Download PDF
Original Article  |  Open Access  |  24 Jun 2023

A pilot study to disentangle the infant gut microbiota composition and identification of bacteria correlates with high fat mass

Views: 520 |  Downloads: 81 |  Cited:   0
Microbiome Res Rep 2023;2:23.
10.20517/mrr.2023.11 |  © The Author(s) 2023.
Author Information
Article Notes
Cite This Article

Abstract

Background: At birth, the human intestine is colonized by a complex community of microorganisms known as gut microbiota. These complex microbial communities that inhabit the gut microbiota are thought to play a key role in maintaining host physiological homeostasis. For this reason, correct colonization of the gastrointestinal tract in the early stages of life could be fundamental for human health. Furthermore, alterations of the infant microbiota are correlated with the development of human inflammatory diseases and disorders. In this context, the possible relationships between intestinal microbiota and body composition during infancy are of great interest.

Methods: In this study, we have performed a pilot study based on 16S rRNA gene profiling and metagenomic approaches on repeatedly measured data on time involving a cohort of 41 Italian newborns, which is aimed to investigate the possible correlation between body fat mass percentage (FM%) and the infant gut microbiota composition.

Results and conclusion: The taxonomical analysis of the stool microbiota of each infant included in the cohort allowed the identification of a specific correlation between intestinal bacteria, such as Bifidobacterium and Veillonella, and the increase in FM%. Moreover, the analysis of the infant microbiome’s metabolic capabilities suggested that the intestinal microbiome functionally impacts the human host and its possible influence on host physiology.

Keywords

Infant gut microbiota, microbiome, fat mass %, body weight

INTRODUCTION

The human gastrointestinal tract is colonized at birth by a variety of microbial communities that together will constitute the human gut microbiota[1]. It is now well established that these microbial communities play important roles in developing and maintaining the host’s physiology, contributing to the immunological, physiological, nutritional, and metabolic functionalities[2-5]. However, the human gut microbiota varies among individuals, and its composition is strongly related to the host genotype and environmental factors[1,6]. In this context, numerous studies concerning the infant’s microbiota have highlighted how different pre- and post-natal factors, such as delivery mode, gestational age, type of feeding and weaning, can influence infant microbiota composition[1,7-11] and consequently its development[12,13]. C-section delivery, for example, is considered one of the most impacting events on the gut microbiota development, contributing to the colonization of the intestinal tract by bacteria derived from the environment and originating from the mother’s skin, which is thought to play a negative impact on the host health status in the mid-long term[14,15].

Similarly, studies based on the analysis of preterm infants’ gut microbiota highlighted an alteration in the bacterial communities, characterized by a decrease of biodiversity, and its possible relation with life-threatening diseases, such as necrotizing enterocolitis (NEC)[16,17] and late-onset sepsis[18], and with the risk of developing inflammatory diseases, such as atopy, asthma, and obesity[19-21]. Moreover, the type of feeding and weaning has been related to the alteration of infant gut microbiota[9,22,23]. In detail, breastfed newborns have been demonstrated to promote a more stable and uniform population compared to formula-fed ones[24].

Furthermore, several studies focused their interest on the association between the gut microbiota composition and the increase in body weight during the growth and development of infants, revealing a positive correlation with bacterial complexity[25] and with bacterial genera, such as Acinetobacter, Bifidobacterium, Collinsella, Enterococcus, Neisseria, Lactobacillus and Parabacteroides[26-28]. Notably, studies focused on body weight gain in the first months of life are mainly based on body mass index (BMI), and only very few are directed at the most accurate measurement of body fat mass percentage (FM%)[29,30]. Therefore, to understand the possible relationships between body weight, FM%, and the infant gut microbiota composition, we have performed a pilot study based on repeatedly measured data on time from a cohort of 41 Italian newborns whose gut microbiota was investigated using 16S rRNA gene profiling and metagenomic approaches. In detail, the stool microbiota of each infant has been assessed through 16S rRNA gene profiling analysis at three different stages of life, i.e., at birth, at the fourth week, and at the thirteenth week, and correlated with the most relevant parameters of the subject, such as length of pregnancy, type of feeding, type of delivery, sex, body weight, and FM%. Furthermore, once the possible microbial markers correlated to the increase in FM% were identified, a metagenomic shotgun analysis was performed on a subset of samples to identify possible peculiar microbiome functional capabilities.

METHODS

Recruitment

In this pilot study, forty-one Italian infants from the NICU Fondazione IRCCS Ca’Granda Ospedale Policlinico of Milan were enrolled, trying to balance the clinical characteristics of the cohort of infants. In detail, all infants enrolled have not been hospitalized in Intensive Care. Moreover, the infants stayed at the nursery for a couple of days immediately after birth. In fact, all the infants included in the study are healthy with no severe pathologies, such as diseases related to the gastrointestinal and respiratory tract, and/or major comorbidities, and no antibiotic treatment. A single fecal sample was collected from each infant at three different time points, at birth (T0), i.e., a few days after birth, at the fourth week (T1), and at the thirteenth week (T2). These three time points were selected to study the evolution of microbiota in the early period of life in parallel to that of body composition. All three stool samples were collected in the onsite clinic.

The study protocol was approved by the Ethics Committee of the Fondazione IRCCS “Ca’Granda” Ospedale Policlinico of Milan (103_2016). Furthermore, written informed consent was obtained from the parents of all recruited infants.

Data collection procedures

Infants were enrolled at birth. At enrolment, basic subject characteristics such as gestational age at birth, anthropometric parameters at each study point (body weight, length, and head circumference), and sex were recorded prospectively [Table 1 and Supplementary Table 1]. Gestational age was based on the last menstrual period and first-trimester ultrasonogram. The maternal characteristics were also collected [Supplementary Table 2].

Table 1

Clinical characteristics of the studied infant population

N samples = 41
SexFemale49%
Male51%
Delivery typeTerm68%
Late preterm32%
Mode of deliveryVaginal56%
C-section44%
FeedingExclusive breast milk68%
Breast milk and formula29%
Exclusively formula2%

Nutritional practices

Infants were fed on demand, and mothers were encouraged to breastfeed or express their milk according to their infant’s clinical condition. When human milk was unavailable or insufficient, formula feeding was started.

Growth and body composition assessment

Anthropometric measurements (body weight, length, and head circumference) were assessed at birth (T0), at the fourth week (T1), and at the thirteenth week (T2). The weight of each baby was measured on an electronic scale accurate to 0.1 g (PEA POD Infant Body Composition System; COSMED, Italy). Body length was measured to the nearest 1 mm on a Harpenden neonatometer (Holtain, Crymych, UK). Furthermore, a non-stretch measuring tape measured the head circumference to the nearest 1 mm. All measurements were assessed by the trained medical staff of the author’s institution. Body composition was assessed using an air-displacement plethysmograph (PEA POD Infant Body Composition System; COSMED, Italy). A detailed description of the PEA POD’s physical design, operating principles, validation, and measurement procedures is provided elsewhere[31]. Briefly, the PEA POD assesses fat mass and fat-free mass by direct measurements of body mass and volume and the application of a classic densitometric model where the percentage of body fat is calculated using body density and pre-determined fat and fat-free mass density values. Body fat was defined as body weight minus fat-free mass. A constant fat mass density value of 0.9007 g/mL is used. Fat-free mass density values are calculated as the sum of the contribution of the various components in the fat-free mass compartment.

Samples collection and DNA extraction

Fresh fecal samples were collected at birth (T0), at the fourth week (T1), and at the thirteenth week (T2), from infants born at the NICU Fondazione IRCCS Ca’ Granda Ospedale Policlinico of Milan and were immediately inactivated with DNA/RNA shield buffer (Zymo Research, USA) and subsequently submitted to the extraction of bacterial DNA using the protocol previously described[32]. The stool samples were collected by spontaneous evacuation and stored at room temperature in preservation tubes containing 2 mL of preservative and inactivating solution DNA/RNA shield buffer (Zymo Research, USA). Samples were then delivered to the laboratory of Probiogenomics, University of Parma, to perform the DNA extraction and sequencing. In detail, Bacterial DNA was extracted from fecal samples using the QIAamp Fast DNA Stool Mini kit following the manufacturer’s instructions (Qiagen Ltd., Strasse, Germany) and quantified using fluorometric Qubit quantification system (Life Technologies, Thermo Fisher Scientific, Waltham, Massachusetts, USA).

16S rRNA sequencing and profiling

Partial 16S rRNA gene sequences were amplified from extracted DNA using primer pair Probio_Uni and/Probio_Rev, targeting the V3 region of the 16S rRNA gene sequence[33]. 16S rRNA gene amplification and amplicon checks were carried out as previously described[33]. 16S rRNA gene sequencing was performed using a MiSeq (Illumina) according to the protocol previously reported[33,34]. Following sequencing, the FASTQ files were processed using a custom script based on the QIIME software suite[35]. Quality control-maintained sequences with a length between 140 and 400 bp and average sequence quality score of > 20, while sequences with homopolymers of > 7 bp and mismatched primers were omitted. 16S rRNA Amplicon sequence variants (ASVs) were defined at 100% sequence homology using DADA2, and ASVs represented by just a single sequence were removed. All reads were classified to the lowest possible taxonomic rank using QIIME 2, and a reference dataset from the SILVA database v.132.

Shotgun metagenomics sequencing

According to the manufacturer’s instructions, DNA library preparation was performed using the Nextera XT DNA sample preparation kit (Illumina, San Diego, CA, USA). First, 1 ng input DNA from each sample was used for the library preparation, which underwent fragmentation, adapter ligation, and amplification. Then, Illumina libraries were pooled equimolarly, denatured, and diluted to a concentration of 1.5 pM. Next, DNA sequencing was performed on a MiSeq instrument (Illumina) using a 2 × 250 bp Output sequencing Kit together with a deliberate spike-in of 1% PhiX control library.

Taxonomic classification of sequence reads

Taxonomic profiling of sequenced reads was performed employing the METAnnotatorX2 bioinformatics platform[36,37]. In detail, the downloaded FASTQ files were filtered to remove reads with a quality of  < 25, and to retain reads with a length of > 100 bp. Subsequently, a human host DNA filtering was performed through bowtie2 software[38,39], following the METAnnotatorX2 manual[37]. Afterward, the taxonomic classification of 100,000 reads was achieved by means of MegaBLAST[40] employing a manually curated and pre-processed database of genomes retrieved from the National Center for Biotechnology Information (NCBI), following the METAnnotatorX2 manual[37].

Functional prediction

Functional profiling of the sequenced reads was performed with the METAnnotatorX2 bioinformatics platform[36,37]. In addition, functional classification of reads was performed to reveal metabolic pathways based on the MetaCyc database (release 24.1)[41] through RAPSearch2 software[42,43].

Statistical analysis

ORIGIN 2021 (https://www.originlab.com/2021) and SPSS software (www.ibm.com/software/it/analytics/spss/) were used to compute statistical analyses. Moreover, the similarities between samples (beta-diversity) were calculated by the Bray-Curtis dissimilarity matrix based on species abundance. Beta diversity was represented through Principal Coordinate Analysis (PCoA) using the function “ape” of the R suite package (http://www.rstudio.com/). Moreover, the available metadata and the various detected bacterial species were tested and plotted on the PCoA using the “envfit” and “plot” functions, respectively, in Rstudios (http://www.rstudio.com/). Permutational analysis of variance (PERMANOVA) analyses were performed using 999 permutations to estimate P-values for population differences in PCoA analyses. Furthermore, a correlation analysis between the available metadata and the various detected bacterial species of all samples at different time points was performed through Spearman’s rank correlation coefficient[44]. The False Discovery Rate (FDR) correction is applicated to all statistical analyses based on Benjamini and Hochberg correction[45].

RESULTS AND DISCUSSIONS

Evaluation of stool microbiota inter-individual variability among subjects

In order to define the possible correlation/s between FM% and intestinal bacterial communities, a total of 123 fecal samples were collected from a heterogeneous infant population (n = 41) [Table 1]. The fecal microbiota of all stool samples collected in the framework of this pilot study was assessed by 16S rRNA gene profiling approach following the procedures reported previously[33]. The analysis of the FASTQ obtained from Illumina sequencing allowed to retrieve a total of 5,442,643 reads with an average per sample of 48,595 ± 12,976 after quality and human sequences filtering [Supplementary Table 3]. Unfortunately, 11 samples did not provide enough reads to obtain a reliable taxonomic profile, resulting in a final number of 112 analyzed samples, i.e., 31 samples at T0, 40 at T1, and 41 at T2 [Supplementary Table 3]. In order to identify possible population stratification, we have performed a beta-diversity analysis correlating the main parameters of the subject, such as duration of pregnancy, type of delivery, type of feeding, sex, body weight, and FM%, with the microbial genera that composed the gut microbiota. In detail, separate PCoA analyses at T0, T1, and T2 revealed a possible stratification of the samples at the different time points based on the type of delivery (PERMANOVA P-value < 0.05) [Figure 1A-C, Supplementary Table 4], suggesting an impact of the delivery mode on the development of the microbiota over time[8,46]. Moreover, PCoA analysis does not appear to show further stratification based on the other subject parameters, such as duration of pregnancy, type of feeding, sex, body weight, and FM%.

A pilot study to disentangle the infant gut microbiota composition and identification of bacteria correlates with high fat mass

Figure 1. Evaluation of microbial beta-diversity. (A) displays the PCoA of the fecal samples at T0, subdivided by the type of delivery; (B) shows the PCoA of the fecal samples at T1, subdivided by the type of delivery; (C) reveals the PCoA of the fecal samples at T2, subdivided by the type of delivery. FDR: False discovery rate; PCoA: principal coordinate analysis; PERMANOVA: permutational analysis of variance.

Inspection of the predicted taxonomic profiles showed that the infant cohort is mainly characterized by the genera Streptococcus (average 4.11% ± 7.70%, prevalence 97%), Bacteroides (average 19.66% ± 24.70%, prevalence 96%), Bifidobacterium (average 7.41% ± 11.75%, prevalence 91%) and Escherichia-Shigella (average 23.41% ± 29.13%, prevalence 88%) [Supplementary Table 5], bacteria typical of the gastrointestinal tract[9,47]. Moreover, a specific descriptive taxonomical comparison between the groups of natural and C-section-delivered infants revealed possible differences in the microbiota composition. Specifically, focusing on the bacterial taxa with a total prevalence > 50 % [Table 2], Escherichia-Shigella, Bacteroides, and Parabacteroides genera seemed to be associated and to be persistent in vaginal delivery samples. In contrast, Streptococcus, Flavobacterium, Lactobacillus, Staphylococcus, Clostridium sensu stricto 1, Rothia, and Enterococcus genera resulted related to C-section delivery [Table 2]. These results are in agreement with previous studies that indicated Bacteroides and Escherichia-Shigella as two of the dominant bacterial genera of vaginally delivered infants’ gut microbiota[9,47,48]. Furthermore, the decrease in babies born by C-section of Bacteroides genus in favor of environmental bacteria, such as Streptococcus, Staphylococcus, and Clostridium, could be related to external contamination during childbirth and a consequent increased competition within the bacteria of the infant’s gut[49]. Moreover, in order to identify the possible correlation between the type of delivery and the other variables, such as duration of pregnancy and sex, a cross-tabulation analysis was performed, highlighting the independence of these variables [Supplementary Table 6].

Table 2

Bacterial taxa with a total prevalence > 50% and persistent higher abundance in vaginal or C-section delivery samples

T0T1T2
AverageStandard deviationHigher abundance inAverageStandard deviationHigher abundance inAverageStandard deviationHigher abundance in
TaxonomyTotal prevalenceVaginalC-sectionVaginalC-sectionVaginalC-sectionVaginalC-sectionVaginalC-sectionVaginalC-section
Bacteroides95.54%34.53%1.19%32.35%1.34%Vaginal23.36%3.58%19.63%9.98%Vaginal34.51%5.16%22.73%14.83%Vaginal
Escherichia-Shigella87.50%47.02%19.98%39.63%40.23%Vaginal23.86%6.39%23.11%14.30%Vaginal22.96%14.82%18.31%21.39%Vaginal
Parabacteroides54.46%3.84%0.38%5.96%0.76%Vaginal5.01%2.54%7.99%10.73%Vaginal3.54%0.53%5.28%2.22%Vaginal
Clostridium sensu stricto 159.82%0.04%10.36%0.12%21.94%C-section3.78%13.68%10.76%18.17%C-section3.25%10.62%7.84%16.68%C-section
Enterococcus57.14%0.07%0.41%0.19%0.83%C-section0.17%0.84%0.50%2.54%C-section0.08%0.30%0.14%0.61%C-section
Flavobacterium57.14%0.04%0.29%0.08%0.38%C-section0.01%0.06%0.02%0.14%C-section0.01%0.02%0.01%0.03%C-section
Lactobacillus59.82%0.04%0.17%0.08%0.20%C-section0.06%0.39%0.14%1.11%C-section0.48%1.47%1.21%5.73%C-section
Rothia71.43%0.06%0.49%0.14%0.90%C-section0.30%0.82%0.36%2.03%C-section0.08%0.16%0.19%0.28%C-section
Staphylococcus59.82%0.07%16.67%0.18%31.71%C-section0.27%1.97%0.52%6.35%C-section0.03%0.47%0.05%1.54%C-section
Streptococcus97.32%1.39%9.71%4.38%16.44%C-section4.16%6.31%5.36%7.53%C-section2.72%3.65%6.30%6.51%C-section

Identification of possible correlation between infant gut microbiota and fat mass

The possible correlation and impact of the gut microbiota composition on the host’s body weight and BMI have been extensively investigated[50-55]. Nevertheless, few studies have investigated the relationship between microbiota and FM% in children and adults[29,30,53]. In this context, the metadata collected in this study enabled a specific correlation analysis between infant FM%, body weight, and intestinal microbiota during the first stage of life [Supplementary Table 1]. In detail, the correlation analysis of all samples at different time points suggested that four bacterial taxa were significantly (P-value < 0.05) positively correlated to the infant’s body weight and FM%. In contrast, three bacterial genera were significantly negatively correlated [Table 3]. Interestingly, the Bifidobacterium genus appears to be related to an increase in body weight and fat mass, suggesting their positive key role in newborn growth that, together with other already well-known functional roles exploited by this microbial group on the human body, reinforces the notion of their importance as a crucial bacterial genus for the foundation of human health[56,57]. Furthermore, among the bacterial taxa identified to exploit a possible role in the modulation of body weight and fat mass, Bifidobacterium, Veillonella, and Klebsiella genera have already been previously described as taxa characteristic and dominant of the infant gut microbiota, also referred to as infant community state types (ICSTs)[9,47]. These results may suggest the importance of the microbiota composition in the first months of life and its consequent effect on the absorption and assimilation of nutrients/fat and the physiology and health of the host.

Table 3

Correlation between bacterial taxa and weight or FM%. Only significant statistical correlations are reported

WeightFM (%)
TaxonomySpearman’s rank correlation coefficientFDR P-value
(Benjamini and Hochberg correction)
Spearman’s rank correlation coefficientFDR P-value (Benjamini and Hochberg correction)
Bifidobacterium0.3390.00230.3620.0009
Veillonella0.44600.4160.0001
Idiomarina0.2530.04350.260.0361
Klebsiella0.2630.03280.3540.0012
Haemophilus0.44200.3580.0011
Alistipes-0.2910.0134-0.260.0353
Phascolarctobacterium-0.2820.0179-0.250.0474

Preliminary evaluation of the functionality of gut microbiome related to FM%

The 16S rRNA gene profiling analysis performed in this study allowed to establish a correlation between body weight and FM% and specific intestinal bacteria [Table 3]. In order to identify the possible metabolic role exploited by these microorganisms on the FM% and body weight of the host, a preliminary shotgun metagenomic analysis was performed. In detail, a preliminary in-depth metagenomic analysis was achieved by selecting a subset of four samples based on their FM% and gut microbiota composition. In particular, samples with a high abundance (> 20%) of at least one of the taxa previously observed positively or negatively correlated with FM% and body weight were selected [Table 3 and Supplementary Table 5]. Therefore, four samples were selected, i.e., S031-T2, S014-T1, S034-T2, and S021-T2, re-named S031-T2-S, S014-T1-S, S034-T2-S, and S021-T2-S, respectively. In detail, sample S031-T2-S, C-section delivered and exclusively breastfed, had a high abundance of bifidobacteria (50.86%) and fat mass % above-average (32.2 compared to the T2 average of 25 ± 5). Furthermore, sample S014-T1-S, vaginally delivered and exclusively breastfed, was characterized by a high abundance of Parabacteroides genus (22.25%) and fat mass % below-average (10.2 compared to the T1 average of 16 ± 5). Thus, samples S034-T2-S, vaginally delivered and exclusively breastfed, and S021-T2-S, C-section delivered and exclusively breastfed, had a high abundance of Veillonella genus (37.85% and 26.05%, respectively), but S034-T2-S had a fat mass % below-average (11.2 compared to the T2 average of 25 ± 5) while S021-T2-S had fat mass % in the average (27.3 compared to the T2 average of 25 ± 5). In order to explore the metabolic capabilities of the microbiome of each selected sample, a screening of metabolic enzymatic reactions based on the MetaCyc database[41] and the Enzyme Commission (EC) classification was performed. In particular, we focused on the EC involved in fatty acid metabolism to investigate the possible correlation between the composition of the intestinal microbiome and FM% [Figure 2A]. Interestingly, a specific descriptive comparison between samples revealed that subjects with a high FM%, i.e., S031-T2-S and S021-T2-S samples, displayed a greater number of reads associated with fatty acid metabolism than infants with low FM%, i.e., S014-T1-S and S034-T2-S samples [Figure 2A], suggesting a possible correlation between individual’s fat mass percentage and microbiome composition. In detail, samples S031-T2-S and S021-T2-S showed a larger number of enzymes involved in fatty acid biosynthesis compared to the two samples with low FM% [Supplementary Table 7], mainly represented by fatty-acyl-CoA synthase system (EC 2.3.1.86), indicating an increased capacity for fatty acid synthesis. Moreover, a specific investigation through METAnnotatorX2 software[37,58] was performed to identify which bacterial species of the different samples possessed genes predicted to encode the EC 2.3.1.86 [Figure 2B]. Remarkably, the analysis revealed that this EC enzyme was encoded in all samples by genes mainly presented in microorganisms belonging to the Bifidobacterium genus, such as B. longum and B. breve species, suggesting a possible impact of this bacterial taxon on FM% in relation to its abundance [Figure 2B]. These preliminary results could confirm the notion that host physiology is closely correlated to the composition of the intestinal microbiome, suggesting the importance of proper bacterial colonization to maintain infant health. In particular, these results could support the hypothesis that the correct colonization of the gastrointestinal tract in the first months of life plays a key role in developing and maintaining the host’s state of health in the short and long term[12,59,60].

A pilot study to disentangle the infant gut microbiota composition and identification of bacteria correlates with high fat mass

Figure 2. Functional capability of fatty acid metabolism. (A) shows a bar plot reporting the percentage abundance of EC enzyme related to each selected sample’s metabolism of fatty acid; (B) indicates the percentage of microorganisms presenting genes related to EC 2.3.1.86 in each sample. EC: Enzyme commission; EC 2.3.1.86: fatty-acyl-CoA synthase system.

In conclusion, the human gut microbiota has been increasingly recognized as playing a role in human health and disease. Recent studies have suggested that gut microbiota may be involved in regulating human body weight and fat mass. In the current study, we have investigated the potential correlation between the infant gut microbiota and FM%, also considering the main parameters of the subject, such as duration of pregnancy, type of feeding, type of delivery, sex, and body weight. Interestingly, the analysis of the most relevant subject parameters allowed to identify possible differences in microbiota composition based on the type of delivery. In detail, vaginal delivery samples were characterized by the presence of Escherichia-Shigella, Bacteroides, and Parabacteroides genera, while C-section delivery samples seemed related to environmental bacteria probably obtained during childbirth. Moreover, the 16S rRNA gene profiling analysis allowed to correlate certain bacterial taxa with increased or decreased body weight and fat mass. Remarkably, some of the bacterial genera identified to exploit a possible role in the modulation of body weight and fat mass are commonly found in the gut microbiota of infants, indicating the significance of early-life gut microbiome composition on nutrient and fat absorption. Furthermore, a prediction of the microbiome metabolic functionality of a subset of infant samples revealed differences in the metabolic capabilities related to fatty acids. Specifically, samples with a higher fat mass % showed a higher number of enzymes involved in fatty acid biosynthesis, indicating the infant gut microbiota’s possible role in the host’s body weight and fat mass. Moreover, this analysis revealed that the Bifidobacterium genus represents the main bacterial taxon providing a genetic repertoire involved in fatty acid biosynthesis. Thus, this pilot study confirms the potential correlation between the infant gut microbiome and fat mass development. Further research is needed to fully understand the molecular mechanisms behind this intriguing relationship and determine whether gut microbiome manipulation may be a useful strategy for managing an individual’s body weight/fat mass. In fact, several studies reported that rapid weight gain in the first months of life could be an important risk factor for overweight and/or obesity and subsequent predisposition to an unfavorable metabolic profile in early adulthood[61-63], suggesting the first months of life as a critical window for adiposity programming. Moreover, further longitudinal investigations regarding the type of feeding in early life could allow to identify/clarify the possible relationship between microbiota, feeding, and fat mass. Thus, identifying specific bacterial biomarkers, such as some bifidobacterial species, can contribute to establishing novel strategies able to influence the individual’s body weight and fat mass through the modulation of the microbiota composition.

DECLARATIONS

Acknowledgments

Part of this research is conducted using the high performance computing facility of the University of Parma.

Authors’ contributions

Data analysis and manuscript writing: Mancabelli L

Samples collection: Liotto N, Tabasso C, Perrone M

DNA extraction and sequencing of samples: Viappiani A

Data curation and data analysis: Lugli GA, Tarracchini C, Fontana F, Alessandri G

Conceptualization, supervision and manuscript editing: Bernasconi S, Roggero P, Mosca F, Turroni F, Milani C, Ventura M

Availability of data and materials

Raw sequences of the Italian 16S rRNA gene and shallow-shotgun sequencing data are accessible through SRA under study accession (NO. PRJNA930371).

Financial support and sponsorship

The authors thank GenProbio srl for the financial support of the Laboratory of Probiogenomics.

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

The study protocol was approved by the Ethics Committee of the Fondazione IRCCS “Ca’ Granda” Ospedale Policlinico of Milan (103_2016). Written informed consent was obtained from the parents of all recruited infants.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2023.

Supplementary Materials

REFERENCES

1. Milani C, Duranti S, Bottacini F, et al. The first microbial colonizers of the human gut: composition, activities, and health implications of the infant gut microbiota. Microbiol Mol Biol Rev 2017;81:e00036-17.

2. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016;16:341-52.

3. Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res 2020;30:492-506.

4. Valdes AM, Walter J, Segal E, Spector TD. Role of the gut microbiota in nutrition and health. Brit Med J 2018;361:k2179.

5. Dahl WJ, Rivero Mendoza D, Lambert JM. Diet, nutrients and the microbiome. Prog Mol Biol Transl Sci 2020;171:237-63.

6. Fassarella M, Blaak EE, Penders J, Nauta A, Smidt H, Zoetendal EG. Gut microbiome stability and resilience: elucidating the response to perturbations in order to modulate gut health. Gut 2021;70:595-605.

7. Reyman M, van Houten MA, van Baarle D, et al. Impact of delivery mode-associated gut microbiota dynamics on health in the first year of life. Nat Commun 2019;10:4997.

8. Zhang C, Li L, Jin B, et al. The effects of delivery mode on the gut microbiota and health: state of art. Front Microbiol 2021;12:724449.

9. Mancabelli L, Tarracchini C, Milani C, et al. Multi-population cohort meta-analysis of human intestinal microbiota in early life reveals the existence of infant community state types (ICSTs). Comput Struct Biotechnol J 2020;18:2480-93.

10. Ventura M, Milani C, Lugli GA, van Sinderen D. Health benefits conferred by the human gut microbiota during infancy. Microb Biotechnol 2019;12:243-8.

11. Turroni F, Milani C, Ventura M, van Sinderen D. The human gut microbiota during the initial stages of life: insights from bifidobacteria. Curr Opin Biotechnol 2022;73:81-7.

12. Yao Y, Cai X, Ye Y, Wang F, Chen F, Zheng C. The role of microbiota in infant health: from early life to adulthood. Front Immunol 2021;12:708472.

13. Browne HP, Shao Y, Lawley TD. Mother-infant transmission of human microbiota. Curr Opin Microbiol 2022;69:102173.

14. Hoang DM, Levy EI, Vandenplas Y. The impact of caesarean section on the infant gut microbiome. Acta Paediatr 2021;110:60-7.

15. Callaway E. C-section babies are missing key microbes. Nature 2019; doi: 10.1038/d41586-019-02807-x.

16. Gregory KE, Deforge CE, Natale KM, Phillips M, Van Marter LJ. Necrotizing enterocolitis in the premature infant: neonatal nursing assessment, disease pathogenesis, and clinical presentation. Adv Neonatal Care 2011;11:155-64; quiz 165.

17. Baranowski JR, Claud EC. Necrotizing enterocolitis and the preterm infant microbiome. Adv Exp Med Biol 2019;1125:25-36.

18. El Manouni El Hassani S, Niemarkt HJ, Berkhout DJC, et al. Profound pathogen-specific alterations in intestinal microbiota composition precede late-onset sepsis in preterm infants: a longitudinal, multicenter, case-control study. Clin Infect Dis 2021;73:e224-32.

19. Liu Y, Du X, Zhai S, Tang X, Liu C, Li W. Gut microbiota and atopic dermatitis in children: a scoping review. BMC Pediatr 2022;22:323.

20. Yu L, Deng YH, Huang YH, Ke HJ, Guo Y, Wu JL. Comparison of gut microbiota between infants with atopic dermatitis and healthy controls in Guangzhou, China. J Asthma Allergy 2021;14:493-500.

21. Reddel S, Del Chierico F, Quagliariello A, et al. Gut microbiota profile in children affected by atopic dermatitis and evaluation of intestinal persistence of a probiotic mixture. Sci Rep 2019;9:4996.

22. Cong X, Judge M, Xu W, et al. Influence of feeding type on gut microbiome development in hospitalized preterm infants. Nurs Res 2017;66:123-33.

23. Parra-Llorca A, Gormaz M, Alcántara C, et al. Preterm gut microbiome depending on feeding type: significance of donor human milk. Front Microbiol 2018;9:1376.

24. Guaraldi F, Salvatori G. Effect of breast and formula feeding on gut microbiota shaping in newborns. Front Cell Infect Microbiol 2012;2:94.

25. Unger S, Stintzi A, Shah P, Mack D, O’Connor DL. Gut microbiota of the very-low-birth-weight infant. Pediatr Res 2015;77:205-13.

26. Alderete TL, Jones RB, Shaffer JP, et al. Early life gut microbiota is associated with rapid infant growth in Hispanics from Southern California. Gut Microbes 2021;13:1961203.

27. Luoto R, Kalliomäki M, Laitinen K, Isolauri E. The impact of perinatal probiotic intervention on the development of overweight and obesity: follow-up study from birth to 10 years. Int J Obes 2010;34:1531-7.

28. Chouraqui JP, Grathwohl D, Labaune JM, et al. Assessment of the safety, tolerance, and protective effect against diarrhea of infant formulas containing mixtures of probiotics or probiotics and prebiotics in a randomized controlled trial. Am J Clin Nutr 2008;87:1365-73.

29. Soderborg TK, Clark SE, Mulligan CE, et al. The gut microbiota in infants of obese mothers increases inflammation and susceptibility to NAFLD. Nat Commun 2018;9:4462.

30. Koleva PT, Bridgman SL, Kozyrskyj AL. The infant gut microbiome: evidence for obesity risk and dietary intervention. Nutrients 2015;7:2237-60.

31. Roggero P, Giannì ML, Amato O, et al. Evaluation of air-displacement plethysmography for body composition assessment in preterm infants. Pediatr Res 2012;72:316-20.

32. Mancabelli L, Milani C, Fontana F, et al. Untangling the link between the human gut microbiota composition and the severity of the symptoms of the COVID-19 infection. Environ Microbiol 2022;24:6453-62.

33. Milani C, Hevia A, Foroni E, et al. Assessing the fecal microbiota: an optimized ion torrent 16S rRNA gene-based analysis protocol. PLoS One 2013;8:e68739.

34. Bolyen E, Rideout JR, Dillon MR, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol 2019;37:852-7.

35. Caporaso JG, Kuczynski J, Stombaugh J, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods 2010;7:335-6.

36. Milani C, Casey E, Lugli GA, et al. Tracing mother-infant transmission of bacteriophages by means of a novel analytical tool for shotgun metagenomic datasets: METAnnotatorX. Microbiome 2018;6:145.

37. Milani C, Lugli GA, Fontana F, et al. METAnnotatorX2: a comprehensive tool for deep and shallow metagenomic data set analyses. mSystems 2021;6:e0058321.

38. Langmead B, Salzberg SL. Fast gapped-read alignment with bowtie 2. Nat Methods 2012;9:357-9.

39. Langmead B, Wilks C, Antonescu V, Charles R. Scaling read aligners to hundreds of threads on general-purpose processors. Bioinformatics 2019;35:421-32.

40. Chen Y, Ye W, Zhang Y, Xu Y. High speed BLASTN: an accelerated MegaBLAST search tool. Nucleic Acids Res 2015;43:7762-8.

41. Caspi R, Billington R, Ferrer L, et al. The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of pathway/genome databases. Nucleic Acids Res 2016;44:D471-80.

42. Zhao Y, Tang H, Ye Y. RAPSearch2: a fast and memory-efficient protein similarity search tool for next-generation sequencing data. Bioinformatics 2012;28:125-6.

43. Ye Y, Choi JH, Tang H. RAPSearch: a fast protein similarity search tool for short reads. BMC Bioinformatics 2011;12:159.

44. Spearman's rank correlation coefficient. BMJ 2018;362:k4131.

45. Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res 2001;125:279-84.

46. Rutayisire E, Huang K, Liu Y, Tao F. The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants’ life: a systematic review. BMC Gastroenterol 2016;16:86.

47. Alessandri G, Fontana F, Mancabelli L, et al. Exploring species-level infant gut bacterial biodiversity by meta-analysis and formulation of an optimized cultivation medium. NPJ Biofilms Microbiomes 2022;8:88.

48. Matharu D, Ponsero AJ, Dikareva E, et al. Bacteroides abundance drives birth mode dependent infant gut microbiota developmental trajectories. Front Microbiol 2022;13:953475.

49. Mitchell CM, Mazzoni C, Hogstrom L, et al. Delivery mode affects stability of early infant gut microbiota. Cell Rep Med 2020;1:100156.

50. Gao X, Zhang M, Xue J, et al. Body mass index differences in the gut microbiota are gender specific. Front Microbiol 2018;9:1250.

51. Lv Y, Qin X, Jia H, Chen S, Sun W, Wang X. The association between gut microbiota composition and BMI in Chinese male college students, as analysed by next-generation sequencing. Br J Nutr 2019;122:986-95.

52. Kim MH, Yun KE, Kim J, et al. Gut microbiota and metabolic health among overweight and obese individuals. Sci Rep 2020;10:19417.

53. Palmas V, Pisanu S, Madau V, et al. Gut microbiota markers associated with obesity and overweight in Italian adults. Sci Rep 2021;11:5532.

54. Ignacio A, Fernandes MR, Rodrigues VA, et al. Correlation between body mass index and faecal microbiota from children. Clin Microbiol Infect 2016;22:258.e1-8.

55. Greathouse KL, White JR, Padgett RN, et al. Gut microbiome meta-analysis reveals dysbiosis is independent of body mass index in predicting risk of obesity-associated CRC. BMJ Open Gastroenterol 2019;6:e000247.

56. Barratt MJ, Nuzhat S, Ahsan K, et al. Bifidobacterium infantis treatment promotes weight gain in Bangladeshi infants with severe acute malnutrition. Sci Transl Med 2022;14:eabk1107.

57. Michael DR, Jack AA, Masetti G, et al. A randomised controlled study shows supplementation of overweight and obese adults with lactobacilli and bifidobacteria reduces bodyweight and improves well-being. Sci Rep 2020;10:4183.

58. Alessandri G, Lugli GA, Tarracchini C, et al. Disclosing the genomic diversity among members of the bifidobacterium genus of canine and feline origin with respect to those from human. Appl Environ Microbiol 2022;88:e0203821.

59. Turroni F, Milani C, Duranti S, et al. The infant gut microbiome as a microbial organ influencing host well-being. Ital J Pediatr 2020;46:16.

60. Tanaka M, Nakayama J. Development of the gut microbiota in infancy and its impact on health in later life. Allergol Int 2017;66:515-22.

61. van Beijsterveldt IALP, de Fluiter KS, Breij LM, van der Steen M, Hokken-Koelega ACS. Fat mass and fat-free mass track from infancy to childhood: new insights in body composition programming in early life. Obesity 2021;29:1899-906.

62. Leunissen RW, Kerkhof GF, Stijnen T, Hokken-Koelega A. Timing and tempo of first-year rapid growth in relation to cardiovascular and metabolic risk profile in early adulthood. J Am Med Assoc 2009;301:2234-42.

63. de Fluiter KS, van Beijsterveldt IALP, Breij LM, Acton D, Hokken-Koelega ACS. Association between fat mass in early life and later fat mass trajectories. JAMA Pediatr 2020;174:1141-8.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Mancabelli L, Milani C, Fontana F, Liotto N, Tabasso C, Perrone M, Lugli GA, Tarracchini C, Alessandri G, Viappiani A, Bernasconi S, Roggero P, Mosca F, Turroni F, Ventura M. A pilot study to disentangle the infant gut microbiota composition and identification of bacteria correlates with high fat mass. Microbiome Res Rep 2023;2:23. http://dx.doi.org/10.20517/mrr.2023.11

AMA Style

Mancabelli L, Milani C, Fontana F, Liotto N, Tabasso C, Perrone M, Lugli GA, Tarracchini C, Alessandri G, Viappiani A, Bernasconi S, Roggero P, Mosca F, Turroni F, Ventura M. A pilot study to disentangle the infant gut microbiota composition and identification of bacteria correlates with high fat mass. Microbiome Research Reports. 2023; 2(3): 23. http://dx.doi.org/10.20517/mrr.2023.11

Chicago/Turabian Style

Mancabelli, Leonardo, Christian Milani, Federico Fontana, Nadia Liotto, Chiara Tabasso, Michela Perrone, Gabriele Andrea Lugli, Chiara Tarracchini, Giulia Alessandri, Alice Viappiani, Sergio Bernasconi, Paola Roggero, Fabio Mosca, Francesca Turroni, Marco Ventura. 2023. "A pilot study to disentangle the infant gut microbiota composition and identification of bacteria correlates with high fat mass" Microbiome Research Reports. 2, no.3: 23. http://dx.doi.org/10.20517/mrr.2023.11

ACS Style

Mancabelli, L.; Milani C.; Fontana F.; Liotto N.; Tabasso C.; Perrone M.; Lugli GA.; Tarracchini C.; Alessandri G.; Viappiani A.; Bernasconi S.; Roggero P.; Mosca F.; Turroni F.; Ventura M. A pilot study to disentangle the infant gut microbiota composition and identification of bacteria correlates with high fat mass. Microbiome. Res. Rep. 2023, 2, 23. http://dx.doi.org/10.20517/mrr.2023.11

About This Article

© The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
520
Downloads
81
Citations
0
Comments
0
2

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 5 clicks
Like This Article 2 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Microbiome Research Reports
ISSN 2771-5965 (Online)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/