healthy
individuals, such as miR-124, miR-320, miR-21, miR-31, and miR-141 [29]. Further, work
done on Crohn’s disease suggests that dysregulation of miRNAs at the level of the
intestinal mucosa may play an important role in the early stages of the disease [30].
Studies in human milk (HM) have shown evidence of the presence of miRNAs, where
they remain stable, even under very low pH conditions. This supports the notion that
miRNAs can remain stable in the acidic conditions of the gastrointestinal tract and
consequently, are able to be absorbed in the gut. Many miRNAs present in HM have been
described, some with unknown function, but others with immunoregulatory function.
Among the miRNAs found in a study of HM using massive sequencing, miR-146b-5p,
miR-200a-3p, and miR-148-3p which were measured in our work, were found among the
10 most abundant [31]. A similar expression of miR-148a-3p, miR-146b-5p, and miR-
200a/c-3p has been reported in different studies discussed in a recent systematic review,
indistinctly of factors like the fraction of HM, lactational age, and health status of the
mother and her offspring [32].
The miRNA expression after birth has been monitored. The expression of miRNA-
146b, analyzed in the lipid and skimmed fraction of HM, was reported at relatively stable
Figure 2.
Comparative expression of selected hsa-miRs in colostrum/milk (n = 60) with respect to
milk formula (n = 18). The graphics show the relative expression of each miRNA with respect to the
internal control cel-miR-39. (
A
) hsa-miR-146, (
B
) hsa-miR-148, (
C
) hsa-miR-155, (
D
) hsa-miR-200. The
Y-axis shows the relative expression of each miRNA, and the X-axis shows the type of sample. Each
dot in the plot represents a sample. The double horizontal lines indicate the standard error of the
mean. Mann-Whitney U (Wilcoxon rank-sum) test was made. ** indicates p< 0.01 and *** indicates
p< 0.001.
4. Discussion
miRNAs play an important role in the regulation of gene expression and contribute to
the pathogenesis of complex diseases, such as IBD [
28
]. Several studies have revealed that
levels of certain miRNAs are altered in IBD patients in comparison to healthy individuals,
such as miR-124, miR-320, miR-21, miR-31, and miR-141 [
29
]. Further, work done on
Crohn’s disease suggests that dysregulation of miRNAs at the level of the intestinal mucosa
may play an important role in the early stages of the disease [30].
Studies in human milk (HM) have shown evidence of the presence of miRNAs, where
they remain stable, even under very low pH conditions. This supports the notion that
miRNAs can remain stable in the acidic conditions of the gastrointestinal tract and con-
sequently, are able to be absorbed in the gut. Many miRNAs present in HM have been
described, some with unknown function, but others with immunoregulatory function.
Among the miRNAs found in a study of HM using massive sequencing, miR-146b-5p,
miR-200a-3p, and miR-148-3p which were measured in our work, were found among the 10
most abundant [
31
]. A similar expression of miR-148a-3p, miR-146b-5p, and miR-200a/c-3p
has been reported in different studies discussed in a recent systematic review, indistinctly
of factors like the fraction of HM, lactational age, and health status of the mother and her
offspring [32].
The miRNA expression after birth has been monitored. The expression of miRNA-146b,
analyzed in the lipid and skimmed fraction of HM, was reported at relatively stable levels
from the first to the second month after delivery [
15
]. In another work evaluating a different
group of miRNAs, the expression of miR-146b-5p is reported stable in samples obtained
BioTech 2022,11, 11 7 of 9
during two, four, and six months after birth [
33
]. On the other hand, miR-148a suffered
a slight decline in its expression in late lactation. Additionally, a differential expression
was found between women with normal weight and women who are overweight or
obese. A decrease of 30% was registered in obesity with respect to the healthy group
during the first month of lactation. This decrease was not observed two months later,
where significant differences in the expression of this miRNA among the groups were not
found [
34
]. However, in another report, differences in the expression of miR-148 in skim
milk (0 to 1 month postpartum) were not observed [
35
]. In another study, the presence of
miR-200a-3p was reported consistently and highly expressed in all human milk samples
evaluated [
36
]. Regarding variations in miRNA expression during the day, miR-146b has
shown little variation in its expression [15].
It is noteworthy that studied miRNAs could be found in IMF despite the processing
involved in their production, however, similar results, yielding lower miRNA in IMF with
respect to HCM have been previously reported [
7
,
37
], despite the fact that miRNAs are
known to be stable and resistant to different adverse conditions, like abrupt temperature
and pH changes and endonuclease activity [
31
,
38
]. It is also important to remark that HCM
samples used in this study were collected in late 2017 and early 2018, so they had been stored
for at least three years at
70
C, while the IMF were prepared and processed immediately
after purchase. To this point, no reports have been found discussing the stability of miRNAs
over prolonged periods of time, and although the long storage time of the samples could
be a limitation of this work, it is interesting to note that there is a higher expression of
the miRNAs studied in HCM, despite the age of the samples. Moreover, consistent with
these results, another study focused on miRNA presence in infant gastric content and
found that miRNA profile of gastric content of formula-fed infants was lower than that of
breastfed infants, nevertheless, their presence indicates miRNA stability and transfer [
39
].
The limitations of this work are the small number of different immunoregulatory miRNAs
and IMF that were monitored.
5. Conclusions
The relative expression of the four immunoregulatory miRNAs analyzed in this study
is comparable in all the human colostrum/milk samples analyzed. The same miRNAs
are not abundantly expressed in the infant milk formulae studied in this work. The
expression of the immunomodulatory miRNAs is higher in human colostrum/milk than
the expression in infant milk formulae. This observation supports the importance of
the human breastfeeding practice. For instance, further studies analyzing human milk
miRNA profile characterization by high-throughput sequencing could be useful. Human
milk miRNA expression could be related with different factors, like breastfeeding period,
mother’s age and weight, for this reason, appropriate studies are needed. Moreover, it is
an important matter to compare human miRNA profile with different health conditions
present in pregnancy, e.g., obesity, metabolic syndrome, and gestational diabetes.
Supplementary Materials:
The following are available online at https://www.mdpi.com/article/
10.3390/biotech11020011/s1, Figure S1. Time frame graphic for sample procurement in the study,
Figure S2. Expression of selected hsa-miRs in colostrum/milk at different time, Figure S3. Expression
of miRNAs in colostrum/milk v.s. the age of mothers. Table S1. Nutrimental information of Infant
Milk Formulae, and Table S2. Primer sequences used in this study.
Author Contributions:
Conceptualization, J.M.V.-I., M.N.R.-C. and J.G.-M.; methodology, J.M.V.-I.,
T.B.-G. and A.A.-H.; validation, J.M.V.-I., T.B.-G. and A.A.-H.; formal analysis, J.M.V.-I.; investigation,
J.M.V.-I., T.B.-G., A.A.-H., H.M.-C., K.C.-C., C.J.J.-C., M.N.R.-C. and J.G.-M.; resources, M.N.R.-C. and
J.G.-M.; data curation, J.M.V.-I., T.B.-G., A.A.-H. and K.C.-C.; writing—original draft preparation,
J.M.V.-I., T.B.-G. and J.G.-M.; writing—review and editing, J.M.V.-I., T.B.-G., A.A.-H., H.M.-C., K.C.-C.,
C.J.J.-C., M.N.R.-C., J.G.-M.; visualization, J.M.V.-I.; supervision, C.J.J.-C., M.N.R.-C. and J.G.-M.;
project administration, M.N.R.-C. and J.G.-M.; funding acquisition, M.N.R.-C. and J.G.-M. All authors
have read and agreed to the published version of the manuscript.
BioTech 2022,11, 11 8 of 9
Funding:
This research was funded by Fondo SEP-Cinvestav-2018-174; and CONACyT FORDECYT-
PRONACES/6669/2020.
Institutional Review Board Statement:
The study was conducted according to the guidelines of the
Declaration of Helsinki, and approved by the Ethics Committee of the General Hospital (Project
identification code: 217B560002018006).
Informed Consent Statement:
Informed consent was obtained from all subjects involved in the
study.
Data Availability Statement: Not applicable.
Acknowledgments:
We express our gratitude to Alberto Piña-Escobedo, Carolina Miranda-Brito,
Rodrigo García-Gutiérrez for support in the laboratory work; to Viridiana Rosas-Ocegueda and
Alma Lemus-Hernández for administrative assistance. We thank the CONACyT M. Sc. Fellowship
for J.M.V.-I. (997152), A.A.-H. (911623), H.M.-C. (1078225); Ph. D. Fellowship for T.B.-G. (635676),
K.C.-C. (777953), and Post-Doctoral Fellowship for C.J.J.-C. (321600). J.G.-M (19815) is Fellow from
the Sistema Nacional de Investigadores (SNI), Mexico.
Conflicts of Interest:
The authors declare no conflict of interest. The funders had no role in the design
of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or
in the decision to publish the results.
References
1.
Vista de Situación de las Prácticas de Lactancia Materna y Alimentación Complementaria en México: Resultados de la Ensanut
2018-19. Available online: https://www.saludpublica.mx/index.php/spm/article/view/11567/11972 (accessed on 6 December
2021).
2.
De Cosío-Martínez, T.G.; Hernández-Cordero, S.; Rivera-Dommarco, J.; Hernández-Ávila, M. Recomendaciones para una política
nacional de promoción de la lactancia materna en México: Postura de la Academia Nacional de Medicina. TT—Recommendations
for a multisectorial national policy to promote breastfeeding in Mexico: Position of the Nationa. Salud Publica Mex.
2017
,59,
106–113. [CrossRef] [PubMed]
3.
Friedman, R.C.; Farh, K.K.H.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome
Res. 2009,19, 92–105. [CrossRef] [PubMed]
4.
Shin, S.; Jung, Y.; Uhm, H.; Song, M.; Son, S.; Goo, J.; Jeong, C.; Song, J.J.; Kim, V.N.; Hohng, S. Quantification of purified
endogenous miRNAs with high sensitivity and specificity. Nat. Commun. 2020,11, 6033. [CrossRef]
5.
Bauman, D.E.; Mather, I.H.; Wall, R.J.; Lock, A.L. Major advances associated with the biosynthesis of milk. J. Dairy Sci.
2006
,89,
1235–1243. [CrossRef]
6. Mosca, F.; Giannì, M.L. Human milk: Composition and health benefits. Pediatr. Med. Chir. 2017,39, 155. [CrossRef] [PubMed]
7.
Golan-Gerstl, R.; Elbaum Shiff, Y.; Moshayoff, V.; Schecter, D.; Leshkowitz, D.; Reif, S. Characterization and biological function of
milk-derived miRNAs. Mol. Nutr. Food Res. 2017,61, 1700009. [CrossRef] [PubMed]
8.
Ballard, O.; Morrow, A.L. Human milk composition: Nutrients and bioactive factors. Pediatr. Clin. N. Am.
2013
,60, 49–74.
[CrossRef] [PubMed]
9.
Carrillo-Lozano, E.; Sebastián-Valles, F.; Knott-Torcal, C. Circulating micrornas in breast milk and their potential impact on the
infant. Nutrients 2020,12, 3066. [CrossRef]
10.
Reif, S.; Elbaum Shiff, Y.; Golan-Gerstl, R. Milk-derived exosomes (MDEs) have a different biological effect on normal fetal colon
epithelial cells compared to colon tumor cells in a miRNA-dependent manner. J. Transl. Med.
2019
,17, 325. [CrossRef] [PubMed]
11.
Pan, W.; Zhu, S.; Yuan, M.; Cui, H.; Wang, L.; Luo, X.; Li, J.; Zhou, H.; Tang, Y.; Shen, N. MicroRNA-21 and microRNA-148a
contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1. J.
Immunol. 2010,184, 6773–6781. [CrossRef]
12.
Kosaka, N.; Izumi, H.; Sekine, K.; Ochiya, T. microRNA as a new immune-regulatory agent in breast milk. Silence
2010
,1, 7.
[CrossRef] [PubMed]
13.
Ventura, A.; Young, A.G.; Winslow, M.M.; Lintault, L.; Meissner, A.; Erkeland, S.J.; Newman, J.; Bronson, R.T.; Crowley, D.; Stone,
J.R.; et al. Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters. Cell
2008,132, 875–886. [CrossRef] [PubMed]
14.
Zhou, Q.; Li, M.; Wang, X.; Li, Q.; Wang, T.; Zhu, Q.; Zhou, X.; Wang, X.; Gao, X.; Li, X. Immune-related microRNAs are abundant
in breast milk exosomes. Int. J. Biol. Sci. 2012,8, 118–123. [CrossRef] [PubMed]
15.
Floris, I.; Billard, H.; Boquien, C.Y.; Joram-Gauvard, E.; Simon, L.; Legrand, A.; Boscher, C.; Rozé, J.C.; Bolaños-Jiménez, F.; Kaeffer,
B. MiRNA analysis by quantitative PCR in preterm human breast milk reveals daily fluctuations of hsa-miR-16-5p. PLoS ONE
2015,10, e0140488. [CrossRef]
16.
Leiferman, A.; Shu, J.; Upadhyaya, B.; Cui, J.; Zempleni, J. Storage of Extracellular Vesicles in Human Milk, and MicroRNA
Profiles in Human Milk Exosomes and Infant Formulas. J. Pediatr. Gastroenterol. Nutr. 2019,69, 235–238. [CrossRef]
BioTech 2022,11, 11 9 of 9
17.
García-González, I.; Corona-Cervantes, K.; Hernández-Quiroz, F.; Villalobos-Flores, L.E.; Galván-Rodríguez, F.; Romano, M.C.;
Miranda-Brito, C.; Piña-Escobedo, A.; Borquez-Arreortúa, F.G.; Rangel-Calvillo, M.N.; et al. The Effect of Holder Pasteurization on
the Diversity of the Human Milk Bacterial Microbiota Using High-Throughput DNA Sequencing. J. Hum. Lact.
2021
. [CrossRef]
18. Kramer, M.F. Stem-loop RT-qPCR for miRNAs. Curr. Protoc. Mol. Biol. 2011,95, 15.10.1–15.10.15. [CrossRef]
19.
R Core Team. R: A Language and Environment for Statistical Computing; R Core Team: Vienna, Austria, 2021. Available online:
https://www.R-project.org/ (accessed on 1 December 2021).
20.
RStudio Team. RStudio: Integrated Development Environment for R. 2021. Available online: https://www.rstudio.com (accessed
on 1 December 2021).
21.
Wickham, H.; Hester, J. Readr: Read Rectangular Text Data. 2020. Available online: https://readr.tidyverse.org/ (accessed on 1
December 2021).
22.
Wickham, H.; François, R.; Henry, L.; Müller, K. Dplyr: A Grammar of Data Manipulation. 2021. Available online: https:
//dplyr.tidyverse.org// (accessed on 1 December 2021).
23.
Wickham, H.; Seidel, D. Scales: Scale Functions for Visualization. 2020. Available online: https://scales.r-lib.org/ (accessed on 1
December 2021).
24. Wickham, H. Tidyr: Tidy Messy Data. 2021. Available online: https://tidyr.tidyverse.org/ (accessed on 1 December 2021).
25.
Schmittgen, T.D.; Livak, K.J. Analyzing real-time PCR data by the comparative CT method. Nat. Protoc.
2008
,3, 1101–1108.
[CrossRef]
26.
Lüdecke, D. sjPlot: Data Visualization for Statistics in Social Science. 2021. Available online: https://strengejacke.github.io/sjPlot/
(accessed on 1 December 2021).
27. Wickham, H. ggplot2: Elegant Graphics for Data Analysis; Springer: New York, NY, USA, 2016; ISBN 978-3-319-24277-4.
28.
Thomas, J.P.; Ölbei, M.; Brooks-Warburton, J.; Korcsmaros, T.; Modos, D. Analysing miRNA-Target Gene Networks in Inflamma-
tory Bowel Disease and Other Complex Diseases Using Transcriptomic Data. Genes 2022,2022, 370. [CrossRef]
29.
Soroosh, A.; Koutsioumpa, M.; Pothoulakis, C.; Iliopoulos, D. Functional role and therapeutic targeting of microRNAs in
inflammatory bowel disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2018,314, G256. [CrossRef]
30.
Cao, B.; Zhou, X.; Ma, J.; Zhou, W.; Yang, W.; Fan, D.; Hong, L. Role of MiRNAs in Inflammatory Bowel Disease. Dig. Dis. Sci.
2017,62, 1426–1438. [CrossRef] [PubMed]
31.
Liao, Y.; Du, X.; Li, J.; Lönnerdal, B. Human milk exosomes and their microRNAs survive digestion
in vitro
and are taken up by
human intestinal cells. Mol. Nutr. Food Res. 2017,61, 1700082. [CrossRef] [PubMed]
32.
Tingö, L.; Ahlberg, E.; Johansson, L.; Pedersen, S.A.; Chawla, K.; Sætrom, P.; Cione, E.; Simpson, M.R. Non-Coding RNAs in
Human Breast Milk: A Systematic Review. Front. Immunol. 2021,12, 725323. [CrossRef] [PubMed]
33.
Alsaweed, M.; Lai, C.T.; Hartmann, P.E.; Geddes, D.T.; Kakulas, F. Human milk miRNAs primarily originate from the mammary
gland resulting in unique miRNA profiles of fractionated milk. Sci. Rep. 2016,6, 20680. [CrossRef]
34.
Shah, K.B.; Chernausek, S.D.; Garman, L.D.; Pezant, N.P.; Plows, J.F.; Kharoud, H.K.; Demerath, E.W.; Fields, D.A. Human
Milk Exosomal MicroRNA: Associations with Maternal Overweight/Obesity and Infant Body Composition at 1 Month of Life.
Nutrients 2021,13, 1091. [CrossRef]
35.
Shiff, Y.E.; Reif, S.; Marom, R.; Shiff, K.; Reifen, R.; Golan-Gerstl, R. MiRNA-320a is less expressed and miRNA-148a more
expressed in preterm human milk compared to term human milk. J. Funct. Foods 2019,57, 68–74. [CrossRef]
36.
Simpson, M.R.; Brede, G.; Johansen, J.; Johnsen, R.; Storrø, O.; Sætrom, P.; Øien, T. Human Breast Milk miRNA, Maternal Probiotic
Supplementation and Atopic Dermatitis in Offspring. PLoS ONE 2015,10, e0143496. [CrossRef]
37.
Chen, X.; Gao, C.; Li, H.; Huang, L.; Sun, Q.; Dong, Y.; Tian, C.; Gao, S.; Dong, H.; Guan, D.; et al. Identification and
characterization of microRNAs in raw milk during different periods of lactation, commercial fluid, and powdered milk products.
Cell Res. 2010,20, 1128–1137. [CrossRef]
38.
Tomé-Carneiro, J.; Fernández-Alonso, N.; Tomás-Zapico, C.; Visioli, F.; Iglesias-Gutierrez, E.; Dávalos, A. Breast milk microRNAs
harsh journey towards potential effects in infant development and maturation. Lipid encapsulation can help. Pharmacol. Res.
2018,132, 21–32. [CrossRef]
39.
Kakimoto, Y.; Matsushima, Y.; Tanaka, M.; Hayashi, H.; Wang, T.; Yokoyama, K.; Ochiai, E.; Osawa, M. MicroRNA profiling of
gastric content from breast-fed and formula-fed infants to estimate last feeding: A pilot study. Abbreviations BM Breast milk FM
Formula milk GC Gastric content GCB GC of breast-fed infant GCF GC of formula-fed infant GJ Gastric juice. Int. J. Leg. Med.
2020,134, 903–909. [CrossRef]
... MicroRNAs (miRNAs) are a group of regulatory RNA species capable of modulating the activation of certain mRNA targets to influence a wide range of biological processes. Segments of mRNA consist of short nucleotide sequences, between 19 and 24, and are derived from hairpin precursors [3]. The biogenesis of miRNAs is controlled by RNA polymerase II to generate primary microRNAs, which are modified by various proteins from the RNAse III family to produce shorter microRNAs. ...
... Recently the presence of other small RNA species in body fluids has been reported, including circular RNAs (circRNAS), long non-coding RNAs, transfer RNAs (tRNAs), and small nuclear RNAs (snRNAs) [3,4]. The role of these non-coding RNA species in breast milk and other fluids remains poorly understood and is the subject of ongoing research on human breast milk. ...
... Velez-Ixta, 2022 (Mexico) [3] To assess the expression of immunoregulatory microRNAs in human breast milk. ...
Article
Full-text available
MicroRNAs are non-coding segments of RNA involved in the epigenetic modulation of various biological processes. Their occurrence in biological fluids, such as blood, saliva, tears, and breast milk, has drawn attention to their potential influence on health and disease development. Hundreds of microRNAs have been isolated from breast milk, yet the evidence on their function remains inconsistent and inconclusive. The rationale for the current scoping review is to map the evidence on the occurrence, characterization techniques, and functional roles of microRNAs in breast milk. The review of the sources of this evidence highlights the need to address methodological challenges to achieve future advances in understanding microRNAs in breast milk, particularly their role in conditions such as neoplasms. Nonetheless, remarkable progress has been made in characterizing the microRNA profiles of human breast milk.
... In recent years, special attention has been paid to the effect of the microRNA (miR) of BM on the health of newborn babies. MicroRNAs are small non-coding RNA molecules containing 18-25 nucleotides [3,4]. Most BM miRs are contained in extracellular vesicles, which contributes to their absorption in the child's digestive tract [5]. ...
... Each miR regulates the expression of several genes. Thereby, miRs influence metabolism, differentiation, and development processes in cells of various tissues; and immune system maturation [3,4,7,8]. MiR-148a is a representative of the highly concentrated miR pool of BM. According to some authors, the share of miR-148a varies from 12 to 35.79 % of the miR transcriptome of BM [5,[9][10][11]. ...
Article
Full-text available
Background. Breast milk (BM) is an optimal nutritional product for newborns and a source of exogenous microRNAs (miR). MiR-148a is one of the most highly expressed miR of BM. Currently, there is a lack of data on the miR-148a effect on the development of necrotizing enterocolitis (NEC) in premature newborns. The purpose of the study was to determine the influence of miR-148a of the mother’s BM on the risk of NEC development in preterm newborns. Materials and methods. We examined 74 newborns, who were treated in neonatal departments. We determined the level of miR-148a in the BM of 44 mothers of exclusively breastfed children. In parallel, we evaluated gene expression of the transcription factor T-bet in scrapings of the buccal mucosa of all the newborns. Three groups for comparison were selected: group 1 consisted of 32 newborns up to 37 weeks of gestation on breastfeeding (BF); group 2 — of 30 preterm newborns on artificial feeding; the control group — 12 full-term newborns on BF. Results. The gestational age median of group 1 children was 33 (31; 34) weeks; group 2 — 32.5 (32; 35) weeks; and it was comparatively higher in the control group (p
... Moreover, immune-related pre-miRNAs are also abundant in human milk [48]. While some studies described similar levels of miRNAs in colostrum and mature milk [49], some show lower miRNA levels in mature milk compared to transition milk (collected 4-5 days after delivery) [50]. However, it has been unequivocally indicated that infant formulas, compared to breast milk, have a much lower content of the most common immune-related dairy miRNAs (miR-125b-5p, miR-146b-5p, miR-148a-3p, miR-155-5p, miR-181a-5p, miR-200a-3p) [49,50]. ...
... While some studies described similar levels of miRNAs in colostrum and mature milk [49], some show lower miRNA levels in mature milk compared to transition milk (collected 4-5 days after delivery) [50]. However, it has been unequivocally indicated that infant formulas, compared to breast milk, have a much lower content of the most common immune-related dairy miRNAs (miR-125b-5p, miR-146b-5p, miR-148a-3p, miR-155-5p, miR-181a-5p, miR-200a-3p) [49,50]. ...
Article
Full-text available
MicroRNAs (miRNAs), that is, short non-coding RNA molecules, have been found in different common foods, like fruits and vegetables, meat and its products, milk (including human breast milk) and dairy products, as well as honey and herbs. Moreover, they are isolated from supernatants from cultures of various mammalian cells. A growing amount of evidence appears to support the idea of using miRNAs as therapeutics. One possible and promising route of administration is oral, which is considered noninvasive and well-tolerated by patients. Association with extracellular vesicles (EVs), nanoparticles, RNA-binding proteins, lipoproteins, or lipid derivatives, protects miRNAs from an unfavorable gastrointestinal environment (including salivary and pancreatic RNases, low pH in the stomach, digestive enzymes, peristaltic activity and microbial enzymes). Such protection likely favors miRNA absorption from the digestive tract. Internalization of miRNA by gastric and intestinal cells as well as effects on the gut microbiota by orally delivered miRNA have recently been described. Furthermore, gene regulation by orally administered miRNAs and their immunomodulatory properties indicate the possibility of cross-species or cross-kingdom communication through miRNA. In addition to the local effects, these molecules may enter the circulatory system and reach distant tissues, and thus cell-free nucleic acids are promising candidates for future selective treatments of various diseases. Nonetheless, different limitations of such a therapy imply a number of questions for detailed investigation.
... A recent Mexican investigation that aimed to compare the expression of five immuneregulatory miRNAs in HM samples of full-term mothers with formula concluded that the expression of the immunomodulatory miRNAs was similar in all the human colostrum and mature milk samples and was higher than in formula milk [48]. MiRNAs are nowadays deeper studied since they are associated with immune system compounds, including TGFβ, T cell receptor, Toll-like receptor signaling, Janus kinase-signal transducer and activator of transcription (JAK-STAT), and Th1 and Th2 cells differentiation. ...
Article
Full-text available
Our narrative review focuses on colostrum components, particularly those that influence the neonatal immune system of newborns. Colostrum is secreted in small volumes by the alveolar cells of the breast during the first two to five days after birth. Colostrum is poor in fat and carbohydrates, with larger protein and bioactive compounds than mature milk. It plays a crucial role in driving neonates’ immunity, transferring those immunological factors which help the correct development of the neonatal immune system and support establishing a healthy gut microbiome. The newborn has an innate and adaptive immune system deficiency, with a consequent increase in infection susceptibility. In particular, neonates born prematurely have reduced immunological competencies due to an earlier break in the maternal trans-placenta transfer of bioactive components, such as maternal IgG antibodies. Moreover, during pregnancy, starting from the second trimester, maternal immune cells are conveyed to the fetus and persist in small quantities post-natal, whereby this transfer is known as microchimerism (MMc). Thus, preterm newborns are deficient in this maternal heritage, and have their own immune system under-developed, but colostrum can compensate for the lack. Early breastfeeding, which should be strongly encouraged in mothers of preterm and full-term babies, provides those immunomodulant compounds that can act as a support, allowing the newborn to face immune needs, including fronting infections and establishing tolerance. Moreover, making mothers aware that administering colostrum helps their infants in building a healthy immune system is beneficial to sustain them in the difficult post-partum period.
... 71 miRNAs have been detected in cow milk-based formula, but at very low concentrations relative to human POM. 70,177 ...
Article
Full-text available
Plain Language Summary Before they begin eating solid foods, infants may be fed a variety of nutritional sources such as breast milk (“parent’s own milk”, fed directly at the breast or pumped and fed from a bottle), commercial infant formula, or sterilized “donor human milk” from a certified milk bank. Early nutrition affects the infant’s gut microbiome (bacteria living in the intestinal tract), development of their immune system, and their health throughout life. However, it is unclear how different forms of early nutrition affect these important processes. Parent’s own milk contains compounds that support gut microbes and stimulate development of the infant immune system, changing over time to meet infant needs. For many of these compounds, donor human milk contains a lesser amount, and formula contains even less – or none at all. Some compounds are also affected by pumping and storing parents’ own milk. This review highlights how differences among these nutritional sources influence gene expression and gut development to shape the infant microbiome and immune system. Current evidence shows that parent’s own milk, fed at the breast, offers unique benefits that are not replicated by other forms of early nutrition. This review also outlines how early life nutrition research can help us understand human development and develop new ways to provide the best possible start to life for all infants.
Article
Full-text available
Background. Patent ductus arteriosus (PDA) is a serious problem in clinical neonatology and pediatrics. Today treatment of PDA remains debatable because of high risk of complications with medical and invasive treatment. The purpose of the study was to determine the dependence of PDA closure on the type of feeding the newborn. Materials and methods. We have made a retrospective analysis of 300 case histories of children in neonatal units. We selected two comparison groups: the main group consisted of breastfed babies born at 37 weeks gestation (n = 102); controls — preterm infants who received artificial feeding since birth (n = 198). Results. At discharge from the hospital, 44 (14.7 ± 2.0 %) children had PDA (p < 0.05). The percentage of infants on artificial feeding with PDA was 2.3 times higher compared to breastfed babies: 18.2 ± 2.7 % versus 7.8 ± 2.7 % (p = 0.017 by χ2 test). Respiratory distress syndrome was more common in the control group as well: 72.7 ± 3.2 % versus 46.1 ± 4.9 % (p < 0.001). Additionally, the group of formula-fed children more often had severe complications of respiratory distress syndrome: respiratory failure type III — 25.8 ± 3.1 % versus12.7 ± 3.3 % (р = 0.009); development of bronchopulmonary dysplasia — 13.1 ± 2.4 % versus 2.9 ± 1.7 % (p = 0.005). The need for invasive mechanical ventilation in the intensive care unit was observed much less often in breastfed children. Moreover, the level of respiratory disorders (type II respiratory failure) in both groups differed insignificantly (p = 0.742): 24.5 ± 4.3 % in the main group and 26.3 ± 3.1 % in controls. Conclusions. Breastfeeding from the first days of life contributes to the closure of the ductus arteriosus and the favorable course of respiratory distress syndrome in premature babies.
Article
Full-text available
Patients with inflammatory bowel disease (IBD) are known to have perturbations in microRNA (miRNA) levels as well as altered miRNA regulation. Although experimental methods have provided initial insights into the functional consequences that may arise due to these changes, researchers are increasingly utilising novel bioinformatics approaches to further dissect the role of miRNAs in IBD. The recent exponential increase in transcriptomics datasets provides an excellent opportunity to further explore the role of miRNAs in IBD pathogenesis. To effectively understand miRNA-target gene interactions from gene expression data, multiple database resources are required, which have become available in recent years. In this technical note, we provide a step-by-step protocol for utilising these state-of-the-art resources, as well as systems biology approaches to understand the role of miRNAs in complex disease pathogenesis. We demonstrate through a case study example how to combine the resulting miRNA-target gene networks with transcriptomics data to find potential disease-specific miRNA regulators and miRNA-target genes in Crohn’s disease. This approach could help to identify miRNAs that may have important disease-modifying effects in IBD and other complex disorders, and facilitate the discovery of novel therapeutic targets.
Article
Full-text available
Breast milk is the primary source of nutrition and hydration for the newborn infant but also plays an important role in the child’s first immune defense. Additionally, several breast milk factors have been implicated in immune-related health outcomes later in life, including immunoglobulins, cytokines, chemokines, growth factors and, more recently, non-coding RNA (ncRNA) species. In this systematic review, we provide a comprehensive summary of the current literature on endogenous ncRNAs found in human breast milk. Thirty (30) relevant studies were identified and, whilst the majority studies focused on microRNAs (miRNAs), there is evidence that breast milk contains high quantities of RNA which also include long-coding RNAs, circular RNAs, as well as other short RNAs and fragmented tRNA and rRNAs. Among studies investigating miRNAs, miR-148a-3p, miR-30a/d-5p, miR-22-3p, miR-146b-5p, miR-200a/c-3p, and the 5p end of the let-7 miRNAs were commonly reported among the top 10 miRNAs in the cell, lipid, and skim milk fractions of breast milk. Methodological difference and small sample sizes limit the possibility of conclusively identifying which maternal and infant characteristics affect the miRNA profile. The highly expressed miRNAs were generally reported to be similar across lactational stage, milk fraction, maternal and infant characteristics, or infant growth and health. All the same, individual studies identify potential differences in miRNA expression levels which should be confirmed by future studies. Stability, uptake, and physiological functions of miRNAs were also considered in several studies. Breast milk miRNAs are relatively resistant to a range of harsh conditions and uptake experiments suggest that extracellular vesicles containing miRNAs and circular RNAs can be taken up by intestinal epithelial cells. Although the evidence regarding the functional effect of breast milk miRNAs is limited, the predicted functions range from metabolic and biosynthetic processes to signaling pathways, cellular adhesion, communication, growth, and differentiation. Finally, this systematic review highlights some of the methodological challenges and knowledge gaps which can help direct future research in this field. In particular, it is important to further investigate the bioavailability of miRNAs in different milk fractions, and to characterize other ncRNAs which are largely unstudied. Systematic Review Registration PROSPERO https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=138989, identifier CRD42020138989.
Article
Full-text available
Among all the body fluids, breast milk is one of the richest sources of microRNAs (miRNAs). MiRNAs packaged within the milk exosomes are bioavailable to breastfeeding infants. The role of miRNAs in determining infant growth and the impact of maternal overweight/obesity on human milk (HM) miRNAs is poorly understood. The objectives of this study were to examine the impact of maternal overweight/obesity on select miRNAs (miR-148a, miR-30b, miR-29a, miR-29b, miR-let-7a and miR-32) involved in adipogenesis and glucose metabolism and to examine the relationship of these miRNAs with measures of infant body composition in the first 6 months of life. Milk samples were collected from a cohort of 60 mothers (30 normal-weight [NW] and 30 overweight [OW]/obese [OB]) at 1-month and a subset of 48 of these at 3 months of lactation. Relative abundance of miRNA was determined using real-time PCR. The associations between the miRNAs of interest and infant weight and body composition at one, three, and six months were examined after adjusting for infant gestational age, birth weight, and sex. The abundance of miR-148a and miR-30b was lower by 30% and 42%, respectively, in the OW/OB group than in the NW group at 1 month. miR-148a was negatively associated with infant weight, fat mass, and fat free mass, while miR-30b was positively associated with infant weight, percent body fat, and fat mass at 1 month. Maternal obesity is negatively associated with the content of select miRNAs in human milk. An association of specific miRNAs with infant body composition was observed during the first month of life, suggesting a potential role in the infant’s adaptation to enteral nutrition.
Article
Full-text available
MicroRNAs (miRNAs) are short (19–24 nt) non-coding RNAs that suppress the expression of protein coding genes at the post-transcriptional level. Differential expression profiles of miRNAs across a range of diseases have emerged as powerful biomarkers, making a reliable yet rapid profiling technique for miRNAs potentially essential in clinics. Here, we report an amplification-free multi-color single-molecule imaging technique that can profile purified endogenous miRNAs with high sensitivity, specificity, and reliability. Compared to previously reported techniques, our technique can discriminate single base mismatches and single-nucleotide 3′-tailing with low false positive rates regardless of their positions on miRNA. By preloading probes in Thermus thermophilus Argonaute (TtAgo), miRNAs detection speed is accelerated by more than 20 times. Finally, by utilizing the well-conserved linearity between single-molecule spot numbers and the target miRNA concentrations, the absolute average copy numbers of endogenous miRNA species in a single cell can be estimated. Thus our technique, Ago-FISH (Argonaute-based Fluorescence In Situ Hybridization), provides a reliable way to accurately profile various endogenous miRNAs on a single miRNA sensing chip.
Article
Full-text available
MicroRNAs (MiRNAs) are small RNA molecules that can exert regulatory functions in gene expression. MiRNAs have been identified in diverse tissues and biological fluids, both in the context of health and disease. Breastfeeding has been widely recognized for its superior nutritional benefits; however, a number of bioactive compounds have been found to transcend these well-documented nutritional contributions. Breast milk was identified as a rich source of miRNAs. There has been increasing interest about their potential ability to transfer to the offspring as well as what their specific involvement is within the benefits of breast milk in the infant. In comparison to breast milk, formula milk lacks many of the benefits of breastfeeding, which is thought to be a result of the absence of some of these bioactive compounds. In recent years, the miRNA profile of breast milk has been widely studied, along with the possible transfer mechanisms throughout the infant’s digestive tract and the role of miRNA-modulated genes and their potential protective and regulatory functions. Nonetheless, to date, the current evidence is not consistent, as many methodological limitations have been identified; hence, discrepancies exits about the biological functions of miRNAs. Further research is needed to provide thorough knowledge in this field.
Article
Full-text available
Recently, we were consulted about a challenging case, where an infant died by poisoning and the drug-dependent mother insisted that she unintentionally gave the toxic drug through breast milk. Accordingly, we investigated the utility of immunoblotting and microRNA (miRNA) profiling of the infant’s gastric content (GC) to differentiate between breast-feeding and formula-feeding. As a pilot study, we sampled the GC from breast-fed (GCB) and formula-fed (GCF) infants, as well as gastric juice (GJ) from fasted adults at autopsy. Breast milk (BM) samples were collected from volunteers within 1 year post-delivery. By immunoblotting, lactoferrin and gross cystic disease fluid protein (GCDEP) were clearly detected in BM, but could not be detected in GCB. Similarly, β-lactoglobulin was detected in formula milk, but could not be detected in GCF. Meanwhile, miRNA sequencing revealed that the miRNA expression profile of GCB was closer to BM than GCF and GJ. Especially, miR-151a and miR-186 were more abundant in BM and GCB than in GCF and GJ. Our study is the first to elucidate the human GJ miRNA profile and demonstrate the possibility that miR-151a and miR-186 in GC may be the biomarker of breast-feeding.
Article
Full-text available
Background: Breastfeeding is the ideal source of infant nutrition. Human milk consists not only of nutrients but also biologically active components. Among these latter compounds, exosomes contain proteins, lipids, mRNAs and miRNAs. Methods: To elucidate the biological effects of milk-derived exosomes (MDEs) on normal colonic epithelial cells compared to colonic tumor cells, we incubated cells with MDEs. MDEs were able to enter into normal and tumor cells and change their miRNA expression profiles. Proliferation, cell morphology and protein expression were analyzed in these cells. Results: Human milk-derived exosomes induced proliferation- and epithelial mesenchymal transformation-related changes, such as collagen type I and twist expression, in normal but not in tumor cells. PTEN, a target of miRNA-148a, was downregulated in normal but not in tumor cells following incubation with MDEs. Moreover, miRNA-148a-3p knockdown cells were used to demonstrate the importance of miRNA in the effect of exosomes on cell proliferation and protein expression. MDEs inhibited proliferation and DNMT1 expression in cells with knockdown of miRNA-148a. Conclusions: In conclusion, the positive effect of exosomes on normal cells without affecting tumor cells may presents an aspect of their safety when considering it use as a nutritional supplement to infant formula.
Article
Background Human milk is the best food for infants; however, when breastfeeding is not possible, pasteurized milk from human milk banks is the best alternative. Little has been reported about variations in the bacterial microbiota composition of human milk after pasteurization. Research aim To characterize and compare the bacterial microbiota composition and diversity within human milk among Mexican mothers before and after the Holder pasteurization process. Methods A cross-sectional, observational, and comparative design was used. The effect of the pasteurization process on the bacterial composition and diversity of human milk samples of donors ( N = 42) from a public milk bank was assessed before and after pasteurization by high throughput deoxyribonucleic acid sequencing of V3-16S rRNA gene libraries. Sequencing data were examined using the Quantitative Insights into Microbial Ecology software and Phyloseq in R environment. Results A varied community of bacteria was found in both raw and pasteurized human milk. The bacterial diversity of the milk samples was increased by the pasteurization, where some thermoduric bacteria of the phyla Proteobacteria, Firmicutes, and Actinobacteria were more abundant. The source tracker analysis indicated that at most 1.0% of bacteria may have come from another source, showing the safety of the process used to treat milk samples. Conclusion The pasteurization process increased the bacterial diversity. We selected taxa capable of surviving the process, which could proliferate after the treatment without being a risk for infants.
Article
The objectives of this study were to lay the methodological groundwork for field studies of microRNA analysis in exosomes from small sample volumes of human milk, and assess exosome and microRNA content in infant formulas. When human milk was stored at 4°C for four weeks, the count of exosome-sized vesicles decreased progressively to 49% ± 13% of that in fresh milk. Exosomes were purified from 1 mL of fresh human milk and their microRNA content was assessed by microRNA-sequencing analysis and compared to that in infant formulas. We identified 221 microRNAs in exosomes from three samples of fresh human milk; 84 microRNAs were present in all three samples. MicroRNAs were not detectable in infant formulas and their exosome-sized vesicles, which appeared to be casein micelles. We conclude that large scale studies of microRNAs in human milk exosomes are feasible, and exosomes and microRNAs are not detectable in formulas.