Next Article in Journal
Dietary Fibers in Healthy Children and in Pediatric Gastrointestinal Disorders: A Practical Guide
Next Article in Special Issue
Probiotics and Human Milk Differentially Influence the Gut Microbiome and NEC Incidence in Preterm Pigs
Previous Article in Journal
Molecular Oxygen Levels and Percentages of DNA Damage in TPN Patients
Previous Article in Special Issue
Iron and DHA in Infant Formula Purchased in the US Fails to Meet European Nutrition Requirements
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Limosilactobacillus fermentum CECT5716: Clinical Potential of a Probiotic Strain Isolated from Human Milk

1
School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Türkiye
2
Department of Paediatric Chronic Diseases, Nantes University Hospital, 44000 Nantes, France
3
HiPP GmbH & Co. Vertrieb KG, 85276 Pfaffenhofen, Germany
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(9), 2207; https://doi.org/10.3390/nu15092207
Submission received: 22 March 2023 / Revised: 28 April 2023 / Accepted: 2 May 2023 / Published: 6 May 2023

Abstract

:
Breastfeeding provides the ideal nutrition for infants. Human milk contains a plethora of functional ingredients which foster the development of the immune system. The human milk microbiota predominantly contributes to this protective effect. This is mediated by various mechanisms, such as an antimicrobial effect, pathogen exclusion and barrier integrity, beneficial effects on the gastrointestinal microbiota, vitamin synthesis, immunity enhancement, secreted probiotic factors, and postbiotic mechanisms. Therefore, human milk is a good source for isolating probiotics for infants who cannot be exclusively breastfed. One such probiotic which was isolated from human milk is Limosilactobacillus fermentum CECT5716. In this review, we give an overview of available interventional studies using Limosilactobacillus fermentum CECT5716 and summarise preclinical trials in several animal models of different pathologies, which have given first insights into its mechanisms of action. We present several randomised clinical studies, which have been conducted to investigate the clinical efficacy of the Limosilactobacillus fermentum CECT5716 strain in supporting the host’s health.

Graphical Abstract

1. Introduction

Breastfeeding is the ideal nutrition for infants, providing the neonate with all the required macro- and micronutrients in an individualised and optimal way. Besides nutrients, human milk (HM) contains a plethora of non-nutritive functional ingredients, e.g., immunoglobulins, human milk oligosaccharides (HMO), microorganisms, and cytokines, supporting the neonate´s specific immune system and tolerogenic immune response [1]. The role of HM in preventing infections is well established [2,3,4]. Therefore, exclusive breastfeeding is recommended in the first six months of life [5]. The mechanisms of this protective effect are not only based on direct immunological effects (anti-infectious substances such as immunoglobulins) but also on the modulation of the microbiota, specifically, by the human milk microbiota (HMM) [6].
The microbial community of HM is transferred to the infant during breastfeeding (vertical transmission) and plays an important role in the development of the infant’s intestinal microbiota, immune response, and pathogen defence. These microbes contribute to the health-promoting effects of HM via various mechanisms, e.g., via the production of antimicrobial compounds [7]. Scientific evidence shows that the HMM is an infant-tailored protective entity influenced by several factors: internal factors (e.g., genetic predisposition regarding HMO) and external factors (e.g., delivery mode, perinatal antibiotics) are associated with alterations in the HMM. Correlations have also been observed for the phase of lactation (more lactobacilli in the neonatal phase and more bifidobacteria in mature milk) and sex of the infant [8,9] or mother´s nutrition and geographic location [10].
The presence of staphylococci, streptococci, corynebacteria, propionibacteria, lactobacilli, and bifidobacteria was detected in the vast majority of HM samples tested in different cohorts via different methods, leading to the hypothesis that these are part of the core microbiota, i.e., the microbial taxa most often found to be part of the HMM in different studies [1,11]. Recent estimations suggest that HM contains approximately 103 colony forming units (CFU) per ml. Previous estimations were higher, possibly due to less stringent disinfection of surrounding skin [7]. Although most studies on the HMM focus on bacteria, some studies detected fungi in HM [12]. Furthermore, a virome unique to HM was identified that may be transmitted to the infant, which will be very interesting for future studies to investigate [13].
While the origin of the HMM is not yet fully understood, several routes have been hypothesised: (1) contamination from skin microbiota, (2) engraftment of microbes from the infant´s mouth via retrograde flow, and (3) translocation of gut-resident microbes, via the entero-mammary pathway. All three factors might synergise in generating a diverse health-promoting HMM.
The isolation of probiotic strains from HM may present a possibility to transfer the HMM-mediated protection to infants who cannot be breastfed [6]. As probiotics are assumed to be host specific, the characterisation of probiotic candidates from human sources is a way to obtain effective probiotic strains with health-promoting properties. Isolation from non-human sources might be less effective, due to the inability of engraftment in the intestinal microbiota [14]. Therefore, the isolation of new strains from HM or from infant faecal sources is considered the ideal approach to obtain probiotic strains for infants’ health [6]. For instance, lactobacilli are regularly found in the HMM [11], and bifidobacteria are abundant in faeces [10].
Per the definition of probiotics, delivery of the microorganism in its viable form and survival in the gastrointestinal tract (GIT) are required. This ensures that the metabolically active microorganisms produce proactive compounds in the final product or in the gut. In addition to probiotics, HM also contains prebiotics that support the establishment of a characteristic infant microbiota, rich in lactobacilli and bifidobacteria. The aim of microbiota modulation is to foster a healthy microbiota mimicking the microbiota of breastfed infants by using probiotics, prebiotics, or a combination of both (synbiotics). Recently, the term postbiotics emerged after it was shown that some of the protective effects of probiotics can also be observed in a non-viable state [15]. Thus, some studies on probiotics, where inactivated forms of bacterial strains were used, describe postbiotic effects.
Limosilactobacillus fermentum (L. fermentum) CECT5716, isolated from HM, is a well-known strain in the fields of nutritional research and consumer health. This interest arises due to the actions of L. fermentum CECT5716 on the immune system and intestinal barrier. Health-promoting effects have been shown in vitro, in vivo, and in human studies. In this review, we describe the mechanisms by which the probiotic strain L. fermentum CECT5716 confers health-promoting effects. We further summarise its safety profile and the results of relevant preclinical and clinical studies with L. fermentum CECT5716.

2. L. fermentum CECT5716

The genus of Lactobacillus has grown very large and diverse. Therefore, the taxonomy has recently been updated, thus introducing new genus names. Based on physiology and genomics, Lactobacillus fermentum was therefore re-categorised as Limosilactobacillus fermentum [16]. It still remains within the family of Lactobacillaceae (order: Lactobacillales; class: Bacilli; phylum: Bacillota (formerly Firmicutes)).
The presence of the L. fermentum species in HM was confirmed in several cohorts and studies performed by various research groups worldwide [7,8,17]. L. fermentum is a normal inhabitant of the human GIT, including that of breastfed infants [18,19,20]. The L fermentum CECT5716 strain was categorised originally as a strain within the species of Lactobacilli, which contains Gram-positive, catalase-negative, non-sporulating, rod-shaped organisms. The abundance of lactobacilli in adults is lower than in infants and seems to be even lower in industrialised countries and in unfavourable dietary habits [21].
Lactobacilli in HM were first discovered in 2003, by analysing bacteria from different habitats of healthy mother–infant pairs during physiological lactation. This discovery prompted the hypothesis that the HMM could have probiotic benefits for the infant [22].
After testing different isolated strains from HM, one strain in particular, catalogued as L. fermentum CECT5716, displayed superior probiotic properties compared to other strains (e.g., GIT survival) [23]. Therefore, this strain was chosen for further investigation in preclinical and clinical studies, as described in Section 5 and Section 6.
As the benefits of probiotic bacteria are strain specific, a better knowledge of the genetic basis of the strain-specific traits was required. Therefore, the whole genome of L. fermentum CECT5716 was sequenced and related to phenotypic traits [24,25], and the fermentation profile of L. fermentum CECT5716 was established [23].

3. Probiotic Properties and Mechanisms of Action of L. fermentum CECT5716

In 2001, an expert meeting of international scientists working on behalf of the Food and Agriculture Organization of the United Nations (FAO) and the WHO discussed the emerging field of probiotics and re-defined the term “probiotics” as “live microorganisms which when administered in adequate amounts confer a health benefit on the host” [26,27]. While the probiotic strain has to be in a viable state throughout the shelf-life in the respective product, it also has to survive the harsh conditions of the GIT. Notably, it was shown that even after 80 min and a low pH of 1.8, L. fermentum CECT5716 displayed 30% viability and thereby exceeded the viability of some other tested strains [23]. Survival conditions in the infant stomach are expected to be less harsh; therefore, the percentage surviving in the infant stomach is estimated to be higher.
Furthermore, prebiotics can support probiotic effects. In vitro, L. fermentum reached higher growth levels when supplemented with prebiotics, indicating that certain prebiotic factors contained in HM may foster probiotic growth in the colon [28]. Probiotic properties are strain dependent, and therefore, probiotic candidates are not chosen solely on their capability to survive in the GIT, as this trait is no indication of true health-promoting capacities. This choice is also based on their ability to inhibit pathogens via mechanisms presented in the following sections. A summary of shown probiotic mechanisms for L. fermentum CECT5716 is presented in Figure 1.

3.1. Antimicrobial Effect

To prevent pathogens from infecting the host, antimicrobial defence by host cells and commensal microbiota is exerted in close proximity to the intestinal epithelial layer, as the first line of defence. Infants fed HM exhibit significantly lower rates of infectious diseases compared to formula-fed infants [3]. Thus, it is hypothesised that probiotics in the HMM may contribute to antimicrobial defence by inhibition of pathogen growth or adhesion. Inhibitory effects of L. fermentum CECT5716 are assumed to be mediated by the production of short-chain fatty acids (SCFA) and lactate [23]. L. fermentum CECT5716 inhibited Salmonella choleraesuis CECT4155 in vitro and in vivo and repressed the growth of Staphylococcus aureus, Listeria spp., and Escherichia coli in vitro [29]. The antimicrobial effect against Staphylococcus aureus was also shown in mothers consuming L. fermentum CECT5716 [30]. Moreover, the authors of the present review have confirmed a strong antimicrobial activity of L. fermentum CECT5716 in recent experiments, where the highest inhibition was observed against Serratia marcescens, Streptococcus pyogenes, and Klebsiella pneumonia (internal data).

3.2. Pathogen Exclusion and Barrier Integrity

The adhesion of probiotics to intestinal epithelial cells is assumed to be an important probiotic trait. The rationale lies within the assumed importance of the close proximity between the probiotic and the epithelium to exert a direct effect on the host cell, e.g., by increasing the amount of mucin [34], or to displace or exclude pathogens from attachment to epithelial cells. L. fermentum CECT5716 displayed superior adhesion ability in vitro to HT-29 as well as Caco-2 cells [23]. In addition, increased mucin expression was demonstrated [29].
Another potential mechanism could be the strengthening of the host´s intestinal barrier function. This effect could be mediated by increasing the expression of barrier proteins such as occludin or zonula occludens 1 (ZO-1). It was shown that L. fermentum CECT5716 prevented the stress-induced loss of intestinal barrier proteins in rats [35]. This observation was confirmed in a rat model of vascular oxidative stress [31]. However, the mechanisms behind this L. fermentum CECT5716-induced protection from barrier loss are yet to be investigated and confirmed in humans.

3.3. Effects on the Gastrointestinal Microbiota

The growth promotion of bifidobacteria is referred to as the “bifidogenic effect” and is associated with a healthy faecal microbiota in infants. Exclusively breastfed infants have high a bifidobacteria abundance in their faeces, despite the fact that the HMM is not dominated by bifidobacteria [11,38]. Supplementation of infant formula with bifidobacteria is not always successful in increasing the abundance of bifidobacteria in the infant gut [39]. Therefore, synbiotic factors, i.e., factors that positively influence the abundance of beneficial bacteria, are assumed to be of greater importance to the bifidogenic effect of HM.
One strategy to increase the abundance of bifidobacteria is the addition of prebiotics to infant formula [40]. Another possibility is the addition of probiotics that enable engraftment and support the growth of bifidobacteria. Especially in the neonatal gut, aerotolerant lactobacilli, which could originate from either the HMM or the mother´s vaginal microbiota, play an important role in setting the stage for the colonisation of the strictly anaerobic bifidobacteria. It is hypothesised that lactobacilli reduce intestinal oxygen and thereby might promote the engraftment of bifidobacteria [41]. It was shown in vitro and in vivo that L. fermentum CECT5716 has a bifidogenic effect [31,32].
In addition to increasing the abundance of bifidobacteria, other positive effects on the overall composition of microbiota were observed for L. fermentum CECT5716, e.g., by increasing the abundance of microbes which produce favourable metabolites or by the reduction in intestinal dysbiosis [42]. In a pig model of formula feeding, a beneficial influence on the faecal microbial composition was demonstrated. In this model, L. fermentum CECT5716 was only administered during the suckling phase, and the pigs were subsequently challenged with a high-energy diet during their weaning phase. The addition of L. fermentum CECT5716 had positive long-term effects on bacterial SCFAs production (especially propionate) and caecal glucagon-like peptide 1 (GLP-1) secretion [43]. GLP-1 has an important role in various metabolic processes, such as insulin secretion and inhibition of gastric emptying and food intake [44]. Interestingly, this L. fermentum CECT5716-induced increase in GLP-1 has been previously observed in a rat model under a high fructose diet challenge [45]. This indicates that probiotic intervention during early phases might have long-term influences on the microbiota composition and host metabolism.

3.4. Effects on Vitamin Synthesis

Although most lactic acid bacteria are auxotrophic for several vitamins, certain strains have the ability to synthesise water-soluble vitamins such as those categorised as B-group vitamins (e.g., folate (B9), riboflavin (B2), and cobalamin (B12)) [46]. L. fermentum CECT5716 produces riboflavin, pyroxidine (B6), and folate in vitro [24]. Folate is particularly interesting due to high folate requirements in the rapidly growing infant. Further, the gut epithelium has a high turnover and is therefore also dependent on folate in the adult state. In particular, the colonic epithelium might benefit from microbially produced folate [47]. However, it is not clear which genes are involved in the synthesis and whether the amount of vitamins produced indeed contributes to the host´s folate status. Few strains have been identified to contribute to the host’s vitamin status [48]. An improvement in the folate status was demonstrated in vivo and suggests that the bifidogenic effect of HM might mediate this improvement [49].

3.5. Effects on Immune Response

Another mechanism by which probiotics mediate their health-promoting effect is via activation of the host’s immune response. In a human trial of anti-influenza vaccination, L. fermentum CECT5716 increased the immunomodulatory response upon vaccination, as discussed in more detail in Section 6.3 [50]. Further, an increased interferon-γ (IFN-γ) response was observed in another study [35]. For L. fermentum CECT5716, a predominant T-helper type 1 (Th1)- and immunoglobulin A (IgA)-driven immune response was observed, compared to other strains [36].
Interestingly, there are indications that the activation of certain immune response characteristics might be linked to the origin of the strain, thereby emphasising the host specificity of probiotics. Compared to saliva-derived strains, the HM-derived strain L. fermentum CECT5716 was demonstrated to induce IL-10 to a larger extent. Further, L. fermentum CECT5716 also preferentially activated CD56+CD8+ natural killer (NK) cells [37], which are assumed to have greater cytotoxic efficacy than their CD8- counterparts [51]. The overall immunomodulatory response, however, was similar when comparing the strains of different origin [37]. As NK cells seem to be important in neonatal immune defence, i.e., before acquired immunity is fully developed, the enhancement of NK cell maturation by the HMM could be of pivotal importance in the neonatal gut, where more NK cells are found than in adults [51,52].

3.6. Secreted Probiotic Factors

Compounds secreted by metabolically active microorganisms include protein- and carbohydrate-degrading enzymes, antimicrobial factors, or immune-modulatory factors (e.g., lactocepin, reuterin, or SCFAs) [53,54,55]. By inducing antioxidants in the GIT, probiotics can reduce intestinal inflammation. Antioxidant activity is perceived as an important probiotic characteristic and might be mediated in part by the tripeptide glutathione (gamma-L-glutamyl-L-cysteinyl-glycine)—the major component of the endogenous non-protein sulfhydryl pool [56]. Glutathione neutralises reactive oxygen species and is therefore an important antioxidant in the maintenance of intestinal barrier integrity and prevention of inflammation. Low glutathione levels in humans are connected with several disease states such as cancer, AIDS, Alzheimer’s disease, and Parkinson’s disease [57]. Of note, secretion of glutathione is not a common feature among lactic acid bacteria, but has been shown for L. fermentum ME-3 [58,59,60]. L. fermentum CECT5716 increases the extracellular glutathione levels by recycling (i.e., oxidation) of reduced glutathione (GSSG) [58,59]. In addition to glutathione, the antioxidant dipeptide ɤ-Glu-Cys is also released by L. fermentum CECT5716. The dipeptide ɤ-Glu-Cys is a precursor of glutathione and might be more efficiently absorbed in the intestine than glutathione [24,61]. Increasing the extracellular glutathione levels is not only beneficial for the host during phases of increased oxidative stress, but may also increase bacterial viability and survival during gastrointestinal passage [56].
Besides glutathione secretion, production of SCFAs such as acetate, propionate, and butyrate is also of central interest in the prevention of diseases and maintenance of gastrointestinal health. SCFAs are ingested through certain foods and produced by intestinal microbes. There is evidence that SCFAs have protective effects against the development of allergies, such as allergic rhinitis, asthma, and food allergies [62].
Probiotics may also play an important role in the improvement in iron uptake and thereby support adequate iron levels. Anaemia resulting from low iron levels is one of the most common nutritional disorders, mostly affecting infants and women [63]. Dietary iron leaves the stomach as Fe(III) and is reduced by ferrireductase (DcytB) in the duodenum to Fe(II), which is subsequently transferred across the apical membrane by divalent metal transporters. While several studies claim facilitation of iron absorption by probiotics, the mechanism is not yet clear. It was proposed that a reduced pH caused by lactic acid increases iron solubility and therefore absorption [64]. Furthermore, L. fermentum CECT5716 secretes p-hydroxyphenyllactic acid (HPLA). In vitro, HPLA from L. fermentum CECT5716 can reduce Fe(III) to Fe(II) and thereby renders it to the essential form that can be taken up by enterocytes [33]. HPLA is also discussed to have antifungal and biopreservative capacities [65]. This intriguing observation aligns with the observation of a low-molecular-weight fraction in HM with iron-reducing capacity, which enhances the iron absorption in newborns [66].

3.7. Postbiotic Mechanisms

The presence of viable bacteria is crucial for some of the beneficial effects of probiotics. However, bacterial factors derived from non-active microorganisms can also be beneficial. For instance, heat-inactivated L. fermentum CECT5716 was demonstrated to reduce inflammation caused by the induction of colitis in a rat model of gut inflammation [15]. Several studies in animals and humans have shown that some strains show health-promoting effects independent of their viability or even cell integrity. These effects may be due to the fact that host cells recognise bacterial motifs (e.g., DNA, cell wall components such as peptidoglycan, or lipopolysaccharides) via Toll-like receptors, which triggers an immunomodulatory effect [67]. These effects have come to be called “postbiotic”, though a recently published definition of “postbiotic” not only includes inanimate cells but also their produced metabolites [68]. By this definition, postbiotic effects have clearly been demonstrated for L. fermentum CECT5716. Postbiotics could be an alternative to probiotics for vulnerable individuals, for whom live probiotics might raise potential safety concerns.

4. Safety of L. fermentum CECT5716

L. fermentum CECT5716 is included in the list of taxonomic units proposed by the European Food Safety Authority (EFSA) for the qualified presumption as safe (QPS) status and is generally recognised as safe (GRAS Notification in 2015) by the US Food and Drug administration (FDA) [69]. However, despite their safe use over a long time and the general assumption of their safety, lactobacilli may theoretically and in rare cases cause negative side effects. Therefore, safety assessments regarding antibiotic susceptibility and virulence properties are recommended. When judging safety of new probiotic strains, criteria such as human origin, long history of safe usage, or absence of pathogenic traits are applied and therefore also tested for probiotic candidates [70,71]. The EFSA has created cut-off values for antibiotic susceptibility for probiotics [72]. Two independent studies have shown that the pattern of susceptibility of L. fermentum CECT5716 met the thresholds referenced by the EFSA, for all antibiotics tested [24,70]. Similarly, based on a systematic review, the European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) committee concluded that probiotic-supplemented formula does not raise safety concerns [73].
Regarding toxicity, even at a concentration 10,000 times higher (expressed per kg of body weight) than those consumed normally by humans, no death, illness, or sign of negative activity was observed in a mouse model. Additionally, no translocation of L. fermentum CECT5716 to blood, liver or spleen was detected [70].
Another potential safety concern is the generation of biogenic amines such as histamine and tyramine, which can be generated by some microorganisms through the activity of amino acid decarboxylase. Ingestion of high amounts of biogenic amines such as histamine and tyramine can be mistaken for allergic reactions due to the similarity of the symptoms, e.g., facial flushing, sweating, rash, diarrhoea, and cramps [74]. However, the production of unwanted biogenic amines (tyramine, histamine, putrescine, and cadaverine) was not detected for L. fermentum CECT5716. Further, the production of prophages (i.e., bacteriophage genomes integrated into the host genome), which is of interest due to their involvement in the transduction of antibiotic resistance genes, could not be identified [23,24,75].
In addition to testing these safety properties in vitro and in vivo, several clinical studies in infants have confirmed the safety of L. fermentum CECT5716 for human use. The details of these randomised trials are further described in Section 6. For instance, a randomised study of 137 infants aged 1 month has shown that the growth of infants who had received either L. fermentum CECT5716-containing formula or the control formula was identical in both groups after 4 months and 6 months. The supplemented infants did not experience any bacterial complication relating to probiotic supplementation [76]. In a 3-year follow-up of this study comparing children who had received either L. fermentum CECT5716-containing formula or the control formula, no differences in growth or incidence of diseases were observed. Thus, the long-term safety of L. fermentum CECT5716 was confirmed [77].
In addition, preclinical studies can provide insight into the probiotic mechanisms of L. fermentum CECT5716 and thus inform on potential safety concerns, as described in the next section.

5. Preclinical Studies

A plethora of probiotic strains are assigned to putative health-promoting effects. Preclinical trials in several animal models of different pathologies have given first insights into mechanisms of action, which is an essential step in determining host- and pathology-tailored probiotics. Currently available animal-free testing models lack the complex interplay of different organ systems as well as the behaviour of the respective strain in the GIT. Nonetheless, the usage of animal models in testing probiotic strains should be strictly controlled and the benefit defended in front of an ethics committee to limit animal numbers. A summary of preclinical studies using L. fermentum CECT5716 is presented in Table 1.

5.1. Gastrointestinal Inflammation

Breastfeeding seems to prevent the development of inflammatory bowel diseases (IBD) such as Crohn´s disease or Ulcerative colitis in later life [87,88]. This effect might be mediated by the HMM. Several murine models for human IBD are available. They vary not only in their way of inducing the intestinal inflammation, but also in the type of immune response, location of inflammation, and affected structures [89]. Therefore, probiotic strains should be tested in different models to verify an IBD-protective capacity.
In a rat model of chemically induced colitis (trinitrobenzenesulfonic acid (TNBS)), the efficacy of various probiotics, including L. fermentum CECT5716, to prevent histological signs of inflammation was tested. The study showed that different probiotics exerted different mechanisms, ultimately resulting in a reduction in histological damage. While both L. fermentum CECT5716 and L. reuteri ATCC55730 prevented histological signs of inflammation and reduced the level of colonic myeloperoxidase infiltration (as marker of neutrophil infiltration), only L. fermentum CECT5716 significantly counteracted the colonic glutathione depletion induced by inflammatory processes. Further, a decrease in cyclooxygenase expression and an increase in SCFAs was only observed for L. fermentum CECT5716, a trait already described and associated with protection against exacerbated inflammatory processes [55,61].
In another chemically induced murine colitis model (dextrane sulphate sodium (DSS)) L. fermentum CECT5716 also reduced colonic histopathology. One possible mechanism of protection might be the preservation of intestinal barrier function. L. fermentum CECT5716 treatment sustained the expression of the barrier proteins occludin and ZO-1, which resulted in reduced intestinal permeability [42]. In another mouse study, L. fermentum CECT5716 attenuated TNBS-induced colitis, even when given in a therapeutic manner, i.e., after induction of pathology [79].
Although these are artificial models, they can help to elucidate probiotic mechanisms. For instance, L. fermentum CECT5716 treatment resulted in amelioration of the inflammatory response by counteracting the colonic glutathione depletion [61]. This antioxidant effect may be especially important in this model of gut inflammation, as TNBS administration is assumed to induce acute colonic injury mainly by the production of highly reactive oxygen species (ROS) in the early phase of colonic damage [79]. Another possible mechanism might be the L. fermentum CECT5716-mediated increase in luminal SCFA levels. SCFAs are serving as the main energy substrate for intestinal epithelial cells and ameliorate the IBD-related excessive immune response, by promoting regulatory T-cells [55,90,91].
Irritable bowel syndrome (IBS) is a widespread intestinal disorder, characterised by abdominal discomfort and pain, changed bowel habits, flatulence, and bloating. The aetiology is still unknown, but differences in the intestinal epithelial barrier function, composition of the gut microbiota, and immune response are discussed as possible pathophysiological mechanisms [92,93,94,95,96]. In a model of experimental IBS induced by deoxycholic acid (DCA) in rats, attenuation of visceral hypersensitivity, changes in gut inflammatory markers, and improvement of the intestinal barrier in experimental IBS were demonstrated. In behavioural studies, L. fermentum CECT5716 also improved anxiety-like behaviour, associated with experimental IBS, showing its potential to improve psychological stress [78].

5.2. Psychological Stress

Psychological stress, e.g., by maternal separation, can induce a profound stress response in infants that could impair the intestinal barrier function. This increased stress response can alter systemic cortisol levels and affect learning processes. Moreover, a stress-induced decrease in intestinal barrier function might be associated with long-term health consequences such as increased incidence of allergies [97,98]. A potential option to ameliorate these stress-induced consequences might be early life administration of probiotics, including L. fermentum CECT5716. A recent animal study investigated this hypothesis by exposing rat pups to psychological stress by maternal separation and water avoidance testing. In both tests, pups receiving L. fermentum CECT5716 showed improved intestinal barrier tightness and reduction in intestinal permeability (i.e., increased ZO-1 expression and decreased permeability to fluorescein sulfonic acid) in a rat model of maternal separation and water avoidance. Interestingly, ex vivo analysis showed spatial differences in the barrier protection effects. L. fermentum CECT5716 especially improved the ileal barrier, while having almost no effect on the proximal colon. Furthermore, they tested the probiotic effects on anxiety-like behaviour, ethological parameters, and locomotor activity in a maze setting. While anxiety-like behaviour did not change, the L. fermentum CECT5716-treated group showed increased exploratory behaviour during the test phase. While behavioural observations are subjective measures, plasma markers of barrier function (sulfonic acid) or stress (corticosterone) were improved by the probiotic treatment as well. The authors therefore concluded that L. fermentum CECT5716 may be a novel method for prevention and treatment of gastrointestinal disorders associated with reduced intestinal barrier function [35].

5.3. Hypertension

Hypertension is one of the main risk factors for cardiovascular events such as myocardial infarct or stroke. Current guidelines for the management of hypertension include dietary interventions with the aim to reduce the number and dosage of necessary medication. As the consumption of fermented milk products historically showed antihypertensive potential, probiotics might play a role in the reduction in hypertension. One proposed mechanism for this effect is the strengthening of the anti-inflammatory capacity, as endothelial inflammation may play an important role in causing endothelial dysfunction [99]. By using a spontaneously hypertensive rat (SHR) model (characterised by elevated blood pressure, arterial remodelling, endothelial dysfunction, vascular inflammation, and dysregulated immune system), orally administered L. fermentum CECT5716 reduced the systolic blood pressure. Along with this, an increased relaxant response was observed in the probiotic groups (as measured by the contractile responses). One of the key mechanisms of endothelial dysfunction is vascular generation of ROS, which is abolished via indirect effects by orally administered L. fermentum CECT5716. Further, the proinflammatory response in the aorta of these rats was reduced, as shown by attenuated tumour necrosis factor alpha (TNF-α) expression [80]. In the same animal model, L. fermentum CECT5716 was demonstrated to prevent the development of hypertension. Furthermore, an increase in butyrate-producing bacteria, improved balance of T-helper and T-regulatory cells, normalised endotoxaemia, and improved endothelial function were observed [81].
On the other hand, L. fermentum CECT5716 was not effective in a rat model of induced hypertension via administration of an inhibitor of nitric oxide synthase (NOS)-induced dysbiosis, endothelial inflammation, and hypertension [31]. However, the early signs in the atherosclerotic process, i.e., dysbiosis (in the respective work defined as increased Firmicutes/Bacteroidetes ratio), vascular oxidative stress, and inflammation (defined by aortic ROS accumulation and Th17 infiltration, respectively) could be attenuated by L. fermentum CECT5716 [31].
In a mouse model of induced hypertension, L. fermentum CECT5716 inhibited the development of hypertension and endothelial dysfunction, which were induced by tacrolimus. Further, the balance between T-helper and T-regulatory cells was restored. The authors hypothesised that these changes were mediated by the prevention of gut dysbiosis [82].
Systemic lupus erythematosus is an autoimmune disorder leading to multiple complications, including endothelial dysfunction and hypertension. In a mouse model of systemic lupus erythematosus, L. fermentum CECT5716 and/or Bifidobacterium breve CECT7263 were administered. Both probiotics prevented hypertension and endothelial dysfunction, reduced the plasma levels of autoantibodies against double-stranded DNA, activated Toll-like receptor 9 (TLR-9) expression, and reduced T-cell activation [83].
Taken together, these results show that although probiotics cannot be the sole treatment for hypertension, certain pathophysiological mechanisms can benefit from probiotic intervention.

5.4. Immunity

L. fermentum CECT5716 displayed beneficial effects on immunity when administered during pregnancy and lactation in rats. In lactating dams, a reduction in T-cytotoxic cells, modulation of intestinal cytokines, and change in fatty acid composition were observed, while their pups exhibited changes in immunoglobulins and fatty acid profile [85].
These results indicate that L. fermentum CECT5716 has beneficial effects on the immune system of mother and offspring.
Among hospitalised patients in intensive care units, sepsis is one of the most common causes of death. In 2016, the Sepsis-3 conference defined sepsis as a “life-threatening organ dysfunction caused by a dysregulated host response to infection” and septic shock as a “subset of sepsis in which circulatory, cellular, and metabolic abnormalities are associated with a greater risk of mortality” [100]. Sepsis is often a secondary complication, e.g., in patients suffering from trauma characterised by a reduction in cellular immune response. An initial event is the release of lipopolysaccharides (LPS) from bacteria into the bloodstream, shown also by the fact that septic shock can be induced by intraperitoneal application of LPS in animals. Interestingly, mice pre-treated with L. fermentum CECT5716 had significantly reduced LPS-induced changes in organ weight and improved biochemical parameters such as TNF-α levels after LPS injection, compared to the control group. The LPS-induced alteration in liver function, as evidenced by the decrease in glutathione levels, was not as marked in L. fermentum CECT5716-pre-treated mice as in the control group, a mechanism that could attenuate the deleterious effect of ROS often observed in sepsis [84].

5.5. Metabolic Disease

Metabolic syndrome is characterised by obesity, insulin resistance, fatty acid profile alterations, and hypertension. Likewise, emerging research suggests that the intestinal microbiota can have a large impact on the emergence of these conditions. Therefore, probiotic interventions could be an attractive therapy in the management of metabolic syndrome. For example, the effect of L. fermentum CECT5716 on metabolic syndrome was analysed in a preclinical study on rats that were fed a high-fructose diet, resulting in insulin resistance, liver steatosis, altered production of SCFAs, and changes in microbiota. Feeding a synbiotic containing L. fermentum CECT5716 and fructooligosaccharides ameliorated the effects of a high-fructose diet on liver steatosis, systemic inflammation, dysbiosis, and barrier function [45]. Similarly, beneficial effects were observed in a study on minipigs. Piglets were fed a probiotic formula with plant lipids, dairy lipids, and L. fermentum CECT5716, before being challenged with a high-energy diet. The probiotic formula had multiple beneficial effects, including modulation of gut microbiota, increase in SCFA concentration, and enhanced GLP-1 secretion [43]. These results indicate that L. fermentum CECT5716 supplementation in infants may decelerate or prevent development of metabolic diseases later in life.

5.6. Asthma and Allergy

Asthma is a bronchial inflammatory disease, causing shortness of breath, chest tightness, and other symptoms, thus presenting a significant impact on the quality of life of asthma patients. A recent study in ovalbumin-induced mice showed that supplementation with L. fermentum CECT5716 decreased the content of inflammatory factors in bronchoalveolar fluid and expression of TLR2/TLR4 in the duodenum and lungs. Further, inflammatory cell infiltration in the airway mucosa was reduced [86]. These results suggest that L. fermentum CECT5716 might be a novel complementary treatment for asthma, although further studies, especially human trials, are needed.
First ex vivo results with the serum of cow’s milk-allergic infants showed that L. fermentum CECT5716 did not show any allergenic potential when tested against specific antibodies (α-S1-casein, α-S2-casein, α-β-casein, α-κ-casein, α-lactalbumin, α-β-lactoglobulin, α-lactoferrin). In the same study, it was shown that in vitro, in combination with a galactooligosaccharide containing extensively hydrolysed infant formula, L. fermentum CECT5716 significantly reduced the allergenic activity of the infant formula. This was shown by a decreased basophil degranulation of rat basophil leukaemia (RBL) cells that express the human high-affinity receptor for immunoglobulin E (FcεRI) and were loaded with serum immunoglobulin E (IgE) from cow’s milk-allergic patients. These results suggest that L. fermentum CECT5716 might be a promising ingredient for hypoallergenic infant formulae [101].
Although preclinical studies have provided convincing evidence for the efficacy of L. fermentum CECT5716 to modulate host responses, specifically those relating to immune and gastrointestinal functions, translating and confirming these findings into clinical populations is of utmost importance. Thus, the following section summarises clinical studies on the efficacy of L. fermentum CECT5716 in humans.

6. Clinical Trials

The first years of human life are a period with a particularly high rate of infections. As explained previously, a relationship between immunity and the microbiota has been established in various studies [37,51,52]. Thus, it is hypothesised that support of the microbiota in early life, e.g., by breastfeeding, might prevent infections in infants and adults.
We have recently published the results of a randomised controlled clinical trial investigating the effect of L. fermentum CECT5716 on the infant microbiota during the first year of life. A total of 540 infants were included in the study and received, for a duration of 12 months, either the synbiotic intervention formula L. fermentum CECT5716 and galactooligosaccharides, or a standard formula without pro- or prebiotics. A non-randomised breastfed group was also included. At the age of 4 months, significantly higher prevalence and abundance of Bifidobacterium spp. and Lactobacillaceae in infants fed with formula supplemented with L. fermentum CECT5716 and galactooligosaccharides were demonstrated. The phylogenetic profiles of these infants were closer to the profiles of breastfed infants, compared to infants fed with the control formula [102].
Other randomised clinical studies have previously been conducted to investigate the safety and efficacy of L. fermentum CECT5716 in preventing infections. A summary of clinical trials in adults is presented in Table 2, and a summary of clinical trials in infants is presented in Table 3.

6.1. Mastitis

Mastitis is a breast infection that commonly occurs during lactation. The effects of oral administration of L. fermentum CECT5716 were studied in several groups of women with mastitis. In the study by Arroyo et al., 352 women with mastitis were either treated with L. fermentum CECT5716, L. salivarius CECT5713 (9 × 1010 log cfu/day, respectively), or antibiotics for a duration of 21 days. Probiotic supplementation was even more effective than antibiotic treatment in lowering bacterial count (Staphylococcus epidermidis, Staphylococcus aureus, and Staphylococcus mitis) in HM. In addition, patients treated with probiotics experienced a higher reduction in pain scores and lower recurrence of mastitis [30].
Two further studies also showed a significant reduction in Staphylococcus spp. in HM of mothers having received perinatal antibiotics or self-reported pain during breastfeeding. Here, mothers were treated with L. fermentum CECT5716 compared to a placebo [103,104]. In women reporting painful breastfeeding, a decrease in pain score was observed after a three-week supplementation with different doses of L. fermentum CECT5716 (3, 6, or 9 × 109 cfu/day, respectively) [104]. Hereby, significantly less pain was observed after 7 days already. When L. fermentum CECT5716 was given in a preventative manner (3 × 109 cfu/day), a 51% lower incidence of mastitis was observed during the four-month observation period [103].
A recent randomised controlled trial analysed the long-term impact of L. fermentum CECT5716 in women with breast abscesses. Three months after the trial, patients were followed up via an online questionnaire. There was no significant difference in the overall rate of stopping breastfeeding, pain relief, or recurrence of mastitis. However, a statistically significant difference in the rate of stopping breastfeeding due to the recurrence of mastitis was observed in the treatment group compared to the placebo group, showing that the supplementation of nursing women with L. fermentum CECT5716 might support exclusive breastfeeding [105].

6.2. Influenza Vaccination

Since influenza presents a large economic burden, controlling yearly influenza waves is of utmost importance. Therefore, vaccination is recommended. Since it was previously demonstrated that lactobacilli can improve immunity, the effect of L. fermentum CECT5716 on the immune response following influenza vaccination was analysed in healthy adults. In the study, 50 healthy adults were vaccinated and received either 1 × 1010 log cfu/day L. fermentum CECT5716 or placebo 2 weeks before and 2 weeks after vaccination. The probiotic group showed a significant increase in NK cells and helper and cytotoxic T-cells, compared to the placebo group. The production of antigen-specific IgA was also significantly enhanced [50]. Thus, probiotic supplementation could be an interesting approach to improving vaccine efficacy by promoting activation of the immune system.

6.3. Infections in Infants

Due to the high rate of infections, the first years of life are favourable for microbiota modification using probiotics. Thus, several clinical studies have been conducted to investigate whether L. fermentum CECT5716 can prevent infection in infants. Hereby, infants were either directly (via infant formula) or indirectly (via intake of probiotic capsules by breastfeeding mothers) supplemented with L. fermentum CECT5716. Interestingly, even the indirect supplementation resulted in the first compelling protective effects: for instance, a 16-week supplementation with L. fermentum CECT5716 in breastfeeding mothers after delivery led to a significantly reduced incidence of conjunctivitis in their infants [107].
Various studies concluded that no relevant changes in infant growth could be observed after supplementing infants with L. fermentum CECT5716 [76,77,106]. Further, the consumption of a formula containing L. fermentum CECT5716 was similar to that of the control formula. Importantly, tolerance and compliance were comparable between formula groups [76]. Thus, clinical trials confirm the safety and tolerability of L. fermentum CECT5716.

6.3.1. Respiratory Tract Infections

Respiratory tract infections are among the most common diseases in infants. In fact, a recent study revealed that over 80% of infants had at least one upper respiratory tract infection during the first year of life [106]. Breastfeeding is known to decrease the incidence of upper and lower respiratory tract infections, which may involve complementary immunological mechanisms of action [108,109].
Similarly, it was demonstrated that the L. fermentum CECT5716-supplemented formula can reduce the incidence of upper respiratory tract infections by 27% in infants aged 6 to 12 months (the incidence rate during the study period: in the probiotic group, 0.969 ± 0.96; in the control group, 1.330 ± 1.23) [32]. This was not confirmed in a different formula trial using L. fermentum CECT5716 in infants aged 1 to 6 months, where no significant difference in incidence of respiratory tract infections was observed [76], probably due to the low incidence of respiratory tract infections in this age group. Another formula study reported no general difference in incidence of respiratory tract infections, although a subgroup analysis of infants born by caesarean section revealed a reduction in upper respiratory tract infections after L. fermentum CECT5716 administration from 1 to 12 months of age [106].

6.3.2. Gastroenteritis

Diarrhoea caused by gastroenteritis is a mostly viral infection affecting predominantly infants and young children. The prevalence in young children (<3 years) is 0.5 to 2 episodes per child [110]. These infections are widespread throughout the year but usually spread in the form of winter epidemics, and they represent a frequent cause of mortality in developing countries, leading to high health and social costs in industrialised areas.
Breastfeeding is the most effective way of reducing the incidence and severity of these infections in infants. One mechanism through which breastfeeding can prevent infections is fostering the development of the infant microbiota. Thus, in addition to developing vaccines, modulating the microbiota of non-breastfed infants has been explored. Several probiotic strains have been shown to significantly reduce the incidence of gastroenteritis. L. fermentum CECT5716 administration for 5 months showed a significant (71%) reduction in gastroenteritis in infants aged 1 to 6 months (the incidence rate during the study period: in probiotic group, 0.082 ± 0.04; in the control group, 0.283 ± 0.07) [76]. In a second clinical trial, supplementing infants aged 6 to 12 months with L. fermentum CECT5716 led to a significant (46%) reduction in the incidence of gastrointestinal infections as well (the incidence rate during the study period: in the probiotic group, 0.196 ± 0.51; in control group, 0.363 ± 0.53) [32]. In both studies, infants in the probiotic as well as the control group received galactooligosaccharides as prebiotics. Furthermore, a third clinical study in infants aged 1 to 2 months concluded that L. fermentum CECT5716 decreased the incidence of diarrhoea by 44% and the duration by 2.5 days [106]. All three studies demonstrated good tolerability of the study formula enriched with L. fermentum CECT5716 [32,76,106].
Furthermore, these three studies were analysed in a recent systematic review and meta-analysis. This review concluded that L. fermentum CECT5716 at doses from 1 × 109 to 8.4 × 108 cfu/day in milk formulas led to a significantly reduced incidence of gastrointestinal infections in infants up to 12 months. The authors suggested that future long-term studies should be conducted [111]. In addition, the effects observed in these three study cohorts were specifically analysed for those infants delivered by caesarean section. The results revealed a statistically significant reduction (73%) in gastrointestinal infections in infants born by caesarean section who received L. fermentum CECT5716 [112]. As subgroup analyses of intervention studies in infants have shown stronger effects in infants born by caesarean section, further studies focussing on these infants will give valuable insights for this specific group. The microbiota of infants born by caesarean section might benefit even more from probiotic intervention, as their microbiota might have vacant niches in their early phase of life [102]. Therefore, a current study recruiting 486 infants (NCT04991792) is being conducted to assess the positive effect of feeding a formula containing L. fermentum CECT5716 and GOS on the composition of the gut microbiota in caesarean section (CS)-born healthy term infants within the first 6 months of life. Current and future studies will give further insights into the probiotic, as well as postbiotic mechanisms performed by L. fermentum CECT5716 in adults with a predisposition for certain diseases (e.g., IBD), in nursing mothers with a risk of mastitis development, as well as in infants who cannot be breastfed.

7. Conclusions

L. fermentum CECT5716 is a probiotic strain isolated from HM. Several preclinical studies have demonstrated the health-promoting effects of L. fermentum CECT5716 in models of gastrointestinal inflammation, psychological stress, hypertension, immunity, metabolic disease, and asthma. Its positive effect on the microbiota in a synbiotic formulation was shown in various clinical trials investigating mastitis, influenza vaccination, respiratory tract infections, and gastroenteritis. The ESPGHAN criteria for safety are met by L. fermentum CECT5716, and several clinical trials have confirmed the good tolerability and safety of an L. fermentum CECT5716-containing formula.
Future evaluations on the long-term health effects of L. fermentum CECT5716 may provide further insight into its use. Therefore, more intervention studies in infants with longer observation periods will give further insights into its health promoting effects. As probiotic effects might be modified by formula compounds, e.g., hydrolysed protein, prebiotics, or fats, studies with formulas differing in these compounds will be important, to specify certain probiotic or postbiotic mechanisms. Currently, several interventional studies are being performed to further support the existing results and explore new probiotic potentials of this strain.

Author Contributions

Conceptualization, M.S.; writing—original draft preparation, M.S. and H.P.; writing—review and editing, M.S., H.P. and M.O.; visualization, M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by HiPP GmbH & Co. Vertrieb KG.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Katharina Wißmiller and Uwe Walschus of Cerner Enviza (an Oracle company) for medical writing assistance.

Conflicts of Interest

Monika Schaubeck is an employee of HiPP GmbH & Co. Vertrieb KG. Metehan Ozen; within the last 5 years, served as a consultant and keynote speaker for Sanofi Pasteur Vaccines, Pfizer Vaccines, Biogaia, HiPP, Nestlé Nutrition, Sanofi CHC, Nobel Pharma, and Abdi Ibrahim Pharma. Hugues Piloquet, within the last 5 years, has served as a consultant and keynote speaker for HiPP, Nestlé Nutrition, Danone, Sodilac, and Mead Johnson.

References

  1. Ojo-Okunola, A.; Nicol, M.; du Toit, E. Human Breast Milk Bacteriome in Health and Disease. Nutrients 2018, 10, 1643. [Google Scholar] [CrossRef] [PubMed]
  2. Meek, J.Y.; Noble, L.; Section on Breastfeeding. Policy Statement: Breastfeeding and the Use of Human Milk. Pediatrics 2022, 150, e2022057988. [Google Scholar] [CrossRef] [PubMed]
  3. Victora, C.G.; Bahl, R.; Barros, A.J.D.; França, G.V.A.; Horton, S.; Krasevec, J.; Murch, S.; Sankar, M.J.; Walker, N.; Rollins, N.C. Breastfeeding in the 21st century: Epidemiology, mechanisms, and lifelong effect. Lancet 2016, 387, 475–490. [Google Scholar] [CrossRef] [PubMed]
  4. Salone, L.R.; Vann, W.F., Jr.; Dee, D.L. Breastfeeding: An overview of oral and general health benefits. J. Am. Dent. Assoc. 2013, 144, 143–151. [Google Scholar] [CrossRef]
  5. Habicht, J.P. Expert consultation on the optimal duration of exclusive breastfeeding: The process, recommendations, and challenges for the future. Adv. Exp. Med. Biol 2004, 554, 79–87. [Google Scholar] [CrossRef]
  6. Lyons, K.E.; Ryan, C.A.; Dempsey, E.M.; Ross, R.P.; Stanton, C. Breast Milk, a Source of Beneficial Microbes and Associated Benefits for Infant Health. Nutrients 2020, 12, 1039. [Google Scholar] [CrossRef]
  7. Fernandez, L.; Langa, S.; Martin, V.; Maldonado, A.; Jimenez, E.; Martin, R.; Rodriguez, J.M. The human milk microbiota: Origin and potential roles in health and disease. Pharmacol. Res. 2013, 69, 1–10. [Google Scholar] [CrossRef]
  8. Soto, A.; Martin, V.; Jimenez, E.; Mader, I.; Rodriguez, J.M.; Fernandez, L. Lactobacilli and bifidobacteria in human breast milk: Influence of antibiotherapy and other host and clinical factors. J. Pediatr. Gastroenterol. Nutr. 2014, 59, 78–88. [Google Scholar] [CrossRef]
  9. Moossavi, S.; Sepehri, S.; Robertson, B.; Bode, L.; Goruk, S.; Field, C.J.; Lix, L.M.; de Souza, R.J.; Becker, A.B.; Mandhane, P.J.; et al. Composition and Variation of the Human Milk Microbiota Are Influenced by Maternal and Early-Life Factors. Cell Host Microbe 2019, 25, 324–335.e324. [Google Scholar] [CrossRef]
  10. Williams, J.E.; Carrothers, J.M.; Lackey, K.A.; Beatty, N.F.; Brooker, S.L.; Peterson, H.K.; Steinkamp, K.M.; York, M.A.; Shafii, B.; Price, W.J.; et al. Strong Multivariate Relations Exist Among Milk, Oral, and Fecal Microbiomes in Mother-Infant Dyads During the First Six Months Postpartum. J. Nutr. 2019, 149, 902–914. [Google Scholar] [CrossRef]
  11. Zimmermann, P.; Curtis, N. Breast milk microbiota: A review of the factors that influence composition. J. Infect. 2020, 81, 17–47. [Google Scholar] [CrossRef] [PubMed]
  12. Boix-Amoros, A.; Puente-Sanchez, F.; du Toit, E.; Linderborg, K.M.; Zhang, Y.; Yang, B.; Salminen, S.; Isolauri, E.; Tamames, J.; Mira, A.; et al. Mycobiome Profiles in Breast Milk from Healthy Women Depend on Mode of Delivery, Geographic Location, and Interaction with Bacteria. Appl. Environ. Microbiol. 2019, 85, e02994-18. [Google Scholar] [CrossRef] [PubMed]
  13. Pannaraj, P.S.; Ly, M.; Cerini, C.; Saavedra, M.; Aldrovandi, G.M.; Saboory, A.A.; Johnson, K.M.; Pride, D.T. Shared and Distinct Features of Human Milk and Infant Stool Viromes. Front. Microbiol 2018, 9, 1162. [Google Scholar] [CrossRef] [PubMed]
  14. Krumbeck, J.A.; Rasmussen, H.E.; Hutkins, R.W.; Clarke, J.; Shawron, K.; Keshavarzian, A.; Walter, J. Probiotic Bifidobacterium strains and galactooligosaccharides improve intestinal barrier function in obese adults but show no synergism when used together as synbiotics. Microbiome 2018, 6, 121. [Google Scholar] [CrossRef] [PubMed]
  15. Rodriguez-Nogales, A.; Algieri, F.; Vezza, T.; Garrido-Mesa, N.; Olivares, M.; Comalada, M.; Riccardi, C.; Utrilla, M.P.; Rodriguez-Cabezas, M.E.; Galvez, J. The viability of Lactobacillus fermentum CECT5716 is not essential to exert intestinal anti-inflammatory properties. Food Funct. 2015, 6, 1176–1184. [Google Scholar] [CrossRef]
  16. 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]
  17. Davoodabadi, A.; Soltan Dallal, M.M.; Rahimi Foroushani, A.; Douraghi, M.; Sharifi Yazdi, M.K.; Amin Harati, F. Antibacterial activity of Lactobacillus spp. isolated from the feces of healthy infants against enteropathogenic bacteria. Anaerobe 2015, 34, 53–58. [Google Scholar] [CrossRef]
  18. Yang, B.; Chen, Y.; Stanton, C.; Ross, R.P.; Lee, Y.K.; Zhao, J.; Zhang, H.; Chen, W. Bifidobacterium and Lactobacillus Composition at Species Level and Gut Microbiota Diversity in Infants before 6 Weeks. Int. J. Mol. Sci. 2019, 20, 3306. [Google Scholar] [CrossRef]
  19. Jomehzadeh, N.; Javaherizadeh, H.; Amin, M.; Saki, M.; Al-Ouqaili, M.T.S.; Hamidi, H.; Seyedmahmoudi, M.; Gorjian, Z. Isolation and identification of potential probiotic Lactobacillus species from feces of infants in southwest Iran. IJID 2020, 96, 524–530. [Google Scholar] [CrossRef]
  20. Kook, S.Y.; Chung, E.C.; Lee, Y.; Lee, D.W.; Kim, S. Isolation and characterization of five novel probiotic strains from Korean infant and children faeces. PLoS ONE 2019, 14, e0223913. [Google Scholar] [CrossRef]
  21. Martínez, I.; Stegen, J.C.; Maldonado-Gómez, M.X.; Eren, A.M.; Siba, P.M.; Greenhill, A.R.; Walter, J. The Gut Microbiota of Rural Papua New Guineans: Composition, Diversity Patterns, and Ecological Processes. Cell Rep. 2015, 11, 527–538. [Google Scholar] [CrossRef] [PubMed]
  22. Martin, R.; Langa, S.; Reviriego, C.; Jiminez, E.; Marin, M.L.; Xaus, J.; Fernandez, L.; Rodriguez, J.M. Human milk is a source of lactic acid bacteria for the infant gut. J. Pediatr. 2003, 143, 754–758. [Google Scholar] [CrossRef] [PubMed]
  23. Martin, R.; Olivares, M.; Marin, M.L.; Fernandez, L.; Xaus, J.; Rodriguez, J.M. Probiotic potential of 3 Lactobacilli strains isolated from breast milk. J. Hum. Lact. 2005, 21, 8–17; quiz 18–21, 41. [Google Scholar] [CrossRef] [PubMed]
  24. Cardenas, N.; Laino, J.E.; Delgado, S.; Jimenez, E.; Juarez del Valle, M.; Savoy de Giori, G.; Sesma, F.; Mayo, B.; Fernandez, L.; LeBlanc, J.G.; et al. Relationships between the genome and some phenotypical properties of Lactobacillus fermentum CECT 5716, a probiotic strain isolated from human milk. Appl. Microbiol. Biotechnol. 2015, 99, 4343–4353. [Google Scholar] [CrossRef]
  25. Jimenez, E.; Langa, S.; Martin, V.; Arroyo, R.; Martin, R.; Fernandez, L.; Rodriguez, J.M. Complete genome sequence of Lactobacillus fermentum CECT 5716, a probiotic strain isolated from human milk. J. Bacteriol. 2010, 192, 4800. [Google Scholar] [CrossRef]
  26. 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]
  27. Food and Agricultural Organization of the United Nations; World Health Organization. Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Live Lactic Acid Bacteria; FAO: Rome, Italy; WHO: Geneva, Switzerland, 2001. [Google Scholar]
  28. Bañuelos, O.; Fernández, L.; Corral, J.M.; Valdivieso-Ugarte, M.; Adrio, J.L.; Velasco, J. Metabolism of prebiotic products containing β(2-1) fructan mixtures by two Lactobacillus strains. Anaerobe 2008, 14, 184–189. [Google Scholar] [CrossRef]
  29. Olivares, M.; Diaz-Ropero, M.P.; Martin, R.; Rodriguez, J.M.; Xaus, J. Antimicrobial potential of four Lactobacillus strains isolated from breast milk. J. Appl. Microbiol. 2006, 101, 72–79. [Google Scholar] [CrossRef]
  30. Arroyo, R.; Martin, V.; Maldonado, A.; Jimenez, E.; Fernandez, L.; Rodriguez, J.M. Treatment of infectious mastitis during lactation: Antibiotics versus oral administration of Lactobacilli isolated from breast milk. Clin. Infect. Dis. 2010, 50, 1551–1558. [Google Scholar] [CrossRef]
  31. Robles-Vera, I.; Toral, M.; de la Visitacion, N.; Sanchez, M.; Romero, M.; Olivares, M.; Jimenez, R.; Duarte, J. The Probiotic Lactobacillus fermentum Prevents Dysbiosis and Vascular Oxidative Stress in Rats with Hypertension Induced by Chronic Nitric Oxide Blockade. Mol. Nutr. Food Res. 2018, 62, e1800298. [Google Scholar] [CrossRef]
  32. Maldonado, J.; Canabate, F.; Sempere, L.; Vela, F.; Sanchez, A.R.; Narbona, E.; Lopez-Huertas, E.; Geerlings, A.; Valero, A.D.; Olivares, M.; et al. Human Milk Probiotic Lactobacillus fermentum CECT5716 Reduces the Incidence of Gastrointestinal and Upper Respiratory Tract Infections in Infants. J. Pediatr. Gastroenterol. Nutr. 2012, 54, 55–61. [Google Scholar] [CrossRef] [PubMed]
  33. González, A.; Gálvez, N.; Martín, J.; Reyes, F.; Pérez-Victoria, I.; Dominguez-Vera, J.M. Identification of the key excreted molecule by Lactobacillus fermentum related to host iron absorption. Food Chem. 2017, 228, 374–380. [Google Scholar] [CrossRef] [PubMed]
  34. Mack, D.R.; Michail, S.; Wei, S.; McDougall, L.; Hollingsworth, M.A. Probiotics inhibit enteropathogenic E. coliadherence in vitro by inducing intestinal mucin gene expression. Am. J. Physiol. Gastrointest. 1999, 276, G941–G950. [Google Scholar] [CrossRef] [PubMed]
  35. Vanhaecke, T.; Aubert, P.; Grohard, P.A.; Durand, T.; Hulin, P.; Paul-Gilloteaux, P.; Fournier, A.; Docagne, F.; Ligneul, A.; Fressange-Mazda, C.; et al. L. fermentum CECT 5716 prevents stress-induced intestinal barrier dysfunction in newborn rats. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2017, 29, e13069. [Google Scholar] [CrossRef]
  36. Diaz-Ropero, M.P.; Martin, R.; Sierra, S.; Lara-Villoslada, F.; Rodriguez, J.M.; Xaus, J.; Olivares, M. Two Lactobacillus strains, isolated from breast milk, differently modulate the immune response. J. Appl. Microbiol. 2007, 102, 337–343. [Google Scholar] [CrossRef]
  37. Perez-Cano, F.J.; Dong, H.; Yaqoob, P. In vitro immunomodulatory activity of Lactobacillus fermentum CECT5716 and Lactobacillus salivarius CECT5713: Two probiotic strains isolated from human breast milk. Immunobiology 2010, 215, 996–1004. [Google Scholar] [CrossRef]
  38. Galazzo, G.; van Best, N.; Bervoets, L.; Dapaah, I.O.; Savelkoul, P.H.; Hornef, M.W.; Lau, S.; Hamelmann, E.; Penders, J. Development of the Microbiota and Associations With Birth Mode, Diet, and Atopic Disorders in a Longitudinal Analysis of Stool Samples, Collected From Infancy Through Early Childhood. Gastroenterology 2020, 158, 1584–1596. [Google Scholar] [CrossRef]
  39. Bazanella, M.; Maier, T.V.; Clavel, T.; Lagkouvardos, I.; Lucio, M.; Maldonado-Gomez, M.X.; Autran, C.; Walter, J.; Bode, L.; Schmitt-Kopplin, P.; et al. Randomized controlled trial on the impact of early-life intervention with bifidobacteria on the healthy infant fecal microbiota and metabolome. Am. J. Clin. Nutr. 2017, 106, 1274–1286. [Google Scholar] [CrossRef]
  40. Ackerman, D.L.; Craft, K.M.; Townsend, S.D. Infant food applications of complex carbohydrates: Structure, synthesis, and function. Carbohydr. Res. 2017, 437, 16–27. [Google Scholar] [CrossRef]
  41. Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L.; et al. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef]
  42. Rodríguez-Nogales, A.; Algieri, F.; Garrido-Mesa, J.; Vezza, T.; Utrilla, M.P.; Chueca, N.; Garcia, F.; Olivares, M.; Rodríguez-Cabezas, M.E.; Gálvez, J. Differential intestinal anti-inflammatory effects of Lactobacillus fermentum and Lactobacillus salivarius in DSS mouse colitis: Impact on microRNAs expression and microbiota composition. Mol. Nutr. Food Res. 2017, 61, 1700144. [Google Scholar] [CrossRef] [PubMed]
  43. Lemaire, M.; Dou, S.; Cahu, A.; Formal, M.; Le Normand, L.; Romé, V.; Nogret, I.; Ferret-Bernard, S.; Rhimi, M.; Cuinet, I.; et al. Addition of dairy lipids and probiotic Lactobacillus fermentum in infant formula programs gut microbiota and entero-insular axis in adult minipigs. Sci. Rep. 2018, 8, 11656. [Google Scholar] [CrossRef]
  44. Müller, T.D.; Finan, B.; Bloom, S.R.; D’Alessio, D.; Drucker, D.J.; Flatt, P.R.; Fritsche, A.; Gribble, F.; Grill, H.J.; Habener, J.F.; et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef] [PubMed]
  45. Rivero-Gutiérrez, B.; Gámez-Belmonte, R.; Suárez, M.D.; Lavín, J.L.; Aransay, A.M.; Olivares, M.; Martínez-Augustin, O.; Sánchez de Medina, F.; Zarzuelo, A. A synbiotic composed of Lactobacillus fermentum CECT5716 and FOS prevents the development of fatty acid liver and glycemic alterations in rats fed a high fructose diet associated with changes in the microbiota. Mol. Nutr. Food Res. 2017, 61, 1600622. [Google Scholar] [CrossRef] [PubMed]
  46. LeBlanc, J.G.; Laino, J.E.; del Valle, M.J.; Vannini, V.; van Sinderen, D.; Taranto, M.P.; de Valdez, G.F.; de Giori, G.S.; Sesma, F. B-group vitamin production by lactic acid bacteria--current knowledge and potential applications. J. Appl. Microbiol. 2011, 111, 1297–1309. [Google Scholar] [CrossRef] [PubMed]
  47. Asrar, F.M.; O’Connor, D.L. Bacterially synthesized folate and supplemental folic acid are absorbed across the large intestine of piglets. J. Nutr. Biochem. 2005, 16, 587–593. [Google Scholar] [CrossRef]
  48. Rossi, M.; Amaretti, A.; Raimondi, S. Folate production by probiotic bacteria. Nutrients 2011, 3, 118–134. [Google Scholar] [CrossRef] [PubMed]
  49. Krause, L.J.; Forsberg, C.W.; O’Connor, D.L. Feeding human milk to rats increases Bifidobacterium in the cecum and colon which correlates with enhanced folate status. J. Nutr. 1996, 126, 1505–1511. [Google Scholar] [CrossRef]
  50. Olivares, M.; Diaz-Ropero, M.P.; Sierra, S.; Lara-Villoslada, F.; Fonolla, J.; Navas, M.; Rodriguez, J.M.; Xaus, J. Oral intake of Lactobacillus fermentum CECT5716 enhances the effects of influenza vaccination. Nutrition 2007, 23, 254–260. [Google Scholar] [CrossRef]
  51. Addison, E.G.; North, J.; Bakhsh, I.; Marden, C.; Haq, S.; Al-Sarraj, S.; Malayeri, R.; Wickremasinghe, R.G.; Davies, J.K.; Lowdell, M.W. Ligation of CD8alpha on human natural killer cells prevents activation-induced apoptosis and enhances cytolytic activity. Immunology 2005, 116, 354–361. [Google Scholar] [CrossRef]
  52. Perez-Cano, F.J.; Castellote, C.; Gonzalez-Castro, A.M.; Pelegri, C.; Castell, M.; Franch, A. Developmental changes in intraepithelial T lymphocytes and NK cells in the small intestine of neonatal rats. Pediatr. Res. 2005, 58, 885–891. [Google Scholar] [CrossRef]
  53. Cleusix, V.; Lacroix, C.; Vollenweider, S.; Duboux, M.; Le Blay, G. Inhibitory activity spectrum of reuterin produced by Lactobacillus reuteri against intestinal bacteria. BMC Microbiol. 2007, 7, 101. [Google Scholar] [CrossRef] [PubMed]
  54. von Schillde, M.A.; Hormannsperger, G.; Weiher, M.; Alpert, C.A.; Hahne, H.; Bauerl, C.; van Huynegem, K.; Steidler, L.; Hrncir, T.; Perez-Martinez, G.; et al. Lactocepin secreted by Lactobacillus exerts anti-inflammatory effects by selectively degrading proinflammatory chemokines. Cell Host Microbe 2012, 11, 387–396. [Google Scholar] [CrossRef] [PubMed]
  55. Peran, L.; Sierra, S.; Comalada, M.; Lara-Villoslada, F.; Bailon, E.; Nieto, A.; Concha, A.; Olivares, M.; Zarzuelo, A.; Xaus, J.; et al. A comparative study of the preventative effects exerted by two probiotics, Lactobacillus reuteri and Lactobacillus fermentum, in the trinitrobenzenesulfonic acid model of rat colitis. Br. J. Nutr. 2007, 97, 96–103. [Google Scholar] [CrossRef] [PubMed]
  56. Pophaly, S.D.; Singh, R.; Pophaly, S.D.; Kaushik, J.K.; Tomar, S.K. Current status and emerging role of glutathione in food grade lactic acid bacteria. Microb. Cell Fact. 2012, 11, 114. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, G.; Fang, Y.Z.; Yang, S.; Lupton, J.R.; Turner, N.D. Glutathione metabolism and its implications for health. J. Nutr. 2004, 134, 489–492. [Google Scholar] [CrossRef]
  58. Pophaly, S.D.; Poonam, S.; Pophaly, S.D.; Kapila, S.; Nanda, D.K.; Tomar, S.K.; Singh, R. Glutathione biosynthesis and activity of dependent enzymes in food-grade lactic acid bacteria harbouring multidomain bifunctional fusion gene (gshF). J. Appl. Microbiol. 2017, 123, 194–203. [Google Scholar] [CrossRef]
  59. Surya, A.; Liu, X.; Miller, M.J. Glutathione Utilization in Lactobacillus fermentum CECT 5716. J. Agric. Food Chem. 2018, 66, 12651–12656. [Google Scholar] [CrossRef]
  60. Mikelsaar, M.; Zilmer, M. Lactobacillus fermentum ME-3—An antimicrobial and antioxidative probiotic. Microb Ecol. Health Dis 2009, 21, 1–27. [Google Scholar] [CrossRef]
  61. Peran, L.; Camuesco, D.; Comalada, M.; Nieto, A.; Concha, A.; Adrio, J.L.; Olivares, M.; Xaus, J.; Zarzuelo, A.; Galvez, J. Lactobacillus fermentum, a probiotic capable to release glutathione, prevents colonic inflammation in the TNBS model of rat colitis. Int. J. Color. Dis. 2006, 21, 737–746. [Google Scholar] [CrossRef]
  62. Roduit, C.; Frei, R.; Ferstl, R.; Loeliger, S.; Westermann, P.; Rhyner, C.; Schiavi, E.; Barcik, W.; Rodriguez-Perez, N.; Wawrzyniak, M.; et al. High levels of butyrate and propionate in early life are associated with protection against atopy. Allergy 2019, 74, 799–809. [Google Scholar] [CrossRef] [PubMed]
  63. Victora, C.G.; Christian, P.; Vidaletti, L.P.; Gatica-Domínguez, G.; Menon, P.; Black, R.E. Revisiting maternal and child undernutrition in low-income and middle-income countries: Variable progress towards an unfinished agenda. Lancet 2021, 397, 1388–1399. [Google Scholar] [CrossRef] [PubMed]
  64. Hoppe, M.; Önning, G.; Berggren, A.; Hulthén, L. Probiotic strain Lactobacillus plantarum 299v increases iron absorption from an iron-supplemented fruit drink: A double-isotope cross-over single-blind study in women of reproductive age. BJN 2015, 114, 1195–1202. [Google Scholar] [CrossRef] [PubMed]
  65. Lavermicocca, P.; Valerio, F.; Evidente, A.; Lazzaroni, S.; Corsetti, A.; Gobbetti, M. Purification and Characterization of Novel Antifungal Compounds from the Sourdough Lactobacillus plantarum Strain 21B. AEM 2000, 66, 4084–4090. [Google Scholar] [CrossRef] [PubMed]
  66. Pullakhandam, R.; Nair, M.K.; Kasula, S.; Kilari, S.; Thippande, T.G. Ferric reductase activity of low molecular weight human milk fraction is associated with enhanced iron solubility and uptake in Caco-2 cells. BBRC 2008, 374, 369–372. [Google Scholar] [CrossRef]
  67. Mosca, A.; Abreu, Y.; Abreu, A.T.; Gwee, K.A.; Ianiro, G.; Tack, J.; Nguyen, T.V.H.; Hill, C. The clinical evidence for postbiotics as microbial therapeutics. Gut Microbes 2022, 14, 2117508. [Google Scholar] [CrossRef]
  68. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef]
  69. EFSA. Statement on the update of the list of QPS-recommended biological agents intentionally added to food or feed as notified to EFSA. 2: Suitability of taxonomic units notified to EFSA until March 2015. EFSA J. 2015, 13, 4138. [Google Scholar] [CrossRef]
  70. Lara-Villoslada, F.; Sierra, S.; Diaz-Ropero, M.P.; Rodriguez, J.M.; Xaus, J.; Olivares, M. Safety assessment of Lactobacillus fermentum CECT5716, a probiotic strain isolated from human milk. J. Dairy Res. 2009, 76, 216–221. [Google Scholar] [CrossRef]
  71. Stefańska, I.; Kwiecień, E.; Jóźwiak-Piasecka, K.; Garbowska, M.; Binek, M.; Rzewuska, M. Antimicrobial Susceptibility of Lactic Acid Bacteria Strains of Potential Use as Feed Additives—The Basic Safety and Usefulness Criterion. Front. Vet. Sci. 2021, 8, 687071. [Google Scholar] [CrossRef]
  72. EFSA. Technical guidance-Update of the criteria used in the assessment of bacterial resistance to antibiotics of human or veterinary importance. EFSA J. 2008, 6, 732. [Google Scholar]
  73. Braegger, C.; Chmielewska, A.; Decsi, T.; Kolacek, S.; Mihatsch, W.; Moreno, L.; Piescik, 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]
  74. Maintz, L.; Novak, N. Histamine and histamine intolerance. AJCN 2007, 85, 1185–1196. [Google Scholar] [CrossRef] [PubMed]
  75. Pei, Z.; Sadiq, F.A.; Han, X.; Zhao, J.; Zhang, H.; Ross, R.P.; Lu, W.; Chen, W. Comprehensive Scanning of Prophages in Lactobacillus: Distribution, Diversity, Antibiotic Resistance Genes, and Linkages with CRISPR-Cas Systems. mSystems 2021, 6, e01211-20. [Google Scholar] [CrossRef] [PubMed]
  76. Gil-Campos, M.; López, M.Á.; Rodriguez-Benítez, M.V.; Romero, J.; Roncero, I.; Linares, M.D.; Maldonado, J.; López-Huertas, E.; Berwind, R.; Ritzenthaler, K.L.; et al. Lactobacillus fermentum CECT 5716 is safe and well tolerated in infants of 1–6 months of age: A Randomized Controlled Trial. Pharm. Res. 2012, 65, 231–238. [Google Scholar] [CrossRef]
  77. Maldonado-Lobón, J.A.; Gil-Campos, M.; Maldonado, J.; López-Huertas, E.; Flores-Rojas, K.; Valero, A.D.; Rodríguez-Benítez, M.V.; Bañuelos, O.; Lara-Villoslada, F.; Fonollá, J.; et al. Long-term safety of early consumption of Lactobacillus fermentum CECT5716: A 3-year follow-up of a randomized controlled trial. Pharm. Res. 2015, 95–96, 12–19. [Google Scholar] [CrossRef]
  78. Rodríguez-Sojo, M.J.; Garcia-Garcia, J.; Ruiz-Malagón, A.J.; Diez-Echave, P.; Hidalgo-García, L.; Molina-Tijeras, J.A.; González-Lozano, E.; López-Escanez, L.; Rodríguez-Cabezas, M.E.; Rodríguez-Sánchez, M.J.; et al. Beneficial Effects of Limosilactobacillus fermentum in the DCA Experimental Model of Irritable Bowel Syndrome in Rats. Nutrients 2023, 15, 24. [Google Scholar] [CrossRef]
  79. Mane, J.; Loren, V.; Pedrosa, E.; Ojanguren, I.; Xaus, J.; Cabre, E.; Domenech, E.; Gassull, M.A. Lactobacillus fermentum CECT 5716 prevents and reverts intestinal damage on TNBS-induced colitis in mice. Inflamm. Bowel Dis. 2009, 15, 1155–1163. [Google Scholar] [CrossRef]
  80. Gomez-Guzman, M.; Toral, M.; Romero, M.; Jimenez, R.; Galindo, P.; Sanchez, M.; Zarzuelo, M.J.; Olivares, M.; Galvez, J.; Duarte, J. Antihypertensive effects of probiotics Lactobacillus strains in spontaneously hypertensive rats. Mol. Nutr. Food Res. 2015, 59, 2326–2336. [Google Scholar] [CrossRef]
  81. Robles-Vera, I.; Toral, M.; de la Visitación, N.; Sánchez, M.; Gómez-Guzmán, M.; Romero, M.; Yang, T.; Izquierdo-Garcia, J.L.; Jiménez, R.; Ruiz-Cabello, J.; et al. Probiotics Prevent Dysbiosis and the Rise in Blood Pressure in Genetic Hypertension: Role of Short-Chain Fatty Acids. Mol. Nutr. Food Res. 2020, 64, 1900616. [Google Scholar] [CrossRef]
  82. Toral, M.; Romero, M.; Rodríguez-Nogales, A.; Jiménez, R.; Robles-Vera, I.; Algieri, F.; Chueca-Porcuna, N.; Sánchez, M.; de la Visitación, N.; Olivares, M.; et al. Lactobacillus fermentum Improves Tacrolimus-Induced Hypertension by Restoring Vascular Redox State and Improving eNOS Coupling. Mol. Nutr. Food Res. 2018, 62, 1800033. [Google Scholar] [CrossRef] [PubMed]
  83. de la Visitación, N.; Robles-Vera, I.; Moleón-Moya, J.; Sánchez, M.; Jiménez, R.; Gómez-Guzmán, M.; González-Correa, C.; Olivares, M.; Toral, M.; Romero, M.; et al. Probiotics Prevent Hypertension in a Murine Model of Systemic Lupus Erythematosus Induced by Toll-Like Receptor 7 Activation. Nutrients 2021, 13, 2669. [Google Scholar] [CrossRef] [PubMed]
  84. Arribas, B.; Rodríguez-Cabezas, M.E.; Comalada, M.; Bailón, E.; Camuesco, D.; Olivares, M.; Xaus, J.; Zarzuelo, A.; Gálvez, J. Evaluation of the preventative effects exerted by Lactobacillus fermentum in an experimental model of septic shock induced in mice. Br. J. Nutr. 2009, 101, 51–58. [Google Scholar] [CrossRef]
  85. Azagra-Boronat, I.; Tres, A.; Massot-Cladera, M.; Franch, À.; Castell, M.; Guardiola, F.; Pérez-Cano, F.J.; Rodríguez-Lagunas, M.J. Lactobacillus fermentum CECT5716 Supplementation in Rats during Pregnancy and Lactation Impacts Maternal and Offspring Lipid Profile, Immune System and Microbiota. Cells 2020, 9, 575. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, W.; Li, Y.; Han, G.; Li, A.; Kong, X. Lactobacillus fermentum CECT5716 Alleviates the Inflammatory Response in Asthma by Regulating TLR2/TLR4 Expression. Front. Nutr 2022, 9, 931427. [Google Scholar] [CrossRef]
  87. Barclay, A.R.; Russell, R.K.; Wilson, M.L.; Gilmour, W.H.; Satsangi, J.; Wilson, D.C. Systematic review: The role of breastfeeding in the development of pediatric inflammatory bowel disease. J. Pediatr. 2009, 155, 421–426. [Google Scholar] [CrossRef] [PubMed]
  88. Gungor, D.; Nadaud, P.; Dreibelbis, C.; LaPergola, C.C.; Wong, Y.P.; Terry, N.; Abrams, S.A.; Beker, L.; Jacobovits, T.; Jarvinen, K.M.; et al. Infant milk-feeding practices and diagnosed celiac disease and inflammatory bowel disease in offspring: A systematic review. Am. J. Clin. Nutr. 2019, 109, 838s–851s. [Google Scholar] [CrossRef]
  89. Hormannsperger, G.; Schaubeck, M.; Haller, D. Intestinal Microbiota in Animal Models of Inflammatory Diseases. Ilar J. 2015, 56, 179–191. [Google Scholar] [CrossRef] [PubMed]
  90. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
  91. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Yu, D.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D.; et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef]
  92. Lacy, B.E.; Mearin, F.; Chang, L.; Chey, W.D.; Lembo, A.J.; Simren, M.; Spiller, R. Bowel Disorders. Gastroenterology 2016, 150, 1393–1407.e1395. [Google Scholar] [CrossRef] [PubMed]
  93. Ford, A.C.; Lacy, B.E.; Talley, N.J. Irritable Bowel Syndrome. N. Engl. J. Med. 2017, 376, 2566–2578. [Google Scholar] [CrossRef] [PubMed]
  94. Black, C.J.; Ford, A.C. Global burden of irritable bowel syndrome: Trends, predictions and risk factors. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 473–486. [Google Scholar] [CrossRef]
  95. Lee, Y.J.; Park, K.S. Irritable bowel syndrome: Emerging paradigm in pathophysiology. WJG 2014, 20, 2456. [Google Scholar] [CrossRef] [PubMed]
  96. Zamani, M.; Alizadeh-Tabari, S.; Zamani, V. Systematic review with meta-analysis: The prevalence of anxiety and depression in patients with irritable bowel syndrome. Aliment. Pharm. 2019, 50, 132–143. [Google Scholar] [CrossRef] [PubMed]
  97. Dupont, C.; Barau, E.; Molkhou, P.; Raynaud, F.; Barbet, J.P.; Dehennin, L. Food-induced alterations of intestinal permeability in children with cow’s milk-sensitive enteropathy and atopic dermatitis. J. Pediatr. Gastroenterol. Nutr. 1989, 8, 459–465. [Google Scholar] [CrossRef]
  98. Niewiem, M.; Grzybowska-Chlebowczyk, U. Assessment of Selected Intestinal Permeability Markers in Children with Food Allergy Depending on the Type and Severity of Clinical Symptoms. Nutrients 2022, 14, 4385. [Google Scholar] [CrossRef]
  99. Khalesi, S.; Sun, J.; Buys, N.; Jayasinghe, R. Effect of probiotics on blood pressure: A systematic review and meta-analysis of randomized, controlled trials. Hypertension 2014, 64, 897–903. [Google Scholar] [CrossRef]
  100. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.-D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef]
  101. Freidl, R.; Garib, V.; Linhart, B.; Haberl, E.M.; Mader, I.; Szépfalusi, Z.; Schmidthaler, K.; Douladiris, N.; Pampura, A.; Varlamov, E.; et al. Extensively Hydrolyzed Hypoallergenic Infant Formula with Retained T Cell Reactivity. Nutrients 2023, 15, 111. [Google Scholar] [CrossRef]
  102. Lagkouvardos, I.; Intze, E.; Schaubeck, M.; Rooney, J.P.K.; Hecht, C.; Piloquet, H.; Clavel, T. Early life gut microbiota profiles linked to synbiotic formula effects: A randomized clinical trial in European infants. AJCN 2022, 117, 326–339. [Google Scholar] [CrossRef] [PubMed]
  103. Hurtado, J.A.; Maldonado-Lobón, J.A.; Díaz-Ropero, M.P.; Flores-Rojas, K.; Uberos, J.; Leante, J.L.; Affumicato, L.; Couce, M.L.; Garrido, J.M.; Olivares, M. Oral administration to nursing women of Lactobacillus fermentum CECT5716 prevents lactational mastitis development: A randomized controlled trial. Breastfeed. Med. 2017, 12, 202–209. [Google Scholar] [CrossRef]
  104. Maldonado-Lobón, J.A.; Díaz-López, M.A.; Carputo, R.; Duarte, P.; Díaz-Ropero, M.P.; Valero, A.D.; Sanudo, A.; Sempere, L.; Ruiz-López, M.D.; Banuelos, O. Lactobacillus fermentum CECT 5716 reduces Staphylococcus load in the breastmilk of lactating mothers suffering breast pain: A randomized controlled trial. Breastfeed. Med. 2015, 10, 425–432. [Google Scholar] [CrossRef]
  105. Zhang, Y.; Gao, Y.; He, X.; Ding, S.; Gao, H. Oral Lactobacillus fermentum CECT5716 in the patients with lactational abscess treated by needle aspiration: The late follow-up of a randomized controlled trial. Medicine 2022, 101, e29761. [Google Scholar] [CrossRef] [PubMed]
  106. Maldonado, J.; Gil-Campos, M.; Maldonado-Lobón, J.A.; Benavides, M.R.; Flores-Rojas, K.; Jaldo, R.; Jiménez del Barco, I.; Bolívar, V.; Valero, A.D.; Prados, E.; et al. Evaluation of the safety, tolerance and efficacy of 1-year consumption of infant formula supplemented with Lactobacillus fermentum CECT5716 Lc40 or Bifidobacterium breve CECT7263: A randomized controlled trial. BMC Pediatr. 2019, 19, 361. [Google Scholar] [CrossRef] [PubMed]
  107. Pastor-Villaescusa, B.; Hurtado, J.; Gil-Campos, M.; Uberos, J.; Maldonado-Lobón, J.; Díaz-Ropero, M.; Bañuelos, O.; Fonollá, J.; Olivares, M.; Group, P. Effects of Lactobacillus fermentum CECT5716 Lc40 on infant growth and health: A randomised clinical trial in nursing women. Benef. Microbes 2020, 11, 235–244. [Google Scholar] [CrossRef]
  108. Tromp, I.; Kiefte-de Jong, J.; Raat, H.; Jaddoe, V.; Franco, O.; Hofman, A.; de Jongste, J.; Moll, H. Breastfeeding and the risk of respiratory tract infections after infancy: The Generation R Study. PLoS ONE 2017, 12, e0172763. [Google Scholar] [CrossRef] [PubMed]
  109. Rouw, E.; von Gartzen, A.; Weißenborn, A. Bedeutung des Stillens für das Kind. Bundesgesundheitsblatt-Gesundh.-Gesundh. 2018, 61, 945–951. [Google Scholar] [CrossRef] [PubMed]
  110. Whyte, L.A.; Al-Araji, R.A.; McLoughlin, L.M. Guidelines for the management of acute gastroenteritis in children in Europe. Arch. Dis Child. 2015, 100, 308. [Google Scholar] [CrossRef] [PubMed]
  111. Pastor-Villaescusa, B.; Blanco-Rojo, R.; Olivares, M. Evaluation of the Effect of Limosilactobacillus fermentum CECT5716 on Gastrointestinal Infections in Infants: A Systematic Review and Meta-Analysis. Microorganisms 2021, 9, 1412. [Google Scholar] [CrossRef]
  112. Blanco-Rojo, R.; Maldonado, J.; Schaubeck, M.; Özen, M.; López-Huertas, E.; Olivares, M. Beneficial Effects of Limosilactobacillus fermentum CECT 5716 Administration to Infants Delivered by Cesarean Section. Front. Pediatr. 2022, 10, 906924. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Probiotic and postbiotic mechanisms of L. fermentum CECT5716. Probiotic effects in the intestinal lumen were seen for antimicrobial effects [23,29,30], bifidogenic effects [31,32], and nutritional support, as seen by improved iron uptake and vitamin synthesis [24,33]. Positive effects in the mucus layer and direct vicinity of epithelial cells were seen by pathogen exclusion and increased mucus production [23,29,34]. Further effects were seen for an increased epithelial barrier integrity [31,35] and support of the immune system [36,37]. Probiotic and postbiotic effects were seen in preclinical models and in human trials in infants and adults (as detailed in Section 5 and Section 6). IgA: Immunoglobulin A; IL-10: Interleukin 10; HPLA: p-hydroxyphenyllactic acid; Th1: T-helper type 1; NK-cells: Natural killer cells.
Figure 1. Probiotic and postbiotic mechanisms of L. fermentum CECT5716. Probiotic effects in the intestinal lumen were seen for antimicrobial effects [23,29,30], bifidogenic effects [31,32], and nutritional support, as seen by improved iron uptake and vitamin synthesis [24,33]. Positive effects in the mucus layer and direct vicinity of epithelial cells were seen by pathogen exclusion and increased mucus production [23,29,34]. Further effects were seen for an increased epithelial barrier integrity [31,35] and support of the immune system [36,37]. Probiotic and postbiotic effects were seen in preclinical models and in human trials in infants and adults (as detailed in Section 5 and Section 6). IgA: Immunoglobulin A; IL-10: Interleukin 10; HPLA: p-hydroxyphenyllactic acid; Th1: T-helper type 1; NK-cells: Natural killer cells.
Nutrients 15 02207 g001
Table 1. Overview of preclinical studies for L. fermentum CECT5716.
Table 1. Overview of preclinical studies for L. fermentum CECT5716.
Model Disease/ConditionAnimal ModelMain ResultsReference
IBS induced by DCARatReduction in signs of intestinal hypersensitivity, reduction in inflammation, and improvement in intestinal barrier integrityRodriguez-Sojo et al., 2022 [78]
Colitis induced by TNBSRatReduction in histological signs of inflammationPeran et al., 2006 [61], Peran et al., 2007 [55]
Colitis induced by TNBSMouseReduction in histological signs of inflammationMane et al., 2009 [79]
Colitis induced by DSSMouseReduction in histological signs of inflammationRodriguez-Nogales et al., 2017 [42]
Maternal separation, Water avoidance stressRatPrevention of stress-induced ZO-1 disorganization in epithelial cells and plasma hypercorticosteronaemia; increased exploratory behaviourVanhaecke et al., 2016 [35]
Hypertension (in SHR)RatReduction in vascular ROS generation, proinflammatory response, and systolic blood pressureGomez-Guzman et al., 2015 [80]
Hypertension (in SHR)RatPrevention of hypertension; increase in butyrate-producing bacteria; improved balance of T-helper and T-regulatory cellsRobles-Vera et al., 2020 [81]
Hypertension induced by NO blockadeRatReduction in dysbiosis, vascular oxidative stress, and inflammationRobles-Vera et al., 2018 [31]
Hypertension induced by tacrolimusMousePrevention of hypertension and endothelial dysfunction; improved balance of T-helper and T-regulatory cellsToral et al., 2018 [82]
Hypertension and systemic lupus erythematosus, induced by TLR-7 agonistMousePrevention of hypertension; reduction in autoantibodies; activation of TLR-9; reduced T-cell activationVisitación et al., 2021 [83]
Septic shock induced by LPSMouseReduction in LPS-induced changes in organ weight, TNF-α levels, and liver functionArribas et al., 2008 [84]
Immunity in pregnancy and lactationRatReduction in T-cytotoxic cells and modulation of intestinal cytokines and fatty acid profile; modulation of immunoglobulins and fatty acid profile in pupsAzagra-Boronat et al., 2020 [85]
Metabolic syndrome induced by HFDRatPrevention of liver steatosis and systemic inflammation; amelioration of dysbiosis and barrier function (synbiotic treatment with L. fermentum CECT5716 + fructooligosaccharides)Rivero-Gutiérrez et al., 2017 [45]
Overweight induced by HEDPigIncrease in SCFAs; improvement in endocrine function, e.g., GLP-1 (treatment with L. fermentum CECT5716 + plant/dairy lipids)Lemaire et al., 2018 [43]
Asthma induced by ovalbuminMouseReduction in inflammatory response and inflammatory cell infiltration in lungWang et al., 2022 [86]
DCA: deoxycholic acid; DSS: dextrane sulphate sodium; GLP-1: glucagon-like peptide 1; HED: high energy diet; HFD: high fructose diet; IBS: irritable bowel syndrome; LPS: lipopolysaccharides; NO: nitric oxide; ROS: reactive oxygen species; SCFA: short-chain fatty acids; SHR: spontaneously hypertensive rats; TLR: Toll-like receptor; TNBS: trinitrobenzenesulfonic acid; TNF-α: tumour necrosis factor alpha; ZO-1: zona occludens 1.
Table 2. Overview of clinical trials for oral administration of L. fermentum CECT5716 in adults.
Table 2. Overview of clinical trials for oral administration of L. fermentum CECT5716 in adults.
ConditionParticipantsStudy DesignMain resultsReference
Mastitis625 lactating mothersRandomised, double blinded, controlled51% reduction in the incidence rate of mastitis in the probiotic groupHurtado et al., 2017 [103]
Mastitis352 lactating mothers with mastitisRandomised, controlledLower bacterial count in HM samples of probiotic group, compared to the control groupArroyo et al., 2010 [30]
Mastitis148 lactating mothers with breast painRandomised, double blinded, controlledSignificant decrease in bacterial load in HM samples of the probiotic groupMaldonado-Lobon et al., 2015 [104]
Mastitis101 lactating mothers with breast abscessMulticentre, randomised, double blinded, controlledSignificant decrease in the rate of stopping breastfeeding due to recurrence of mastitisZhang et al., 2022 [105]
Influenza vaccination50 healthy adults
(31 male, 19 female)
Randomised, double blinded, placebo controlledSignificantly lower incidence of influenza-like illness; significant increase in antigen-specific IgA in the probiotic groupOlivares et al., 2007 [50]
IgA: Immunoglobulin A, HM: Human milk.
Table 3. Overview of clinical trials for oral administration of L. fermentum CECT5716 in infants.
Table 3. Overview of clinical trials for oral administration of L. fermentum CECT5716 in infants.
ConditionParticipantsStudy DesignMain ResultsReference
Gastrointestinal and upper respiratory tract infections215 infantsRandomised, double blinded, controlledSignificant reduction in the incidence rate of gastrointestinal and upper respiratory tract infections in the probiotic groupMaldonado et al., 2012 [32]
Gastrointestinal infection and safety137 infantsRandomised, double blinded, controlledSignificant reduction in gastrointestinal infection;
normal growth and weight gain; normal consumption of formula; no symptoms relating to formula
Gil-Campos et al., 2012 [76]
Modulation of infant microbiota540 infantsRandomised, double blinded, controlled, multicentreSignificant effects on microbiota states: phylogenetic profiles of the infants receiving synbiotic infant formula were closer to reference profiles of those fed with HMLagkouvardos et al., 2022 [102]
Infection and safety236 infantsRandomised, double blinded, controlled44% lower incidence of diarrhoea and 2.5 days reduction in duration of diarrhoea;
lower incidence of respiratory tract infections among infants born by caesarean section;
normal growth
Maldonado et al., 2019 [106]
Long-term safety110 infants3-year follow-up studyNo significant difference in growth and incidence of infectious and non-infectious gastrointestinal diseasesMaldonado-Lobon et al., 2015 [77]
Infection and growth625 mother–infant pairsRandomised, double blinded, placebo controlledLower incidence of conjunctivitis in infants in the probiotic group;
higher weight of infants in probiotic group
Pastor-Villaescusa et al., 2020 [107]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ozen, M.; Piloquet, H.; Schaubeck, M. Limosilactobacillus fermentum CECT5716: Clinical Potential of a Probiotic Strain Isolated from Human Milk. Nutrients 2023, 15, 2207. https://doi.org/10.3390/nu15092207

AMA Style

Ozen M, Piloquet H, Schaubeck M. Limosilactobacillus fermentum CECT5716: Clinical Potential of a Probiotic Strain Isolated from Human Milk. Nutrients. 2023; 15(9):2207. https://doi.org/10.3390/nu15092207

Chicago/Turabian Style

Ozen, Metehan, Hugues Piloquet, and Monika Schaubeck. 2023. "Limosilactobacillus fermentum CECT5716: Clinical Potential of a Probiotic Strain Isolated from Human Milk" Nutrients 15, no. 9: 2207. https://doi.org/10.3390/nu15092207

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