Skip to main content
Erschienen in: BMC Pediatrics 1/2023

Open Access 01.12.2023 | Research

A cross-sectional study on stool- and gastrointestinal-related outcomes of Mexican infants consuming different formulae

verfasst von: Carlijn M. Maasakkers, Jeske H.J. Hageman, Olivia Balcazar Muñoz, Tomás Gómez Tamayo, Andrés Blanco Montero, Luis Gerardo Garza Lara, Regina Flores-López, Miriam Contreras Fernández, Sofía Morán Ramos, Tim T. Lambers

Erschienen in: BMC Pediatrics | Ausgabe 1/2023

Abstract

Background

Immaturities present at birth, such as in the gut microbiome and digestive, nervous, and immune system, resolve with time. Nevertheless, this may result in mild digestive symptoms early in life, particularly in formula-fed infants. Formula composition and processing may impact this discomfort. This study therefore aimed to assess stool characteristics and gastrointestinal symptoms of healthy infants fed different formulae.

Methods

A multicenter, cross-sectional, observational trial was performed in Mexico between November 2019 and January 2022, where exclusively formula-fed infants (n = 342, aged 1–4 months) were studied in four groups based on their existing formula use. Feeding was continued per practice following label instructions. For 7 days, parents/caregivers were requested to record fecal characteristics, using the Amsterdam Infant Stool Scale, and rate gastrointestinal symptoms. Stool samples were collected to determine pH, dry matter content, and fecal calprotectin levels.

Results

Most infants had a soft/formed stool consistency, although odds for hard stools were different between groups. Gastrointestinal symptom scores revealed significant differences for burping and diarrhea, while other symptoms did not differ between groups. No significant differences between groups were found for stool frequency, dry matter content, and fecal pH. Although calprotectin was within the expected healthy ranges, significant differences among groups were seen. Furthermore, calprotectin significantly correlated with the severity of the gastrointestinal symptoms burping, flatulence, abdominal distension, and diarrhea.

Conclusions

Differences in stool characteristics and specific differences in gastrointestinal symptoms were observed between different formula brand users. This may potentially be explained by the different composition and processing of the formulae, although there are multiple factors that influence the assessed outcomes.

Trial registration

The study was registered in the Netherlands Trial Registry (NL7805), linked to https://​trialsearch.​who.​int/​, on 11/06/2019.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12887-023-04426-y.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

The gastrointestinal system of infants is still underdeveloped. For example, the production of pancreatic enzymes is still low and the small intestine is relatively short [1]. Therefore, the capacity of digestion and absorption of nutrients is lower in infants, compared to the adult situation [2]. Furthermore, the gut microbiome, nervous, and immune system of the gastrointestinal tract is in an immature state [3]. These immaturities resolve with time as the gastrointestinal tract matures. However, early in life these immaturities may contribute to the development of mild gastrointestinal symptoms including colic, regurgitation, diarrhea, and constipation, particularly in formula-fed infants [4, 5]. These symptoms could result in distress for the infant. Roughly 50% of the infants develop at least one of these symptoms, of which regurgitation, infantile colic, and functional constipation are the most common [5]. Diet is an important factor in developing these gastrointestinal complications [6]. This is one of the many reasons why it is universally accepted that the optimal nutrition for a newborn infant is breastmilk. When a mother is unable to breastfeed her infant, infant formulae are the only food products that fulfill the nutritional requirements of infants during the first months of life [7]. With respect to formula-fed infants, particularly the protein source [810] and the optional ingredients pre- and probiotics [11, 12] have been considered as factors influencing these symptoms. It has been shown that formula-fed infants have a higher excretion of proteins compared to breastfed infants, which is caused by a reduced efficacy of protein absorption from the intestinal lumen [13, 14]. Besides general compositional differences between human milk and infant formula, such as protein, lipid, and carbohydrate complexity, the decreased absorption of infant formula is likely related to the processing of infant formulae [1, 15]. Predominantly heating during the processing of infant formulae can impact protein digestion [15]. Variations in formula composition and processing may thus impact gastrointestinal symptoms in infants. This study therefore aimed to assess the performance of four different formulae, that varied in composition and processing, with regard to stool- and gastrointestinal-related outcomes in a real-life setting. It was hypothesized that the different formulae would result in different stool- and gastrointestinal-related outcomes.

Methods

Study design

A multicenter, cross-sectional, observational study was performed, in which exclusively formula-fed infants were studied in four different groups based on their existing use of infant formula (IF A (FrieslandCampina, Amersfoort, The Netherlands), IF B, IF C, and IF D). The four different IFs were selected based on differences in composition and processing, the latter as illustrated by different blocked-lysine levels (shown in Additional Table 1), that were analyzed as indicators for protein glycation (as a result of the Maillard reaction) [16]. Feeding was continued per practice following the label instructions. Additional Table 1 displays the composition of the IFs, as presented on the labels, and from analyses.
This study was approved by the Ethical Committee of Hospital SMIQ S de R.L. de C.V. (CONBIOÉTICA-22-CEI-003-20171130, #16CI22014059) and by the Ethical Committee of Sociedad de Beneficencia Española, l.A.P. (CONBIOÉTICA-09-CEI-009-20170421, #15CI09016029) and was performed in accordance with the Declaration of Helsinki and its later amendments [17]. The study has been registered in the Netherlands Trial Registry, NL7805, on 11/06/2019 and was registered and approved by COFEPRIS (193300410D0017/2019) in Mexico.

Participants

In total 364 subjects were enrolled between November 2019 and January 2022 by four different sites in Mexico; Clínica Pediátrica Pigüi and Hospital Star Médica Hip in Mexico City, Clínica VITALI in Guadalajara, and Hospital Christus Muguerza Sur in Monterrey. Healthy, term infants (gestational age 37–42 weeks, birth weight 2.5-4 kg), aged 1–4 months, were included. They were exclusively fed with a commercially available IF of interest for at least 3 weeks prior to inclusion in the study. For an infant to be included, parents/caregivers of the subjects had to agree to offer no weaning foods over the course of the study, had to be 18 years or older, should be fluent in Spanish, and own a smartphone with internet access. Subjects were excluded from the study if they: (I) had a birth weight-for-length (WFL) percentile > 85 or < 5, (II) had a congenital condition and/or previous or current illness that could interfere with the study as judged by the healthcare provider, (III) had a known or increased risk of cow’s milk allergy and/or lactose intolerance (i.e. one of the biological parents/caregivers and or siblings diagnosed with cow’s milk allergy, asthma, fever, etc.), (IV) were participating in another survey or trial, (V) had parents/caregivers that seemed unable or unwilling to comply with the protocol requirements, (VI) used antibiotics and/or other medication that influences the study outcomes as judged by the healthcare provider, as well as probiotic and/or prebiotic supplements (other than those found in the formula itself), at the time of screening and/or two weeks prior to the start of the study.

Study procedures and questionnaires

After the study was properly explained, parents/caregivers were asked to sign the informed consent form. Subsequently, a screening visit took place in which anthropometric measurements were performed, and screening questionnaires were conducted to check the in- and exclusion criteria. Once enrolled in the study, the parents/caregivers received instructions on how to access and use the app which was used to fill in the questionnaires. Parents/caregivers were able to login with their individual account and see a listing of the task per day. They could open each of the questionnaires to insert the corresponding data. The app would prompt parents/caregivers to complete the required activities on a daily basis. Furthermore, stool sample kits were provided. To check the type of IF that was consumed, the parents/caregivers were requested to upload a picture of the IF used for feeding into the app as well.
Parents/caregivers were requested to complete daily diaries, to record time and description of each bowel movement, according to the Amsterdam Infant Stool Scale [18], for one week. This entailed the characterization of each stool on consistency (watery, soft, formed, hard), amount (four categories), and color (six categories). The primary endpoint of this study was stool consistency of the first stool per day for 7 days, where the categories soft and formed were grouped together as beneficial, compared to the categories watery and hard. Furthermore, stool frequency was extracted from these diaries based on the number of entries. These diaries were split in daily tasks to minimize recall bias.
Parents/caregivers were also asked to rate certain gastrointestinal symptoms in the daily diaries (score per day absent = 0 – very severe = 5), including abdominal distension, burping, flatulence, diarrhea, constipation, colic, diaper dermatitis, arching of the back, vomiting, and spit ups/regurgitation.
On day 7 parents/caregivers were requested to fill in the IGSQ (Infant Gastrointestinal Symptoms Questionnaire). The score was computed by summing the scores of all 13 items (range 13–65) [19].

Stool sampling and analyses

Parents/caregivers were asked to collect stool samples from their child’s diaper on three different days, place these samples in a sterile plastic container, keep them at -20 °C, and bring them to the last visit on day 8. At the site, samples were stored at -20 °C and transported to the laboratory where they were stored at -80 °C until further analysis. Fecal samples were thawed at room temperature and homogenized for analyses. Standard pH strips (Merck-Milipore) were used to measure pH of diluted (1:10, sterile water) samples. Dry matter content was measured by lyophilizing a 100–200 mg aliquot for 24 h and weighing it. Levels of fecal calprotectin, an inflammatory marker [20, 21], were determined with a calprotectin ELISA kit (EK-CAL; Bühlmann Laboratories AG) according to the manufacturer’s instructions.

Statistical analysis

A sample size of 100 subjects per group was based on a similar study comparing formulae on stool characteristics [22]. Accounting for a 10% drop-out rate this resulted in 440 subjects to be recruited. For continuous variables, means with standard deviations or medians with interquartile ranges were calculated, while for categorical variables percentages were computed. Stool consistency and stool color were analyzed with General Estimating Equation (GEE) applying multinomial logistic regression with repeated measurements, including all study days and IF groups. For stool amount and gastrointestinal symptoms, a similar GEE model but with ordinal multinomial accumulated responses was applied. If the variable IF type had an influence on the outcome measures (i.e. was significant in the GEE model) pair-wise post-hoc product comparisons were performed using Bonferroni adjustments.
Data normality of continuous variables was assessed with the Shapiro-Wilcoxon test. Since all the variables analyzed showed a nonparametric distribution, outliers were identified using 1.5xIQR. For comparison among IF groups of IGSQ scores, pH, dry matter content, and calprotectin levels, a Kruskal–Wallis test was used with Dunn’s post-hoc test to determine significant differences between groups, and Bonferroni adjusted p-values were used to correct for multiple comparisons. For IGSQ scores the Quade test was used as a nonparametric alternative to adjust for covariates. All analyses were performed unadjusted, and adjusted for sex (male/female), type of birth (C-section/natural), and age.
Intention to Treat (ITT) population entailed all included subjects. For the Per Protocol (PP) dataset all randomized subjects without protocol violations or deviations (e.g. out-of-window visit, prohibited medication use, or formula incompliance), and that had complete primary outcome data available were included. Due to the real-life setting of this study and the absence of large differences between ITT and PP populations, only ITT analyses are presented, with differences to PP indicated where applicable. Missing data were not imputed. Statistical analyses were performed in SPSS, SAS, and R, with p-values < 0.05 being considered statistically significant.

Results

Baseline characteristics and demographics

Of the 367 subjects screened, 364 were enrolled in the study, see flow diagram in Additional Fig. 1. In total 342 subjects completed the study; IF A: 102 subjects, IF B: 105 subjects, IF C: 100 subjects, and IF D: 35 subjects. The recruitment target of IF D was lowered (35 vs. 100) given the difficulties in recruitment due to a lower use of this product at the study locations in Mexico. In the PP analysis set, 296 subjects were taken along. Stool samples were available for 326 subjects, where 20 subjects were excluded due to protocol violations/deviations. Furthermore, for some subjects, not enough material was present for all analyses to be performed, therefore the objective measurements of pH, dry matter content, and calprotectin were conducted in 306, 302, and 304 samples respectively.
Table 1 displays the demographic characteristics of the four study groups. All groups contained slightly more boys than girls, ranging from 57 to 60%. The average age at enrollment was about 3 months. Small differences were found in weight and length.
Table 1
Demographics
 
IF A
IF B
IF C
IF D
N enrolled
105
112
108
39
N completed
102
105
100
35
Sex (% male)
57.1%
60.7%
57.4%
59%
Age at enrolment (months)
3.0 ± 1.2
2.9 ± 1.1
3.2 ± 1.0
3.1 ± 1.1
Weight at enrolment (kg)*
5.5 ± 1.2b
5.7 ± 1.2b
6.2 ± 1.2a
5.7 ± 1.2a,b
Length at enrolment (cm)*
57.9 ± 4.5b
59.6 ± 4.4a
60.5 ± 4.4a
59.2 ± 4.6a,b
Head circumference at enrolment (cm)#
39.1 ± 2.2b
39.7 ± 2.0a,b
40.4 ± 2.1a
39.8 ± 4.0a,b
Mode of delivery (% vaginal)
34.3%
44.6%
49.1%
38.5%
Gestational age (weeks)#
38.6 ± 1.0b
39.0 ± 1.2a
38.9 ± 1.2a,b
38.9 ± 1.2a,b
Birth weight (kg)
3.1 ± 0.4
3.1 ± 0.4
3.1 ± 0.4
3.1 ± 0.5
Birth length (cm)
49.7 ± 2.2
50.2 ± 2.0
50.0 ± 2.6
49.0 ± 2.7
Twins/triplets (%)
10.5%
4.5%
0%
15.4%
Mean values ± SD are shown for continuous variables.*Indicates statistically significant ANOVA, #Indicates statistically significant Kruskal Wallis test. Superscript letters indicate significant differences by post hoc Bonferroni or Dunns test respectively, adjusted for multiple comparisons.

Stool characteristics

No significant differences between groups were reported in stool frequency, dry matter content, or fecal pH (data not shown). Figure 1 shows the stool characteristics per formulae. For stool amount it was found that infants consuming IF A had lower odds of high amounts of stool compared to infants consuming IF B (OR 0.6 (95%CI 0.4–0.8)). Most stools were of a soft/formed consistency. Infants on IF D had a higher odds of hard stools vs. soft/formed compared to infants consuming IF B and IF C (OR 8.2 (95% CI 1.3–51.7) and OR 11.2 (95% CI 1.2-105.1) respectively). Infants consuming IF A had higher odds of having stool color I vs. III compared to the other IFs. All these differences remained significant after adjusting for sex, delivery mode, and age.
No differences were observed in general gastrointestinal symptoms between the four IFs, since the average IGSQ scores were similar for the four groups, between 23 and 24. For most individual gastrointestinal symptoms no differences between the four IFs were observed. Except for burping and flatulence, all gastrointestinal symptoms were reported to be absent in the majority of the registrations (percentages can be found in Additional Table 2). Infants consuming IF A had lower odds of more severe burping compared to infants consuming IF C (Table 2). Furthermore, infants consuming IF A had lower odds of having more severe diarrhea compared to the groups consuming IF B and IF D. These significances remained after adjustment with covariates, but in the PP analysis the difference between IF A and IF B on diarrhea was not found to be significance anymore. In the adjusted model the tendency of IF C having lower odds of more severe diarrhea compared to IF D became significant in both ITT and PP populations.
Table 2
Gastrointestinal symptom severity odd ratio’s
 
IF A vs. IF B
IF A vs. IF C
IF A vs. IF D
IF B vs. IF C
IF B vs. IF D
IF C vs. IF D
 
OR (95% IC)
P-value
OR (95% IC)
P-value
OR (95% IC)
P-value
OR (95% IC)
P-value
OR (95% IC)
P-value
OR (95% IC)
P-value
Abdominal distention
1.25 (0.84;1.88)
1.00
1.63 (1.04;2.55)
0.20
1.18 (0.65;2.14)
1.00
1.30 (0.84;2.02)
1.00
0.94 (0.52;1.70)
1.00
0.72 (0.39;1.35)
1.00
Arching of the back
1.35 (0.83;2.22)
1.00
1.22 (0.75;1.98)
1.00
0.78 (0.40;1.50)
1.00
0.90 (0.54;1.50)
1.00
0.57 (0.29;1.13)
0.65
0.64 (0.33;1.25)
1.00
Burping
0.65 (0.43;0.96)
0.20
0.55 (0.37;0.83)
0.03*
0.58 (0.31;1.06)
0.45
0.86 (0.58;1.27)
1.00
0.89 (0.49;1.62)
1.00
1.04 (0.57;1.90)
1.00
Colic
1.12 (0.73;1.71)
1.00
1.26 (0.80;1.97)
1.00
1.14 (0.62;2.09)
1.00
1.13 (0.73;1.73)
1.00
1.02 (0.57;1.84)
1.00
0.91 (0.50;1.66)
1.00
Constipation
1.20 (0.80;1.81)
1.00
1.74 (1.14;2.67)
0.06
0.87 (0.45;1.68)
1.00
1.45 (0.95;2.22)
0.50
0.72 (0.37;1.40)
1.00
0.50 (0.26;0.97)
0.24
Diaper dermatitis
0.98 (0.57;1.69)
1.00
1.18 (0.67;2.10)
1.00
0.76 (0.37;1.58)
1.00
1.20 (0.67;2.15)
1.00
0.77 (0.37;1.61)
1.00
0.64 (0.30;1.37)
1.00
Diarrhea
0.44 (0.25;0.76)
0.02*
0.63 (0.36;1.12)
0.71
0.26 (0.12;0.54)
0.002*
1.45 (0.85;2.48)
1.00
0.59 (0.29;1.20)
0.88
0.41 (0.20;0.84)
0.09
Flatulence
0.92 (0.60;1.39)
1.00
0.79 (0.52;1.20)
1.00
0.50 (0.29;0.88)
0.09
0.87 (0.58;1.30)
1.00
0.55 (0.32;0.95)
0.19
0.63 (0.37;1.10)
0.63
Unadjusted odds ratio’s ITT population GEE model, WALD Type 3 test, p value with Bonferroni adjustment.*P < 0.05
Gastrointestinal scores were also correlated to fecal calprotectin levels. More severe ratings of burping, flatulence, and diarrhea correlated significantly with higher calprotectin levels (Fig. 2B). Although calprotectin levels were in the expected range for healthy infants [23], levels were lower in fecal samples from infants on IF A compared to infants on IF D (Fig. 2A). This effect was not shown when outliers were excluded.

Discussion

In this observational, real-life setting study in Mexico, gastrointestinal and digestion-related outcomes of different commercially available infant formulae were studied in healthy term subjects. Most infants had soft or formed stools, although odds for having hard stools were different between groups. Further significant differences were found in stool amount and color. No significant differences between groups were reported on stool frequency, dry matter content, or fecal pH. Gastrointestinal symptom scores revealed significant differences for burping and diarrhea, while other gastrointestinal symptoms did not differ. Interestingly, although calprotectin levels were within the expected ranges for healthy infants, differences among groups could be established. Furthermore, calprotectin levels correlated with the severity of the gastrointestinal symptoms burping, flatulence, abdominal distension, and diarrhea.
Several gastrointestinal disorders are associated with changes in stool characteristics, causing them to be monitored by parents and in clinical practice [18]. Odds for having hard stools were higher for infants on IF D compared to IF B and IF C, although differences in dry matter content were not found. This could relate to the limited sample size in the current study, since dry matter content (as determined by lyophilization) was different among the consistency categories, as similarly demonstrated previously in adults [24]. Breastfed infants generally have softer stools compared to formula-fed infants, with formula composition affecting factors such as fecal soap formation, that may explain variations among formula-fed infants [25]. Also fecal color from healthy infants varies, ranging from yellow to green, where breastfed infants more often have a yellow color compared to formula-fed infants [26]. The current study showed that infants consuming IF A had higher odds of having a yellow stool color compared to the other IFs. The physiological relevance of color variations within the healthy range is unknown, and clinically less relevant than discriminating category 5 and/or 6 from others. However, the current study thus suggests that fecal color may differ per the consumed formula type, similar as stool consistency.
Next to the stool characteristics, gastrointestinal symptoms were also studied. As the study population consisted of healthy infants, it was in line with expectations that many gastrointestinal symptoms were reported to be absent or very mild in many of the registrations. This is in line with observations of several countries where mild gastrointestinal disorders have previously been observed in otherwise healthy infants [4]. In this study, only burping and flatulence were reported more often. Due to the low prevalence of GI symptoms, the differences between study products were expected to be limited. Nonetheless, the current study revealed that infants consuming IF A had lower odds of more severe burping and diarrhea compared to infants consuming IF C and infants on IF B and IF D, respectively. Significant differences were thus found in otherwise healthy infants.
The inflammatory marker calprotectin was lower in fecal samples of infants consuming IF A compared to IF D. Interestingly, fecal calprotectin correlated with the gastrointestinal symptoms burping, flatulence, and diarrhea. With respect to gastrointestinal complications, fecal calprotectin has limited use in standard clinical practice [20]. Fecal calprotectin is, for example, only informative in the diagnosis and monitoring of inflammatory bowel disease and to distinguish it from functional gastrointestinal disorders, whereas application in the diagnosis of colic and constipation is limited [20]. Although clinical relevance will therefore be limited in healthy infants, the correlation of fecal calprotectin with the symptoms burping, flatulence, and diarrhea may highlight a potential mechanistic interaction which may be interesting for future studies.
Combined, the differences in gastrointestinal symptoms and fecal calprotectin might partly be related to the different formulae consumed, with diet impacting digestive challenges that may be caused by the immature gastrointestinal system in early life [4].
Different infant formulae are available on the market, with variation in composition, predominantly related to the protein and fat source, and the presence of optional ingredients. With respect to the protein source, hydrolyzed protein formulae have previously been demonstrated to reduce digestive complications mostly associated with regurgitation [8]. Furthermore, fat sources, i.e. bovine milk or plant oil-derived and modified plant oils, such as beta-palmitate, can impact gastrointestinal outcomes through their effect on fecal soap formation. This may impact clinical outcomes, such as bone strength, gut microbiome composition, and crying in infants [9, 10, 27, 28]. Pre- and probiotics are considered for the management of pediatric gastrointestinal disorders as well, as has been reviewed in detail by the ESPGHAN taskforce for pre- and probiotics [11, 12]. Recommendations for the use of specific probiotic strains were made by this taskforce for the management of functional abdominal pain disorders and infant colic, among others [12]. More recently, characteristics related to processing [15, 29] and casein mineralization [30] have also been suggested to impact digestion and gastrointestinal-related outcomes. For example, protein denaturation and glycation have been demonstrated to impact overall IF protein digestion [15, 31, 32] and may thus warrant further investigation. All formulae in this study contained non-hydrolyzed protein sources and three of the four formulae contained different oligosaccharides (IF A, C, D). Three IFs included a human milk oligosaccharide (IF A, B, D), and one was supplemented with a probiotic (IF B) (see Additional Table 1). Moreover, formulae varied in indicators of glycation, ranging from a blocked lysin percentage of 6.1% (IF A) to 18.5% (IF C). These levels are indicative for differences in heating applied during formula production and may impact digestion and absorption as indicated by both in vitro and in vivo studies [29, 33, 34]. Combined, the differences in formula composition (particularly related to optional ingredients such as oligosaccharides and probiotics) and processing might therefore have contributed to the differences seen in this study. Theoretically, this effect could be either direct and/or through alterations in the gut microbiome. However, a direct association between IF characteristics with clinical outcomes in this observational study is not possible, as assessed outcomes are multifactorial and can be affected by many factors.
Strengths of this study include the observational setting, which enables real-world data collection. Subjects were recruited in different centers across Mexico, making the results generalizable to the wider population of, at least, Mexican infants. A limitation of the study was that outcomes were assessed based on parental reports, likely increasing the variation. Therefore, several objective and potential mechanistic measures, and the validated Amsterdam Infant Stool Scale [18] were also included in the current study. Furthermore, the lower number of subjects in one of the four groups, although representative of the frequency of use, could have resulted in reduced statistical power to determine significant differences. Additionally, no breastfed reference was included. Lastly, due to the observational nature of the study and cross-sectional assessment of the outcome measures no longitudinal data was obtained meaning no causal conclusions can be drawn.

Conclusion

Exclusively formula-fed infants, using different commercially available formulae, were compared in this study. Although only healthy subjects were included, differences in stool characteristics and gastrointestinal symptoms were observed. Despite the fact that assessed outcomes are multifactorial, variations may potentially be explained by the different compositions and processing conditions of the formulae. Interestingly, fecal calprotectin correlated with the severity of the gastrointestinal symptoms burping, flatulence, abdominal distention, and diarrhea which may be a subject for further studies.

Acknowledgements

The authors would like to thank all parents/caregivers and their infants that participated in this study. Furthermore, they want to acknowledge the Clinical Research Organisation, SPRIM Américas Mexico, as well as the four sites and their employees, Clínica Pediátrica Pigüi, Hospital Star Médica Hip, Hospital Christus Muguerza Sur, and Clínica VITALI, for their help in the recruitment of participants.

Declarations

This study was approved by the Ethical Committee of Hospital SMIQ S de R.L. de C.V. (CONBIOÉTICA-22-CEI-003-20171130, #16CI22014059) and by the Ethical Committee of Sociedad de Beneficencia Española, l.A.P. (CONBIOÉTICA-09-CEI-009-20170421, #15CI09016029). After the study was properly explained, parents/caregivers were asked to sign the informed consent form. The study was performed in accordance with the Declaration of Helsinki and its later amendments.
Not applicable.

Competing interests

CMM, JHJH, MCF, and TTL are employed by FrieslandCampina. For the remaining authors, none is declared. This study was funded by FrieslandCampina, Amersfoort, The Netherlands.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
2.
Zurück zum Zitat Bourlieu C, Ménard O, Bouzerzour K, Mandalari G, Macierzanka A, Mackie AR et al. Specificity of infant digestive conditions: some clues for developing relevant in vitro models. Crit Rev Food Sci Nutr [Internet]. 2014 [cited 2022 Jan 6];54(11):1427–57. Available from: https://pubmed.ncbi.nlm.nih.gov/24580539/. Bourlieu C, Ménard O, Bouzerzour K, Mandalari G, Macierzanka A, Mackie AR et al. Specificity of infant digestive conditions: some clues for developing relevant in vitro models. Crit Rev Food Sci Nutr [Internet]. 2014 [cited 2022 Jan 6];54(11):1427–57. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​24580539/​.
3.
Zurück zum Zitat Indrio F, Neu J, Pettoello-Mantovani M, Marchese F, Martini S, Salatto A et al. Development of the Gastrointestinal Tract in Newborns as a Challenge for an Appropriate Nutrition: A Narrative Review. Nutrients [Internet]. 2022 Apr 1 [cited 2023 Aug 25];14(7). Available from: https://pubmed.ncbi.nlm.nih.gov/35406018/. Indrio F, Neu J, Pettoello-Mantovani M, Marchese F, Martini S, Salatto A et al. Development of the Gastrointestinal Tract in Newborns as a Challenge for an Appropriate Nutrition: A Narrative Review. Nutrients [Internet]. 2022 Apr 1 [cited 2023 Aug 25];14(7). Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​35406018/​.
4.
5.
Zurück zum Zitat Vandenplas Y, Hauser B, Salvatore S. Functional Gastrointestinal Disorders in Infancy: Impact on the Health of the Infant and Family. Pediatr Gastroenterol Hepatol Nutr [Internet]. 2019 May 1 [cited 2023 Aug 25];22(3):207. Available from: /pmc/articles/PMC6506429/. Vandenplas Y, Hauser B, Salvatore S. Functional Gastrointestinal Disorders in Infancy: Impact on the Health of the Infant and Family. Pediatr Gastroenterol Hepatol Nutr [Internet]. 2019 May 1 [cited 2023 Aug 25];22(3):207. Available from: /pmc/articles/PMC6506429/.
9.
Zurück zum Zitat Manios Y, Karaglani E, Thijs-Verhoeven I, Vlachopapadopoulou E, Papazoglou A, Maragoudaki E et al. Effect of milk fat-based infant formulae on stool fatty acid soaps and calcium excretion in healthy term infants: two double-blind randomised cross-over trials. BMC Nutr [Internet]. 2020 Dec [cited 2023 Feb 13];6(1). Available from: https://pubmed.ncbi.nlm.nih.gov/32944265/. Manios Y, Karaglani E, Thijs-Verhoeven I, Vlachopapadopoulou E, Papazoglou A, Maragoudaki E et al. Effect of milk fat-based infant formulae on stool fatty acid soaps and calcium excretion in healthy term infants: two double-blind randomised cross-over trials. BMC Nutr [Internet]. 2020 Dec [cited 2023 Feb 13];6(1). Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​32944265/​.
10.
Zurück zum Zitat Stroebinger N, Rutherfurd SM, Henare SJ, Hernandez JFP, Moughan PJ. Fatty Acids from Different Fat Sources and Dietary Calcium Concentration Differentially Affect Fecal Soap Formation in Growing Pigs. J Nutr [Internet]. 2021 May 1 [cited 2023 Jan 24];151(5):1102–10. Available from: https://pubmed.ncbi.nlm.nih.gov/33880549/. Stroebinger N, Rutherfurd SM, Henare SJ, Hernandez JFP, Moughan PJ. Fatty Acids from Different Fat Sources and Dietary Calcium Concentration Differentially Affect Fecal Soap Formation in Growing Pigs. J Nutr [Internet]. 2021 May 1 [cited 2023 Jan 24];151(5):1102–10. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​33880549/​.
11.
Zurück zum Zitat Braegger C, Chmielewska A, Decsi T, Kolacek S, Mihatsch W, Moreno L et al. Supplementation of infant formula with probiotics and/or prebiotics: a systematic review and comment by the ESPGHAN committee on nutrition. J Pediatr Gastroenterol Nutr [Internet]. 2011 Feb [cited 2022 Nov 28];52(2):238–50. Available from: https://pubmed.ncbi.nlm.nih.gov/21150647/. Braegger C, Chmielewska A, Decsi T, Kolacek S, Mihatsch W, Moreno L et al. Supplementation of infant formula with probiotics and/or prebiotics: a systematic review and comment by the ESPGHAN committee on nutrition. J Pediatr Gastroenterol Nutr [Internet]. 2011 Feb [cited 2022 Nov 28];52(2):238–50. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​21150647/​.
12.
Zurück zum Zitat Szajewska H, Canani RB, Domellöf M, Guarino A, Hojsak I, Indrio F et al. Probiotics for the management of pediatric gastrointestinal disorders: position paper of the ESPGHAN Special Interest Group on Gut Microbiota and modifications. J Pediatr Gastroenterol Nutr. 2022. Szajewska H, Canani RB, Domellöf M, Guarino A, Hojsak I, Indrio F et al. Probiotics for the management of pediatric gastrointestinal disorders: position paper of the ESPGHAN Special Interest Group on Gut Microbiota and modifications. J Pediatr Gastroenterol Nutr. 2022.
13.
Zurück zum Zitat Kennedy K, Fewtrell MS, Morley R, Abbott R, Quinlan PT, Wells JCK et al. Double-blind, randomized trial of a synthetic triacylglycerol in formula-fed term infants: effects on stool biochemistry, stool characteristics, and bone mineralization. Am J Clin Nutr [Internet]. 1999 [cited 2022 Nov 28];70(5):920–7. Available from: https://pubmed.ncbi.nlm.nih.gov/10539755/. Kennedy K, Fewtrell MS, Morley R, Abbott R, Quinlan PT, Wells JCK et al. Double-blind, randomized trial of a synthetic triacylglycerol in formula-fed term infants: effects on stool biochemistry, stool characteristics, and bone mineralization. Am J Clin Nutr [Internet]. 1999 [cited 2022 Nov 28];70(5):920–7. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​10539755/​.
14.
Zurück zum Zitat Chappell JE, Clandinin MT, Kearney-Volpe C, Reichman B, Swyer PW. Fatty acid balance studies in premature infants fed human milk or formula: effect of calcium supplementation. J Pediatr [Internet]. 1986 [cited 2022 Nov 28];108(3):439–47. Available from: https://pubmed.ncbi.nlm.nih.gov/3512812/. Chappell JE, Clandinin MT, Kearney-Volpe C, Reichman B, Swyer PW. Fatty acid balance studies in premature infants fed human milk or formula: effect of calcium supplementation. J Pediatr [Internet]. 1986 [cited 2022 Nov 28];108(3):439–47. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​3512812/​.
15.
Zurück zum Zitat van Lieshout GAA, Lambers TT, Bragt MCE, Hettinga KA. How processing may affect milk protein digestion and overall physiological outcomes: A systematic review. Crit Rev Food Sci Nutr [Internet]. 2020 Aug 5 [cited 2022 Nov 28];60(14):2422–45. Available from: https://pubmed.ncbi.nlm.nih.gov/31437019/. van Lieshout GAA, Lambers TT, Bragt MCE, Hettinga KA. How processing may affect milk protein digestion and overall physiological outcomes: A systematic review. Crit Rev Food Sci Nutr [Internet]. 2020 Aug 5 [cited 2022 Nov 28];60(14):2422–45. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​31437019/​.
16.
Zurück zum Zitat Zenker HE, Van Lieshout GAA, Van Gool MP, Bragt MCE, Hettinga KA. Lysine blockage of milk proteins in infant formula impairs overall protein digestibility and peptide release. Food Funct. 2020;11(1):358–69.CrossRefPubMed Zenker HE, Van Lieshout GAA, Van Gool MP, Bragt MCE, Hettinga KA. Lysine blockage of milk proteins in infant formula impairs overall protein digestibility and peptide release. Food Funct. 2020;11(1):358–69.CrossRefPubMed
17.
Zurück zum Zitat World Medical Association. World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191–4.CrossRef World Medical Association. World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191–4.CrossRef
18.
Zurück zum Zitat Bekkali N, Hamers SL, Reitsma JB, Van Toledo L, Benninga MA. Infant Stool Form Scale: development and results. J Pediatr. 2009;154(4):521–526e1.CrossRefPubMed Bekkali N, Hamers SL, Reitsma JB, Van Toledo L, Benninga MA. Infant Stool Form Scale: development and results. J Pediatr. 2009;154(4):521–526e1.CrossRefPubMed
19.
Zurück zum Zitat Riley AW, Trabulsi J, Yao M, Bevans KB, DeRusso PA. Validation of a parent report questionnaire: the infant gastrointestinal Symptom Questionnaire. Clin Pediatr (Phila). 2015;54(12):1167–74.CrossRefPubMed Riley AW, Trabulsi J, Yao M, Bevans KB, DeRusso PA. Validation of a parent report questionnaire: the infant gastrointestinal Symptom Questionnaire. Clin Pediatr (Phila). 2015;54(12):1167–74.CrossRefPubMed
20.
Zurück zum Zitat Koninckx CR, Donat E, Benninga MA, Broekaert IJ, Gottrand F, Kolho KL et al. The Use of Fecal Calprotectin Testing in Paediatric Disorders: A Position Paper of the European Society for Paediatric Gastroenterology and Nutrition Gastroenterology Committee. J Pediatr Gastroenterol Nutr [Internet]. 2021 Apr 1 [cited 2023 Jan 24];72(4):617–40. Available from: https://pubmed.ncbi.nlm.nih.gov/33716293/. Koninckx CR, Donat E, Benninga MA, Broekaert IJ, Gottrand F, Kolho KL et al. The Use of Fecal Calprotectin Testing in Paediatric Disorders: A Position Paper of the European Society for Paediatric Gastroenterology and Nutrition Gastroenterology Committee. J Pediatr Gastroenterol Nutr [Internet]. 2021 Apr 1 [cited 2023 Jan 24];72(4):617–40. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​33716293/​.
22.
Zurück zum Zitat Sheng XY, Buthmanaban V, Van Lieshout GAA, Parikh P. Reduced Occurrence of Gastrointestinal Symptoms in Chinese Infants Fed Minimally Processed Commercially Available Formula: A Cross-Sectional Observational Study. J Nutr Metab [Internet]. 2020 [cited 2023 Jan 26];2020. Available from: https://pubmed.ncbi.nlm.nih.gov/32300486/. Sheng XY, Buthmanaban V, Van Lieshout GAA, Parikh P. Reduced Occurrence of Gastrointestinal Symptoms in Chinese Infants Fed Minimally Processed Commercially Available Formula: A Cross-Sectional Observational Study. J Nutr Metab [Internet]. 2020 [cited 2023 Jan 26];2020. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​32300486/​.
23.
Zurück zum Zitat Li F, Ma J, Geng S, Wang J, Liu J, Zhang J et al. Fecal Calprotectin Concentrations in Healthy Children Aged 1–18 Months. PLoS One [Internet]. 2015 Mar 5 [cited 2023 Jan 19];10(3):119574. Available from: /pmc/articles/PMC4351193/. Li F, Ma J, Geng S, Wang J, Liu J, Zhang J et al. Fecal Calprotectin Concentrations in Healthy Children Aged 1–18 Months. PLoS One [Internet]. 2015 Mar 5 [cited 2023 Jan 19];10(3):119574. Available from: /pmc/articles/PMC4351193/.
24.
Zurück zum Zitat Nordin E, Hellström PM, Brunius C, Landberg R. Modest Conformity Between Self-Reporting of Bristol Stool Form and Fecal Consistency Measured by Stool Water Content in Irritable Bowel Syndrome and a FODMAP and Gluten Trial. Am J Gastroenterol [Internet]. 2022 Oct 1 [cited 2023 Mar 13];117(10):1668–74. Available from: https://pubmed.ncbi.nlm.nih.gov/36087104/. Nordin E, Hellström PM, Brunius C, Landberg R. Modest Conformity Between Self-Reporting of Bristol Stool Form and Fecal Consistency Measured by Stool Water Content in Irritable Bowel Syndrome and a FODMAP and Gluten Trial. Am J Gastroenterol [Internet]. 2022 Oct 1 [cited 2023 Mar 13];117(10):1668–74. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​36087104/​.
25.
Zurück zum Zitat Quinlan PT, Lockton S, Irwin J, Lucas AL. The relationship between stool hardness and stool composition in breast- and formula-fed infants. J Pediatr Gastroenterol Nutr. 1995;20(1):81–90.CrossRefPubMed Quinlan PT, Lockton S, Irwin J, Lucas AL. The relationship between stool hardness and stool composition in breast- and formula-fed infants. J Pediatr Gastroenterol Nutr. 1995;20(1):81–90.CrossRefPubMed
26.
Zurück zum Zitat Den Hertog J, Van Leengoed E, Kolk F, Van Den Broek L, Kramer E, Bakker EJ et al. The defecation pattern of healthy term infants up to the age of 3 months. Arch Dis Child Fetal Neonatal Ed [Internet]. 2012 Nov 1 [cited 2023 Feb 13];97(6):F465–70. Available from: https://fn.bmj.com/content/97/6/F465. Den Hertog J, Van Leengoed E, Kolk F, Van Den Broek L, Kramer E, Bakker EJ et al. The defecation pattern of healthy term infants up to the age of 3 months. Arch Dis Child Fetal Neonatal Ed [Internet]. 2012 Nov 1 [cited 2023 Feb 13];97(6):F465–70. Available from: https://​fn.​bmj.​com/​content/​97/​6/​F465.
28.
Zurück zum Zitat Sheng XY, Buthmanaban V, Vonk MM, Feitsma AL, Parikh P. Reduced crying and favourable stool characteristics in Chinese infants fed milk fat-based formula. Asia Pac J Clin Nutr [Internet]. 2020 [cited 2023 Aug 28];29(1):144–51. Available from: https://pubmed.ncbi.nlm.nih.gov/32229453/. Sheng XY, Buthmanaban V, Vonk MM, Feitsma AL, Parikh P. Reduced crying and favourable stool characteristics in Chinese infants fed milk fat-based formula. Asia Pac J Clin Nutr [Internet]. 2020 [cited 2023 Aug 28];29(1):144–51. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​32229453/​.
29.
Zurück zum Zitat Nyakayiru J, Van Lieshout GAA, Trommelen J, Van Kranenburg J, Verdijk LB, Bragt MCE et al. The glycation level of milk protein strongly modulates post-prandial lysine availability in humans. Br J Nutr [Internet]. 2020 [cited 2023 Feb 13];123(5):545–52. Available from: https://pubmed.ncbi.nlm.nih.gov/31727194/. Nyakayiru J, Van Lieshout GAA, Trommelen J, Van Kranenburg J, Verdijk LB, Bragt MCE et al. The glycation level of milk protein strongly modulates post-prandial lysine availability in humans. Br J Nutr [Internet]. 2020 [cited 2023 Feb 13];123(5):545–52. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​31727194/​.
30.
Zurück zum Zitat Huppertz T, Timmer C. Salt equilibria in nutritional formulae for infants and young children. Int Dairy J. 2020;110:104805.CrossRef Huppertz T, Timmer C. Salt equilibria in nutritional formulae for infants and young children. Int Dairy J. 2020;110:104805.CrossRef
31.
Zurück zum Zitat Bavaro SL, Mamone G, Picariello G, Callanan MJ, Chen Y, Brodkorb A et al. Thermal or membrane processing for Infant Milk Formula: Effects on protein digestion and integrity of the intestinal barrier. Food Chem [Internet]. 2021 Jun 15 [cited 2023 Jan 24];347. Available from: https://pubmed.ncbi.nlm.nih.gov/33484955/. Bavaro SL, Mamone G, Picariello G, Callanan MJ, Chen Y, Brodkorb A et al. Thermal or membrane processing for Infant Milk Formula: Effects on protein digestion and integrity of the intestinal barrier. Food Chem [Internet]. 2021 Jun 15 [cited 2023 Jan 24];347. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​33484955/​.
32.
Zurück zum Zitat Huppertz T, Lambers TT. Influence of micellar calcium phosphate on in vitro gastric coagulation and digestion of milk proteins in infant formula model systems. Int Dairy J. 2020;107. Huppertz T, Lambers TT. Influence of micellar calcium phosphate on in vitro gastric coagulation and digestion of milk proteins in infant formula model systems. Int Dairy J. 2020;107.
33.
Zurück zum Zitat Zenker HE, Wichers HJ, Tomassen MMM, Boeren S, De Jong NW, Hettinga KA. Peptide Release after Simulated Infant In Vitro Digestion of Dry Heated Cow’s Milk Protein and Transport of Potentially Immunoreactive Peptides across the Caco-2 Cell Monolayer. Nutrients [Internet]. 2020 Aug 1 [cited 2023 Feb 13];12(8):1–24. Available from: https://pubmed.ncbi.nlm.nih.gov/32824739/. Zenker HE, Wichers HJ, Tomassen MMM, Boeren S, De Jong NW, Hettinga KA. Peptide Release after Simulated Infant In Vitro Digestion of Dry Heated Cow’s Milk Protein and Transport of Potentially Immunoreactive Peptides across the Caco-2 Cell Monolayer. Nutrients [Internet]. 2020 Aug 1 [cited 2023 Feb 13];12(8):1–24. Available from: https://​pubmed.​ncbi.​nlm.​nih.​gov/​32824739/​.
Metadaten
Titel
A cross-sectional study on stool- and gastrointestinal-related outcomes of Mexican infants consuming different formulae
verfasst von
Carlijn M. Maasakkers
Jeske H.J. Hageman
Olivia Balcazar Muñoz
Tomás Gómez Tamayo
Andrés Blanco Montero
Luis Gerardo Garza Lara
Regina Flores-López
Miriam Contreras Fernández
Sofía Morán Ramos
Tim T. Lambers
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Pediatrics / Ausgabe 1/2023
Elektronische ISSN: 1471-2431
DOI
https://doi.org/10.1186/s12887-023-04426-y

Weitere Artikel der Ausgabe 1/2023

BMC Pediatrics 1/2023 Zur Ausgabe

Mit dem Seitenschneider gegen das Reißverschluss-Malheur

03.06.2024 Urologische Notfallmedizin Nachrichten

Wer ihn je erlebt hat, wird ihn nicht vergessen: den Schmerz, den die beim Öffnen oder Schließen des Reißverschlusses am Hosenschlitz eingeklemmte Haut am Penis oder Skrotum verursacht. Eine neue Methode für rasche Abhilfe hat ein US-Team getestet.

Reanimation bei Kindern – besser vor Ort oder während Transport?

29.05.2024 Reanimation im Kindesalter Nachrichten

Zwar scheint es laut einer Studie aus den USA und Kanada bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Alter der Mutter beeinflusst Risiko für kongenitale Anomalie

28.05.2024 Kinder- und Jugendgynäkologie Nachrichten

Welchen Einfluss das Alter ihrer Mutter auf das Risiko hat, dass Kinder mit nicht chromosomal bedingter Malformation zur Welt kommen, hat eine ungarische Studie untersucht. Sie zeigt: Nicht nur fortgeschrittenes Alter ist riskant.

Begünstigt Bettruhe der Mutter doch das fetale Wachstum?

Ob ungeborene Kinder, die kleiner als die meisten Gleichaltrigen sind, schneller wachsen, wenn die Mutter sich mehr ausruht, wird diskutiert. Die Ergebnisse einer US-Studie sprechen dafür.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.