Next Article in Journal
Bacteroides thetaiotaomicron Fosters the Growth of Butyrate-Producing Anaerostipes caccae in the Presence of Lactose and Total Human Milk Carbohydrates
Next Article in Special Issue
Prevention of Clostridium difficile Infection and Associated Diarrhea: An Unsolved Problem
Previous Article in Journal
Caffeine Stabilises Fission Yeast Wee1 in a Rad24-Dependent Manner but Attenuates Its Expression in Response to DNA Damage
Previous Article in Special Issue
Application of an O-Linked Glycosylation System in Yersinia enterocolitica Serotype O:9 to Generate a New Candidate Vaccine against Brucella abortus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Postbiotics against Pathogens Commonly Involved in Pediatric Infectious Diseases

by
Anastasia Mantziari
1,*,
Seppo Salminen
1,
Hania Szajewska
2 and
Jeadran Nevardo Malagón-Rojas
3,4
1
Functional Foods Forum, Faculty of Medicine, University of Turku, 20520 Turku, Finland
2
Department of Paediatrics at the Medical University of Warsaw, 02091 Warsaw, Poland
3
Facultad de Medicina, Universidad El Bosque, 110121 Bogotá, Colombia
4
Instituto Nacional de Salud de Colombia, 111321 Bogotá, Colombia
*
Author to whom correspondence should be addressed.
Microorganisms 2020, 8(10), 1510; https://doi.org/10.3390/microorganisms8101510
Submission received: 25 August 2020 / Revised: 22 September 2020 / Accepted: 29 September 2020 / Published: 30 September 2020
(This article belongs to the Special Issue Infectious Diseases, New Approaches to Old Problems)

Abstract

:
The Sustainable Development goals for 2020 included reducing all causes associated with infant and perinatal mortality in their priorities. The use of compounds with bioactive properties has been proposed as a therapeutic strategy due to their stimulating effect on the host’s immune system. Additionally, biotherapeutic products such as postbiotics, tentatively defined as compounds produced during a fermentation process that support health and well-being, promote intestinal barrier integrity without posing considerable risks to children’s health. Although this is a concept in development, there are increasing studies in the field of nutrition, chemistry, and health that aim to understand how postbiotics can help prevent different types of infections in priority populations such as minors under the age of five. The present review aims to describe the main mechanisms of action of postbiotics. In addition, it presents the available current evidence regarding the effects of postbiotics against pathogens commonly involved in pediatric infections. Postbiotics may constitute a safe alternative capable of modulating the cellular response and stimulating the host’s humoral response.

1. Introduction

Saving children’s lives and improving child health are two critical global priorities and represent essential targets of Goal 3 (Good Health and Well-Being) of the United Nations Sustainable Development Goals. Since 1990, there has been a significant reduction in the mortality rates of children aged one month to five years old [1]. However, this is not the case for newborn mortality rates that are falling behind, accounting for the more significant proportion of all deaths among children younger than five years old. To understand this pattern, one must look closer at the causes of newborn deaths. The vast majority of infant deaths occur within the first month of life and are caused by being born too soon, complications during delivery, and infectious diseases [2]. Among infectious diseases, diarrheal disease is one of the leading causes of death in children under five years old [3]. Common causes of bacterial diarrhea include diarrhoeagenic Escherichia coli (DEC; E. coli), Shigella species, Salmonella species, Campylobacter, and Yersinia enterocolitica. Besides E. coli, rotavirus (RV) is the other most common etiological agent of moderate-to-severe diarrhea in low-income countries. Fortunately, most of these causes are preventable and treatable but require global attention, increased access to quality healthcare, and cost-effective nutritional interventions.
Various factors contribute to infectious disease risk, including low birth weight, genetics, and diet, among others. The establishment of a healthy gut microbiome plays a pivotal role in the development of the immune system and could potentially lower the risk of infectious diseases in infants and children. One way to shape and modulate the gut microbiome is through diet. Although breast milk is the optimal source of nutrition for developing a diverse and healthy microbiota composition, there are cases that it may not always be available or adequate for the infant. For this reason, infant formulas are manufactured and are continuously being improved as they aim to resemble the nutritional composition of breast milk. Over the past decades, different preparations and interventions, such as supplementation with probiotics, have been explored, aiming to improve the composition of infant formulas. Probiotics are defined as ‘’live microorganisms that, when administered in adequate amounts, confer a health benefit on the host’’ [4]. The rationale for using probiotics to fight infections is that probiotics demonstrate a good ability to bind to intestinal mucus and compete with specific pathogens for the same adhesion sights. They therefore block pathogen adherence and consequently make pathogen propagation challenging [5]. The adhesion of probiotics to intestinal mucus or epithelial cells is mediated by surface proteins of probiotic bacteria and can inhibit pathogen adhesion through competitive exclusion. Some of these bacterial surface compounds include proteins, glycoproteins, lipoproteins, lipoteichoic acids, lipopolysaccharides, adhensins, and flagellins [6]. These properties support supplementing infant formulas with probiotics. However, at present, there is insufficient data to recommend the routine use of probiotic-supplemented formulas [7,8,9].
Besides providing possible protection against infections, probiotics may be a good new alternative antimicrobial solution when an infant is already infected by a pathogen. One intervention aimed to treat bacterial diarrhea is antibiotic therapy, although it is generally not recommended (with some exceptions). Moreover, many pathogenic strains have developed resistance to antibiotics [10]. On the other hand, there is the increasing concern that some probiotics harvest antibiotic-resistant genes in their genome that can be transferred to other potentially pathogenic bacteria [11,12]. In addition, many physicians are still skeptical with regards to the use of probiotics for pediatric care due to rare case reports of probiotic adverse effects [13,14]. Growing evidence suggests that many probiotic strains do not have to be alive to exert potential beneficial effects on host health [15]. For instance, it has been shown that, depending on the inactivation method, inactivated probiotics can adhere better to intestinal mucus compared to viable bacteria [16]. On the basis of these facts, new research is exploring the use of fermented infant formulas containing inactivated probiotics and/or their metabolic products and their role in protecting against infections [17,18,19].
Recently, new terms such as postbiotics, paraprobiotics, metabiotics, proteobiotics, pharmabiotics, and ghost probiotics have emerged [20,21,22,23,24,25]. Interestingly, evidence from the literature suggests that by using the term postbiotics, one refers to either inactivated probiotic strains or their metabolic products or both. Currently, a definition is under development; however, tentatively, postbiotics have been defined as bioactive ingredients from food-grade microorganisms resulting from a fermentation process that supports health and/or well-being [26]. The term postbiotic includes inactivated microbes and/or cell structures or metabolites that are released after bacterial lysis or that are secreted during fermentation. In most studies, postbiotics derive from the fermented culture medium that is being filtered or heated after the growth of the microorganism, resulting in a liquid called cell-free supernatant (CFS; known also as spent culture supernatant (SCS) or cell-free spent medium (CFSM)) [27]. This means that a combination of bioactive molecules is present rather than one specific purified compound [27]. Other ways to obtain postbiotics include the inactivation of probiotics using heat, filtration, sonication, centrifugation, and ultraviolet radiation (UV), among others [28]. In this case, bacterial lysis can occur, releasing a large array of compounds such as DNA, enzymes, lipoteichoic acids, and other intracellular metabolites that can serve as possible postbiotics [28].
A 2020 systematic review evaluated the clinical effects of postbiotics for preventing and treating common infectious diseases in children [29]. Given that there is limited research on postbiotics, coupled with their potential beneficial effects, this review investigates and reports the possible preventive mechanisms and uses of postbiotics against pediatric infectious diseases on the basis of current evidence.

2. Potential Mechanisms of Action

The action mechanism of postbiotics and their role in improving host health are not yet clearly defined. However, two mechanisms might explain how postbiotics can stimulate and modulate the host’s immunological response, involving both elicit immune and acquired host response. The initial response is related to the innate immune system, and it consists of a series of pattern recognition receptors able to associate with microorganisms. Two of these pattern recognition receptors involved in the host response to postbiotics are the nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) and the toll-like receptors (TLRs) (Figure 1).
The NOD domain of the NLRs functions as an intracellular receptor and is associated with the innate immune response [30]. On the basis of their terminal domain, NLRs are divided into four functional categories: inflammasome assembly, signaling transduction, transcription activation, and autophagy [31]. The NLRs can recognize different ligands from microbial pathogens such as viral RNA; peptidoglycan; and flagellin, the main component of flagella. For instance, muramyl dipeptide (MDP), a bioactive peptidoglycan motif common to all bacteria, activates the NACHT [(which comes from present in NAIP (neuronal apoptosis inhibitory protein), CIITA (MHC class II transcription activator), HET-E (incompatibility locus protein from Podospora anserina) and TP1 (telomerase-associated protein)] domain-, leucine-rich repeat-, and PYRIN-containing protein 1 (NLRP1) [32]. Upon activation, NLRP1 forms a multi protein complex known as the NLRP1 inflammasome that is expressed in innate and adaptive immunity cells including epithelial cells, T lymphocytes, macrophages, and dendritic cells (DCs) [33]. This allows the activation of caspase-1, leading to the secretion of pro-inflammatory cytokines interleukin (IL)-1β and IL-18.
The host’s innate immune system can also be stimulated via activation of the TLRs, a family of receptors that can recognize associated pathogens [34]. Each type of TLR can bind to a specific bacterial structure: lipopolysaccharides (LPS) are recognized by TLR4; lipoproteins, lipoteichoic acid, and peptidoglycan are recognized by TLR2; and flagellin is recognized by TLR5 [35]. Moreover, TLR3, TLR7, and TLR8 can bind bacterial RNA, in contrast to TLR9, which has been identified as the DNA-recognizing receptor [36]. Altogether, the activation of TLRs modulates the cytokine profile (signaling molecules) in immune cells. For instance, TLR9 is responsible for recognizing bacterial CpG site (cytosines followed by guanine residues) DNA released from probiotics. Since the C+G content between bacterial species is different, their affinity to bind to and activate TLR9 may vary [37]. It is also worth mentioning that the location of TLR9 on the cell membrane plays an essential role in its function and subsequent activation of signaling pathways. For example, TLR9 present on the basolateral membrane of intestinal epithelial cells activates the nuclear factor kappa B (NF-ĸB) pathway compared to the apical membrane that inhibits it [38]. The c-Jun N-terminal kinase (JNK) and NF-κB pathways are essential due to their role in directing the synthesis of pro-inflammatory cytokines [39].
During infection, pro-inflammatory cytokines are produced and released from various cell types, including immune cells, thus initiating cell inflammation. Cytokines can have anti-inflammatory or pro-inflammatory properties. However, some cytokines may promote or suppress inflammation, depending on the immunological situation. Interestingly, anti-inflammatory cytokines are required to inhibit pro-inflammatory cytokine production, consequently suppressing inflammation, whereas pro-inflammatory cytokines are necessary for the activation of immune cell function. Thus, it is evident that a balance between the two has to be maintained in order to avoid immunosuppression or excessive pro-inflammatory response [40]. Many lactic acid bacteria (LAB) produce metabolites that can induce cytokine production against pathogens [41]. For example, inactivated LAB cells have been found to induce the production of various cytokines such as interleukin-12 (IL-12) and IL-10 in macrophages and DCs, respectively [42,43].
Bacterial metabolites such as lactic acids, organic acids, bacteriocins, proteases, peroxides, and exopolysaccharides are known to have antibacterial and antifungal properties [44,45]. Organic acids such as short-chain fatty acids (SCFAs; mainly acetate, propionate, butyrate), formic acid, and propionic acid act against pathogenic bacteria by interfering with the cytoplasmic membrane structure and nutrient transport, as well as by influencing macromolecular synthesis [46]. Nevertheless, the effectiveness of organic acids is primarily determined by the pH. At low pH values, organic acids are found in non-dissociated forms and can penetrate the bacteria’s hydrophobic cell membranes [47]. Once they are inside the bacterial cell, SFCAs dissociate into anions and protons. However, the bacterial cell is trying to export the extra protons since the cytoplasm has a neutral pH, leading to the depletion of cellular energy [48].
Bioactive peptides such as bacteriocins have been reported to display inhibitory activity against various pathogens such as Listeria monocytogenes, Clostridium perfringens, Salmonella enterica, and Escherichia coli [49,50]. Bacteriocins can either have a bactericidal or bacteriostatic effect, inhibiting cell growth, and have lately been found to exert an antibiofilm activity [51]. Other antimicrobial molecules released by probiotic bacteria are hydrogen peroxide (H2O2) and extracellular high molecular weight sugar polymers known as exopolysaccharides (EPS). Hydrogen peroxide produced by lactobacilli may oxidize different compounds (proteins, nucleic acids, and lipids) of pathogenic microorganisms, thus leading to significant impairments in structure and possibly to the loss of cell viability [52,53]. On the other hand, although the mechanism of action is not yet clear, it has been shown that EPS from lactobacilli and bifidobacteria plays an important role in protecting pathogenic bacteria such as enterotoxigenic E. coli and Citrobacter rodentium [54,55].

2.1. Effects against Pathogenic Bacteria

Although it is both preventable and treatable, diarrheal disease is one of the leading causes of death among children under the age of five. One of its major causes is inadequate sanitation or contamination of water that can lead to either bacterial, viral, or parasitic infections [3]. The potential infectious disease targets for postbiotics discussed in the current review are summarized in Table 1.

2.1.1. Escherichia coli

Infection with pathogenic strains of E. coli is one of the most common etiological agents of moderate-to-severe diarrhea in low-income countries and is considered a significant public health problem [84,85]. Accumulated evidence from in vitro studies supports the use of postbiotics for the protection against infectious diseases. One prominent attempt was made by Abdelhamid and coworkers, showing that six CFS were able to reduce the growth of two multi-resistant clinical E. coli isolates (WW1 and IC2) [56]. Furthermore, all six CFS derived from Man–Rogosa–Sharpe (MRS) broth or skim milk were effective in inhibiting the biofilm formation of both E. coli strains, except for skim milk CFS that exhibited zero or slight E. coli IC2 biofilm inhibition.
A recent study demonstrated that CFS from Bifidobacterium bifidum BBA1 and Bifidobacterium crudilactis FR/62/B/3 grown in media supplemented with 3′-sialyllactose, a major bovine milk oligosaccharide, induced a significant decrease in the expression of genes mainly implicated in the virulence mechanism of E. coli O157:H7 [57]. Another Bifidobacterium-producing postbiotic metabolite that acts against E. coli O157:H7 is Bifidobacterium bifidum ATCC 29521 [58]. In vitro experiments indicated that CFSM fractions from Bifidobacterium bifidum ATCC 29521 were able to reduce the attachment of the pathogen to intestinal cells by 70%. Moreover, heat-killed Lactobacillus acidophilus strain LB (Lactobacillus Boucard) with its SCS displayed a high adherence to Caco-2 cells and was able to inhibit the cell association and cell entry of different diarrheagenic bacteria including enterotoxigenic (ETEC) and enteropathogenic (EPEC) E. coli [59,60,86]. It can be postulated that postbiotics present in the SCS are responsible for the increased adhesion to Caco-2 cells. For instance, the ability of L. acidophilus LB to adhere to Caco-2 cells was significantly decreased when the CFS was discarded or replaced by fresh culture medium. This observation suggested that a secreted metabolic product produced by L. acidophilus LB was responsible for its increased adhesion to Caco-2 cells. By treating the supernatant with trypsin and pronase, the adhesion of L. acidophilus LB was greatly decreased, leading the authors to conclude that this metabolic product was an extracellular protein. In the same study, a second factor, namely, a non-proteinaceous cell surface component, also seemed to mediate the adhesion of Lactobacillus strain LB to Caco-2 cells [87]. Recently, the three novel probiotics Lactobacillus paracasei (reclassified to Lacticaseibacillus paracasei) CNCM I-4034, Bifidobacterium breve CNCM I-4035, and Lactobacillus rhamnosus (reclassified to Lacticaseibacillus rhamnosus) CNCM I-4036 were isolated from the stool of breastfed infants by Muñoz-Quezada and coworkers [83,88]. In addition, the same group evaluated the ability of CFSs derived from the three probiotics to inhibit the growth of E. coli ETEC and EPEC [61]. The authors reported that all CFSs were able to inhibit the growth of E. coli up to 40%. However, when the CFSs were neutralized, this antimicrobial activity was either maintained or lost. Another study has also reported that the neutralization of CFS from lactobacilli had a negative effect on its antimicrobial activity against E. coli ETEC [62].
Another group investigated the potential postbiotic use against neonatal E. coli K1 infection [63]. The CFS from Lactobacillus rhamnosus GG (LGG) was pre-incubated with Caco-2 cells or administered to neonatal rats and then exposed to E. coli [83]. This pre-treatment with the CFS could inhibit adhesion, invasion, and translocation of E. coli to Caco-2 monolayer. In the case of neonatal rats, the group exposed to CFS reduced the bacterial intestinal colonization, translocation, dissemination, and systemic infection. Moreover, pre-incubation with CFS could promote the maturation of neonatal intestinal defense as treated rats had higher expression of immunoglobulin A (IgA) compared to untreated rats. Moreover, two recent projects from the same group identified a novel secreted protein (HM0539) from the CFS of LGG that may promote the development of neonatal intestinal defense and prevent against E. coli K1 and O157:H7 infection [64,89]. More specifically, HM0539 treatment significantly increased the expression of tight junction proteins and inhibited their destruction by E. coli O157:H7 in HT-29 cells and the jejunum of mice. Overall, these findings contribute to the fact that postbiotics instead of probiotics could be good candidates for the prophylactic treatment against E. coli K1 and O157:H7 infection, thus potentially protecting against neonatal sepsis and meningitis.

2.1.2. Cronobacter sakazakii

Growing concerns over emerging antibiotic resistance coupled with the high risk of Cronobacter sakazakii infections in formula-fed infants have led to new studies investigating the efficacy of postbiotics produced by different probiotic strains against C. sakazakii [90]. For instance, lyophilized and heat-treated probiotic Lactobacillus acidophilus INMIA 9602 Er 317/402 (strain Narine) proved to inhibit the growth of C. sakazakii in contaminated reconstructed powdered infant formula after coculture for 6 h at 37 °C [65]. However, when employing the agar well diffusion method, CFS Narine was the most effective in inhibiting C. sakazakii growth compared to lyophilized and heat-treated Narine. Indeed, when examining the morphology by transmission electron microscopy, it was evident that CFS Narine was able to damage the cell membrane of C. sakazakii. On the other hand, and in agreement with previous studies, when the CFS was neutralized, this in vitro antimicrobial activity was diminished. Four more CFS (filtered and filtered + heat inactivated) from Lactobacillus casei strain Shirota (Yakult); Lactobacillus sporongenes, Streptococcus faecalis, Clostridium butyricum, Bacillus mesentericus (Bifilac; cells); Streptococcus faecalis, Lactobacillus sporongenes, Clostridium butyricum, Bacillus mesentericus (Vibact; spores); and Lactobacillus sporongenes (Caplac; spores) were successful in inhibiting C. sakazakii growth. The pathogenic potential of this pathogen was also increased due to its high biofilm-forming capacity. Additionally, in this study, a higher biofilm inhibitory activity (> 80%) was observed at the initial stages of biofilm formation. However, the same group reported that when the four CFSs were neutralized, the possessed antimicrobial activity was lost [66].

2.1.3. Clostridioides difficile

Postbiotics may also be a new alternative to prevent or treat Clostridioides difficile (formerly known as Clostridium difficile) infections (CDI). Although considered extremely rare in neonates and infants, further studies report the occurrence of diarrhea in this specific population [91,92]. Routine testing for CDI in infants is currently not recommended due to the high asymptomatic colonization in this group and should be performed only when other causes of diarrhea, such as rotavirus, have been excluded [93,94]. However, toxigenic C. difficile has been identified alone or in co-infection with rotavirus, suggesting that it could be a contributing factor to diarrhea in infants [92,95]. New evidence shows that CFS from various lactic acid bacteria (LAB) cocultured with multi-resistant C. difficile can reduce the cytotoxic effects of clostridial toxins on different cell lines compared to pure C. difficile CFS. Additionally, neutralized CFS from LAB + C. difficile can reduce the level of IL-8 and tumor necrosis factor α (TNF-α), two pro-inflammatory cytokines released by intestinal epithelial cells, caused by the CFS of C. difficile [67,68].

2.1.4. Salmonella spp.

Postbiotics could also be a potential treatment against Salmonella infections. CFSs from Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035, and Lactobacillus rhamnosus CNCM I-4036 were able to inhibit the growth of Salmonella typhimurium and/or Salmonella typhi but in most cases this antimicrobial activity was lost when the CFSs were neutralized [61]. In addition, CFS from Bifidobacterium bifidum ATCC 29521 proved to inhibit the multiplication of Salmonella typhimurium in macrophages, thus affecting its ability to colonize the gastrointestinal tract [58]. Besides interfering with its growth, a study on Caco-2 cells indicated that heat-killed Lactobacillus acidophilus LB with its SCS had a protective effect against Salmonella typhimurium by means of blocking the cell entry of the pathogen into the cell line [59]. Moreover, a study on a novel organ culture system of human intestinal mucosa showed that the supernatant from Lactobacillus paracasei B21060 grown in MRS inhibited the inflammatory potential of Salmonella and conditioned the epithelium against Salmonella invasion [69].
On the other hand, postbiotics could increase the inflammatory response in the presence of pathogens. One interesting study used an in vitro bilayer system of human intestinal epithelial cells (IEC) and intestinal-like dendritic cells (DC) to study the immunomodulatory properties of Lactobacillus paracasei CNCM I-4034 CFS [83]. The authors reported that the CFS increased the production of pro-inflammatory cytokines [IL-1β, IL-6, transforming growth factor β2 (TGF-β2), and interferon-γ-inducible protein 10 (IP-10)] when Salmonella typhi was present, thus stimulating the innate immune system. This increase in cytokine production was associated with the downregulation of TLR genes, except TLR9, which was upregulated. Due to increased concerns that probiotics could augment ongoing inflammation, Zagato and coworkers decided to investigate whether postbiotics could be an alternative option in suppressing inflammation. Indeed, the in vitro experiment indicated that when co-incubated with Salmonella typhimurium, only the CFS and not the live cells from Lactobacillus paracasei CBA L74 could reduce the pro-inflammatory IL-12p70 and increase anti-inflammatory IL-10 in human DCs [43]. Moreover, in the same study, mice treated with L. paracasei CBA L74 CFS were protected against dextran sulfate sodium (DSS)-induced colitis since they demonstrated reduced weight loss compared to the mice treated only with live cells. Trying to evaluate the properties of the fermented infant formula by L. paracasei CBA L74, the authors found that it displayed protective effects against colitis, showed a slightly longer survival to a lethal dose of Salmonella, and increased the production of anti-inflammatory cytokines. On the other hand, the administration of live L. paracasei CBA L74 led to the death of the mice. Overall, these results indicated that it is essential to consider the use of postbiotics in infant formulas, especially when targeting their administration to immunocompromised infants. Another animal study was carried out to determine the protective capacity of autochthonous Lactobacillus strains to prevent Salmonella Typhimurium infection in mice. The strains used in the study were (Lactobacillus helveticus (Lh 05, Lh 06, Lh 07, and Lh 08), Lactobacillus delbrueckii subsp. bulgaricus (Lb 03, Lb 09, Lb 10, Lb 11, and Lb 12), and L. delbrueckii subsp. lactis (Ll 210 and Ll 133). The cell-free supernatant was spray-dried to inactivate it. The authors reported that the administration of postbiotics was effective in protecting the mice from Salmonella typhimurium infection, increasing the survival in the postbiotic group [96].

2.2. Effects against Viruses

2.2.1. Influenza

Influenza virus infections contribute significantly to respiratory hospitalizations among children, with those aged less than five years old being at a higher risk for influenza complications [97,98]. This not only has detrimental health outcomes for the children but is considered a financial burden to society, employees, and parents who have to stay home to attend to their children [99,100]. Apart from vaccines and antiviral medications, postbiotics could be a useful alternative for the early treatment of seasonal flu. New evidence reveals that heat-killed Lactobacillus paracasei MCC1849 is efficient in increasing the IgA production in the small intestine and serum, facilitating protection against influenza virus infection in mice [81]. IgA plays an essential role in the host defense mechanism as it can inhibit the adherence of pathogenic bacteria and viruses to epithelial cells. Of note, an increased IL-10 gene expression in MCC1849-fed mice compared to the control was confirmed, possibly positively affecting the production of IgA. Finally, heat-killed Lactobacillus paracasei MCC1849 increased the gene expression of IL-12p40, IL-10, IL-21, signal transducer and activator of transcription 4 (STAT4), and B cell lymphoma 6 (BCL6) associated with follicular helper T (TFH) cell differentiation Peyer’s patches, where IgA production occurs. Additionally, postbiotic compounds produced by Lactobacillus plantarum (reclassified to Lactiplantibacillus plantarum) YML009 and Leuconostoc mesenteroides YML003, two strains isolated from kimchi, exhibited an antiviral effect on various types of influenza virus, including H1N1 and H9N2, respectively [71,72]. More specifically, the antiviral efficacy of L. plantarum YML009 CFS was increased when compared with Tamiflu, an antiviral medication, and it was found to revoke viral infection entirely.

2.2.2. Rotavirus

A significant cause of diarrhea-related child mortality worldwide is infection with RV [101]. Apart from E. coli, RV is the other most common cause of moderate-to-severe diarrhea, particularly in low-income countries. Some studies have demonstrated that postbiotics are effective in preventing and treating RV diarrhea. For instance, heat-inactivated L. acidophilus LB proved to be effective in treating children with diarrhea by decreasing its duration [102,103]. This decrease was more prominent in children that did not receive antibiotics before inclusion. However, other studies found no effect on the duration of diarrhea. A randomized clinical trial compared the impact of viable and heat-inactivated LGG in children under the age of four with rotaviral diarrhea. The findings indicated that the group of children treated with viable LGG had higher sera anti-RV IgA response to rotavirus than children in the postbiotic group. Nevertheless, in the clinical outcome, there were no differences in the duration of diarrhea, which was short (2.5–3 days) and equal between probiotic and postbiotic groups [73]. Another study, conducted in rats, evaluated the effect of a prebiotic-enriched, heat-treated fermented milk formula on the prevention of rotavirus-associated diarrhea [18]. This dietary intervention reduced two of the clinical symptoms of diarrhea (incidence and severity) and improved the immune response against RV by increasing sera anti-RV IgG and intestinal anti-RV IgA. These findings demonstrate that postbiotics, produced during the fermentation of infant formulas, might also have the potential to prevent diarrhea in children.

2.2.3. Human Immunodeficiency Virus

Human immunodeficiency virus (HIV) transmission from mother to child remains a significant public health problem. In particular, in countries of Southern Africa, more than one in five pregnant women are HIV-infected, and additionally, regarding children, infectious diseases are the leading cause of infant mortality [104,105]. Until 2010, the World Health Organization (WHO) advised HIV-positive mothers to avoid breastfeeding and instead use infant formula for their child’s nutrition to prevent transmission of HIV [106,107,108]. Since then, new evidence has surfaced, suggesting that the combination of breastfeeding and antiretroviral treatment can significantly reduce the risk of postnatal HIV transmission to infants through breastfeeding [109]. Over the years, breastmilk use has received growing attention due to its immunomodulatory and antimicrobial properties. More recently, 38 strains of heat-killed commensal breastmilk bacteria and their CFS were evaluated for their capacity to constrain HIV-1 infection in vitro [74]. Findings showed that the isolated strains effectively inhibited HIV-1 infection, with heat-inactivated cells exerting an increased antiviral activity compared to the CFS. This might be due to the association of the virus with bacterial surface components, such as peptidoglycans, exopolysaccharides, and lipopolysaccharide moieties. Consequently, postbiotics from human breastmilk may be essential for protecting against HIV-1 in the breastfeeding infant.

2.3. Effects against Candida spp.

Another major cause of pediatric-associated infections is the prevalence of candidiasis. In the United States, invasive candidiasis in the pediatric population mainly occurs in neonates and infants less than one year of age and can often lead to high mortality rates [110]. Prematurity and admission to the intensive care unit are considered the main predisposing factors for candidiasis [111]. Prevention against fungal infection in preterm infants can be achieved through antifungal medication, but there are strong concerns over potential adverse effects and the possible emergence of resistant strains [112]. In light of this, researchers have focused on finding alternatives, and through in vitro studies have demonstrated that SCS of L. rhamnosus and L. casei exhibit antifungal activity against blastoconidia and biofilm of C. albicans [77]. Moreover, CFS from honey and vaginal LAB were proven to be interesting sources of antifungals [75,76]. Compounds present in the supernatant inhibited the adhesion and decreased the biofilm formation of five Candida spp. on polystyrene and HeLa cells. It can be hypothesized that these compounds could help prevent adhesion on medical devices, thus decreasing the risk of potential candidiasis in hospitalized children.

2.4. Effect against Common Pediatric Infectious Diseases with Unknown Cause

Postbiotics from L. paracasei CBA L74 derived from fermented formula can also prevent common pediatric infectious diseases. For instance, Corsello and coworkers reported that when the fermented product was given to healthy children aged 1–4 years in daycare or preschool, its consumption was associated with a reduction in the incidence of common infectious diseases [17]. However, the cause of pediatric infectious diseases was not evaluated. Additionally, the same product induced an increase in fecal biomarkers of innate (α- and β-defensins, cathelicidin) and acquired immunity (secretory IgA) after 3 months. Cow’s milk or rice fermented with L. paracasei CBA L74 was also found to prevent common pediatric infectious diseases in children attending daycare after 3 months of consumption [78]. This could be explained by the higher levels of α-defensin, β-defensin, cathelicidin, and IgA in stools that were higher in cow’s milk and rice group compared to the placebo group. These findings underline the importance of fermented foods and their role in the modulation of innate and acquired immunity.
Another recently developed infant formula fermented with Bifidobacterium breve C50 and Streptococcus thermophilus 065 was assessed for its preventive effect against the incidence of acute diarrhea in healthy infants aged 4–6 months [79]. While consumption lasted for 5 months, the outcome showed that there was no difference on the incidence and duration of diarrhea episodes between the control (standard infant formula) and the fermented formula. However, the fermented formula was shown to alleviate the severity of diarrhea episodes.

2.5. Effects against Neonatal Necrotizing Enterocolitis

Prematurity is considered a predisposing factor for necrotizing enterocolitis (NEC), a disease with a 30% mortality rate in extremely and very low birth weight infants [113]. Although the pathogenesis of this disease is not fully understood, scientific evidence points out that dysbiosis is playing an important role [114]. Thus far, a limited amount of clinical trials using postbiotics have been conducted, but findings from in vitro or in vivo studies look promising. For instance, a group of scientists reported that DNA derived and released from inactivated Lactobacillus rhamnosus HN001 was sufficient in attenuating NEC severity in newborn mice and premature piglets [82]. Interestingly, it was shown that activation of TLR9 by L. rhamnosus HN001 DNA was an important mediator for the protection against NEC. Over the years, clinical studies have shown that the administration of bifidobacteria could prevent NEC development in premature infants [115]. It is also important to mention that various studies have linked clostridia with the development of NEC [116,117]. Therefore, postbiotics derived from whey cow’s milk retentate fermented by bifidobacteria might be effective in decreasing intestinal clostridia [94]. The difference in the gut microbiome of preterm infants compared to the one of term infants could be considered an important factor in NEC development [118]. Since concerns over the development of sepsis in infants after probiotic administration are raised, the use of postbiotics instead of probiotics in this vulnerable group might be a useful strategy in shaping their gut microbiome and thus preventing or treating NEC [119,120,121]. However, it is essential to add that specific probiotics with documented efficacy are recommended for preventing NEC by both the European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) and the American Gastroenterological Association (AGA) [122,123]. Over the years, a different array of postbiotic compounds, including reuterin; SCFAs; tryptophan; and various organic acids, such as taurine, histamine, and spermine, are found to either inhibit bacterial growth or promote activation of immune responses [124,125]. In contrast, another study focused on the use of postbiotics and their impact on the gut microbiota composition of preterm infants. According to Campeotto and coworkers, heat-treated fermented preterm infant formula by Bifidobacterium breve C50 and Streptococcus thermophilus 065 did not significantly modulate the bacterial colonization, but it was well tolerated, meaning that infants did not lose weight [126]. This is an important finding as reduction in weight gain can negatively affect the neurodevelopment of preterm infants [127]. Finally, in the same study, the fermented formula reduced the fecal calprotectin (increased levels of calprotectin are associated with inflammatory gastrointestinal diseases in children) and increased secretory IgA, suggesting immunomodulatory properties [126,128,129].

3. Discussion

Currently, the majority of the studies describing mechanisms of postbiotic action have been performed in vitro or in animal models by focusing only on mechanisms targeting either the pathogens or the host. Therefore, considerable research is required to establish the clinical relevance of postbiotics. In addition, clinical studies targeting the mechanism behind the postbiotic effect on host–pathogen interaction are required to understand the effects of postbiotics in humans.
Overall, the majority of clinical studies that tested postbiotic administration for the prevention of common infectious diseases in children did not find any significant differences in adverse effects between the intervention and placebo groups [17,78,79]. Additionally, when postbiotics were used for the treatment of mild RV diarrhea in children, no major side effects were noted [103]. However, there is a need for more randomized controlled trials that focus on a specific metabolite or component and establish its effectiveness and safety for preventing common pediatric infectious diseases measuring a pre-specified clinical outcome. Besides safety and effectiveness concerns, another challenge is the lack of a consensus regarding the definition of postbiotics that delays their commercialization and regulation [26]. Agreeing on a proper definition will also help increase their study and allow health benefits to be linked to different classes of postbiotics.
Besides pathogen infections, childhood obesity is another disease with increasing prevalence, especially in developed countries, but that is very challenging in its management. Promising preliminary studies show that SCFAs may play a role in energy metabolism and stimulate secretions of hormones that enhance food absorption [130]. However, the role of SCFAs in obesity still remains controversial due to mixed results from clinical studies [131,132]. On the other hand, in developing countries, malnutrition affects many children that become predisposed to infections [133]. A promising strategy targeting alterations in the functional activity of the immune system in malnourished children could be bioactive compounds that regulate host immune responses. Recently, researchers from Mexico showed that pre-treatment with heat-killed L. casei IMAU60214 can induce cytokine secretion on monocyte-derived macrophages isolated from malnourished children [134]. This is a positive first step on elucidating the role of postbiotics in malnutrition management, but more studies are needed. Due to their immunomodulatory properties, postbiotics could also be explored as a novel therapeutic approach for the prevention and treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A recent study proposed the role of the microbiome in the development of severe cases of coronavirus disease 2019 (COVID-19) [135], while others suggested that metabolites produced by the intestinal microbiome could be used for developing a drug for individuals who are susceptible to SARS-CoV-2 infection [136,137]. However, well controlled studies in humans are urgently needed to demonstrate whether postbiotics would have any effect against these diseases.

4. Conclusions

In summary, recently accumulated evidence from cell cultures and animal models indicates that postbiotics may be a promising strategy to prevent or protect against infectious diseases in children under the age of five. Postbiotics can exert immunomodulatory, antimicrobial, and antibiofilm effects similar to their parent probiotics; have a longer shelf life; and could be introduced in developing countries where certain infectious diseases are more prevalent. However, further studies are necessary to characterize different postbiotic compounds and to shed more light on their exact mechanisms of action and the way in which they protect the host from diseases. Finally, even if current findings from in vitro and in vivo experiments support the use of postbiotics, more clinical trials are required to validate their protective effects against infectious diseases.

Author Contributions

All authors contributed to the design of the study. A.M. and J.N.M.-R. reviewed the articles included. All authors contributed to the development of the manuscript and agreed upon the final draft. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. UN IGME Levels & Trends in Child Mortality: Report 2019, Estimates Developed by the UN Inter-Agency Group for Child Mortality Estimation; UN: New York, NY, USA, 2019.
  2. WHO Maternal and Child Epidemiology Estimation Group Child Causes of Death 2000–2017; WHO: Geneva, Switzerland, 2018.
  3. Troeger, C.; Blacker, B.F.; Khalil, I.A.; Rao, P.C.; Cao, S.; Zimsen, S.R.; Albertson, S.B.; Stanaway, J.D.; Deshpande, A.; Abebe, Z.; et al. Estimates of the global, regional, and national morbidity, mortality, and aetiologies of diarrhoea in 195 countries: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Infect. Dis. 2018, 18, 1211–1228. [Google Scholar] [CrossRef] [Green Version]
  4. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  5. Vuotto, C.; Longo, F.; Donelli, G. Probiotics to counteract biofilm-associated infections: Promising and conflicting data. Int. J. Oral Sci. 2014, 6, 189–194. [Google Scholar] [CrossRef] [Green Version]
  6. Do Carmo, F.L.R.; Rabah, H.; De Oliveira Carvalho, R.D.; Gaucher, F.; Cordeiro, B.F.; da Silva, S.H.; Le Loir, Y.; Azevedo, V.; Jan, G. Extractable Bacterial Surface Proteins in Probiotic–Host Interaction. Front. Microbiol. 2018, 9. [Google Scholar] [CrossRef]
  7. Kolaček, S.; Hojsak, I.; Berni Canani, R.; Guarino, A.; Indrio, F.; Orel, R.; Pot, B.; Shamir, R.; Szajewska, H.; Vandenplas, Y.; et al. Commercial Probiotic Products: A Call for Improved Quality Control. A Position Paper by the ESPGHAN Working Group for Probiotics and Prebiotics. J. Pediatr. Gastroenterol. Nutr. 2017, 65, 117–124. [Google Scholar] [CrossRef]
  8. Braegger, C.; Chmielewska, A.; Decsi, T.; Kolacek, S.; Mihatsch, W.; Moreno, L.; Pieścik, M.; Puntis, J.; Shamir, R.; Szajewska, H.; et al. Supplementation of infant formula with probiotics and/or prebiotics: A systematic review and comment by the ESPGHAN committee on nutrition. J. Pediatr. Gastroenterol. Nutr. 2011, 52, 238–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Skórka, A.; Pieścik-Lech, M.; Kołodziej, M.; Szajewska, H. To add or not to add probiotics to infant formulae? An updated systematic review. Benef. Microbes 2017, 8, 717–725. [Google Scholar] [CrossRef] [PubMed]
  10. Puccetti, M.; Xiroudaki, S.; Ricci, M.; Giovagnoli, S. Postbiotic-Enabled Targeting of the Host-Microbiota-Pathogen Interface: Hints of Antibiotic Decline? Pharmaceutics 2020, 12, 624. [Google Scholar] [CrossRef] [PubMed]
  11. Courvalin, P. Antibiotic resistance: The pros and cons of probiotics. Dig. Liver Dis. 2006, 38, S261–S265. [Google Scholar] [CrossRef]
  12. Zheng, M.; Zhang, R.; Tian, X.; Zhou, X.; Pan, X.; Wong, A. Assessing the Risk of Probiotic Dietary Supplements in the Context of Antibiotic Resistance. Front. Microbiol. 2017, 8, 908. [Google Scholar] [CrossRef] [PubMed]
  13. Embleton, N.D.; Zalewski, S.; Berrington, J.E. Probiotics for prevention of necrotizing enterocolitis and sepsis in preterm infants. Curr. Opin. Infect. Dis. 2016, 29, 256–261. [Google Scholar] [CrossRef] [PubMed]
  14. Dani, C.; Coviello, C.C.; Corsini, I.I.; Arena, F.; Antonelli, A.; Rossolini, G.M. Lactobacillus Sepsis and Probiotic Therapy in Newborns: Two New Cases and Literature Review. AJP Rep. 2016, 6, e25–e29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Lahtinen, S.J. Probiotic viability – does it matter? Microb. Ecol. Health Dis. 2012, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ouwehand, A.C.; Tölkkö, S.; Kulmala, J.; Salminen, S.; Salminen, E. Adhesion of inactivated probiotic strains to intestinal mucus. Lett. Appl. Microbiol. 2000, 31, 82–86. [Google Scholar] [CrossRef]
  17. Corsello, G.; Carta, M.; Marinello, R.; Picca, M.; De Marco, G.; Micillo, M.; Ferrara, D.; Vigneri, P.; Cecere, G.; Ferri, P.; et al. Preventive Effect of Cow’s Milk Fermented with Lactobacillus paracasei CBA L74 on Common Infectious Diseases in Children: A Multicenter Randomized Controlled Trial. Nutrients 2017, 9, 669. [Google Scholar] [CrossRef]
  18. Del Rigo-Adrover, M.M.; Knipping, K.; Garssen, J.; van Limpt, K.; Knol, J.; Franch, À.; Castell, M.; Rodríguez-lagunas, M.J.; Pérez-Cano, F.J. Prevention of Rotavirus Diarrhea in Suckling Rats by a Specific Fermented Milk Concentrate with Prebiotic Mixture. Nutrients 2019, 11, 189. [Google Scholar] [CrossRef] [Green Version]
  19. Rudkowski, Z.; Bromirska, J. [Reduction of the duration of salmonella excretion in infants with Hylak forte]. Padiatr. Padol. 1991, 26, 111–114. [Google Scholar]
  20. Nordeste, R.; Tessema, A.; Sharma, S.; Kovač, Z.; Wang, C.; Morales, R.; Griffiths, M.W. Molecules produced by probiotics prevent enteric colibacillosis in pigs. BMC Vet. Res. 2017, 13. [Google Scholar] [CrossRef] [Green Version]
  21. Aguilar-Toalá, J.E.; Garcia-Varela, R.; Garcia, H.S.; Mata-Haro, V.; González-Córdova, A.F.; Vallejo-Cordoba, B.; Hernández-Mendoza, A. Postbiotics: An evolving term within the functional foods field. Trends Food Sci. Technol. 2018, 75, 105–114. [Google Scholar] [CrossRef]
  22. Kanauchi, O.; Andoh, A.; AbuBakar, S.; Yamamoto, N. Probiotics and Paraprobiotics in Viral Infection: Clinical Application and Effects on the Innate and Acquired Immune Systems. Curr. Pharm. Des. 2018, 24, 710–717. [Google Scholar] [CrossRef]
  23. Shenderov, B.A. Metabiotics: Novel idea or natural development of probiotic conception. Microb. Ecol. Health Dis. 2013, 24. [Google Scholar] [CrossRef]
  24. Bajpai, V.K.; Chandra, V.; Kim, N.-H.; Rai, R.; Kumar, P.; Kim, K.; Aeron, A.; Kang, S.C.; Maheshwari, D.K.; Na, M.; et al. Ghost probiotics with a combined regimen: A novel therapeutic approach against the Zika virus, an emerging world threat. Crit. Rev. Biotechnol. 2018, 38, 438–454. [Google Scholar] [CrossRef] [PubMed]
  25. Lee, E.-S.; Song, E.-J.; Nam, Y.-D.; Lee, S.-Y. Probiotics in human health and disease: From nutribiotics to pharmabiotics. J. Microbiol. 2018, 56, 773–782. [Google Scholar] [CrossRef]
  26. Collado, M.C.; Vinderola, G.; Salminen, S. Postbiotics: Facts and open questions. A position paper on the need for a consensus definition. Benef. Microbes 2019, 10, 711–719. [Google Scholar] [CrossRef] [PubMed]
  27. Żółkiewicz, J.; Marzec, A.; Ruszczyński, M.; Feleszko, W. Postbiotics—A Step Beyond Pre- and Probiotics. Nutrients 2020, 12, 2189. [Google Scholar] [CrossRef]
  28. Taverniti, V.; Guglielmetti, S. The immunomodulatory properties of probiotic microorganisms beyond their viability (ghost probiotics: Proposal of paraprobiotic concept). Genes Nutr. 2011, 6, 261–274. [Google Scholar] [CrossRef] [Green Version]
  29. Malagón-Rojas, J.N.; Mantziari, A.; Salminen, S.; Szajewska, H. Postbiotics for Preventing and Treating Common Infectious Diseases in Children: A Systematic Review. Nutrients 2020, 12, 389. [Google Scholar] [CrossRef] [Green Version]
  30. Jayamani, E.; Mylonakis, E. Effector triggered manipulation of host immune response elicited by different pathotypes of Escherichia coli. Virulence 2014, 5, 733–739. [Google Scholar] [CrossRef] [Green Version]
  31. Kim, Y.K.; Shin, J.-S.; Nahm, M.H. NOD-Like Receptors in Infection, Immunity, and Diseases. Yonsei Med. J. 2016, 57, 5–14. [Google Scholar] [CrossRef] [Green Version]
  32. Fenini, G.; Contassot, E.; French, L.E. Potential of IL-1, IL-18 and Inflammasome Inhibition for the Treatment of Inflammatory Skin Diseases. Front. Pharmacol. 2017, 8. [Google Scholar] [CrossRef]
  33. Kummer, J.A.; Broekhuizen, R.; Everett, H.; Agostini, L.; Kuijk, L.; Martinon, F.; van Bruggen, R.; Tschopp, J. Inflammasome Components NALP 1 and 3 Show Distinct but Separate Expression Profiles in Human Tissues Suggesting a Site-specific Role in the Inflammatory Response. J. Histochem. Cytochem. 2007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Warshakoon, H.J.; Hood, J.D.; Kimbrell, M.R.; Malladi, S.; Wu, W.Y.; Shukla, N.M.; Agnihotri, G.; Sil, D.; David, S.A. Potential adjuvantic properties of innate immune stimuli. Hum. Vaccin. 2009, 5, 381–394. [Google Scholar] [CrossRef] [PubMed]
  35. Chandler, C.E.; Ernst, R.K. Bacterial lipids: Powerful modifiers of the innate immune response. F1000Research 2017, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Petes, C.; Odoardi, N.; Gee, K. The Toll for Trafficking: Toll-Like Receptor 7 Delivery to the Endosome. Front. Immunol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  37. Dalpke, A.; Frank, J.; Peter, M.; Heeg, K. Activation of Toll-Like Receptor 9 by DNA from Different Bacterial Species. Infect. Immun. 2006, 74, 940–946. [Google Scholar] [CrossRef] [Green Version]
  38. Yu, S.; Gao, N. Compartmentalizing Intestinal Epithelial Cell Toll-like Receptors for Immune Surveillance. Cell. Mol. Life Sci. CMLS 2015, 72, 3343–3353. [Google Scholar] [CrossRef] [Green Version]
  39. Wojdasiewicz, P.; Poniatowski, Ł.A.; Szukiewicz, D. The Role of Inflammatory and Anti-Inflammatory Cytokines in the Pathogenesis of Osteoarthritis. Mediators Inflamm. 2014, 2014. [Google Scholar] [CrossRef] [Green Version]
  40. Srinivasan, L.; Harris, M.C.; Kilpatrick, L.E. 128-Cytokines and Inflammatory Response in the Fetus and Neonate. In Fetal and Neonatal Physiology, Polin, R.A., Abman, S.H., Rowitch, D.H., Benitz, W.E., Fox, W.W., Eds.; 5th ed.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 1241–1254. ISBN 978-0-323-35214-7. [Google Scholar]
  41. Azad, M.A.K.; Sarker, M.; Wan, D. Immunomodulatory Effects of Probiotics on Cytokine Profiles. Available online: https://www.hindawi.com/journals/bmri/2018/8063647/ (accessed on 7 April 2019).
  42. Miyazawa, K.; He, F.; Kawase, M.; Kubota, A.; Yoda, K.; Hiramatsu, M. Enhancement of immunoregulatory effects of Lactobacillus gasseri TMC0356 by heat treatment and culture medium. Lett. Appl. Microbiol. 2011, 53, 210–216. [Google Scholar] [CrossRef]
  43. Zagato, E.; Mileti, E.; Massimiliano, L.; Fasano, F.; Budelli, A.; Penna, G.; Rescigno, M. Lactobacillus paracasei CBA L74 Metabolic Products and Fermented Milk for Infant Formula Have Anti-Inflammatory Activity on Dendritic Cells In Vitro and Protective Effects against Colitis and an Enteric Pathogen In Vivo. PLoS ONE 2014, 9. [Google Scholar] [CrossRef]
  44. Wang, H.; Yan, Y.; Wang, J.; Zhang, H.; Qi, W. Production and Characterization of Antifungal Compounds Produced by Lactobacillus plantarum IMAU10014. PLoS ONE 2012, 7, e29452. [Google Scholar] [CrossRef] [Green Version]
  45. Servin, A.L. Antagonistic activities of lactobacilli and bifidobacteria against microbial pathogens. FEMS Microbiol. Rev. 2004, 28, 405–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ricke, S.C. Perspectives on the use of organic acids and short chain fatty acids as antimicrobials. Poult. Sci. 2003, 82, 632–639. [Google Scholar] [CrossRef] [PubMed]
  47. Suiryanrayna, M.V.A.N.; Ramana, J.V. A review of the effects of dietary organic acids fed to swine. J. Anim. Sci. Biotechnol. 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Slonczewski, J.L.; Fujisawa, M.; Dopson, M.; Krulwich, T.A. Cytoplasmic pH Measurement and Homeostasis in Bacteria and Archaea. In Advances in Microbial Physiology; Poole, R.K., Ed.; Academic Press: Cambridge, MA, USA, 2009; Volume 55, pp. 1–317. [Google Scholar]
  49. Patel, R.M.; Denning, P.W. Therapeutic Use of Prebiotics, Probiotics, and Postbiotics to Prevent Necrotizing Enterocolitis: What is the Current Evidence? Clin. Perinatol. 2013, 40, 11–25. [Google Scholar] [CrossRef] [Green Version]
  50. Kareem, K.Y.; Hooi Ling, F.; Teck Chwen, L.; May Foong, O.; Anjas Asmara, S. Inhibitory activity of postbiotic produced by strains of Lactobacillus plantarum using reconstituted media supplemented with inulin. Gut Pathog. 2014, 6, 23. [Google Scholar] [CrossRef] [Green Version]
  51. Sharma, V.; Harjai, K.; Shukla, G. Effect of bacteriocin and exopolysaccharides isolated from probiotic on P. aeruginosa PAO1 biofilm. Folia Microbiol. (Praha) 2018, 63, 181–190. [Google Scholar] [CrossRef]
  52. Li, P.; Gu, Q. Antimicrobial Effects of Probiotics and Novel Probiotic-Based Approaches for Infectious Diseases. Probiotics Curr. Knowl. Future Prospects 2018. [Google Scholar] [CrossRef] [Green Version]
  53. Pridmore, R.D.; Pittet, A.-C.; Praplan, F.; Cavadini, C. Hydrogen peroxide production by Lactobacillus johnsonii NCC 533 and its role in anti-Salmonella activity. FEMS Microbiol. Lett. 2008, 283, 210–215. [Google Scholar] [CrossRef] [Green Version]
  54. Wang, Y.; Gänzle, M.G.; Schwab, C. Exopolysaccharide Synthesized by Lactobacillus reuteri Decreases the Ability of Enterotoxigenic Escherichia coli To Bind to Porcine Erythrocytes. Appl Environ. Microbiol. 2010, 76, 4863–4866. [Google Scholar] [CrossRef] [Green Version]
  55. Fanning, S.; Hall, L.J.; Cronin, M.; Zomer, A.; MacSharry, J.; Goulding, D.; O’Connell Motherway, M.; Shanahan, F.; Nally, K.; Dougan, G.; et al. Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection. Proc. Natl. Acad. Sci. USA 2012, 109, 2108–2113. [Google Scholar] [CrossRef] [Green Version]
  56. Abdelhamid, A.G.; Esaam, A.; Hazaa, M.M. Cell free preparations of probiotics exerted antibacterial and antibiofilm activities against multidrug resistant E. coli. Saudi Pharm. J. SPJ 2018, 26, 603–607. [Google Scholar] [CrossRef]
  57. Bondue, P.; Crèvecoeur, S.; Brose, F.; Daube, G.; Seghaye, M.-C.; Griffiths, M.W.; LaPointe, G.; Delcenserie, V. Cell-Free Spent Media Obtained from Bifidobacterium bifidum and Bifidobacterium crudilactis Grown in Media Supplemented with 3′-Sialyllactose Modulate Virulence Gene Expression in Escherichia coli O157:H7 and Salmonella Typhimurium. Front. Microbiol. 2016, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Bayoumi, M.A.; Griffiths, M.W. In vitro inhibition of expression of virulence genes responsible for colonization and systemic spread of enteric pathogens using Bifidobacterium bifidum secreted molecules. Int. J. Food Microbiol. 2012, 156, 255–263. [Google Scholar] [CrossRef] [PubMed]
  59. Chauvière, G.; Coconnier, M.H.; Kerneis, S.; Darfeuille-Michaud, A.; Joly, B.; Servin, A.L. Competitive exclusion of diarrheagenic Escherichia coli (ETEC) from human enterocyte-like Caco-2 cells by heat-killed Lactobacillus. FEMS Microbiol. Lett. 1992, 70, 213–217. [Google Scholar] [CrossRef]
  60. Coconnier, M.H.; Bernet, M.F.; Chauvière, G.; Servin, A.L. Adhering heat-killed human Lactobacillus acidophilus, strain LB, inhibits the process of pathogenicity of diarrhoeagenic bacteria in cultured human intestinal cells. J. Diarrhoeal Dis. Res. 1993, 11, 235–242. [Google Scholar] [PubMed]
  61. Muñoz-Quezada, S.; Bermudez-Brito, M.; Chenoll, E.; Genovés, S.; Gomez-Llorente, C.; Plaza-Diaz, J.; Matencio, E.; Bernal, M.J.; Romero, F.; Ramón, D.; et al. Competitive inhibition of three novel bacteria isolated from faeces of breast milk-fed infants against selected enteropathogens. Br. J. Nutr. 2013, 109, S63–S69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Tsai, C.-C.; Lin, P.-P.; Hsieh, Y.-M. Three Lactobacillus strains from healthy infant stool inhibit enterotoxigenic Escherichia coli grown in vitro. Anaerobe 2008, 14, 61–67. [Google Scholar] [CrossRef]
  63. He, X.; Zeng, Q.; Puthiyakunnon, S.; Zeng, Z.; Yang, W.; Qiu, J.; Du, L.; Boddu, S.; Wu, T.; Cai, D.; et al. Lactobacillus rhamnosus GG supernatant enhance neonatal resistance to systemic Escherichia coli K1 infection by accelerating development of intestinal defense. Sci. Rep. 2017, 7, 43305. [Google Scholar] [CrossRef]
  64. Gao, J.; Li, Y.; Wan, Y.; Hu, T.; Liu, L.; Yang, S.; Gong, Z.; Zeng, Q.; Wei, Y.; Yang, W.; et al. A Novel Postbiotic From Lactobacillus rhamnosus GG With a Beneficial Effect on Intestinal Barrier Function. Front. Microbiol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
  65. Charchoghlyan, H.; Kwon, H.; Hwang, D.-J.; Lee, J.S.; Lee, J.; Kim, M. Inhibition of Cronobacter sakazakii by Lactobacillus acidophilus n.v. Er2 317/402. Korean J. Food Sci. Anim. Resour. 2016, 36, 635–640. [Google Scholar] [CrossRef] [Green Version]
  66. Jamwal, A.; Sharma, K.; Chauhan, R.; Bansal, S.; Goel, G. Evaluation of commercial probiotic lactic cultures against biofilm formation by Cronobacter sakazakii. Intest. Res. 2018. [Google Scholar] [CrossRef] [PubMed]
  67. Dowdell, P.; Chankhamhaengdecha, S.; Panbangred, W.; Janvilisri, T.; Aroonnual, A. Probiotic Activity of Enterococcus faecium and Lactococcus lactis Isolated from Thai Fermented Sausages and Their Protective Effect Against Clostridium difficile. Probiotics Antimicrob. Proteins 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Ephraim, E.; Schultz, R.D.; Safdar, N. Lactobacillus rhamnosus GG Protects Cells from Clostridium difficile Toxins. Br. Microbiol. Res. J. 2013, 3, 165–175. [Google Scholar] [CrossRef]
  69. Tsilingiri, K.; Barbosa, T.; Penna, G.; Caprioli, F.; Sonzogni, A.; Viale, G.; Rescigno, M. Probiotic and postbiotic activity in health and disease: Comparison on a novel polarised ex-vivo organ culture model. Gut 2012, 61, 1007–1015. [Google Scholar] [CrossRef] [Green Version]
  70. Bermudez-Brito, M.; Munoz-Quezada, S.; Gomez-Llorente, C.; Matencio, E.; Romero, F.; Gil, A. Lactobacillus paracasei CNCM I-4034 and its culture supernatant modulate Salmonella-induced inflammation in a novel transwell co-culture of human intestinal-like dendritic and Caco-2 cells. Bmc Microbiol. 2015, 15, 79. [Google Scholar] [CrossRef] [Green Version]
  71. Rather, I.A.; Choi, K.-H.; Bajpai, V.K.; Park, Y.-H. Antiviral mode of action of Lactobacillus plantarum YML009 on Influenza virus H1N1. Bangladesh J. Pharmacol. 2015, 10, 475. [Google Scholar] [CrossRef] [Green Version]
  72. Seo, B.J.; Rather, I.A.; Kumar, V.J.R.; Choi, U.H.; Moon, M.R.; Lim, J.H.; Park, Y.H. Evaluation of Leuconostoc mesenteroides YML003 as a probiotic against low-pathogenic avian influenza (H9N2) virus in chickens. J. Appl. Microbiol. 2012, 113, 163–171. [Google Scholar] [CrossRef]
  73. Kaila, M.; Isolauri, E.; Saxelin, M.; Arvilommi, H.; Vesikari, T. Viable versus inactivated lactobacillus strain GG in acute rotavirus diarrhoea. Arch. Dis. Child. 1995, 72, 51–53. [Google Scholar] [CrossRef] [Green Version]
  74. Martín, V.; Maldonado, A.; Fernández, L.; Rodríguez, J.M.; Connor, R.I. Inhibition of Human Immunodeficiency Virus Type 1 by Lactic Acid Bacteria from Human Breastmilk. Breastfeed. Med. 2010, 5, 153–158. [Google Scholar] [CrossRef]
  75. Hassan, Z. Anti-Adhesion Activity of Lactic Acid Bacteria Supernatant against Human Pathogenic Candida Species Biofilm. Health Sci. J. 2015, 9. [Google Scholar]
  76. Parolin, C.; Marangoni, A.; Laghi, L.; Foschi, C.; Palomino, R.A.Ñ.; Calonghi, N.; Cevenini, R.; Vitali, B. Isolation of Vaginal Lactobacilli and Characterization of Anti-Candida Activity. PLoS ONE 2015, 10, e0131220. [Google Scholar] [CrossRef] [PubMed]
  77. Song, Y.-G.; Lee, S.-H. Inhibitory effects of Lactobacillus rhamnosus and Lactobacillus casei on Candida biofilm of denture surface. Arch. Oral Biol. 2017, 76, 1–6. [Google Scholar] [CrossRef] [PubMed]
  78. Nocerino, R.; Paparo, L.; Terrin, G.; Pezzella, V.; Amoroso, A.; Cosenza, L.; Cecere, G.; Marco, G.D.; Micillo, M.; Albano, F.; et al. Cow’s milk and rice fermented with Lactobacillus paracasei CBA L74 prevent infectious diseases in children: A randomized controlled trial. Clin. Nutr. 2017, 36, 118–125. [Google Scholar] [CrossRef] [PubMed]
  79. Thibault, H.; Aubert-Jacquin, C.; Goulet, O. Effects of Long-term Consumption of a Fermented Infant Formula (with Bifidobacterium breve c50 and Streptococcus thermophilus 065) on Acute Diarrhea in Healthy Infants. J. Pediatr. Gastroenterol. Nutr. 2004, 39, 147. [Google Scholar] [CrossRef] [PubMed]
  80. Romond, M.B.; Ais, A.; Yazourh, A.; Romond, C. Cell-free Wheys From Bifidobacteria Fermented Milks Exert a Regulatory Effect on the Intestinal Microflora of Mice and Humans. Anaerobe 1997, 3, 137–143. [Google Scholar] [CrossRef]
  81. Arai, S.; Iwabuchi, N.; Takahashi, S.; Xiao, J.; Abe, F.; Hachimura, S. Orally administered heat-killed Lactobacillus paracasei MCC1849 enhances antigen-specific IgA secretion and induces follicular helper T cells in mice. PLoS ONE 2018, 13, e0199018. [Google Scholar] [CrossRef]
  82. Good, M.; Sodhi, C.P.; Ozolek, J.A.; Buck, R.H.; Goehring, K.C.; Thomas, D.L.; Vikram, A.; Bibby, K.; Morowitz, M.J.; Firek, B.; et al. Lactobacillus rhamnosus HN001 decreases the severity of necrotizing enterocolitis in neonatal mice and preterm piglets: Evidence in mice for a role of TLR9. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 306, G1021–G1032. [Google Scholar] [CrossRef] [Green Version]
  83. Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J.; et al. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef]
  84. Kotloff, K.L.; Nataro, J.P.; Blackwelder, W.C.; Nasrin, D.; Farag, T.H.; Panchalingam, S.; Wu, Y.; Sow, S.O.; Sur, D.; Breiman, R.F.; et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): A prospective, case-control study. Lancet 2013, 382, 209–222. [Google Scholar] [CrossRef]
  85. Kotloff, K.L.; Nasrin, D.; Blackwelder, W.C.; Wu, Y.; Farag, T.; Panchalingham, S.; Sow, S.O.; Sur, D.; Zaidi, A.K.M.; Faruque, A.S.G.; et al. The incidence, aetiology, and adverse clinical consequences of less severe diarrhoeal episodes among infants and children residing in low-income and middle-income countries: A 12-month case-control study as a follow-on to the Global Enteric Multicenter Study (GEMS). Lancet Glob. Health 2019, 7, e568–e584. [Google Scholar] [CrossRef] [Green Version]
  86. Liévin-Le Moal, V. A gastrointestinal anti-infectious biotherapeutic agent: The heat-treated Lactobacillus LB. Ther. Adv. Gastroenterol. 2016, 9, 57–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Chauvière, G.; Coconnier, M.H.; Kernéis, S.; Fourniat, J.; Servin, A.L. Adhesion of human Lactobacillus acidophilus strain LB to human enterocyte-like Caco-2 cells. J. Gen. Microbiol. 1992, 138, 1689–1696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Muñoz-Quezada, S.; Chenoll, E.; Vieites, J.M.; Genovés, S.; Maldonado, J.; Bermúdez-Brito, M.; Gomez-Llorente, C.; Matencio, E.; Bernal, M.J.; Romero, F.; et al. Isolation, identification and characterisation of three novel probiotic strains (Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035 and Lactobacillus rhamnosus CNCM I-4036) from the faeces of exclusively breast-fed infants. Br. J. Nutr. 2013, 109 (Suppl. 2), S51–S62. [Google Scholar] [CrossRef] [Green Version]
  89. Zhang, H.; Gao, J.; He, X.; Gong, Z.; Wan, Y.; Hu, T.; Li, Y.; Cao, H. The postbiotic HM0539 from Lactobacillus rhamnosus GG prevents intestinal infection by enterohemorrhagic E. coli O157: H7 in mice. J. South. Med. Univ. 2020, 40, 211–218. [Google Scholar] [CrossRef]
  90. Agostoni, C.; Axelsson, I.; Goulet, O.; Koletzko, B.; Michaelsen, K.F.; Puntis, J.W.L.; Rigo, J.; Shamir, R.; Szajewska, H.; Turck, D.; et al. Preparation and Handling of Powdered Infant Formula: A Commentary by the ESPGHAN Committee on Nutrition. J. Pediatr. Gastroenterol. Nutr. 2004, 39, 320–322. [Google Scholar] [CrossRef]
  91. Borali, E.; De Giacomo, C. Clostridium Difficile Infection in Children: A Review. J. Pediatr. Gastroenterol. Nutr. 2016, 63, e130–e140. [Google Scholar] [CrossRef]
  92. Plants-Paris, K.; Bishoff, D.; Oyaro, M.O.; Mwinyi, B.; Chappell, C.; Kituyi, A.; Nyangao, J.; Mbatha, D.; Darkoh, C. Prevalence of Clostridium difficile infections among Kenyan children with diarrhea. Int. J. Infect. Dis. 2019, 81, 66–72. [Google Scholar] [CrossRef] [Green Version]
  93. Jangi, S.; Lamont, J. Asymptomatic Colonization by Clostridium difficile in Infants: Implications for Disease in Later Life. J. Pediatr. Gastroenterol. Nutr. 2010, 51, 2–7. [Google Scholar] [CrossRef]
  94. Schäffler, H.; Breitrück, A. Clostridium difficile–From Colonization to Infection. Front. Microbiol. 2018, 9, 646. [Google Scholar] [CrossRef] [Green Version]
  95. Fiedoruk, K.; Daniluk, T.; Rozkiewicz, D.; Zaremba, M.L.; Oldak, E.; Sciepuk, M.; Leszczynska, K. Conventional and molecular methods in the diagnosis of community-acquired diarrhoea in children under 5 years of age from the north-eastern region of Poland. Int. J. Infect. Dis. 2015, 37, 145–151. [Google Scholar] [CrossRef] [Green Version]
  96. Dunand, E.; Burns, P.; Binetti, A.; Bergamini, C.; Peralta, G.H.; Forzani, L.; Reinheimer, J.; Vinderola, G. Postbiotics produced at laboratory and industrial level as potential functional food ingredients with the capacity to protect mice against Salmonella infection. J. Appl. Microbiol. 2019, 127, 219–229. [Google Scholar] [CrossRef] [PubMed]
  97. Lafond, K.E.; Nair, H.; Rasooly, M.H.; Valente, F.; Booy, R.; Rahman, M.; Kitsutani, P.; Yu, H.; Guzman, G.; Coulibaly, D.; et al. Global Role and Burden of Influenza in Pediatric Respiratory Hospitalizations, 1982–2012: A Systematic Analysis. PLoS Med. 2016, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. El Guerche-Séblain, C.; Moureau, A.; Schiffler, C.; Dupuy, M.; Pepin, S.; Samson, S.I.; Vanhems, P.; Schellevis, F. Epidemiology and burden of influenza in healthy children aged 6 to 35 months: Analysis of data from the placebo arm of a phase III efficacy trial. BMC Infect. Dis. 2019, 19, 308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. de Francisco (Shapovalova), N.; Donadel, M.; Jit, M.; Hutubessy, R. A systematic review of the social and economic burden of influenza in low- and middle-income countries. Vaccine 2015, 33, 6537–6544. [Google Scholar] [CrossRef]
  100. Thorrington, D.; Balasegaram, S.; Cleary, P.; Hay, C.; Eames, K. Social and Economic Impacts of School Influenza Outbreaks in England: Survey of Caregivers. J. Sch. Health 2017, 87, 209–216. [Google Scholar] [CrossRef]
  101. Hagbom, M.; Sharma, S.; Lundgren, O.; Svensson, L. Towards a human rotavirus disease model. Curr. Opin. Virol. 2012, 2, 408–418. [Google Scholar] [CrossRef]
  102. Simakachorn, N.; Pichaipat, V.; Rithipornpaisarn, P.; Kongkaew, C.; Tongpradit, P.; Varavithya, W. Clinical evaluation of the addition of lyophilized, heat-killed Lactobacillus acidophilus LB to oral rehydration therapy in the treatment of acute diarrhea in children. J. Pediatr. Gastroenterol. Nutr. 2000, 30, 68–72. [Google Scholar] [CrossRef]
  103. Salazar-Lindo, E.; Figueroa-Quintanilla, D.; Caciano, M.; Reto-Valiente, V.; Chauviere, G.; Colin, P. Effectiveness and Safety of Lactobacillus LB in the Treatment of Mild Acute Diarrhea in Children. J. Pediatr. Gastroenterol. Nutr. 2007, 44, 571–576. [Google Scholar] [CrossRef]
  104. Osano, B.O.; Were, F.; Mathews, S. Mortality among 5-17 year old children in Kenya. Pan Afr. Med. J. 2017, 27. [Google Scholar] [CrossRef]
  105. Buvé, A.; Bishikwabo-Nsarhaza, K.; Mutangadura, G. The spread and effect of HIV-1 infection in sub-Saharan Africa. Lancet 2002, 359, 2011–2017. [Google Scholar] [CrossRef]
  106. WHO Antiretroviral Drugs for Treating Pregnant Women and Preventing HIV Infection in Infants. Available online: https://apps.who.int/iris/bitstream/handle/10665/75236/9789241599818_eng.pdf?sequence=1 (accessed on 17 August 2020).
  107. WHO Guidelines on HIV and Infant Feeding. 2010. Available online: http://www.who.int/maternal_child_adolescent/documents/9789241599535/en/ (accessed on 17 August 2020).
  108. WHO HIV and Infant Feeding: Update. Available online: https://apps.who.int/iris/bitstream/handle/10665/43747/9789241595964_eng.pdf;jsessionid=3D425244271D458E9F10E85E311DFBCD?sequence=1 (accessed on 17 August 2020).
  109. White, A.B.; Mirjahangir, J.F.; Horvath, H.; Anglemyer, A.; Read, J.S. Antiretroviral interventions for preventing breast milk transmission of HIV. Cochrane Database Syst. Rev. 2014. [Google Scholar] [CrossRef] [PubMed]
  110. Toda, M. Population-Based Active Surveillance for Culture-Confirmed Candidemia — Four Sites, United States, 2012–2016. MMWR Surveill. Summ. 2019, 68. [Google Scholar] [CrossRef] [PubMed]
  111. Noni, M.; Stathi, A.; Vaki, I.; Velegraki, A.; Zachariadou, L.; Michos, A. Changing Epidemiology of Invasive Candidiasis in Children during a 10-Year Period. J. Fungi 2019, 5, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kaufman, D.; Boyle, R.; Hazen, K.C.; Patrie, J.T.; Robinson, M.; Donowitz, L.G. Fluconazole prophylaxis against fungal colonization and infection in preterm infants. N. Engl. J. Med. 2001, 345, 1660–1666. [Google Scholar] [CrossRef]
  113. Gephart, S.M.; McGrath, J.M.; Effken, J.A.; Halpern, M.D. Necrotizing Enterocolitis Risk: State of the Science. Adv. Neonatal Care 2012, 12, 77–87. [Google Scholar] [CrossRef] [Green Version]
  114. Coggins, S.A.; Wynn, J.L.; Weitkamp, J.-H. Infectious causes of necrotizing enterocolitis. Clin. Perinatol. 2015, 42, 133. [Google Scholar] [CrossRef]
  115. Wu, W.; Wang, Y.; Zou, J.; Long, F.; Yan, H.; Zeng, L.; Chen, Y. Bifidobacterium adolescentis protects against necrotizing enterocolitis and upregulates TOLLIP and SIGIRR in premature neonatal rats. BMC Pediatr. 2017, 17, 1. [Google Scholar] [CrossRef] [Green Version]
  116. Schönherr-Hellec, S.; Klein, G.L.; Delannoy, J.; Ferraris, L.; Rozé, J.C.; Butel, M.J.; Aires, J. Clostridial Strain-Specific Characteristics Associated with Necrotizing Enterocolitis. Appl Environ. Microbiol 2018, 84, e02428-17. [Google Scholar] [CrossRef] [Green Version]
  117. Alfa, M.J.; Robson, D.; Davi, M.; Bernard, K.; Caeseele, P.V.; Harding, G.K.M. An Outbreak of Necrotizing Enterocolitis Associated with a Novel Clostridium Species in a Neonatal Intensive Care Unit. Clin. Infect. Dis. 2002, 35, S101–S105. [Google Scholar] [CrossRef] [Green Version]
  118. Denning, N.-L.; Prince, J.M. Neonatal intestinal dysbiosis in necrotizing enterocolitis. Mol. Med. 2018, 24, 4. [Google Scholar] [CrossRef]
  119. Land, M.H.; Rouster-Stevens, K.; Woods, C.R.; Cannon, M.L.; Cnota, J.; Shetty, A.K. Lactobacillus Sepsis Associated With Probiotic Therapy. Pediatrics 2005, 115, 178–181. [Google Scholar] [CrossRef] [PubMed]
  120. Patel, R.M.; Underwood, M.A. Probiotics and Necrotizing Enterocolitis. Semin. Pediatr. Surg. 2018, 27, 39–46. [Google Scholar] [CrossRef] [PubMed]
  121. Chiang, M.-C.; Chen, C.-L.; Feng, Y.; Chen, C.-C.; Lien, R.; Chiu, C.-H. Lactobacillus rhamnosus sepsis associated with probiotic therapy in an extremely preterm infant: Pathogenesis and a review for clinicians. J. Microbiol. Immunol. Infect. 2020. [Google Scholar] [CrossRef] [PubMed]
  122. Su, G.L.; Ko, C.W.; Bercik, P.; Falck-Ytter, Y.; Sultan, S.; Weizman, A.V.; Morgan, R.L. AGA Clinical Practice Guidelines on the Role of Probiotics in the Management of Gastrointestinal Disorders. Gastroenterology 2020. [Google Scholar] [CrossRef]
  123. van den Akker, C.H.P.; van Goudoever, J.B.; Shamir, R.; Domellöf, M.; Embleton, N.D.; Hojsak, I.; Lapillonne, A.; Mihatsch, W.A.; Berni Canani, R.; Bronsky, J.; et al. Probiotics and Preterm Infants: A Position Paper by the European Society for Paediatric Gastroenterology Hepatology and Nutrition Committee on Nutrition and the European Society for Paediatric Gastroenterology Hepatology and Nutrition Working Group for Probiotics and Prebiotics. J. Pediatr. Gastroenterol. Nutr. 2020, 70, 664–680. [Google Scholar] [CrossRef] [PubMed]
  124. Vieira, A.T.; Fukumori, C.; Ferreira, C.M. New insights into therapeutic strategies for gut microbiota modulation in inflammatory diseases. Clin. Transl. Immunol. 2016, 5, e87. [Google Scholar] [CrossRef] [PubMed]
  125. Mosca, F.; Gianni, M.L.; Rescigno, M. Can Postbiotics Represent a New Strategy for NEC? Adv. Exp. Med. Biol. 2019. [Google Scholar] [CrossRef]
  126. Campeotto, F.; Suau, A.; Kapel, N.; Magne, F.; Viallon, V.; Ferraris, L.; Waligora-Dupriet, A.-J.; Soulaines, P.; Leroux, B.; Kalach, N.; et al. A fermented formula in pre-term infants: Clinical tolerance, gut microbiota, down-regulation of faecal calprotectin and up-regulation of faecal secretory IgA. Br. J. Nutr. 2011, 105, 1843–1851. [Google Scholar] [CrossRef]
  127. Ehrenkranz, R.A.; Dusick, A.M.; Vohr, B.R.; Wright, L.L.; Wrage, L.A.; Poole, W.K. Growth in the Neonatal Intensive Care Unit Influences Neurodevelopmental and Growth Outcomes of Extremely Low Birth Weight Infants. Pediatrics 2006, 117, 1253–1261. [Google Scholar] [CrossRef] [Green Version]
  128. Jeong, S.J. The role of fecal calprotectin in pediatric disease. Korean J. Pediatr. 2019, 62, 287–291. [Google Scholar] [CrossRef]
  129. Mullié, C.; Yazourh, A.; Thibault, H.; Odou, M.-F.; Singer, E.; Kalach, N.; Kremp, O.; Romond, M.-B. Increased Poliovirus-Specific Intestinal Antibody Response Coincides with Promotion of Bifidobacterium longum-infantis and Bifidobacterium breve in Infants: A Randomized, Double-Blind, Placebo-Controlled Trial. Pediatr. Res. 2004, 56, 791–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Barczyńska, R.; Litwin, M.; Sliżewska, K.; Szalecki, M.; Berdowska, A.; Bandurska, K.; Libudzisz, Z.; Kapuśniak, J. Bacterial Microbiota and Fatty Acids in the Faeces of Overweight and Obese Children. Pol. J. Microbiol. 2018, 67, 339–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Riva, A.; Borgo, F.; Lassandro, C.; Verduci, E.; Morace, G.; Borghi, E.; Berry, D. Pediatric obesity is associated with an altered gut microbiota and discordant shifts in F irmicutes populations. Environ. Microbiol. 2017, 19, 95–105. [Google Scholar] [CrossRef]
  133. Ibrahim, M.K.; Zambruni, M.; Melby, C.L.; Melby, P.C. Impact of Childhood Malnutrition on Host Defense and Infection. Clin. Microbiol. Rev. 2017, 30, 919–971. [Google Scholar] [CrossRef] [Green Version]
  134. Rocha-Ramírez, L.M.; Hernández-Ochoa, B.; Gómez-Manzo, S.; Marcial-Quino, J.; Cárdenas-Rodríguez, N.; Centeno-Leija, S.; García-Garibay, M. Impact of Heat-Killed Lactobacillus casei Strain IMAU60214 on the Immune Function of Macrophages in Malnourished Children. Nutrients 2020, 12, 2303. [Google Scholar] [CrossRef]
  135. van der Lelie, D.; Taghavi, S. COVID-19 and the Gut Microbiome: More than a Gut Feeling. mSystems 2020, 5, e00453-20. [Google Scholar] [CrossRef]
  136. Gou, W.; Fu, Y.; Yue, L.; Chen, G.; Cai, X.; Shuai, M.; Xu, F.; Yi, X.; Chen, H.; Zhu, Y.J.; et al. Gut microbiota may underlie the predisposition of healthy individuals to COVID-19. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  137. Kalantar-Zadeh, K.; Ward, S.A.; Kalantar-Zadeh, K.; El-Omar, E.M. Considering the Effects of Microbiome and Diet on SARS-CoV-2 Infection: Nanotechnology Roles. ACS Nano 2020, 14, 5179–5182. [Google Scholar] [CrossRef]
Figure 1. Potential mechanisms of action of postbiotics. (a) Modulation of the host’s immune response. (b) Acting on the pathogen. MDP: muramyl dipeptide; NLRP1: NACHT [NAIP (neuronal apoptosis inhibitory protein), CIITA (MHC class II transcription activator), HET-E (incompatibility locus protein from Podospora anserina) and TP1 (telomerase-associated protein)] domain-, leucine-rich repeat-, and PYRIN containing protein 1; IL-1β and IL-18: interleukin 1 beta and 18, respectively; LPS: lipopolysaccharide; TLR 4, TLR 2, and TLR 9: toll-like receptor 4, 2, and 9, respectively; NF-κB: nuclear factor kappa B; SCFAs: short-chain fatty acids.
Figure 1. Potential mechanisms of action of postbiotics. (a) Modulation of the host’s immune response. (b) Acting on the pathogen. MDP: muramyl dipeptide; NLRP1: NACHT [NAIP (neuronal apoptosis inhibitory protein), CIITA (MHC class II transcription activator), HET-E (incompatibility locus protein from Podospora anserina) and TP1 (telomerase-associated protein)] domain-, leucine-rich repeat-, and PYRIN containing protein 1; IL-1β and IL-18: interleukin 1 beta and 18, respectively; LPS: lipopolysaccharide; TLR 4, TLR 2, and TLR 9: toll-like receptor 4, 2, and 9, respectively; NF-κB: nuclear factor kappa B; SCFAs: short-chain fatty acids.
Microorganisms 08 01510 g001
Table 1. Potential infectious disease targets for postbiotics based on current evidence.
Table 1. Potential infectious disease targets for postbiotics based on current evidence.
Targeted PathogenProbiotic BacteriaPreparationType of StudyBioactivity or EffectReference
Escherichia coliLactobacillus acidophilus EMCC 1324 (La) *, Lactobacillus helveticus EMCC 1654 (Lh), Lactobacillus plantarum ss. plantarum EMCC 1027 (Lp), Lactobacillus rhamnosus EMCC 1105 (Lr), Bifidobacterium longum EMCC 1547 (BL), Bifidobacterium bifidum EMCC 1334 (Bb)Cell-free supernatant (CFS; filtered) of MRS or milk fermented by probioticsIn vitroCFS of MRS fermented by all probiotics resulted in inhibition of two multiresistant E. coli (WW1 and IC2) biofilm formation; CFSM of milk fermented by B. longum and L. helveticus reduced E. coli WW1 biofilm[56]
Bifidobacterium bifidum BBA1 and Bifidobacterium crudilactis FR/62/B/3Cell-free supernatant (CFS; filtered) from probiotics grown in media supplemented with 3′-sialyllactose (major bovine milk oligosaccharide)In vitroBoth CFSs induced a significant decrease in virulence gene expression of E. coli O157:H7[57]
Bifidobacterium bifidum ATCC 29521Cell-free supernatant (CFS; filtered) of modified MRS fermented by probioticHeLa and macrophage cell linesDownregulated virulence genes in E. coli O157:H7; inhibited its adhesion to HeLa cell line[58]
Lactobacillus acidophilus LBHeat-inactivated probiotic with its spent culture supernatant (SCS)Caco-2 cell cultureInhibited E. coli ETEC adhesion[59]
Lactobacillus acidophilus LBHeat-inactivated probiotic with its spent culture supernatant (SCS)Caco-2 cell cultureInhibited cell association and cell invasion of E. coli ETEC, DAEC, and EPEC[60]
Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035, Lactobacillus rhamnosus CNCM I-4036)Cell-free supernatant (CFS; filtered) of MRS fermented by probioticsMonitoring bacterial growthInhibited the growth of E. coli ETEC and EPEC[61]
Lactobacillus acidophilus RY2, Lactobacillus salivarius MM1, Lactobacillus paracasei En4Spent culture supernatant (SCS) from bacteria grown in MRS + cysteinColony count assayInhibited growth of E. coli ETEC[62]
Lactobacillus rhamnosus GGCell-free supernatant (CFS; filtered) of MRS fermented by probioticCaco-2 cell cultureBlocked adhesion, invasion, and translocation of E. coli K1[63]
Lactobacillus rhamnosus GGNovel secreted protein (HM0539) present in the CFSInfected Sprague-Dawley neonatal ratsPromoted the development of neonatal intestinal defense and prevented against E. coli K1 pathogenesis[64]
Cronobacter sakazakiiLactobacillus acidophilus INMIA 9602 Er 317/402 strain NarineHeat-inactivated probioticAgar well diffusion methodInhibited the growth of C. sakazakii in contaminated reconstructed powdered infant formula[65]
Lactobacillus casei strain Shirota (Yakult); Lactobacillus sporongenes, Streptococcus faecalis, Clostridium butyricum, Bacillus mesentericus (Bifilac; cells); Streptococcus faecalis, Lactobacillus sporongenes, Clostridium butyricum, Bacillus mesentericus (Vibact; spores); Lactobacillus sporongenes (Caplac; spores)Cell-free supernatant (CFS; filtered) from isolated probioticsIn vitroFour probiotic-derived CFS (filtered and filtered + heat inactivated) possessed antimicrobial activity against C. sakazakii; a higher biofilm inhibitory activity (> 80%) was observed at initial stages of biofilm formation[66]
Clostridioides difficileEnterococcus faecium, Lactococcus lactisCell-free supernatant (CFS; filtered) of MRS fermented by lactic acid bacteriaCaco-2 cell cultureDiminished the expression level of proinflammatory cytokines induced by the cell-free supernatant of C. difficile[67]
Lactobacillus rhamnosus GGCell-free supernatant (CFS; filtered) and cell lysate (sonicated and filtered) of co-cultured toxigenic C. difficile with LGGVero cell cultureDecreased the cytotoxic effect of both extracellular and intracellular clostridial toxins resulting in up to 30% increase in cell viability[68]
Salmonella spp.Bifidobacterium bifidum ATCC 29521Cell-free supernatant (CFS; filtered) of modified MRS fermented by probioticHeLa and macrophage cell linesDownregulated virulence genes in S. Typhimurium; inhibited its adhesion to HeLa cell line; diminished the ability of Salmonella to survive and multiply within macrophages[58]
Lactobacillus acidophilus LBHeat-inactivated probiotic with its spent culture supernatant (SCS)Caco-2 cell cultureInhibited cell association and cell invasion of S. typhimurium[60]
Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035, Lactobacillus rhamnosus CNCM I-4036)Cell-free supernatant (CFS; filtered) of MRS fermented by probioticsMonitoring bacterial growthInhibited the growth of Salmonella typhimurium and/or Salmonella typhi[61]
Lactobacillus paracasei B21060Supernatant of lactic acid bacteria grown in MRSEx vivo organ culture modelInhibited the inflammatory potential of Salmonella and conditioned the epithelium against Salmonella invasion[69]
Lactobacillus paracasei CNCM I-4034Cell-free supernatant (CFS; filtered) of MRS fermented by probioticsCoculture of dendritic and Caco-2 cellsIncreased the production of pro-inflammatory cytokines in the presence of Salmonella typhi[70]
InfluenzaLactobacillus plantarum YML009Cell-free supernatant (CFS; filtered) of MRS fermented by lactic acid bacteriaMDCK cells and hemagglutination assayDisplayed a significant antiviral activity against influenza virus H1N1 and was more effective than Tamiflu[71]
Leuconostoc mesenteroides YML003Cell-free supernatant (CFS; filtered) of MRS fermented by lactic acid bacteriaMDCK cells and hemagglutination assayDemonstrated antiviral activity against low-pathogenic avian influenza (H9N2)[72]
RotavirusBifidobacterium breve C50 and Streptococcus thermophilus 065Heat-treated fermented milk containing the prebiotic mixture scGOS/lcFOSRotavirus-infected Lewis ratsReduced the incidence and severity of the rotaviral diarrhea; enhanced the host’s immune system[18]
Lactobacilus rhamnosus GGOrally administered heat-inactivated probioticChildren under the age of 4 with rotavirus diarrheaRotavirus diarrhea recovery was equal for both viable and heat-inactivated probiotic-receiving groups[73]
Human immunodeficiency virusLactic acid bacteria isolated from human milkHeat-inactivated lactic acid bacteriaTZM-bl cellsSignificantly inhibited R5-tropic HIV-1 infection[74]
Candida spp.Four LAB isolated from honey (Lactobacillus plantarum HS, Pediococcus acidilactici HC, Lactobacillus curvatus HH, and Pediococcus pentosaceus HM)Cell-free supernatant (CFS; filtered) of MRS fermented by probioticsMicrotiter plate methodDecreased the biofilm formation by Candida spp.[75]
Lactobacillus crispatus B1-BC8, Lactobacillus gasseri BC9-BC14, Lactobacillus vaginalis BC15-BC17Cell-free supernatant (CFS; filtered) of MRS fermented by probioticsHeLa cellsReduced the adhesion of Candida spp.[76]
Lactobacillus casei ATCC 334, Lactobacilus rhamnosus GG (ATCC 53103)Cell-free supernatant (CFS; filtered) of MRS fermented by lactic acid bacteriaKirby–Bauer disk diffusion susceptibility testExhibited antifungal activity against blastoconidia and biofilm of Candida albicans[77]
Common pediatric infectious diseases with unknown causeL. paracasei CBA L74Heat-treated fermented cow’s skim milk powderHealthy children aged 1–4 years in daycare or preschoolConsumption was associated with a reduction of common infectious disease[17]
L. paracasei CBA L74Heat-treated fermented cow’s milk or riceHealthy children aged 1–4 years attending daycare or preschoolPrevented common pediatric infectious diseases in children after 3 months of consumption[78]
Bifidobacterium breve C50, Streptococcus thermophilus 065Heat-treated fermented infant formulaHealthy children aged 4–6 monthsDid not prevent the incidence of acute diarrhea in healthy infants but reduced its severity[79]
Necrotizing enterocolitisSix human strains of Bifidobacterium breveWhey retentate from fermented cow’s milkSPF C3H male mice, healthy volunteers aged 21–35 yearsLed to a decrease in clostridia, bacilli, B. fragilis, and fecal pH, as well as to an increase in bifidobacteria[80]
Lactobacillus paracasei CBA L74Heat-treated fermented milk powderC57/BL6 miceStrong anti-inflammatory activity in vitro and protected against colitis or enteric pathogens in vivo[43]
Lactobacillus paracasei MCC1849Orally administered heat-inactivated probioticMale SPF BALB/c miceEnhanced antigen-specific IgA secretion and induced follicular helper T cells[81]
Lactobacillus rhamnosus HN001Orally administered UV-killed probioticNewborn mice and premature pigletsAttenuated necrotizing enterocolitis severity[82]
* Of note, the genus of Lactobacillus has been recently reclassified into 25 genera, which includes 23 novel genera [83].

Share and Cite

MDPI and ACS Style

Mantziari, A.; Salminen, S.; Szajewska, H.; Malagón-Rojas, J.N. Postbiotics against Pathogens Commonly Involved in Pediatric Infectious Diseases. Microorganisms 2020, 8, 1510. https://doi.org/10.3390/microorganisms8101510

AMA Style

Mantziari A, Salminen S, Szajewska H, Malagón-Rojas JN. Postbiotics against Pathogens Commonly Involved in Pediatric Infectious Diseases. Microorganisms. 2020; 8(10):1510. https://doi.org/10.3390/microorganisms8101510

Chicago/Turabian Style

Mantziari, Anastasia, Seppo Salminen, Hania Szajewska, and Jeadran Nevardo Malagón-Rojas. 2020. "Postbiotics against Pathogens Commonly Involved in Pediatric Infectious Diseases" Microorganisms 8, no. 10: 1510. https://doi.org/10.3390/microorganisms8101510

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