Skip to main content
Erschienen in: BMC Pregnancy and Childbirth 1/2022

Open Access 01.12.2022 | Research

Effect of breast milk with or without bacteria on infant gut microbiota

verfasst von: Ting Huang, Zichun Zeng, Xinyuan Liang, Xiaomei Tang, Huijuan Luo, Dongju Wang, Juan Zhou, Xiaomin Xiao

Erschienen in: BMC Pregnancy and Childbirth | Ausgabe 1/2022

Abstract

Background

The breast milk microbiome could be a source of infant intestinal microbiota. Several studies have found that some breast milk is extremely low in bacteria or is even sterile. There are limited studies on the effect of milk without bacteria on the infant gut microbiota. The purpose of this study was to investigate the gut microbiota of infants fed with bacterial milk or sterile milk. Meanwhile, we attempted to find the cause of undetectable bacteria in milk.

Methods

A total of 17 healthy pregnant women and 17 infants were enrolled in this study. Fecal samples were collected from full-term pregnant women. Milk samples and infant fecal samples were collected on the 14th postnatal day. Breast milk and fecal samples were examined using 16S rRNA sequencing technology. Pregnant women and infants were grouped according to milk with or without bacteria. To compare the differences in gut microbiota and clinical characteristics between groups.

Results

Bacteria were detected in 11 breast milk samples, and the bacterial detection rate was 64.7%. Infants fed with bacterial milk showed higher Shannon index and Simpson index (P = 0.020, P = 0.048), and their relative abundance of Lachnospirales, Lachnospiraceae and Eggerthellaceae was markedly higher. In addition, there were more bacterial associations in the co-occurrence network of infants fed with bacterial milk. Pregnant women with sterile and bacterial breast milk showed no significant differences in their clinical characteristics, and microbial composition and diversity.

Conclusions

Some breast milk from healthy postpartum women failed to be sequenced due to low microbial DNA quantities or is sterile. Research is needed to explore the reasons for this phenomenon. Infants fed with bacterial milk had higher Alpha diversity and more complex microbiota networks. These findings provide novel insight into milk microbiota and infant gut microbiota.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12884-022-04930-6.
Ting Huang and Zichun Zeng contributed equally to this study.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
OTUs
Operational taxonomic units
PCOA
Principal Coordinates Analysis
LEfSe
Linear discriminant analysis effect size

Background

Human milk provides the best nutrition for infants in early life. The sugar, lipids, proteins, macronutrients, and micronutrients in breast milk provide rich nourishment for infants. With the development of high-throughput sequencing technology, it was discovered that microorganisms are also present in human breast milk. Milk microbes can colonize the infant gut, affecting the establishment and development of the gut microbiome [1]. Early development of the infant gut microbiome is related to allergic disorders, atopic dermatitis, and asthma [25]. As a result, breast milk plays a critical role in the health of infants and the colonization of the intestinal microbial community.
In our previous study, 11 out of 25 milk samples from healthy pregnant women failed to be sequenced due to an insufficient quantity of bacteria [6]. This phenomenon was also observed in other studies [713]. It indicated that not all healthy lactating women’s milk contains microbiota. Currently, it is unclear what causes this phenomenon and whether infants fed with sterile milk have different microbial communities. Most studies on infant intestinal microbiota have focused on the effects of delivery modes [14], feeding modes [15], bioactive components of milk [16], and maternal diet [17, 18]. Limited data are available on the comparison of gut microbiota in infants fed with bacterial milk and sterile milk. A number of studies reported that delivery mode, geographical location, postpartum period, gestational age, feeding methods, BMI, and infant gender may cause individual differences in the composition of milk microbiota [12, 1922]. In addition, it has been suggested that there may be an entero-mammary pathway in the human body, whereby dendritic cells were able to open tight junctions between intestinal epithelial cells and transported some bacteria from the intestine to the breast [23, 24]. This bacterial translocation occurs more frequently in women during late pregnancy and lactation [25]. Therefore, we speculated that gut microbiota may influence the construction of breast milk microbiota.
In this study, we detected the microbial communities from mother-neonate pairs’ feces and maternal milk. Our goal was to investigate whether bacterial milk and sterile milk have different effects on infant gut microbiota and whether gut microbiota influences the construction of the milk microbiota.

Material and methods

Participants

The participants in this observational study were 20–35 years old women who achieved singleton pregnancy spontaneously. Furthermore, women with pregnancy complications, infectious diseases or chronic diseases, as well as women who had used antibiotics or probiotics during pregnancy were excluded from the study. According to the above criteria, 23 pregnant women from the First Affiliated Hospital of Jinan University (Guangzhou, China) were enrolled in our study. All of the pregnant women delivered vaginally, and their infants’ Apgar scores were all normal. Meanwhile, their infants were also included in this study. All pregnant women who participated in the study signed an informed consent form.

Collection of sample and clinical data

Fecal samples were taken from pregnant women after full term of pregnancy. On the 14th postnatal day, breast milk and infant feces were collected. Fecal samples were collected by professionals using sterile spoons and sterile boxes. Before collecting the milk, the mothers cleaned the nipple and surrounding skin with soap and water. Women wore sterile gloves to squeeze out the milk and filled in sterile tubes. All samples were transported on ice and then stored in the refrigerator at -80℃ until DNA extraction. We also collected clinical information about pregnant women and infants, such as maternal age, height, weight, gestational weeks, neonatal length, weight, and gender.

DNA Extraction and sequencing

The CTAB/SDS technique was used to extract total bacterial DNA from samples. DNA concentration and purity were monitored on a 1% agarose gel. According to the concentration, DNA was diluted to 1 ng/μL with sterile water. The V3-V4 hypervariable region of 16S rRNA gene was amplified using a specific primer (341F-806R) with the barcode. All PCR reactions were performed with Phusion® High-Fidelity PCR Master Mix (New England Biolabs). The PCR products were mixed with the same volume of 1X loading buffer (containing SYB Green) and electrophoresed on a 2 percent agarose gel. PCR products were mixed in equal density ratio. The mixed PCR products were then purified using Qiagen Gel Extraction Kit (Qiagen, Germany). Sequencing libraries were generated by using TruSeq® DNA PCR-Free Sample Preparation Kit. The library was sequenced on a NovaSeq6000 platform after assessing on the Qubit@ 2.0 Fluorometer.

OTU cluster and Species annotation

Each read was stripped of the barcode and the primer, and then they were spliced by using FLASH (V1.2.7, http://​ccb.​jhu.​edu/​software/​FLASH/​) [26]. Next, effective tags were generated by intercepting, filtering, and removing chimeric sequences from the raw tags. The Uparse algorithm (Uparse v7.0.1001,http://​www.​drive5.​com/​uparse/​) was used to cluster all effective tags into sequences, and then all sequences were clustered into operational taxonomic units (OTUs) with 97% identity. Finally, representative sequences from each OTU were aligned to the SSUrRNA database (http://​www.​arb-silva.​de/​).

Statistical and microbial analysis

Alpha diversity was used to assess diversity within samples, whereas Beta diversity was used to compare the composition of microbial communities between samples. The Simpson index and Shannon index were calculated in QIIME software (Version 1.7.0) to estimate Alpha diversity. A higher Shannon and Simpson index implies a community with greater diversity. Comparison of Alpha diversity indices between groups using the Wilcoxon test with GraphPad Prism software (Version 8.0.2). Beta diversity was demonstrated using Principal Coordinates Analysis (PCoA) based on weighted Unifrac distances. To estimate the correlation between genera, we calculated the Spearman coefficients for the top 100 genera by relative abundance. Genera with absolute Spearman correlation coefficients greater than 0.6 and P values less than 0.05 were considered for construction of the network. The visualization of each network and calculation of topological properties were performed using Gephi software (Version 0.9.2). The relative abundance histogram and the Venn diagram were plotted using R software. To further investigate biomarkers between different groups, we conducted linear discriminant analysis effect size (LEfSe). The clinical data were compared between groups using independent-samples t test or Chi-square test. A P value less than 0.05 was considered statistically significant.

Results

Sample sequencing results and participant characteristics

Seventeen of 23 mother-infant pairs were eventually enrolled in our study. Five pairs were eliminated due to lack of breast milk or personal reasons, and one pair was excluded because the mother suffered from mastitis. Six of the 17 milk samples could not be sequenced due to low bacterial loads or the absence of bacteria. The detection rate of microbe in milk was 64.7%. Breast milk that failed to be sequenced in this study was tentatively considered sterile milk. Based on the sequencing results of breast milk, the subjects were divided into four groups: pregnant women with bacterial milk (PBM, n = 11), pregnant women with sterile milk (PSM, n = 6), infants fed with bacterial milk (IBM, n = 11), infants fed with sterile milk (ISM, n = 6). The rarefaction curves of samples in each group tended to be flat as the sequence number increased, indicating that the sequencing depth was sufficient (Fig. 1a).
There were no significant differences in age, height, weight, BMI, and gestational week between pregnant women, and there were no significant differences in birth weight, length, and gender between infants (Table 1).
Table 1
Characteristics of participants
 
Bacterial milk
Sterile milk
P
Maternal characteristics
 Number of women
11
6
 
 Maternal age (year)
26.6 ± 1.91
26.8 ± 1.60
0.834
 Height (m)
1.6 ± 0.59
1.6 ± 0.06
0.788
 Gestational weight (kg)
64.6 ± 8.45
62.5 ± 5.81
0.599
 Gestational BMI (kg/m2)
25.0 ± 2.52
24.7 ± 3.45
0.804
 Gestational week (weeks)
39.5 ± 0.96
38.9 ± 1.17
0.232
Infant characteristics
 Number of infants
11
6
 
 Birth weight (g)
3200.0 ± 246.98
3083.3 ± 320.42
0.414
 Birth body length (cm)
49.5 ± 1.04
49.0 ± 1.67
0.496
Gender (%)
  
0.627
 Male
54.5
66.7
 
 Female
45.5
33.3
 

Microbiota profile of breast milk

The microbial composition of maternal breast milk was shown in Fig. 1b-c and Table 2. Among the microbial communities of breast milk, Firmicutes predominated (73.1%), followed by Proteobacteria (9.2%), Actinobacteria (6.3%), and Bacteroidetes (2.2%), while the relative abundance of the rest of the phylum was less than 1%. At the genus level, eight genera had mean relative abundances greater than 1%. They were Streptococcus (30.1%), Staphylococcus (20.2%), Bacillus (7.4%), Gemella (5.7%), Rothia (5.4%), Acinetobacter (4.3%), Lactobacillus (2.5%), and Clostridium_sensu_stricto_1 (1.1%). Among the above genera, 73.1% of the bacteria were aerobic or facultative anaerobes, and 3.6% were anaerobes (Table 3).
Table 2
Relative abundances of the top four taxonomy at the phylum level for each group (%)
 
HM
PBM
PSM
IBM
ISM
Firmicutes
73.1
66.5
57.0
20.2
16.4
Proteobacteria
9.2
3.3
4.0
32.8
38.0
Actinobacteria
6.3
2.5
6.1
31.0
37.4
Bacteroidetes
2.2
24.6
30.8
15.0
8.2
HM Human milk, PBM Pregnant women with bacterial milk, PSM Pregnant women with sterile milk, IBM Infants fed with bacterial milk, ISM Infants fed with sterile milk
Table 3
Milk bacteria with relative abundance greater than 1% and their relationship with oxygen
 
Relative abundance (%)
Relationship with oxygen
Streptococcus
30.1
Facultative anaerobic
Staphylococcus
20.2
Facultative anaerobic
Bacillus
7.4
Aerobic or facultative anaerobic
Gemella
5.7
Aerobic or facultative anaerobic
Rothia
5.4
Aerobic
Acinetobacter
4.3
Obligate aerobic
Lactobacillus
2.5
Anaerobic
Clostridium_sensu_stricto_1
1.1
Strict anaerobic
Other
23.3
-
The relationship between genera and oxygen was obtained by searching the eighth edition of Berger’s Handbook of Bacteria
It was noted that specific gut bacteria such as Bifidobacterium and Bacteroidetes were present in 10 out of 11 milk samples. The presence of Bifidobacterium and Bacteroides was 90.9%, but their relative abundances were less than 1.0% (Supplementary Table S1).

The influence of milk microbiota on infant gut microbiota

Infants fed bacterial milk and infants fed sterile milk had similar intestinal microbial compositions, but microbial diversity varied. Proteobacteria and Actinobacteria constituted the dominant phylum in infant feces (Fig. 1b-c, Table 2). The most abundant genera were Bifidobacterium, Escherichia-Shigella, and Bacteroides (Fig. 1c). However, the IBM group had significantly higher Shannon and Simpson indices than the ISM group (P = 0.020, P = 0.048), revealing that infants fed bacterial milk have a richer microbial community (Fig. 2a-b). Beta diversity was estimated by using PCoA to seek interindividual differences in microbial communities. PCoA indicated similar clustering of microbiota among the IBM and ISM groups (P = 0.264, Fig. 2c).
We then drew a Venn diagram to visualize the shared and unique OTUs of the IBM and ISM groups. There were 273 shared OTUs between groups. Meanwhile, the IBM group has 1063 unique OTUs, which far exceeds those in the ISM group (Fig. 2d). This suggested that infants fed bacterial milk were more enriched with microbes.
The LEfSe analysis was performed to find biomarkers between the two groups. As illustrated in Fig. 2e, we found that Eggerthellaceae, Lachnospiraceae, and Lachnospiraceae were enriched in the IBM group. The abundance comparison diagram showed that Eggerthellaceae are absent from the ISM group, and Lachnospiraceae are only found in one infant gut (Fig. 2f-g).
Network analysis can reveal the internal relationships of a microbial community. We constructed the co-occurrence network and calculated the topological properties. The number of nodes, edges, and average degree in microbial network were higher in the IBM group (Fig. 2h-i, Table 4), which indicated the microbial community in infants fed with bacterial milk was more connected and complex.
Table 4
Topological properties of co-occurrence network
 
IBM (n = 11)
ISM (n = 6)
Network diameter
8.0
3.0
Clustering coefficient
0.6
0.8
Average degree
6.2
2.5
Modularity
0.6
0.5
Nodes
44.0
25.0
Edges
136.0
31.0
Network diameter: The maximum distance between any two nodes in a network. Clustering coefficient: The percentage of neighbors of a node that can reach another neighbor without passing through it. Average degree: Edges of all nodes divided by total number of nodes. A network with a higher average degree is more complex. Modularity: A parameter that assesses whether the network can be divided into several modules. IBM Infants fed with bacterial milk, ISM Infants fed with sterile milk

Microbial analysis of maternal gut microbiota

As there were no significant differences in clinical characteristics between pregnant women, the gut microbiota was further examined. The microbial composition of PBM and PSM groups was similar. The primary phyla were Firmicutes and Bacteroidetes, while the main genera were Faecalibacterium and Bacteroides. In addition, Bifidobacterium was more abundant in the PSM group (5.3%) than in the PBM group (1.5%) (Supplementary Table S2).
To evaluate variations in the community structure and diversity between pregnant women with or without bacterial milk, we performed Alpha and Beta diversity. Shannon and Simpson indices revealed no significant differences between the PBM and PSM groups (P = 0.591, P = 0.180) (Fig. 3a-b). Meanwhile, PCoA showed no separation of communities between the PBM and PSM groups, suggesting that the two bacterial communities had similar compositions and structures (Fig. 3c).
To further search for specific taxa between the two groups, we performed a LEfSe analysis. A total of five biomarkers were identified when LDA score = 4. The PSM group had significantly more Actinobacteria, Bifidobacteriales, Bifidobacteriaceae, Prevotellaceae, and Bifidobacterium than the PBM group (Fig. 3d). The evolutionary branch diagram demonstrated that Actinobacteria, Bifidobacteriales, Bifidobacteriaceae and Bifidobacterium belong to the same branch (Fig. 3e). Co-occurrence networks were constructed to exhibit the relationship between microbes of pregnant women, focusing on the relationship between Bifidobacterium and other genera. It is worth noting that Bifidobacterium negatively correlated with six genera in the PSM group, while it negatively correlated with only one genus in the PBM group (Fig. 3f-g).

Discussion

In the current study, we found some breast milk collected from healthy women does not contain any microbial community. We then analyzed the microbiota in breast milk and feces from infants and pregnant women. Our study showed that bacterial milk and sterile milk have different effects on infant gut microbiota. We did not prove that the gut microbiota of pregnant women affected the presence of microbes in milk. Nonetheless, as far as we know, this is the first report to compare intestinal microbiota in infants fed with bacterial milk and sterile milk.

Healthy women milk microbial profile

In this study, microbes were detected in 11 of 17 breast milk samples, with a positive detection rate of 64.7%. This result was close to the detection rate of our previous study [6] and the study of Kordy et al. [7]. Many researchers have detected microorganisms in human breast milk. Firmicutes was reported to be the most abundant phylum in milk [27, 28]. The common genera in breast milk included Staphylococcus, Streptococcus, Pseudomonas, and Lactobacillus [20, 2932]. In this study, Firmicutes were abundant in breast milk. Meanwhile, Streptococcus and Staphylococcus had the highest abundance, which was generally consistent with other studies [20, 27, 31, 32]. Furthermore, the milk was also rich in Lactobacillus in our study. However, the abundance of Pseudomonas in breast milk was low, and we detected the commensal skin bacterium Propionibacterium in only one milk sample. A previous research [19] comparing the breast milk bacterial communities of mothers from Spain, Finland, South Africa, and China, has discovered higher levels of Pseudomonas and Propionibacterium in the breast milk of Spanish mothers who delivered by cesarean section than those from other countries. Furthermore, Pseudomonas was much lower in Chinese women who delivered vaginally compared to those who delivered by cesarean section [19]. Therefore, we attribute differences in results to geographic location and delivery mode.
The detection rate of Bifidobacterium and Bacteroides in milk was as high as 90.9%. These bacteria are strict anaerobes that often reside in the gut and are unlikely to originate from the skin. A study has found the existence of gut-associated bacteria such as Bifidobacterium in maternal and infant feces and breast milk [25]. Moreover, Jost et al. [33] found a strain of Bifidobacterium in all three ecosystems of a mother-neonate pair. These findings indicate that some gut microbes can enter milk and then colonize neonatal intestinal.

Milk microbiota may affect the formation of neonatal gut microbiota

It has been reported that delivery modes, feeding modes, gestational age at birth and timing of solid food introduction may affect bacterial communities in infant feces [1, 14, 15, 34]. Our study manifested that the presence or absence of milk microbes has a minor effect on infant gut microbial composition but has a greater impact on microbial diversity and microbial network relationships. Proteobacteria and Actinobacteria constituted the dominant phylum in infant feces, which was in accordance with Pannaraj’s report [1]. After being fed with bacterial milk, Alpha diversity and microbial network associations increased. By contrast, infants fed with sterile milk had fewer microbial species and lower microbial network complexity. Alpha diversity has been regarded as a marker of microbiome health, and a reduction in Alpha diversity is seen as a manifestation of intestinal malnutrition [35]. Low diversity is thought to be related to metabolic disorders [36]. In addition, it has been reported that microbial network stability is related to its complexity, and the complexity of the network will contribute to its stability [37]. Lachnospiraceae, which was abundant in the IBM group, is described as a potential beneficial bacteria [38]. Lachnospiraceae can generate butyrate by hydrolyzing amylase and sugar. Butyrate provides energy to colon cells and maintains the integrity of the intestinal barrier. It appears that bacterial milk may help infants shape a healthy gut microbiome. However, long-term follow up is required to track infants’ health outcomes.
Notably, the microbial composition of the IBM and ISM groups was similar, although some breast milk was not detected for bacteria. Bifidobacterium and Bacteroides were also enriched in the ISM group, which indicated that they could be transmitted to newborns in multiple ways. The researchers identified DNA and cellular structure of intestinal bacteria such as Bifidobacterium in the placenta, amniotic fluid, and fetal membrane [3941], suggesting that the intestinal microbiota such as Bifidobacterium may have colonized the fetal intestine during pregnancy. In addition, prebiotics such as human milk oligosaccharides can promote the proliferation and growth of beneficial bacteria such as Bifidobacterium and Lactobacillus [16]. Therefore, we still recommend breastfeeding even if maternal milk contains extremely low bacteria or is even sterile. It is imperative to carefully consider whether microbiota interventions are necessary for infants fed with sterile milk.

The reason why the bacteria were not detected in the milk is unclear

Geographical location, delivery mode, maternal BMI, infant gender and feeding mode could affect milk microbiota composition [20, 22, 27, 42, 43]. To figure out why some milk had very low bacterial content or was sterile, the participants’ characteristics were compared. The pregnant women all live in Guangzhou, China. Meanwhile, they delivered vaginally and fed their newborns exclusively with breast milk. There were no significant differences between the two groups in geographical location, delivery mode and feeding mode. Other clinical characteristics, such as age, BMI, gestational week and infant gender, showed no significant differences.
Since some researchers have proposed the entero-mammary pathway theory, we analyzed the gut microbiota of the PBM group and the PSM group. Gut microbiota composition and diversity were similar between the two groups. The main difference was that Prevotellaceae and Bifidobacterium were abundant in the PSM group, and Bifidobacterium had more negative correlations. Several in vitro and in vivo studies validated that Bifidobacterium was able to affect the rearrangement of tight junction proteins and strengthen the intestinal barrier [4446]. We speculated that Bifidobacterium might inhibit bacterial growth and improve intestinal permeability, leading to a decrease in the migration of microbes entering milk via the entero-mammary pathway. In contrast, Simpson et al. investigated the effect of probiotic supplementation on maternal breast milk and reported that no significant changes occurred in milk microbial community despite maternal supplementation [10]. Of note, only a few samples could be sequenced in both the probiotic and placebo groups due to low bacterial DNA quantities [10]. It has been suggested that newborns’ oral cavity and breast skin may provide additional microbes for milk. Regrettably, we didn’t collect oral samples and skin samples. However, if there are multiple sources of milk microbes, there should be more bacteria in milk. Taken together, further research is needed to determine the cause of undetectable bacteria in milk.

Limitations

Many factors affect milk and gut microbes, such as skin microbes, environmental factors, and diet. Although we attempted to control for differences in race, age, BMI, and delivery mode among participants, it cannot rule out the possibility that other factors influenced the study's results. Our sample size is small, and studies with a larger sample size are needed to reduce the bias. Nevertheless, this study found distinct differences in Alpha diversity and network associations among infants fed with bacterial milk and sterile milk. This result deserves to be of interest.

Conclusions

Overall, some breast milk from healthy postpartum women failed to be sequenced due to low microbial DNA quantities or is sterile. Research is needed to explore the reasons for this phenomenon. Infants fed with bacterial milk had higher Alpha diversity and more complex microbiota networks. These findings provide novel insight into milk microbiota and infant gut microbiota.

Acknowledgements

We thank the Natural Science Foundation of China for funding this research. We also thank pregnant women and their babies for participating in this study. In addition, we thank Biobank of The first affiliated hospital of Jinan University for sample storage.

Declarations

The study project was authorized by the Institutional Review Board (IRB) for Human Subject Research at the First Affiliated Hospital of Jinan University and the approval number was: 2019–011. All methods were performed consistent with the relevant guidelines and regulations of the Declaration of Helsinki. All participants provided written informed consent prior to beginning the study.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis 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.
Literatur
1.
Zurück zum Zitat Pannaraj PS, Li F, Cerini C, Bender JM, Yang S, Rollie A, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017;171:647–54.PubMedPubMedCentralCrossRef Pannaraj PS, Li F, Cerini C, Bender JM, Yang S, Rollie A, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017;171:647–54.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Johnson CC, Ownby DR. The infant gut bacterial microbiota and risk of pediatric asthma and allergic diseases. Transl Res. 2017;179:60–70.PubMedCrossRef Johnson CC, Ownby DR. The infant gut bacterial microbiota and risk of pediatric asthma and allergic diseases. Transl Res. 2017;179:60–70.PubMedCrossRef
3.
Zurück zum Zitat Wang M, Karlsson C, Olsson C, Adlerberth I, Wold AE, Strachan DP, et al. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol. 2008;121:129–34.PubMedCrossRef Wang M, Karlsson C, Olsson C, Adlerberth I, Wold AE, Strachan DP, et al. Reduced diversity in the early fecal microbiota of infants with atopic eczema. J Allergy Clin Immunol. 2008;121:129–34.PubMedCrossRef
4.
Zurück zum Zitat Ismail IH, Oppedisano F, Joseph SJ, Boyle RJ, Licciardi PV, Robins-Browne RM, et al. Reduced gut microbial diversity in early life is associated with later development of eczema but not atopy in high-risk infants. Pediatr Allergy Immunol. 2012;23:674–81.PubMedCrossRef Ismail IH, Oppedisano F, Joseph SJ, Boyle RJ, Licciardi PV, Robins-Browne RM, et al. Reduced gut microbial diversity in early life is associated with later development of eczema but not atopy in high-risk infants. Pediatr Allergy Immunol. 2012;23:674–81.PubMedCrossRef
5.
Zurück zum Zitat Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy. 2014;44:842–50.PubMedCrossRef Abrahamsson TR, Jakobsson HE, Andersson AF, Björkstén B, Engstrand L, Jenmalm MC. Low gut microbiota diversity in early infancy precedes asthma at school age. Clin Exp Allergy. 2014;44:842–50.PubMedCrossRef
6.
Zurück zum Zitat Li YM. Preliminary study on the influencing factors of intestinal flora establishment in 6-month-old infants. MD thesis. Guangzhou: Jinan University; 2020. Li YM. Preliminary study on the influencing factors of intestinal flora establishment in 6-month-old infants. MD thesis. Guangzhou: Jinan University; 2020.
7.
Zurück zum Zitat Kordy K, Gaufin T, Mwangi M, Li F, Cerini C, Lee DJ, et al. Contributions to human breast milk microbiome and enteromammary transfer of Bifidobacterium breve. PLoS One. 2020;15:e0219633.PubMedPubMedCentralCrossRef Kordy K, Gaufin T, Mwangi M, Li F, Cerini C, Lee DJ, et al. Contributions to human breast milk microbiome and enteromammary transfer of Bifidobacterium breve. PLoS One. 2020;15:e0219633.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Padilha M, Brejnrod A, Danneskiold-Samsøe NB, Hoffmann C, de Melo IJ, Cabral VP, et al. Response of the human milk microbiota to a maternal prebiotic intervention is individual and influenced by maternal age. Nutrients. 2020;12:E1081.PubMedCrossRef Padilha M, Brejnrod A, Danneskiold-Samsøe NB, Hoffmann C, de Melo IJ, Cabral VP, et al. Response of the human milk microbiota to a maternal prebiotic intervention is individual and influenced by maternal age. Nutrients. 2020;12:E1081.PubMedCrossRef
9.
Zurück zum Zitat Shin DY, Park J, Yi DY. Comprehensive analysis of the effect of probiotic intake by the mother on human breast milk and infant fecal microbiota. J Korean Med Sci. 2021;36:e58.PubMedPubMedCentralCrossRef Shin DY, Park J, Yi DY. Comprehensive analysis of the effect of probiotic intake by the mother on human breast milk and infant fecal microbiota. J Korean Med Sci. 2021;36:e58.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Simpson MR, Avershina E, Storrø O, Johnsen R, Rudi K, Øien T. Breastfeeding-associated microbiota in human milk following supplementation with Lactobacillus rhamnosus GG, Lactobacillus acidophilus La-5, and Bifidobacterium animalis ssp. lactis Bb-12. J Dairy Sci. 2018;101:889–99.PubMedCrossRef Simpson MR, Avershina E, Storrø O, Johnsen R, Rudi K, Øien T. Breastfeeding-associated microbiota in human milk following supplementation with Lactobacillus rhamnosus GG, Lactobacillus acidophilus La-5, and Bifidobacterium animalis ssp. lactis Bb-12. J Dairy Sci. 2018;101:889–99.PubMedCrossRef
11.
Zurück zum Zitat LeMay-Nedjelski L, Asbury MR, Butcher J, Ley SH, Hanley AJ, Kiss A, et al. Maternal diet and infant feeding practices are associated with variation in the human milk microbiota at 3 months postpartum in a cohort of women with high rates of gestational glucose intolerance. J Nutr. 2020;151:320–9.PubMedCentralCrossRef LeMay-Nedjelski L, Asbury MR, Butcher J, Ley SH, Hanley AJ, Kiss A, et al. Maternal diet and infant feeding practices are associated with variation in the human milk microbiota at 3 months postpartum in a cohort of women with high rates of gestational glucose intolerance. J Nutr. 2020;151:320–9.PubMedCentralCrossRef
12.
Zurück zum Zitat Williams JE, Carrothers JM, Lackey KA, Beatty NF, York MA, Brooker SL, et al. Human milk microbial community structure is relatively stable and related to variations in macronutrient and micronutrient intakes in healthy lactating women. J Nutr. 2017;147:1739–48.PubMedPubMedCentral Williams JE, Carrothers JM, Lackey KA, Beatty NF, York MA, Brooker SL, et al. Human milk microbial community structure is relatively stable and related to variations in macronutrient and micronutrient intakes in healthy lactating women. J Nutr. 2017;147:1739–48.PubMedPubMedCentral
13.
Zurück zum Zitat Padilha M, Danneskiold-Samsøe NB, Brejnrod A, Hoffmann C, Cabral VP, de Iaucci J M, et al. The Human Milk Microbiota is Modulated by Maternal Diet. Microorganisms. 2019;7:502.PubMedCentralCrossRef Padilha M, Danneskiold-Samsøe NB, Brejnrod A, Hoffmann C, Cabral VP, de Iaucci J M, et al. The Human Milk Microbiota is Modulated by Maternal Diet. Microorganisms. 2019;7:502.PubMedCentralCrossRef
14.
Zurück zum Zitat Yang B, Chen Y, Stanton C, Ross RP, Lee Y-K, Zhao J, et al. Bifidobacterium and lactobacillus composition at species level and gut microbiota diversity in infants before 6 weeks. Int J Mol Sci. 2019;20:E3306.PubMedCrossRef Yang B, Chen Y, Stanton C, Ross RP, Lee Y-K, Zhao J, et al. Bifidobacterium and lactobacillus composition at species level and gut microbiota diversity in infants before 6 weeks. Int J Mol Sci. 2019;20:E3306.PubMedCrossRef
15.
Zurück zum Zitat Li N, Yan F, Wang N, Song Y, Yue Y, Guan J, et al. Distinct gut microbiota and metabolite profiles induced by different feeding methods in healthy Chinese infants. Front Microbiol. 2020;11:714.PubMedPubMedCentralCrossRef Li N, Yan F, Wang N, Song Y, Yue Y, Guan J, et al. Distinct gut microbiota and metabolite profiles induced by different feeding methods in healthy Chinese infants. Front Microbiol. 2020;11:714.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Borewicz K, Gu F, Saccenti E, Arts ICW, Penders J, Thijs C, et al. Correlating infant faecal microbiota composition and human milk oligosaccharide consumption by microbiota of one-month old breastfed infants. Mol Nutr Food Res. 2019;63(13):e1801214.CrossRef Borewicz K, Gu F, Saccenti E, Arts ICW, Penders J, Thijs C, et al. Correlating infant faecal microbiota composition and human milk oligosaccharide consumption by microbiota of one-month old breastfed infants. Mol Nutr Food Res. 2019;63(13):e1801214.CrossRef
17.
Zurück zum Zitat Lundgren SN, Madan JC, Emond JA, Morrison HG, Christensen BC, Karagas MR, et al. Maternal diet during pregnancy is related with the infant stool microbiome in a delivery mode-dependent manner. Microbiome. 2018;6:109.PubMedPubMedCentralCrossRef Lundgren SN, Madan JC, Emond JA, Morrison HG, Christensen BC, Karagas MR, et al. Maternal diet during pregnancy is related with the infant stool microbiome in a delivery mode-dependent manner. Microbiome. 2018;6:109.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Chu DM, Antony KM, Ma J, Prince AL, Showalter L, Moller M, et al. The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med. 2016;8:77.PubMedPubMedCentralCrossRef Chu DM, Antony KM, Ma J, Prince AL, Showalter L, Moller M, et al. The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med. 2016;8:77.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Kumar H, du Toit E, Kulkarni A, Aakko J, Linderborg KM, Zhang Y, et al. Distinct patterns in human milk microbiota and fatty acid profiles across specific geographic locations. Front Microbiol. 2016;7:1619.PubMedPubMedCentral Kumar H, du Toit E, Kulkarni A, Aakko J, Linderborg KM, Zhang Y, et al. Distinct patterns in human milk microbiota and fatty acid profiles across specific geographic locations. Front Microbiol. 2016;7:1619.PubMedPubMedCentral
20.
Zurück zum Zitat Li S-W, Watanabe K, Hsu C-C, Chao S-H, Yang Z-H, Lin Y-J, et al. Bacterial composition and diversity in breast milk samples from mothers living in Taiwan and Mainland China. Front Microbiol. 2017;8:965.PubMedPubMedCentralCrossRef Li S-W, Watanabe K, Hsu C-C, Chao S-H, Yang Z-H, Lin Y-J, et al. Bacterial composition and diversity in breast milk samples from mothers living in Taiwan and Mainland China. Front Microbiol. 2017;8:965.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Lundgren SN, Madan JC, Karagas MR, Morrison HG, Hoen AG, Christensen BC. Microbial communities in human milk relate to measures of maternal weight. Front Microbiol. 2019;10:2886.PubMedPubMedCentralCrossRef Lundgren SN, Madan JC, Karagas MR, Morrison HG, Hoen AG, Christensen BC. Microbial communities in human milk relate to measures of maternal weight. Front Microbiol. 2019;10:2886.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Zimmermann P, Curtis N. Breast milk microbiota: A review of the factors that influence composition. J Infect. 2020;81:17–47.PubMedCrossRef Zimmermann P, Curtis N. Breast milk microbiota: A review of the factors that influence composition. J Infect. 2020;81:17–47.PubMedCrossRef
23.
Zurück zum Zitat Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, Bonasio R, et al. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2001;2:361–7.PubMedCrossRef Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, Bonasio R, et al. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2001;2:361–7.PubMedCrossRef
24.
Zurück zum Zitat Rodríguez JM. The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv Nutr. 2014;5:779–84.PubMedPubMedCentralCrossRef Rodríguez JM. The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv Nutr. 2014;5:779–84.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Perez PF, Dore J, Leclerc M, Levenez F, Benyacoub J, Serrant P, et al. Bacterial imprinting of the neonatal immune system: lessons from maternal cells? Pediatrics. 2007;119:e724–32.PubMedCrossRef Perez PF, Dore J, Leclerc M, Levenez F, Benyacoub J, Serrant P, et al. Bacterial imprinting of the neonatal immune system: lessons from maternal cells? Pediatrics. 2007;119:e724–32.PubMedCrossRef
27.
Zurück zum Zitat Ojo-Okunola A, Claassen-Weitz S, Mwaikono KS, Gardner-Lubbe S, Stein DJ, Zar HJ, et al. Influence of socio-economic and psychosocial profiles on the human breast milk bacteriome of South African women. Nutrients. 2019;11:E1390.PubMedCrossRef Ojo-Okunola A, Claassen-Weitz S, Mwaikono KS, Gardner-Lubbe S, Stein DJ, Zar HJ, et al. Influence of socio-economic and psychosocial profiles on the human breast milk bacteriome of South African women. Nutrients. 2019;11:E1390.PubMedCrossRef
28.
Zurück zum Zitat Jost T, Lacroix C, Braegger C, Chassard C. Assessment of bacterial diversity in breast milk using culture-dependent and culture-independent approaches. Br J Nutr. 2013;110:1253–62.PubMedCrossRef Jost T, Lacroix C, Braegger C, Chassard C. Assessment of bacterial diversity in breast milk using culture-dependent and culture-independent approaches. Br J Nutr. 2013;110:1253–62.PubMedCrossRef
29.
Zurück zum Zitat Hunt KM, Foster JA, Forney LJ, Schütte UME, Beck DL, Abdo Z, et al. Characterization of the diversity and temporal stability of bacterial communities in human milk. PLoS One. 2011;6:e21313.PubMedPubMedCentralCrossRef Hunt KM, Foster JA, Forney LJ, Schütte UME, Beck DL, Abdo Z, et al. Characterization of the diversity and temporal stability of bacterial communities in human milk. PLoS One. 2011;6:e21313.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Murphy K, Curley D, O’Callaghan TF, O’Shea C-A, Dempsey EM, O’Toole PW, et al. The composition of human milk and infant faecal microbiota over the first three months of life: a pilot study. Sci Rep. 2017;7:40597.PubMedPubMedCentralCrossRef Murphy K, Curley D, O’Callaghan TF, O’Shea C-A, Dempsey EM, O’Toole PW, et al. The composition of human milk and infant faecal microbiota over the first three months of life: a pilot study. Sci Rep. 2017;7:40597.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Urbaniak C, Angelini M, Gloor GB, Reid G. Human milk microbiota profiles in relation to birthing method, gestation and infant gender. Microbiome. 2016;4:1.PubMedPubMedCentralCrossRef Urbaniak C, Angelini M, Gloor GB, Reid G. Human milk microbiota profiles in relation to birthing method, gestation and infant gender. Microbiome. 2016;4:1.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Williams JE, Carrothers JM, Lackey KA, Beatty NF, Brooker SL, Peterson HK, et al. Strong multivariate relations exist among milk, oral, and fecal microbiomes in mother-infant dyads during the first six months postpartum. J Nutr. 2019;149:902–14.PubMedPubMedCentralCrossRef Williams JE, Carrothers JM, Lackey KA, Beatty NF, Brooker SL, Peterson HK, et al. Strong multivariate relations exist among milk, oral, and fecal microbiomes in mother-infant dyads during the first six months postpartum. J Nutr. 2019;149:902–14.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Jost T, Lacroix C, Braegger CP, Rochat F, Chassard C. Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding. Environ Microbiol. 2014;16:2891–904.PubMedCrossRef Jost T, Lacroix C, Braegger CP, Rochat F, Chassard C. Vertical mother-neonate transfer of maternal gut bacteria via breastfeeding. Environ Microbiol. 2014;16:2891–904.PubMedCrossRef
34.
Zurück zum Zitat Hill CJ, Lynch DB, Murphy K, Ulaszewska M, Jeffery IB, O’Shea CA, et al. Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET Cohort. Microbiome. 2017;5:4.PubMedPubMedCentralCrossRef Hill CJ, Lynch DB, Murphy K, Ulaszewska M, Jeffery IB, O’Shea CA, et al. Evolution of gut microbiota composition from birth to 24 weeks in the INFANTMET Cohort. Microbiome. 2017;5:4.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Mosca A, Leclerc M, Hugot JP. Gut microbiota diversity and human diseases: should we reintroduce key predators in our ecosystem? Front Microbiol. 2016;7:455.PubMedPubMedCentralCrossRef Mosca A, Leclerc M, Hugot JP. Gut microbiota diversity and human diseases: should we reintroduce key predators in our ecosystem? Front Microbiol. 2016;7:455.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Wei S, Bahl MI, Baunwall SMD, Hvas CL, Licht TR. Determining gut microbial dysbiosis: a review of applied indexes for assessment of intestinal microbiota imbalances. Appl Environ Microbiol. 2021;87:e00395-e421.PubMedPubMedCentral Wei S, Bahl MI, Baunwall SMD, Hvas CL, Licht TR. Determining gut microbial dysbiosis: a review of applied indexes for assessment of intestinal microbiota imbalances. Appl Environ Microbiol. 2021;87:e00395-e421.PubMedPubMedCentral
37.
Zurück zum Zitat Yuan MM. Climate warming enhances microbial network complexity and stability. Nat Clim Chang. 2021;11:18.CrossRef Yuan MM. Climate warming enhances microbial network complexity and stability. Nat Clim Chang. 2021;11:18.CrossRef
38.
Zurück zum Zitat Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, 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-e117.PubMedPubMedCentralCrossRef Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, 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-e117.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Satokari R, Grönroos T, Laitinen K, Salminen S, Isolauri E. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol. 2009;48:8–12.PubMedCrossRef Satokari R, Grönroos T, Laitinen K, Salminen S, Isolauri E. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol. 2009;48:8–12.PubMedCrossRef
41.
Zurück zum Zitat Jiménez E, Fernández L, Marín ML, Martín R, Odriozola JM, Nueno-Palop C, et al. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol. 2005;51:270–4.PubMedCrossRef Jiménez E, Fernández L, Marín ML, Martín R, Odriozola JM, Nueno-Palop C, et al. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol. 2005;51:270–4.PubMedCrossRef
42.
Zurück zum Zitat Cortés-Macías E, Selma-Royo M, Martínez-Costa C, Collado MC. Breastfeeding practices influence the breast milk microbiota depending on pre-gestational maternal BMI and weight gain over pregnancy. Nutrients. 2021;13:1518.PubMedPubMedCentralCrossRef Cortés-Macías E, Selma-Royo M, Martínez-Costa C, Collado MC. Breastfeeding practices influence the breast milk microbiota depending on pre-gestational maternal BMI and weight gain over pregnancy. Nutrients. 2021;13:1518.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Moossavi S, Sepehri S, Robertson B, Bode L, Goruk S, Field CJ, et al. Composition and variation of the human milk microbiota are influenced by maternal and early-life factors. Cell Host Microbe. 2019;25:324-335.e4.PubMedCrossRef Moossavi S, Sepehri S, Robertson B, Bode L, Goruk S, Field CJ, et al. Composition and variation of the human milk microbiota are influenced by maternal and early-life factors. Cell Host Microbe. 2019;25:324-335.e4.PubMedCrossRef
44.
Zurück zum Zitat Al-Sadi R, Dharmaprakash V, Nighot P, Guo S, Nighot M, Do T, et al. Bifidobacterium bifidum Enhances the Intestinal Epithelial Tight Junction Barrier and Protects against Intestinal Inflammation by Targeting the Toll-like Receptor-2 Pathway in an NF-κB-Independent Manner. Int J Mol Sci. 2021;22:8070.PubMedPubMedCentralCrossRef Al-Sadi R, Dharmaprakash V, Nighot P, Guo S, Nighot M, Do T, et al. Bifidobacterium bifidum Enhances the Intestinal Epithelial Tight Junction Barrier and Protects against Intestinal Inflammation by Targeting the Toll-like Receptor-2 Pathway in an NF-κB-Independent Manner. Int J Mol Sci. 2021;22:8070.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Hsieh C-Y, Osaka T, Moriyama E, Date Y, Kikuchi J, Tsuneda S. Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum. Physiol Rep. 2015;3:e12327.PubMedPubMedCentralCrossRef Hsieh C-Y, Osaka T, Moriyama E, Date Y, Kikuchi J, Tsuneda S. Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum. Physiol Rep. 2015;3:e12327.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Ewaschuk JB, Diaz H, Meddings L, Diederichs B, Dmytrash A, Backer J, et al. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am J Physiol Gastrointest Liver Physiol. 2008;295:G1025-1034.PubMedCrossRef Ewaschuk JB, Diaz H, Meddings L, Diederichs B, Dmytrash A, Backer J, et al. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am J Physiol Gastrointest Liver Physiol. 2008;295:G1025-1034.PubMedCrossRef
Metadaten
Titel
Effect of breast milk with or without bacteria on infant gut microbiota
verfasst von
Ting Huang
Zichun Zeng
Xinyuan Liang
Xiaomei Tang
Huijuan Luo
Dongju Wang
Juan Zhou
Xiaomin Xiao
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Pregnancy and Childbirth / Ausgabe 1/2022
Elektronische ISSN: 1471-2393
DOI
https://doi.org/10.1186/s12884-022-04930-6

Weitere Artikel der Ausgabe 1/2022

BMC Pregnancy and Childbirth 1/2022 Zur Ausgabe

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Gestationsdiabetes: In der zweiten Schwangerschaft folgenreicher als in der ersten

13.05.2024 Gestationsdiabetes Nachrichten

Das Risiko, nach einem Gestationsdiabetes einen Typ-2-Diabetes zu entwickeln, hängt nicht nur von der Zahl, sondern auch von der Reihenfolge der betroffenen Schwangerschaften ab.

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.