Next Article in Journal
Dietary Patterns of Nurses on Rotational Shifts Are Marked by Redistribution of Energy into the Nightshift
Next Article in Special Issue
The Impact of Human Milk on Necrotizing Enterocolitis: A Systematic Review and Meta-Analysis
Previous Article in Journal
Shift of Volatile Organic Compounds (VOCs) in Gluten-Free Hemp-Enriched Sourdough Bread: A Metabolomic Approach
Previous Article in Special Issue
Feeding Formula Eliminates the Necessity of Bacterial Dysbiosis and Induces Inflammation and Injury in the Paneth Cell Disruption Murine NEC Model in an Osmolality-Dependent Manner
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Understanding the Elements of Maternal Protection from Systemic Bacterial Infections during Early Life

Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
*
Author to whom correspondence should be addressed.
Nutrients 2020, 12(4), 1045; https://doi.org/10.3390/nu12041045
Submission received: 12 March 2020 / Revised: 6 April 2020 / Accepted: 8 April 2020 / Published: 10 April 2020

Abstract

:
Late-onset sepsis (LOS) and other systemic bloodstream infections are notable causes of neonatal mortality, particularly in prematurely born very low birth weight infants. Breastfeeding in early life has numerous health benefits, impacting the health of the newborn in both the short-term and in the long-term. Though the known benefits of an exclusive mother’s own milk diet in early life have been well recognized and described, it is less understood how breastfed infants enjoy a potential reduction in risk of LOS and other systemic infections. Here we review how gut residing pathogens within the intestinal microbiota of infants can cause a subset of sepsis cases and the components of breastmilk that may prevent the dissemination of pathogens from the intestine.

Graphical Abstract

1. Introduction

Bloodstream infections (BSIs) resulting from bacterial dissemination can be extremely harmful to neonates, particularly preterm and very low birth weight (VLBW, <1500 g) newborns. Late-onset neonatal sepsis (LOS) is defined as sepsis occurring 72 h after delivery. LOS has an incidence rate of 10% in preterm infants and is associated with long-term neurological development deficiencies [1,2]. Cases resulting from bacterial BSIs account for 26% of all deaths in preterm infants. LOS will continue to be an important issue among preterm infants as there is a constant reduction of the age of viability resulting from increased medical technology for treating babies born extremely preterm and at a VLBW, those that are most at risk for neonatal BSIs [3,4].
Antibiotics are currently the first line of defense against LOS, but are possibly causing more harm than good. Empirical antibiotics are given to a majority of preterm neonates, regardless of if the infant has a positive blood culture or not, as a preemptive measure of reducing sepsis [2]. This practice may have the opposite intended effect as multiple studies [5,6] have shown an association between prolonged empirical antibiotic administration in premature babies and increased likelihood of developing LOS, necrotizing enterocolitis (NEC), and/or death. Central line placement, used for administration of parenteral nutrition and antibiotics, risks the introduction of pathogens to the bloodstream and is the likely cause of many BSIs. As such, increased hygienic practices implemented in hospitals have resulted in a stark decrease in LOS caused by normal skin commensals [7]. However, despite these efforts, LOS rates in neonates remain unchanged among cases caused by gut commensals, suggesting the bacteria are entering the bloodstream through another mechanism [7].

2. Breastfeeding and LOS

A preterm infant’s diet plays a crucial role in disease development or avoidance. Parenteral feedings are often the only option for delivering nutrients to VLBW infants in the days immediately following birth, but long term use is strongly associated with increased risk of LOS development [8,9]. When an infant’s organs become mature enough to handle partial or full enteral nutrition, mother’s own milk (MOM) is the preferred source of nutrition [10], though preterm infants once were formula-fed at higher rates compared to newborns delivered at term. It is logical to hypothesize ailing infants physically unable to be enterally-fed are more likely to develop LOS in connection with a frail condition [8]. When MOM is unavailable, human donor milk and/or formula are given to infants instead. A number of clinical studies have demonstrated a clear connection between feeding with MOM and protection against LOS in premature infants, in addition to the benefits of a faster transition to enteral feedings, decreased likelihood of mortality, and reduced length of hospital stay [11,12]. Further, a historical clinical observation showed an LOS incidence of 57% amongst the formula-fed infants compared to an LOS incidence of 7% in MOM-fed infants, which included partial-MOM fed infants [13]. Reduced risk of LOS was correlated with increased consumption of human milk, with the odds of LOS in a NICU cohort decreasing 19% for every 10 mL/kg dose per day of human milk [11]. While this cohort pooled infants receiving donor milk with those receiving MOM into a single human milk- fed group, more than 90% of the infants in that cohort were given MOM exclusively [11]. Recent systemic data analysis suggested a possible, though not-significant, 23% risk reduction in developing LOS among exclusively breastfed infants as compared to exclusively formula-fed infants [14]. Additional clinical observations showed similar significant results where 25% of formula-fed infants developed LOS compared to 14% of MOM-fed infants [12], supporting an initiative to promote exclusive breastfeeding as the preferred protective diet in early days of life of any enterally-fed infant. To date, use of donor milk has not shown a reduction of risk of LOS, in contrast to MOM diets [15], though mechanisms of protection unique to MOM remain unclear. More clinical data should be gathered comparing the outcomes of MOM-fed infants to those fed donor milk or fortified formula as better alternatives become available to those infants unable to be fed MOM. In extremely rare cases, LOS may be the result of contaminated breast milk [16], but in the vast majority of cases, MOM-fed infants have overall better outcomes than those who require parenteral nutrition, or other enteral diets.

3. Enteric Origin of Pathogens

3.1. Pathogens in LOS

The potential mechanism of how breastmilk may protect from bacterial BSIs and LOS initially became elucidated around 10 years ago when multiple groups observed the pathogens residing in the gastrointestinal (GI) tract prior to sepsis events [17,18,19,20]. Following birth, the gastrointestinal tract becomes colonized by commensal bacteria in a dynamic process that is initially pioneered by facultative anaerobic proteobacteria and lactobacilli [21,22,23,24,25]. Common causative pathogens of LOS can be found residing in the gut, including Gram-positive members of the lactobacillales order such as Group B Streptococcus (GBS) or Enterococcus faecalis; and Gram-negative bacilli (GNB) of the gammaproteobacteria class such as Klebsiella pneumonia [18], Escherichia coli [18], Pseudomonas aeruginosa [26], or Enterobacteriaceae species [27]. Due to the low density of the commensal flora in neonates, such pathobionts [28], bacterial species that can reside in the gut microbiota as a commensal but also have the potential to become pathogenic, can colonize the GI tract [29]. By acting as a reservoir for potential pathogens, the gut microbiota poses a risk to the neonate if it becomes dysbiotic, resulting in pathogen expansion [30,31].
Preventing enteric bacterial dissemination of pathobionts may prove to be more difficult than preventing intravenous dissemination, where increased hygienic practices have reduced LOS rates [7,32]. Additionally, animal work suggests the enteral route of infection may contribute to the virulence of sepsis pathogens when compared to the intraperitoneal route of infection [33], underlining the importance of developing therapeutics preventing enteric dissemination. Prophylactic use of oral antibiotics rarely target only potential pathogens, but also disrupts the normal developing microbiota contributing to dysbiosis in the microbial community [34,35,36]. Dysbiosis, in turn, can result in enteric infection and dissemination as pathobionts gain an increased foothold in the microbial community [30]. Antibiotic resistance is a growing concern in neonatal sepsis cases, and as such, dependence and overuse of antibiotics should be avoided [37,38].

3.2. Modification of the Infant Gut Microbiota

Introduction of probiotic strains of commensal bacteria has been proposed as a therapeutic strategy to treat or prevent dysbiosis of the gut microbiota and prevent a number of diseases including enteric infections and LOS [39]. Live strains of commensal bacteria such as Lactobacillus and Bifidobacterium species may improve gut health by preventing pathogen colonization and promoting the development of a healthy microbiota [40,41]. Modest improvements in gut health following single probiotic strains such as Saccharomyces bourlardii, Lactobacillus reuteri, Lactobacillus acidophilus and Bifidobacterium lactis have been observed [42], including a reduction in the time needed for progression to full enteral feeding in preterm infants given Saccharomyces boulardii or B. lactis, though this effect is less pronounced in exclusively formula-fed infants [43].
Probiotic mixes containing multiple strains have shown the most success in reducing the risk of LOS in enterally-fed infants [44]. Such formulations can range from a mix of three strains: L. acidophilus, E. faecium and Bifidobacterium infantum [45] to a mix of eight strains: Streptococcus thermophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, L. acidophilus, Lactobacillus plantarum, Lactobacillus paracasei and Lactobacillus delbrueckii spp bulgaricus [46], suggesting multiple strains may have complementary roles in combination to restore intestinal health and provide protection. However, probiotics such as Lactobacillus rhamnosus GG, S. boulardii, L. reuteri, Lactobacillus sporogenes, or B. breve as single strains, or even mixes of multiple strains show a limited effect in reducing LOS in formula-fed infants [47]. This apparent limitation of probiotics in improving outcomes in formula-fed infants as compared to breastfed infants suggests a greater deficit in the gut health of formula-fed infants that is harder to overcome with therapeutic interventions [48,49].
The shaping of the gut microbiome by breastmilk has been repeatedly observed and formula-fed infants have a gut microbiota distinct from the community found in the GI tract of breastfed infants [50,51,52,53]. Diet during early life can influence bacterial translocation as intestinal permeability was found to be significantly decreased [54] and barrier function matured quicker in breastfed preterm infants [55] when compared to formula-fed preterm infants. In animal models, formula feeding resulted in increased bacterial translocation [56,57,58]. VLBW babies in intensive care units, those most at risk of developing sepsis, often lack access to breastmilk. Therefore, identification of protective components of breastmilk that prevent dissemination represents fertile ground for the development of therapeutics to prevent bacterial infections and LOS.

4. Components of Breastmilk

Breastmilk is a complex formulation of nutrients, proteins, and growth factors providing neonates with benefits beyond the incomparable nourishment. Figure 1 shows maternal protection against intestinal pathogens. Several biologically active factors promote gut health and confer protection from enteric infections to the neonate. Breastmilk changes substantially throughout lactation from colostrum in the days initially following delivery to transitional milk and then mature milk approximately two weeks following delivery. Proteins such as antibodies and growth factors are present in higher concentrations in colostrum and transitional milk compared to mature milk [59,60]. Given the biological role for such proteins, breastmilk is therefore considered one component of the “mother-breastmilk-infant triad” [61] and perhaps synchronized between mother and child to afford age-appropriate nutrition and protection to neonates provided with MOM in the first weeks of life. Such factors and proteins present in breastmilk are discussed in this section. The key components of breastmilk reviewed here all can be found in higher concentrations in colostrum and transitional milk early in lactation, asking the question that for breastmilk to have protective effects in neonates, is timing everything?

4.1. Antibodies

Maternal antibodies, including IgM, IgG, and IgA subtypes, offer superior protection to neonates both within the intestinal lumen, and systemically. These antibodies can provide passive immunity within the neonate to any potential systemic infections [67]. Within the lumen of the neonatal GI tract, IgA is particularly important in providing protection from invasive enteric pathogens by directly binding and preventing adherence and evasion [62,67]. Beyond protection from pathogen translocation from the intestinal lumen, maternal IgG antibodies transferred to the neonates also offered protection from E. coli within the circulation [62]. There has been modest clinical evidence that IgM administration may offer systemic protection from bacterial infections [68], though neither IgM nor IgG administration reduced LOS mortality [69]. Animal work has shown maternal antibodies educate the neonatal immune responses by dampening T cell-mediated responses in early life [70], potentially quieting inflammatory responses that may precede or accompany LOS [71,72,73]. Thus, maternal antibodies protect the neonate by preventing enteric pathogens from translocating from the intestinal lumen, and potentially limiting systemic infections and intestinal inflammation.

4.2. Growth Factors

Growth factors present in the breastmilk include a family of ligands that neonates can sense through the epidermal growth factor receptor (EGFR) expressed on intestinal epithelial cells [25,63,64]: epidermal growth factor (EGF), amphiregulin (AREG), heparin-binding epidermal growth factor-like factor (HB-EGF), and tumor-growth factor-alpha (TGF-α) [74]. All are found in temporal gradients, with the highest concentration in colostrum [74,75,76,77,78]. EGF is one of the most abundantly concentrated growth factors in breastmilk, though EGFR ligands perform redundant functions [79], suggesting these ligands have a necessary role in early life. EGF passes through the digestive tract resisting low pH and enzymatic degradation [80,81] and can be measured in the stool of breastfed children [64], suggesting it has a biological effect throughout the intestines. EGFR activation in the neonate results in epithelial cell division, nutrient uptake, improved intestinal barrier function, reduced bacterial translocation, and reduced Toll-like receptor signaling [63,80,81,82,83]. Recent animal work modeling decreased EGFR ligands within the GI tract of neonatal mice observed translocation of enteric pathogens resulting in a systemic infection in a model of LOS, which was reversed by oral administration of recombinant EGF [64]. Thus growth factors, through the activation of EGFR on neonatal epithelial cells can limit enteric pathogens from disseminating and potentially prevent systemic infections.

4.3. Lactoferrin

Lactoferrin, also present in increased concentrations in the first weeks of lactation, can remain a significant component of breastmilk for months after lactation begins [84,85,86]. The primary function of lactoferrin is to bind iron for transfer to the neonate through epithelial cell absorption, which the growing neonate utilizes as an important nutrient. Therefore, lactoferrin has anti-microbial properties primarily through the iron-binding capacity as iron sequestration can limit the amount of free iron available for bacterial growth [65,87,88]. Lactoferricin, a derivative of lactoferrin, may be directly bacteriostatic and can bind bacterial wall components, potentially limiting luminal pathogens [89,90]. Lactoferrin may also promote the development of the mucosal immune system by stimulating dendritic cells that shape intestinal immune responses and enhancing IgA production in Peyer’s patches [91]. Lactoferrin has been the target of several clinical trials as a supplement to enteral diets to protect against enteric infections and LOS [92]. While therapeutic lactoferrin may have beneficial effects on modifying the neonatal microbiome and reducing potential pathobionts [93], the efficacy of lactoferrin in reducing LOS mortality remains controversial [94,95,96]. Thus, lactoferrin protects the neonate with direct anti-microbial effects, potentially limiting pathogen colonization within the microbiota.

4.4. Human Milk Oligosaccharides

Oligosaccharides in breastmilk, known as human milk oligosaccharides (HMOs), support the maturation of the normal infant microbiota, which in turn provides colonization resistance to enteric pathogens [66,97,98,99,100]. These glycans are dynamically produced in the first weeks of lactation [101,102] and are essentially undigested by the infant [103], but instead utilized by the developing microbiota [52]. HMOs can be utilized as a nutrient source by commensal members of the microbiota, and also probiotic strains such as B. breve, as colonization of the intestinal tract of infants was associated with HMO concentration and fucosylation [104]. HMOs may also modulate the growth of potential pathogens, and have been shown to have a direct effect against the formation of GBS biofilms [105]. Additionally, observations suggest HMOs may have a direct impact on infant’s epithelial cells by increasing mucus expression [106] and promoting goblet cell maturation [107], both of which can improve barrier function by enhancing the mucus layer covering the intestinal epithelium that can prevent bacterial encroachment and translocation. Finally HMOs may have an impact on the infant’s immune system by binding c-type lectins, siglecs, galectins and selectins expressed by phagocytic and antigen-presenting cells, such as dendritic cells, monocytes and neutrophils [108,109,110]. These interactions could modulate immune responses through regulating leukocyte trafficking, influencing cytokine responses and inhibiting TLR-mediated inflammation, all of which could affect LOS development and outcomes [66,111,112]. Clinically, infants born to mothers unable to produce some forms of HMOs trended toward an increased risk of LOS [113]. Similarly, a low diversity of HMOs from mothers was associated with an increase in NEC, though was not significantly associated with an increase is LOS cases [114]. These clinical data suggest more work needs to be compiled regarding supplementation of infant diets with HMOs to potentially improve gut function and sepsis outcomes [115]. Thus, HMOs may protect the neonate by shaping the microbiota, which may protect from pathogen colonization.

5. Future Directions

5.1. Supplements and MOM Alternatives

Donor milk represents a worthy alternative when MOM is unavailable, though reports have shown the processing of donor milk, including pasteurization and potentially multiple freeze-thaw cycles, may reduce the concentrations of the beneficial proteins, particularly immunoglobulins and lactoferrin [74,116]. Pasteurization of MOM showed a non-significant trend of increased infectious LOS morbidity [117], suggesting the reduction of these beneficial components in MOM could lead to an increased risk of LOS. Pasteurization is an imperative step in donor milk processing to prevent potential transmission of pathogens through contaminated breast milk [16], and work optimizing pasteurization processes to remove pathogenic threat while maintaining beneficial factors is currently being completed [118,119]. The Holder pasteurization method, the recommended pasteurization method of donor milk, sterilizes bacteria present in the milk, and does not degrade growth factors such as EGF and TGF-β [119]. Immunoglobulins and lactoferrin are reduced following Holder pasteurization, though may be protected following high temperature short time (HTST) treatments, an experimental pasteurization method [120,121]. Additionally, the increased concentrations of these proteins early in lactation suggests age-matched donor milk or supplements containing a combination of immunoglobulins, growth factors, lactoferrin, and HMOs at concentrations found in colostrum may represent the next steps in the progression toward an appropriate alternative when MOM cannot be provided.

5.2. Animal Modeling

While clinical interventions are quickly being brought to the NICU, animal models are revealing potential mechanisms of acquisition and protection against LOS. Therefore, the development and use of animal models that represent how LOS is clinically acquired is essential [30,62,64]. Traditional models of LOS have been limited to intravenous injection of cecal contents or single bacterial components, such as lipopolysaccharide. While these models may help elucidate how neonates respond to systemic bacterial insults, these models lack insight into the enteric route of entry pathogens may utilize. Injection of cecal contents may introduce too many bacterial elements as most LOS patients are infected with only one bacterial species at a time. Similarly, intravenous injection of lipopolysaccharide, a component of some bacterial cell walls, may reduce relevance as there can be strain variation in LOS pathology within the same bacterial species [64]. As increased hygienic practices reduce the number of intravenous-acquired LOS cases, similar interest should be placed on reducing the number of LOS cases resulting from an enteric route of origin, with animal work modeling the oral route of pathogen entry. Understanding how MOM and the developing microbiota protect the neonate from enteric pathogens will provide clear directions for future therapeutics.

6. Conclusions

While the clinical connection between breastfeeding and reduced LOS risk is currently a potential, though logical, correlation, the many components within breastmilk offer observable benefits to the developing neonate, particularly within the intestinal environment. If enteric pathogens continue to threaten infants and cause a substantial portion of LOS cases, factors present in breastmilk may provide exceptional protection to the neonate, representing strong candidates for supplementation of breast milk that could prevent of translocating pathogens. Clinical measures such as the reduction of unnecessary antibiotics to protect the intestinal microbiome, promotion of an exclusive MOM diet when available, and sophistication of supplements combining immunoglobulins, growth factors, lactoferrin, and HMOs to formula or donor milk could result in further reduction of LOS cases by focusing on protection from enteric pathogens.

Author Contributions

Conceptualization, original draft preparation, review and editing, S.A.K., K.A.K.; funding acquisition, K.A.K. All authors have read and agreed to the published version of the manuscript.

Funding

Supported by grants: DK109006-K.A.K., AI144721-K.A.K., AI095542 MIST Scholars Award–K.A.K.

Acknowledgments

The authors wish to acknowledge Kara Greenfield, Kathryn Lin, and Lila Yokanovich for their advice with this manuscript.

Conflicts of Interest

K.A.K. is an inventor on U.S. Nonprovisional Application Serial No. 15/880,658 Compositions and Methods for Modulation of Dietary and Microbial Exposure.

References

  1. Cortese, F.; Scicchitano, P.; Gesualdo, M.; Filaninno, A.; De Giorgi, E.; Schettini, F.; Laforgia, N.; Ciccone, M.M. Early and late infections in newborns: Where do we stand? A review. Pediatr. Neonatol. 2016, 57, 265–273. [Google Scholar] [CrossRef] [Green Version]
  2. Raymond, S.L.; Rincon, J.C.; Wynn, J.L.; Moldawer, L.L.; Larson, S.D. Impact of early-life exposures to infections, antibiotics, and vaccines on perinatal and long-term health and disease. Front. Immunol. 2017, 8. [Google Scholar] [CrossRef]
  3. Stoll, B.J.; Gordon, T.; Korones, S.B.; Shankaran, S.; Tyson, J.E.; Bauer, C.R.; Fanaroff, A.A.; Lemons, J.A.; Donovan, E.F.; Oh, W.; et al. Late-onset sepsis in very low birth weight neonates: A report from the National Institute of Child Health and Human Development Neonatal Research Network. J. Pediatr. 1996, 129, 63–71. [Google Scholar] [CrossRef]
  4. Stoll, B.J.; Hansen, N.; Fanaroff, A.A.; Wright, L.L.; Carlo, W.A.; Ehrenkranz, R.A.; Lemons, J.A.; Donovan, E.F.; Stark, A.R.; Tyson, J.E.; et al. Late-onset sepsis in very low birth weight neonates: The experience of the NICHD neonatal research network. Pediatrics 2002, 110, 285. [Google Scholar] [CrossRef]
  5. Cotten, C.M.; Taylor, S.; Stoll, B.; Goldberg, R.N.; Hansen, N.I.; Sánchez, P.J.; Ambalavanan, N.; Benjamin, D.K., Jr.; Network, N.N.R. Prolonged duration of initial empirical antibiotic treatment is associated with increased rates of necrotizing enterocolitis and death for extremely low birth weight infants. Pediatrics 2009, 123, 58–66. [Google Scholar] [CrossRef] [Green Version]
  6. Kuppala, V.S.; Meinzen-Derr, J.; Morrow, A.L.; Schibler, K.R. Prolonged initial empirical antibiotic treatment is associated with adverse outcomes in premature infants. J. Pediatr. 2011, 159, 720–725. [Google Scholar] [CrossRef] [Green Version]
  7. Pharande, P.; Lindrea, K.B.; Smyth, J.; Evans, M.; Lui, K.; Bolisetty, S. Trends in late-onset sepsis in a neonatal intensive care unit following implementation of infection control bundle: A 15-year audit. J. Paediatr. Child Health 2018, 54, 1314–1320. [Google Scholar] [CrossRef]
  8. el Manouni el Hassani, S.; Berkhout, D.J.C.; Niemarkt, H.J.; Mann, S.; de Boode, W.P.; Cossey, V.; Hulzebos, C.V.; van Kaam, A.H.; Kramer, B.W.; van Lingen, R.A.; et al. Risk factors for late-onset sepsis in preterm infants: A multicenter case-control study. Neonatology 2019, 116, 42–51. [Google Scholar] [CrossRef]
  9. Kung, Y.-H.; Hsieh, Y.-F.; Weng, Y.-H.; Lien, R.-I.; Luo, J.; Wang, Y.; Huang, Y.-C.; Chen, C.-L.; Chen, C.-J. Risk factors of late-onset neonatal sepsis in Taiwan: A matched case-control study. J. Microbiol. Immunol. Infect. 2016, 49, 430–435. [Google Scholar] [CrossRef] [Green Version]
  10. Moro, G.E.; Arslanoglu, S.; Bertino, E.; Corvaglia, L.; Montirosso, R.; Picaud, J.-C.; Polberger, S.; Schanler, R.J.; Steel, C.; van Goudoever, J.; et al. XII. Human milk in feeding premature infants: Consensus statement. J. Pediatr. Gastroenterol. Nutr. 2015, 61, S16–S19. [Google Scholar] [CrossRef]
  11. Patel, A.L.; Johnson, T.J.; Engstrom, J.L.; Fogg, L.F.; Jegier, B.J.; Bigger, H.R.; Meier, P.P. Impact of early human milk on sepsis and health-care costs in very low birth weight infants. J. Perinatol. 2013, 33, 514–519. [Google Scholar] [CrossRef] [Green Version]
  12. Cortez, J.; Makker, K.; Kraemer, D.F.; Neu, J.; Sharma, R.; Hudak, M.L. Maternal milk feedings reduce sepsis, necrotizing enterocolitis and improve outcomes of premature infants. J. Perinatol. 2018, 38, 71–74. [Google Scholar] [CrossRef]
  13. Ashraf, R.N.; Jalil, F.; Zaman, S.; Karlberg, J.; Khan, S.R.; Lindblad, B.S.; Hanson, L.A. Breast feeding and protection against neonatal sepsis in a high risk population. Arch. Dis. Child. 1991, 66, 488–490. [Google Scholar] [CrossRef] [Green Version]
  14. Miller, J.; Tonkin, E.; Damarell, A.R.; McPhee, J.A.; Suganuma, M.; Suganuma, H.; Middleton, F.P.; Makrides, M.; Collins, T.C. A systematic review and meta-analysis of human milk feeding and morbidity in very low birth weight infants. Nutrients 2018, 10, 707. [Google Scholar] [CrossRef] [Green Version]
  15. Meier, P.; Patel, A.; Esquerra-Zwiers, A. Donor human milk update: Evidence, mechanisms, and priorities for research and practice. J. Pediatr. 2017, 180, 15–21. [Google Scholar] [CrossRef] [Green Version]
  16. Widger, J.; O’Connell, N.H.; Stack, T. Breast milk causing neonatal sepsis and death. Clin. Microbiol. Infect. 2010, 16, 1796–1798. [Google Scholar] [CrossRef]
  17. Carl, M.A.; Ndao, I.M.; Springman, A.C.; Manning, S.D.; Johnson, J.R.; Johnston, B.D.; Burnham, C.A.; Weinstock, E.S.; Weinstock, G.M.; Wylie, T.N.; et al. Sepsis from the gut: The enteric habitat of bacteria that cause late-onset neonatal bloodstream infections. Clin. Infect. Dis. 2014, 58, 1211–1218. [Google Scholar] [CrossRef] [Green Version]
  18. Almuneef, M.A.; Baltimore, R.S.; Farrel, P.A.; Reagan-Cirincione, P.; Dembry, L.M. Molecular typing demonstrating transmission of gram-negative rods in a neonatal intensive care unit in the absence of a recognized epidemic. Clin. Infect. Dis. 2001, 32, 220–227. [Google Scholar] [CrossRef] [Green Version]
  19. Graham, P.L., 3rd; Della-Latta, P.; Wu, F.; Zhou, J.; Saiman, L. The gastrointestinal tract serves as the reservoir for Gram-negative pathogens in very low birth weight infants. Pediatr. Infect. Dis. J. 2007, 26, 1153–1156. [Google Scholar] [CrossRef]
  20. Smith, A.; Saiman, L.; Zhou, J.; Della-Latta, P.; Jia, H.; Graham, P.L., 3rd. Concordance of gastrointestinal tract colonization and subsequent bloodstream infections with gram-negative bacilli in very low birth weight infants in the neonatal intensive care unit. Pediatr. Infect. Dis. J. 2010, 29, 831–835. [Google Scholar] [CrossRef] [Green Version]
  21. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the Human Intestinal Microbial Flora. Science (N. Y.) 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Koenig, J.E.; Spor, A.; Scalfone, N.; Fricker, A.D.; Stombaugh, J.; Knight, R.; Angenent, L.T.; Ley, R.E. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl. Acad. Sci. USA 2011, 108, 4578–4585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef] [PubMed]
  24. Azad, M.B.; Konya, T.; Persaud, R.R.; Guttman, D.S.; Chari, R.S.; Field, C.J.; Sears, M.R.; Mandhane, P.J.; Turvey, S.E.; Subbarao, P.; et al. Impact of maternal intrapartum antibiotics, method of birth and breastfeeding on gut microbiota during the first year of life: A prospective cohort study. BJOG 2016, 123, 983–993. [Google Scholar] [CrossRef]
  25. Knoop, K.A.; Gustafsson, J.K.; McDonald, K.G.; Kulkarni, D.H.; Coughlin, P.E.; McCrate, S.; Kim, D.; Hsieh, C.S.; Hogan, S.P.; Elson, C.O.; et al. Microbial antigen encounter during a preweaning interval is critical for tolerance to gut bacteria. Sci. Immunol. 2017, 2. [Google Scholar] [CrossRef] [Green Version]
  26. Foca, M.; Jakob, K.; Whittier, S.; Latta, P.D.; Factor, S.; Rubenstein, D.; Saiman, L. Endemic Pseudomonas aeruginosa Infection in a Neonatal Intensive Care Unit. N. Engl. J. Med. 2000, 343, 695–700. [Google Scholar] [CrossRef]
  27. Chen, H.N.; Lee, M.L.; Yu, W.K.; Lin, Y.W.; Tsao, L.Y. Late-onset Enterobacter cloacae sepsis in very-low-birth-weight neonates: Experience in a medical center. Pediatr. Neonatol. 2009, 50, 3–7. [Google Scholar] [CrossRef] [Green Version]
  28. Hornef, M. Pathogens, commensal symbionts, and pathobionts: discovery and functional effects on the host. ILAR J. 2015, 56, 159–162. [Google Scholar] [CrossRef]
  29. Kolter, J.; Henneke, P. Codevelopment of microbiota and innate immunity and the risk for Group B streptococcal disease. Front. Immunol. 2017, 8, 1497. [Google Scholar] [CrossRef] [Green Version]
  30. Singer, J.R.; Blosser, E.G.; Zindl, C.L.; Silberger, D.J.; Conlan, S.; Laufer, V.A.; DiToro, D.; Deming, C.; Kumar, R.; Morrow, C.D.; et al. Preventing dysbiosis of the neonatal mouse intestinal microbiome protects against late-onset sepsis. Nat. Med. 2019, 25, 1772–1782. [Google Scholar] [CrossRef]
  31. Sanidad, K.Z.; Zeng, M.Y. LOS in the dysbiotic gut. Cell Host Microbe 2020, 27, 11–13. [Google Scholar] [CrossRef] [PubMed]
  32. Bizzarro, M.J.; Raskind, C.; Baltimore, R.S.; Gallagher, P.G. Seventy-Five years of neonatal sepsis at Yale: 1928–2003. Pediatrics 2005, 116, 595. [Google Scholar] [CrossRef] [PubMed]
  33. Cole, B.K.; Scott, E.; Ilikj, M.; Bard, D.; Akins, D.R.; Dyer, D.W.; Chavez-Bueno, S. Route of infection alters virulence of neonatal septicemia Escherichia coli clinical isolates. PLoS ONE 2017, 12, e0189032. [Google Scholar] [CrossRef] [Green Version]
  34. Sekirov, I.; Tam, N.M.; Jogova, M.; Robertson, M.L.; Li, Y.; Lupp, C.; Finlay, B.B. Antibiotic-induced perturbations of the intestinal microbiota alter host susceptibility to enteric infection. Infect. Immun. 2008, 76, 4726–4736. [Google Scholar] [CrossRef] [Green Version]
  35. Deshmukh, H.S.; Liu, Y.; Menkiti, O.R.; Mei, J.; Dai, N.; O’Leary, C.E.; Oliver, P.M.; Kolls, J.K.; Weiser, J.N.; Worthen, G.S. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat. Med. 2014, 20, 524–530. [Google Scholar] [CrossRef] [Green Version]
  36. Vangay, P.; Ward, T.; Gerber, J.S.; Knights, D. Antibiotics, pediatric dysbiosis, and disease. Cell Host Microbe 2015, 17, 553–564. [Google Scholar] [CrossRef] [Green Version]
  37. Bizzarro, M.J.; Dembry, L.-M.; Baltimore, R.S.; Gallagher, P.G. Changing patterns in neonatal Escherichia coli; sepsis and ampicillin resistance in the era of intrapartum antibiotic prophylaxis. Pediatrics 2008, 121, 689. [Google Scholar] [CrossRef]
  38. Mohsen, L.; Ramy, N.; Saied, D.; Akmal, D.; Salama, N.; Abdel Haleim, M.M.; Aly, H. Emerging antimicrobial resistance in early and late-onset neonatal sepsis. Antimicrob. Resist. Infect. Control 2017, 6, 63. [Google Scholar] [CrossRef]
  39. Panigrahi, P.; Parida, S.; Nanda, N.C.; Satpathy, R.; Pradhan, L.; Chandel, D.S.; Baccaglini, L.; Mohapatra, A.; Mohapatra, S.S.; Misra, P.R.; et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature 2017, 548, 407. [Google Scholar] [CrossRef]
  40. Garrido, D.; Barile, D.; Mills, D.A. A Molecular basis for bifidobacterial enrichment in the infant gastrointestinal tract. Adv. Nutr. 2012, 3, 415S–421S. [Google Scholar] [CrossRef]
  41. Sassone-Corsi, M.; Raffatellu, M. No vacancy: How beneficial microbes cooperate with immunity to provide colonization resistance to pathogens. J. Immunol. 2015, 194, 4081. [Google Scholar] [CrossRef] [Green Version]
  42. Mugambi, M.N.; Musekiwa, A.; Lombard, M.; Young, T.; Blaauw, R. Probiotics, prebiotics infant formula use in preterm or low birth weight infants: A systematic review. Nutr. J. 2012, 11, 58. [Google Scholar] [CrossRef] [Green Version]
  43. Aceti, A.; Gori, D.; Barone, G.; Callegari, M.L.; Fantini, M.P.; Indrio, F.; Maggio, L.; Meneghin, F.; Morelli, L.; Zuccotti, G.; et al. Probiotics and time to achieve full enteral feeding in human milk-fed and formula-fed preterm infants: systematic review and meta-analysis. Nutrients 2016, 8, 471. [Google Scholar] [CrossRef] [Green Version]
  44. Rao, S.C.; Athalye-Jape, G.K.; Deshpande, G.C.; Simmer, K.N.; Patole, S.K. Probiotic supplementation and late-onset sepsis in preterm infants: A meta-analysis. Pediatrics 2016, 137, e20153684. [Google Scholar] [CrossRef] [Green Version]
  45. Kanic, Z.; Micetic Turk, D.; Burja, S.; Kanic, V.; Dinevski, D. Influence of a combination of probiotics on bacterial infections in very low birthweight newborns. Wien. Klin. Wochenschr. 2015, 127, 210–215. [Google Scholar] [CrossRef]
  46. Sinha, A.; Gupta, S.S.; Chellani, H.; Maliye, C.; Kumari, V.; Arya, S.; Garg, B.S.; Gaur, S.D.; Gaind, R.; Deotale, V.; et al. Role of probiotics VSL#3 in prevention of suspected sepsis in low birthweight infants in India: A randomised controlled trial. BMJ Open 2015, 5, e006564. [Google Scholar] [CrossRef]
  47. Aceti, A.; Maggio, L.; Beghetti, I.; Gori, D.; Barone, G.; Callegari, M.L.; Fantini, M.P.; Indrio, F.; Meneghin, F.; Morelli, L.; et al. Probiotics prevent late-onset sepsis in human milk-fed, very low birth weight preterm infants: Systematic review and meta-analysis. Nutrients 2017, 9, 904. [Google Scholar] [CrossRef] [Green Version]
  48. Jakaitis, B.M.; Denning, P.W. Human breast milk and the gastrointestinal innate immune system. Clin. Perinatol. 2014, 41, 423–435. [Google Scholar] [CrossRef] [Green Version]
  49. Yeruva, L.; Spencer, N.E.; Saraf, M.K.; Hennings, L.; Bowlin, A.K.; Cleves, M.A.; Mercer, K.; Chintapalli, S.V.; Shankar, K.; Rank, R.G.; et al. Formula diet alters small intestine morphology, microbial abundance and reduces VE-cadherin and IL-10 expression in neonatal porcine model. BMC Gastroenterol. 2016, 16, 40. [Google Scholar] [CrossRef] [Green Version]
  50. Guaraldi, F.; Salvatori, G. Effect of breast and formula feeding on gut microbiota shaping in newborns. Front. Cell. Infect. Microbiol. 2012, 2, 94. [Google Scholar] [CrossRef] [Green Version]
  51. Goldsmith, F.; O’Sullivan, A.; Smilowitz, J.T.; Freeman, S.L. Lactation and intestinal microbiota: How early diet shapes the infant gut. J. Mammary Gland Biol. Neoplasia 2015, 20, 149–158. [Google Scholar] [CrossRef]
  52. Davis, J.C.C.; Lewis, Z.T.; Krishnan, S.; Bernstein, R.M.; Moore, S.E.; Prentice, A.M.; Mills, D.A.; Lebrilla, C.B.; Zivkovic, A.M. Growth and morbidity of gambian infants are influenced by maternal milk oligosaccharides and infant gut microbiota. Sci. Rep. 2017, 7, 40466. [Google Scholar] [CrossRef]
  53. Baumann-Dudenhoeffer, A.M.; D’Souza, A.W.; Tarr, P.I.; Warner, B.B.; Dantas, G. Infant diet and maternal gestational weight gain predict early metabolic maturation of gut microbiomes. Nat. Med. 2018, 24, 1822–1829. [Google Scholar] [CrossRef]
  54. Taylor, S.N.; Basile, L.A.; Ebeling, M.; Wagner, C.L. Intestinal permeability in preterm infants by feeding type: Mother’s milk versus formula. Breastfeed. Med. 2009, 4, 11–15. [Google Scholar] [CrossRef] [Green Version]
  55. Saleem, B.; Okogbule-Wonodi, A.C.; Fasano, A.; Magder, L.S.; Ravel, J.; Kapoor, S.; Viscardi, R.M. Intestinal barrier maturation in very low birthweight infants: Relationship to feeding and antibiotic exposure. J. Pediatr. 2017, 183, 31–36.e31. [Google Scholar] [CrossRef] [Green Version]
  56. Go, L.L.; Albanese, C.T.; Watkins, S.C.; Simmons, R.L.; Rowe, M.I. Breast milk protects the neonate from bacterial translocation. J. Pediatr. Surg. 1994, 29, 1059–1064. [Google Scholar] [CrossRef]
  57. Yajima, M.; Nakayama, M.; Hatano, S.; Yamazaki, K.; Aoyama, Y.; Yajima, T.; Kuwata, T. Bacterial translocation in neonatal rats: The relation between intestinal flora, translocated bacteria, and influence of milk. J. Pediatr. Gastroenterol. Nutr. 2001, 33, 592–601. [Google Scholar] [CrossRef]
  58. Nakayama, M.; Yajima, M.; Hatano, S.; Yajima, T.; Kuwata, T. Intestinal adherent bacteria and bacterial translocation in breast-fed and formula-fed rats in relation to susceptibility to infection. Pediatr. Res. 2003, 54, 364. [Google Scholar] [CrossRef] [Green Version]
  59. Goldsmith, S.J.; Dickson, J.S.; Barnhart, H.M.; Toledo, R.T.; Eiten-Miller, R.R. IgA, IgG, IgM and lactoferrin contents of human milk during early lactation and the effect of processing and storage. J. Food Prot. 1983, 46, 4–7. [Google Scholar] [CrossRef]
  60. Castellote, C.; Casillas, R.; Ramírez-Santana, C.; Pérez-Cano, F.J.; Castell, M.; Moretones, M.G.; López-Sabater, M.C.; Franch, À. Premature delivery influences the immunological composition of colostrum and transitional and mature human milk. J. Nutr. 2011, 141, 1181–1187. [Google Scholar] [CrossRef]
  61. Bode, L.; Raman, A.S.; Murch, S.H.; Rollins, N.C.; Gordon, J.I. Understanding the mother-breastmilk-infant “triad”. Science 2020, 367, 1070. [Google Scholar] [CrossRef] [PubMed]
  62. Zheng, W.; Zhao, W.; Wu, M.; Song, X.; Caro, F.; Sun, X.; Gazzaniga, F.; Stefanetti, G.; Oh, S.; Mekalanos, J.J.; et al. Microbiota-targeted maternal antibodies protect neonates from enteric infection. Nature 2020, 577, 543–548. [Google Scholar] [CrossRef] [PubMed]
  63. Miettinen, P.J.; Berger, J.E.; Meneses, J.; Phung, Y.; Pedersen, R.A.; Werb, Z.; Derynck, R. Epithelial immaturity and multiorgan failure in mice lacking epidermal growth factor receptor. Nature 1995, 376, 337–341. [Google Scholar] [CrossRef]
  64. Knoop, K.A.; Coughlin, P.E.; Floyd, A.N.; Ndao, I.M.; Hall-Moore, C.; Shaikh, N.; Gasparrini, A.J.; Rusconi, B.; Escobedo, M.; Good, M.; et al. Maternal activation of the EGFR prevents translocation of gut-residing pathogenic Escherichia coli in a model of late-onset neonatal sepsis. Proc. Natl. Acad. Sci. USA 2020. [Google Scholar] [CrossRef] [Green Version]
  65. Vogel, H.J. Lactoferrin, a bird’s eye view. Biochem. Cell Biol. 2012, 90, 233–244. [Google Scholar] [CrossRef] [PubMed]
  66. Smilowitz, J.T.; Lebrilla, C.B.; Mills, D.A.; German, J.B.; Freeman, S.L. Breast milk oligosaccharides: structure-function relationships in the neonate. Annu. Rev. Nutr. 2014, 34, 143–169. [Google Scholar] [CrossRef] [Green Version]
  67. Van de Perre, P. Transfer of antibody via mother’s milk. Vaccine 2003, 21, 3374–3376. [Google Scholar] [CrossRef]
  68. Capasso, L.; Borrelli, A.; Cerullo, J.; Pisanti, R.; Figliuolo, C.; Izzo, F.; Paccone, M.; Ferrara, T.; Lama, S.; Raimondi, F. Role of immunoglobulins in neonatal sepsis. Transl. Med. Unisa 2014, 11, 28–33. [Google Scholar]
  69. Li, Y.; Yang, S.; Wang, G.; Liu, M.; Zhang, Z.; Liu, H.; Yu, K.; Wang, C. Effects of immunotherapy on mortality in neonates with suspected or proven sepsis: A systematic review and network meta-analysis. BMC Pediatr. 2019, 19, 270. [Google Scholar] [CrossRef] [Green Version]
  70. Koch Meghan, A.; Reiner Gabrielle, L.; Lugo Kyler, A.; Kreuk Lieselotte, S.M.; Stanbery Alison, G.; Ansaldo, E.; Seher Thaddeus, D.; Ludington William, B.; Barton Gregory, M. Maternal IgG and IgA antibodies dampen mucosal t helper cell responses in early life. Cell 2016, 165, 827–841. [Google Scholar] [CrossRef] [Green Version]
  71. Collins, A.; Weitkamp, J.-H.; Wynn, J.L. Why are preterm newborns at increased risk of infection? Arch. Dis. Child. Fetal Neonatal Ed. 2018, 103, F391–F394. [Google Scholar] [CrossRef]
  72. Raymond, S.L.; López, M.C.; Baker, H.V.; Larson, S.D.; Efron, P.A.; Sweeney, T.E.; Khatri, P.; Moldawer, L.L.; Wynn, J.L. Unique transcriptomic response to sepsis is observed among patients of different age groups. PLoS ONE 2017, 12, e0184159. [Google Scholar] [CrossRef]
  73. Wynn, J.L.; Guthrie, S.O.; Wong, H.R.; Lahni, P.; Ungaro, R.; Lopez, M.C.; Baker, H.V.; Moldawer, L.L. Postnatal age is a critical determinant of the neonatal host response to sepsis. Mol. Med. 2015, 21, 496–504. [Google Scholar] [CrossRef] [PubMed]
  74. Ballard, O.; Morrow, A.L. Human milk composition: Nutrients and bioactive factors. Pediatr. Clin. N. Am. 2013, 60, 49–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kobata, R.; Tsukahara, H.; Ohshima, Y.; Ohta, N.; Tokuriki, S.; Tamura, S.; Mayumi, M. High levels of growth factors in human breast milk. Early Hum. Dev. 2008, 84, 67–69. [Google Scholar] [CrossRef] [PubMed]
  76. Michalsky, M.P.; Lara-Marquez, M.; Chun, L.; Besner, G.E. Heparin-binding EGF-like growth factor is present in human amniotic fluid and breast milk. J. Pediatr. Surg. 2002, 37, 1–6. [Google Scholar] [CrossRef]
  77. Nojiri, T.; Yoshizato, T.; Fukami, T.; Obama, H.; Yagi, H.; Yotsumoto, F.; Miyamoto, S. Clinical significance of amphiregulin and epidermal growth factor in colostrum. Arch. Gynecol. Obs. 2012, 286, 643–647. [Google Scholar] [CrossRef]
  78. Matsuoka, Y.; Idota, T. The concentration of epidermal growth factor in Japanese mother’s milk. J. Nutr. Sci. Vitaminol. 1995, 41, 241–251. [Google Scholar] [CrossRef] [Green Version]
  79. Luetteke, N.C.; Qiu, T.H.; Fenton, S.E.; Troyer, K.L.; Riedel, R.F.; Chang, A.; Lee, D.C. Targeted inactivation of the EGF and amphiregulin genes reveals distinct roles for EGF receptor ligands in mouse mammary gland development. Development 1999, 126, 2739. [Google Scholar]
  80. Read, L.C.; Upton, F.M.; Francis, G.L.; Wallace, J.C.; Dahlenberg, G.W.; Ballard, F.J. Changes in the growth-promoting activity of human milk during lactation. Pediatr. Res. 1984, 18, 133–139. [Google Scholar] [CrossRef] [Green Version]
  81. Chang, C.-J.; Chao, J.C.-J. Effect of human milk and epidermal growth factor on growth of human intestinal Caco-2 cells. J. Pediatr. Gastroenterol. Nutr. 2002, 34, 394–401. [Google Scholar] [CrossRef] [PubMed]
  82. Okuyama, H.; Urao, M.; Lee, D.; Drongowski, R.A.; Coran, A.G. The effect of epidermal growth factor on bacterial translocation in newborn rabbits. J. Pediatr. Surg. 1998, 33, 225–228. [Google Scholar] [CrossRef]
  83. Good, M.; Sodhi, C.P.; Egan, C.E.; Afrazi, A.; Jia, H.; Yamaguchi, Y.; Lu, P.; Branca, M.F.; Ma, C.; Prindle, T., Jr.; et al. Breast milk protects against the development of necrotizing enterocolitis through inhibition of Toll-like receptor 4 in the intestinal epithelium via activation of the epidermal growth factor receptor. Mucosal Immunol. 2015. [Google Scholar] [CrossRef] [PubMed]
  84. Villavicencio, A.; Rueda, M.S.; Turin, C.G.; Ochoa, T.J. Factors affecting lactoferrin concentration in human milk: How much do we know? Biochem. Cell Biol. 2017, 95, 12–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Yang, Z.; Jiang, R.; Chen, Q.; Wang, J.; Duan, Y.; Pang, X.; Jiang, S.; Bi, Y.; Zhang, H.; Lönnerdal, B.; et al. Concentration of lactoferrin in human milk and its variation during lactation in different Chinese populations. Nutrients 2018, 10, 1235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Czosnykowska-Łukacka, M.; Orczyk-Pawiłowicz, M.; Broers, B.; Królak-Olejnik, B. Lactoferrin in human milk of prolonged lactation. Nutrients 2019, 11, 2350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Bullen, J.J.; Rogers, H.J.; Leigh, L. Iron-binding proteins in milk and resistance to Escherichia coli infection in infants. Br. Med. J. 1972, 1, 69–75. [Google Scholar] [CrossRef] [Green Version]
  88. Sherman, M.P.; Miller, M.M.; Sherman, J.; Niklas, V. Lactoferrin and necrotizing enterocolitis. Curr. Opin. Pediatr. 2014, 26, 146–150. [Google Scholar] [CrossRef] [Green Version]
  89. Elass-Rochard, E.; Roseanu, A.; Legrand, D.; Trif, M.; Salmon, V.; Motas, C.; Montreuil, J.; Spik, G. Lactoferrin-lipopolysaccharide interaction: Involvement of the 28-34 loop region of human lactoferrin in the high-affinity binding to Escherichia coli 055B5 lipopolysaccharide. Biochem. J. 1995, 312, 839–845. [Google Scholar] [CrossRef] [Green Version]
  90. Dijkshoorn, L.; Brouwer, C.P.; Bogaards, S.J.; Nemec, A.; Van Den Broek, P.J.; Nibbering, P.H. The synthetic N-terminal peptide of human lactoferrin, hLF (1-11), is highly effective against experimental infection caused by multidrug-resistant Acinetobacter baumannii. Antimicrob. Agents Chemother. 2004, 48, 4919–4921. [Google Scholar] [CrossRef] [Green Version]
  91. Sherman, M.P.; Adamkin, D.H.; Radmacher, P.G.; Sherman, J.; Niklas, V. Protective Proteins in mammalian milks. NeoReviews 2012, 13, e293. [Google Scholar] [CrossRef]
  92. He, Y.; Cao, L.; Yu, J. Prophylactic lactoferrin for preventing late-onset sepsis and necrotizing enterocolitis in preterm infants: A PRISMA-compliant systematic review and meta-analysis. Med. (Baltim.) 2018, 97, e11976. [Google Scholar] [CrossRef] [PubMed]
  93. Sherman, M.P.; Sherman, J.; Arcinue, R.; Niklas, V. Randomized control trial of human recombinant lactoferrin: A substudy reveals effects on the fecal microbiome of very low birth weight infants. J. Pediatr. 2016, 173, S37–S42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ochoa, T.J.; Zegarra, J.; Bellomo, S.; Carcamo, C.P.; Cam, L.; Castañeda, A.; Villavicencio, A.; Gonzales, J.; Rueda, M.S.; Turin, C.G.; et al. Randomized controlled trial of bovine lactoferrin for prevention of sepsis and neurodevelopment impairment in infants weighing less than 2000 grams. J. Pediatr. 2020. [Google Scholar] [CrossRef] [PubMed]
  95. Manzoni, P.; Militello, M.A.; Rizzollo, S.; Tavella, E.; Messina, A.; Pieretto, M.; Boano, E.; Carlino, M.; Tognato, E.; Spola, R.; et al. Is lactoferrin more effective in reducing late-onset sepsis in preterm neonates fed formula than in those receiving mother’s own milk? Secondary analyses of two multicenter randomized controlled trials. Am. J. Perinatol. 2019, 36, S120–S125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Doyle, L.W.; Cheong, J.L.Y. Does bovine lactoferrin prevent late-onset neonatal sepsis? Lancet 2019, 393, 382–384. [Google Scholar] [CrossRef] [Green Version]
  97. Blanton, L.V.; Charbonneau, M.R.; Salih, T.; Barratt, M.J.; Venkatesh, S.; Ilkaveya, O.; Subramanian, S.; Manary, M.J.; Trehan, I.; Jorgensen, J.M.; et al. Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children. Science 2016, 351. [Google Scholar] [CrossRef] [Green Version]
  98. Kim, Y.-G.; Sakamoto, K.; Seo, S.-U.; Pickard, J.M.; Gillilland, M.G.; Pudlo, N.A.; Hoostal, M.; Li, X.; Wang, T.D.; Feehley, T.; et al. Neonatal acquisition of Clostridiaspecies protects against colonization by bacterial pathogens. Science 2017, 356, 315. [Google Scholar] [CrossRef] [Green Version]
  99. Feng, L.; Raman, A.S.; Hibberd, M.C.; Cheng, J.; Griffin, N.W.; Peng, Y.; Leyn, S.A.; Rodionov, D.A.; Osterman, A.L.; Gordon, J.I. Identifying determinants of bacterial fitness in a model of human gut microbial succession. Proc. Natl. Acad. Sci. USA 2020, 117, 2622. [Google Scholar] [CrossRef] [Green Version]
  100. Moossavi, S.; Atakora, F.; Miliku, K.; Sepehri, S.; Robertson, B.; Duan, Q.L.; Becker, A.B.; Mandhane, P.J.; Turvey, S.E.; Moraes, T.J.; et al. Integrated analysis of human milk microbiota with oligosaccharides and fatty acids in the child cohort. Front. Nutr. 2019, 6. [Google Scholar] [CrossRef]
  101. Thurl, S.; Munzert, M.; Henker, J.; Boehm, G.; Müller-Werner, B.; Jelinek, J.; Stahl, B. Variation of human milk oligosaccharides in relation to milk groups and lactational periods. Br. J. Nutr. 2010, 104, 1261–1271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Kunz, C.; Meyer, C.; Collado, M.C.; Geiger, L.; García-Mantrana, I.; Bertua-Ríos, B.; Martínez-Costa, C.; Borsch, C.; Rudloff, S. Influence of gestational age, secretor, and lewis blood group status on the oligosaccharide content of human milk. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 789–798. [Google Scholar] [CrossRef] [PubMed]
  103. Engfer, M.B.; Stahl, B.; Finke, B.; Sawatzki, G.; Daniel, H. Human milk oligosaccharides are resistant to enzymatic hydrolysis in the upper gastrointestinal tract. Am. J. Clin. Nutr. 2000, 71, 1589–1596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Underwood, M.A.; Davis, J.C.C.; Kalanetra, K.M.; Gehlot, S.; Patole, S.; Tancredi, D.J.; Mills, D.A.; Lebrilla, C.B.; Simmer, K. Digestion of human milk oligosaccharides by bifidobacterium breve in the premature infant. J. Pediatr. Gastroenterol. Nutr. 2017, 65, 449–455. [Google Scholar] [CrossRef] [PubMed]
  105. Ackerman, D.L.; Doster, R.S.; Weitkamp, J.-H.; Aronoff, D.M.; Gaddy, J.A.; Townsend, S.D. Human milk oligosaccharides exhibit antimicrobial and antibiofilm properties against group b streptococcus. ACS Infect. Dis. 2017, 3, 595–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wu, R.Y.; Li, B.; Koike, Y.; Määttänen, P.; Miyake, H.; Cadete, M.; Johnson-Henry, K.C.; Botts, S.R.; Lee, C.; Abrahamsson, T.R.; et al. Human milk oligosaccharides increase mucin expression in experimental necrotizing enterocolitis. Mol. Nutr. Food Res. 2019, 63, 1800658. [Google Scholar] [CrossRef]
  107. Cheng, L.; Kong, C.; Walvoort, M.T.C.; Faas, M.M.; de Vos, P. Human milk oligosaccharides differently modulate goblet cells under homeostatic, proinflammatory conditions and ER stress. Mol. Nutr. Food Res. 2020, 64, e1900976. [Google Scholar] [CrossRef] [Green Version]
  108. Bode, L.; Kunz, C.; Muhly-Reinholz, M.; Mayer, K.; Seeger, W.; Rudloff, S. Inhibition of monocyte, lymphocyte, and neutrophil adhesion to endothelial cells by human milk oligosaccharides. Thromb. Haemost. 2004, 92, 1402–1410. [Google Scholar] [CrossRef]
  109. Bode, L.; Rudloff, S.; Kunz, C.; Strobel, S.; Klein, N. Human milk oligosaccharides reduce platelet-neutrophil complex formation leading to a decrease in neutrophil beta 2 integrin expression. J. Leukoc. Biol. 2004, 76, 820–826. [Google Scholar] [CrossRef]
  110. Noll, A.J.; Yu, Y.; Lasanajak, Y.; Duska-McEwen, G.; Buck, R.H.; Smith, D.F.; Cummings, R.D. Human DC-SIGN binds specific human milk glycans. Biochem. J. 2016, 473, 1343–1353. [Google Scholar] [CrossRef] [Green Version]
  111. Donovan, S.M.; Comstock, S.S. Human milk oligosaccharides influence neonatal mucosal and systemic immunity. Ann. Nutr. Metab. 2016, 69 (Suppl. 2), 42–51. [Google Scholar] [CrossRef] [PubMed]
  112. Triantis, V.; Bode, L.; van Neerven, R.J.J. Immunological effects of human milk oligosaccharides. Front. Pediatr. 2018, 6, 190. [Google Scholar] [CrossRef] [PubMed]
  113. Morrow, A.L.; Meinzen-Derr, J.; Huang, P.; Schibler, K.R.; Cahill, T.; Keddache, M.; Kallapur, S.G.; Newburg, D.S.; Tabangin, M.; Warner, B.B.; et al. Fucosyltransferase 2 non-secretor and low secretor status predicts severe outcomes in premature infants. J. Pediatr. 2011, 158, 745–751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wejryd, E.; Martí, M.; Marchini, G.; Werme, A.; Jonsson, B.; Landberg, E.; Abrahamsson, T.R. Low diversity of human milk oligosaccharides is associated with necrotising enterocolitis in extremely low birth weight infants. Nutrients 2018, 10, 1556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Bering, S.B. Human milk oligosaccharides to prevent gut dysfunction and necrotizing enterocolitis in preterm neonates. Nutrients 2018, 10, 1461. [Google Scholar] [CrossRef] [Green Version]
  116. Demers-Mathieu, V.; Huston, R.K.; Markell, A.M.; McCulley, E.A.; Martin, R.L.; Spooner, M.; Dallas, D.C. Differences in maternal immunoglobulins within mother’s own breast milk and donor breast milk and across digestion in preterm infants. Nutrients 2019, 11, 920. [Google Scholar] [CrossRef] [Green Version]
  117. Cossey, V.; Cossey, V.; Vanhole, C.; Eerdekens, A.; Rayyan, M.; Fieuws, S.; Schuermans, A. Pasteurization of mother’s own milk for preterm infants does not reduce the incidence of late-onset sepsis. Neonatology 2013, 103, 170–176. [Google Scholar] [CrossRef]
  118. Czank, C.; Prime, D.K.; Hartmann, B.; Simmer, K.; Hartmann, P.E. Retention of the immunological proteins of pasteurized human milk in relation to pasteurizer design and practice. Pediatr. Res. 2009, 66, 374–379. [Google Scholar] [CrossRef] [Green Version]
  119. Peila, C.; Moro, G.E.; Bertino, E.; Cavallarin, L.; Giribaldi, M.; Giuliani, F.; Cresi, F.; Coscia, A. The effect of holder pasteurization on nutrients and biologically-active components in donor human milk: A review. Nutrients 2016, 8, 477. [Google Scholar] [CrossRef] [Green Version]
  120. Baro, C.; Giribaldi, M.; Arslanoglu, S.; Giuffrida, M.G.; Dellavalle, G.; Conti, A.; Tonetto, P.; Biasini, A.; Coscia, A.; Fabris, C.; et al. Effect of two pasteurization methods on the protein content of human milk. Front. Biosci. (Elite Ed.) 2011, 3, 818–829. [Google Scholar] [CrossRef] [Green Version]
  121. Escuder-Vieco, D.; Espinosa-Martos, I.; Rodríguez, J.M.; Fernández, L.; Pallás-Alonso, C.R. Effect of HTST and holder pasteurization on the concentration of immunoglobulins, growth factors, and hormones in donor human milk. Front. Immunol. 2018, 9, 2222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Maternal protection from enteric pathogens. (A) Components in breastmilk can limit enteric pathogen dissemination. (1) Maternal antibodies (IgG, IgA) can bind bacteria and directly inhibit pathogen adherence and invasion [62]. (2) Growth factors [epidermal growth factor (EGF), amphiregulin (AREG), heparin-binding epidermal growth factor-like factor (HB-EGF), and tumor-growth factor-alpha (TGF-α)] bind the epidermal growth factor receptor (EGFR) on epithelial cells to promote barrier function by cell proliferation and growth [63], and by limiting translocation via goblet cells [64]. (3) Lactoferrin sequesters iron which limits pathogen growth [65]. (4) Human milk oligosaccharides (HMOs) promote the development of the intestinal microbiota [66], which can offer colonization resistance to enteric pathogens [30]. (B) In the absence of these factors, pathogens can colonize the intestine lumen, cross the epithelium potentially through goblet cells [64], and disseminate to organs through the system, including the mesenteric lymph node (MLN) and spleen, resulting in late-onset sepsis (LOS).
Figure 1. Maternal protection from enteric pathogens. (A) Components in breastmilk can limit enteric pathogen dissemination. (1) Maternal antibodies (IgG, IgA) can bind bacteria and directly inhibit pathogen adherence and invasion [62]. (2) Growth factors [epidermal growth factor (EGF), amphiregulin (AREG), heparin-binding epidermal growth factor-like factor (HB-EGF), and tumor-growth factor-alpha (TGF-α)] bind the epidermal growth factor receptor (EGFR) on epithelial cells to promote barrier function by cell proliferation and growth [63], and by limiting translocation via goblet cells [64]. (3) Lactoferrin sequesters iron which limits pathogen growth [65]. (4) Human milk oligosaccharides (HMOs) promote the development of the intestinal microbiota [66], which can offer colonization resistance to enteric pathogens [30]. (B) In the absence of these factors, pathogens can colonize the intestine lumen, cross the epithelium potentially through goblet cells [64], and disseminate to organs through the system, including the mesenteric lymph node (MLN) and spleen, resulting in late-onset sepsis (LOS).
Nutrients 12 01045 g001

Share and Cite

MDPI and ACS Style

Kleist, S.A.; Knoop, K.A. Understanding the Elements of Maternal Protection from Systemic Bacterial Infections during Early Life. Nutrients 2020, 12, 1045. https://doi.org/10.3390/nu12041045

AMA Style

Kleist SA, Knoop KA. Understanding the Elements of Maternal Protection from Systemic Bacterial Infections during Early Life. Nutrients. 2020; 12(4):1045. https://doi.org/10.3390/nu12041045

Chicago/Turabian Style

Kleist, Sierra A., and Kathryn A. Knoop. 2020. "Understanding the Elements of Maternal Protection from Systemic Bacterial Infections during Early Life" Nutrients 12, no. 4: 1045. https://doi.org/10.3390/nu12041045

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop