Open access peer-reviewed chapter

Antimicrobial Effects of Probiotics and Novel Probiotic-Based Approaches for Infectious Diseases

Written By

Ping Li and Qing Gu

Submitted: 14 July 2017 Reviewed: 29 November 2017 Published: 04 July 2018

DOI: 10.5772/intechopen.72804

From the Edited Volume

Probiotics - Current Knowledge and Future Prospects

Edited by Shymaa Enany

Chapter metrics overview

1,696 Chapter Downloads

View Full Metrics

Abstract

Probiotics are live microorganisms, which confer health benefits on host when administered in adequate amounts. Probiotics exert their beneficial effects by maintenance flora healthy, enhancement of mucosal barrier integrity and modulation of immune responses. Antimicrobial substances including bacteriocins, hydrogen peroxide, organic acids, and short-chain fatty acids (SCFAs) produced by probiotics allow them to inhibit mucosal and epithelial adherence of pathogens and compete for limiting resources, thus suppress the growth of bacterial and fungal pathogens. Probiotics effect the colonization of fungal pathogen Candida to host surfaces, suppress Candida growth and biofilm development in vitro. Clinical results have shown that some probiotics can reduce oral, vaginal, and enteric colonization of Candida, alleviate clinical signs and symptoms, and potentially reduce the incidence of invasive fungal infection. Therefore, probiotics may be potential antifungals for prevention and treatment of candidiasis.

Keywords

  • probiotics
  • mechanism of action
  • antimicrobial activity
  • candidiasis
  • safety

1. Introduction

Probiotics are “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host,” which was defined by the Food and Drug Organization of the United Nations (FAO) and World Health Organization (WHO) [1, 2, 3]. Probiotics should have some fundamental characteristics, such as human origin, nonpathogenic in nature, resistance to destruction by technical processing, acid and bile tolerances, adequate adherence and colonization on epithelial surfaces, antagonistic activity against pathogens, regulation of immune response, and influence human metabolic activities [4, 5, 6, 7].

Bacteria belonging to the genera Lactobacillus and Bifidobacterium are the most frequently used probiotics. Besides, Enterococcus, Streptococcus, Saccharomyces, and Bacillus are also commonly used probiotics (representative species are listed in Table 1). The administration of probiotics has been confirmed as an alternative biological approach to combat bacterial and fungal pathogens in the oral cavity, GI tract, and urogenital system [4, 5, 7, 8, 9, 10, 11, 12, 13, 14]. It has been reported that probiotics could reduce Candida, which cause fungal infections in different organ systems of the human body and prevent bacterial infectious diseases [9, 10, 15]. Probiotics were capable of preventing cancers [16], modulating blood pressure [17, 18], and repressing cholesterol levels [19]. Recently, species of Akkermansia muciniphila, Eubacterium hallii, and Faecalibacterium prausnitzii are identified as new potential probiotics because of their great benefits to the microbial metabolic networks and human health, especially the effects on correcting the imbalance of gut microbiota composition [7, 20, 21, 22]. A combination of probiotics with traditional treatment has been thought to be a potential approach for treatment of certain diseases.

GeneraSpecies
LactobacillusLactobacillus rhamnosus, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus acidophilus, Lactobacillus reuteri, Lactobacillus paracasei, Lactobacillus sporogenes, Lactobacillus lactis, Lactobacillus helveticus, and Lactobacillus fermentium
LactococcusLactococcus lactis, Lactococcus lactis subsp. lactis, Lactococcus lactis subsp. diacetylactis, and Lactococcus Lactis subsp. cremoris
BifidobacteriumBifidobacterium longum, Bifidobacterium bifidum, Bifidobacterium bifidus, and Bifidobacterium lactis
EnterococcusEnterococcus faecalis and Enterococcus faecium
SaccharomycesSaccharomyces cerevisiae and Saccharomyces boulardii
StreptococcusStreptococcus thermophiles
BacillusBacillus coagulans and Bacillus subtilis
OthersAkkermansia muciniphila, Eubacterium hallii, and Faecalibacterium prausnitzii

Table 1.

Representative microbe commonly considered as probiotics.

It is noteworthy that health benefits of probiotic bacteria are strain specific, which cannot be generalized to other strains, not even the same species, although some properties may be common for different strains because of the similarities in the metabolism of ecological functionality [5, 6]. Thus, the selection of certain probiotics for therapeutic purposes should be targeted for specific pathogens. Probiotics effects are dose specific [5, 6]. It has been suggested that a daily intake of 106–109 colony-forming units (CFUs) of probiotic microorganisms is the minimum effective dose for therapeutic purposes [5, 6, 8].

A number of probiotics are currently commercially available, and they have been categorized into single-strain or multi-strain/multispecies products [7, 23, 24]. Multi-strain/multispecies probiotics exhibited better effects than single-strain probiotics. The multispecies probiotic consortium VSL#3 (Streptococcus thermophilus, Eubacterium faecium, Bifidobacterium breve, Bifidobacterium infantis, Bifidobacterium longum, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus casei, and Lactobacillus delbrueckii subsp. bulgaricus) was proven more effective than single-strain probiotics for the treatment of ulcerative colitis [23]. The multispecies probiotic consortium, Ecologic AAD (Bifidobacterium bifidum W23, Bifidobacterium lactis W18, Bifidobacterium longum W51, Enterococcus faecium W54, Lactobacillus acidophilus W37 and W55, Lactobacillus paracasei W72, Lactobacillus plantarum W62, Lactobacillus rhamnosus W71, and Lactobacillus salivarius W24), combined with amoxicillin, could reduce diarrhea-like bowel movements, while the single strain could not [25]. Thus, the combination-specific probiotic effects from diverse strains can lead to synergistic effects.

Among the most frequently used probiotics, the genera Lactobacillus, Bifidobacterium, Lactococcus, and Saccharomyces have been included in the category of “generally regarded as safe” (GRAS) [4, 6]; however, other probiotic organisms such as Enterococcus, Bacillus, and Streptococcus are not generally regarded as safe. Since probiotics have been applied in food production, disease treatment, and others, it is important to undergo safety evaluation of probiotics before human consumption.

In this chapter, we briefly review the mechanisms of action of probiotics, the safety concern of probiotics, and their potentials for prevention and treatment of diseases. Here, we discuss the application of probiotics in the fungal Candida-infected and invasion candidiasis.

Advertisement

2. Probiotics mechanism of action

Probiotics mechanism of action is with important differences among different species and strain, examples are listed in Table 2.

Mechanism of actionProbioticsStudy outcomesReferences
Maintenance flora healthy by reduction the growth and colonization of pathogensL. rhamnosus GG, L. casei Shirota, L. reuteri SD2112 and L. brevis CD2L. rhamnosus GG showed the strongest inhibitory activity in fructose and glucose medium against C. albicans, followed by L. casei Shirota, L. reuteri SD2112 and L. brevis CD2[26]
L. plantarum, commercial preparation LactoLevure®Increased survival of mice infected by multidrug resistant P. aeruginosa and E. coli[27]
B. breve, L. casei (randomized controlled trial, RCT)Levels of beneficial organic acids significantly increased in the gut, and the incidences of infectious (pneumonia and bacteremia) complications were significantly lower in the probiotic group[32]
Synbiotic (Lactobacillus, Bifidobacterium, and galactooligosaccharides) for 8 weeks (RCT)Acetic acid concentration significantly increased (100 times), pH value decreased, Gram-negative rod (1/10) in the gut decreased, and P. aeruginosa decreased in the probiotic group[33]
Multi-strain synbiotic for 7 days (RCT)Synbiotic group had lower pathogenic bacteria (43% versus 75%) and multiple organisms (39% versus 75%) in nasogastric aspirates than controls[34]
B. lactis Bb12 for 7–21 days (RCT)Probiotic group had great higher counts of Bifidobacterium (P = 0.001) and lower counts of Enterobacteriaceae (P = 0.015) and Clostridium spp. (P = 0.014) than in placebo group[35]
L. casei subsp. rhamnosus for 6 weeks (RCT)Colonization of Candida in gut was reduced in probiotic group (P = 0.01)[28]
Enhancement of mucosal barrier integrityL. plantarum 299v for 8 days (RCT)Bacterial translocation in mesenteric lymph nodes and liver was reduced to 0 and 12%, respectively[29]
Microencapsulated BifidobacteriaBacterial translocation to mesenteric lymph nodes was reduced by encapsulated Bifidobacteria (P < 0.05)[30]
VSL#3 (RCT)Decreased incidence of bacterial translocation in VSL#3 group than in water group (8% versus 50%; P = 0.03)[31]
Immune modulationVSL#3 (Lactobacillus, Bifidobacterium, and S. thermophilus) for 7 days (RCT)Reduced acute physiology and chronic health evaluation II score; reduced sequential organ failure assessment, IL-6, procalcitonin, and protein[36]
L. plantarum 299v (RCT)Late attenuating effect (after 15 days), serum IL-6 levels reduced[37]

Table 2.

Mechanism of action of probiotics.

2.1. Maintenance flora healthy by reduction the growth and colonization of pathogens

The ability of probiotics to establish in the gastrointestinal (GI) tract, maintain flora healthy, and reduce the growth of pathogens and colonization is enhanced by their ability to eliminate competitors. Probiotic strains release different antimicrobial molecules such as organic acids, hydrogen peroxide (Н2О2), and antimicrobial peptide bacteriocins into the intestinal environment to limit the growth of bacterial and fungal pathogens [6, 39, 40, 41, 42, 43].

Lactic acid and acetic acid are the main metabolites formed by lactic acid bacteria (LAB). Both lactic acid and acetic acid could result in acidity environment and thus inhibit the growth of various microorganisms. Acetic acid has a broader spectrum of antimicrobial activity when compared to lactic acid. Moreover, it is known that a synergistic effect exists between the two acids: mixtures of acetic and lactic acids suppress the growth of the pathogenic enteric bacterium Salmonella typhimurium [44].

LAB can also produce Н2О2, the antimicrobial activity of which is linked to the strong oxidizing effect. Hydrogen peroxide showed a bactericidal effect on most pathogens when in combination with lactoperoxidase-thiocyanate milk system [45]. L. johnsonii NCC933 and L. gasseri KS120.1 killed enteric uropathogenic and vaginosis-associated pathogens due to the production of lactic acid and hydrogen peroxide [46].

Bacteriocins are ribosomally synthesized antimicrobial peptides, which have broad spectrum of inhibitory effect against Gram-positive and Gram-negative bacteria, viruses, and fungi [47, 48, 49, 50]. L. plantarum 2.9, a bacteriocinogenic strain, inhibited a set of foodborne pathogens including B. cereus, E. coli O157:H7, and S. enterica [51]. Bacteriocin-producing strains identified in our lab, e.g., L. plantarum ZJ316, L. plantarum LZ95, L. plantarum ZJ008, and L. plantarum ZJ005, showed antimicrobial activity against various pathogens in vitro such as S. aureus, E. coli, S. enterica, L. monocytogenes, and C. albicans [42, 52, 53, 54].

2.2. Enhancement of mucosal barrier integrity

Probiotics have been shown to improve barrier function and the mechanisms of barrier function including alteration of tight junction protein expression and/or localization, induction of mucus secretion, increased production of cytoprotective molecules such as heat-shock proteins, inhibition of apoptosis of epithelial cells, and promoting cell survival [29, 55, 56]. They compete with pathogens and prevent their invasion through the epithelium by the ability of adherence to the intestinal epithelium and mucus. L. plantarum has been shown to enhance mucosal barrier by adhering to the mucosal membrane and reducing Gram-negative bacteria [29]. Probiotics also compete for limiting resources, thus suppressing the growth of bacterial and fungal pathogens. The probiotic E. coli Nissle 1917 is able to effectively take up multiple limited environmental irons and simultaneously competitively inhibit the growth of other intestinal microbes and pathogens [57].

Furthermore, butyrate, a short-chain fatty acid (SCFA), could reduce bacterial translocation, improve the organization of tight junctions, modulate intestinal motility in addition to being an energy source for colonocytes, and maintain the integrity of the intestinal epithelium [29, 30, 31, 58, 59, 60]. E. hallii is an important anaerobic butyrate producer resident in our gut, which influences the intestinal metabolic balance and enhances the host-gut microbiota homeostasis [61]. Thus, the administration of probiotics with butyrate-producing bacteria, in particular, could be an effective way to achieve health benefits.

2.3. Immune modulation

Probiotics are reported to enhance phagocytic activity of granulocytes and cytokine excretion in lymphocytes, increase immunoglobulin-secreting cells, and attenuate inflammasome activation. They are able to affect cells involved in immune responses, including epithelial cells, dendritic cells (DCs), T cells, regulatory T (Treg) cells, monocytes/macrophages, immunoglobulin A (IgA)-producing B cells, and natural killer cells [62, 63].

Probiotic bacteria have an effect on intestinal DCs, which have the ability to recognize and respond to different bacteria by linking the innate immune system to the adaptive immune response and to develop T- and B-cell responses. Badia et al. found that the immunomodulatory role of S. boulardii in the DCs prior to infection was related to the upregulation of tumor necrosis factor alpha (TNFα) and C─C chemokine receptor type 7 mRNAs, which might make the DCs more effective in antagonizing bacteria [64, 65]. Smith et al. reported that S. boulardii stimulated the production of cytokines TNFα, IL-1, IL-12, IL-6, and IL-10 in DCs and also induced high levels of costimulatory molecules CD80 and CD86, thus modulated the immune system and led to an efficient clearing of enteropathogenic bacteria from the blood stream coupled with a faster cytokine response [65, 66].

Probiotics also influence intestinal epithelial cells through interaction with Toll-like receptors (TLRs) and downregulate the expression of NF-κB and proinflammatory cytokines [67, 68]. This effect is supported by the following studies: the supernatant of probiotic Faecalibacterium prausnitzii inhibited the NF-κB pathway in vitro and in vivo and showed protective effects in different models such as dinitrobenzene sulfate (DNBS)-induced colitis model and dextran sodium sulfate (DSS)-induced colitis [69]; the probiotic strain L. rhamnosus GG prevented cytokine-induced apoptosis in intestinal epithelial cells [70]; and L. rhamnosus GR-1 reduced the adhesion of E. coli by promoting TLR2 and NOD1 synergism and attenuating ASC-independent NLRP3 inflammasome activation [71].

Advertisement

3. Probiotic as antifungals for prevention and treatment of candidiasis

Candida is an opportunistic pathogen, causing mucosal infections including infections in the oral cavity, oropharynx, esophagus, and vagina, and potentially life-threatening systemic candidiasis. Candida albicans is the most common fungal pathogen in humans responsible for causing superficial as well as deep invasive candidiasis, which are essentially caused by Candida biofilms attached to body surfaces. Other Candida species such as Candida tropicalis, Candida guilliermondii, Candida krusei, and Candida glabrata are less frequently isolated in healthy and diseased humans [72, 73, 74]. Probiotics are known to reduce Candida infection in different organs and are generally considered to be beneficial for overall health. They appear to assist the host combat the pathogen by suppressing filamentation formation and reducing biofilm development, the mechanism of which may be related to expression of genes associated with biofilm formation and filamentation in Candida species. In vitro and in vivo studies have demonstrated the role of probiotics in the prevention of Candida colonization and invasive candidiasis [38, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86].

3.1. In vitro evidences: probiotics in prevention/treatment of Candida infections

Several in vitro studies have addressed the antifungal effects of probiotics against Candida isolated from the human oral cavity, GI tract, and genitourinary tract [77, 78, 79, 80, 81, 86, 87]. The probiotics that have been investigated against Candida species include Lactobacillus (e.g., L. rhamnosus, L. plantarum, L. fermentum, L. acidophilus, L. paracasei, L. johnsonii, and L. salivarius), Bifidobacterium (e.g., B. bifidum and B. infantis), Saccharomyces (e.g., S. boulardii), and Streptococcus (e.g., S. thermophilus). Table 3 shows candidacidal activity of probiotic strains in different studies. C. albicans appears to be more susceptible to the antifungal effect of Lactobacillus than C. pseudotropicalis [81], and the probiotics exhibited growth inhibitory activities against C. glabrata, C. krusei, and C. parapsilosis [79, 87].

ProbioticsTarget pathogenStudy outcomeReferences
14 strains:
L. fermentum, L. rhamnosus, L. plantarum, and L. acidophilus
C. albicans and C. pseudotropicalisAll probiotics inhibited the growth of C. albicans by H2O2 production and alternative mechanism[81]
S. boulardiiC. albicans SC5314S. boulardii inhibited the affecting hyphae formation, Candida adhesion, and biofilm formation by capric acid production[87]
L. paracasei IMC 502C. glabrata, C. krusei, C. parapsilosis, and C. tropicalisHigh activity toward Candida strains except C. glabrata and C. tropicalis[79]
L. plantarum ATCC 8014 and L. johnsonii enriched or not with SeNPsC. albicans ATCC 14053Strong inhibition of C. albicans by supernatant of selenium-enriched Lactobacillus spp.[86]
L. acidophilus, L. rhamnosus, L. salivarius, B. bifidum, S. thermophiles, and B. infantisC. albicans 10341Significant inhibitory effect on biofilm formation and reduce viability of Candida[80]
L. rhamnosus GR-1 and L. reuteri RC-14C. albicans SC5314Visible inhibition zones of fungal C. albicans by probiotic treatment; low pH environment caused by lactic acid and the H2O2 production may be anti-Candida factors[77]
L. acidophilus ATCC 4356C. albicans ATCC 18804Reduce growth of C. albicans cells by 45.1%[78]
L. casei subsp. rhamnosusCandida spp.80 preterm neonates with a very low birth weight: probiotic reduced incidence and intensity of enteric colonization by Candida spp. (RCT)[28]
L. rhamnosus GG, L. rhamnosus LC705, P. freudenreichii subsp. shermanii JSCandida spp.276 elderly people: probiotic intervention reduced the risk of high yeast counts by 75% and the prevalence of hyposalivation (RCT)[76]
L. rhamnosus GR-1 and L. reuteri RC-14Candida spp.55 women: probiotics significant reduced vaginal discharge, itching, and/or burning vaginal feeling, dyspareunia, and/or dysuria, and reduced the presence of Candida spp. (RCT)[82]
L. acidophilus, L. rhamnosus, B. longum, B. bifidum, S. boulardii, and S. thermophilusCandida spp.150 children (aged 3 month to 12 year) on broad-spectrum antibiotics for at least 48 h: probiotic therapy avoided a significant increase in the number of patients colonized by Candida spp., significantly reduced the presence of Candida in the urine (RCT)[83]
L. bulgaricus, B. longum, and S. thermophilusCandida spp.65 patients with Candida-associated stomatitis: detection rate of Candida spp. was reduced in the probiotic group; significant relief of clinical signs and symptoms after probiotic administration (RCT)[84]
L. acidophilus, B. lactis, B. longum, and B. bifidumCandida spp.112 preterm neonates (gestational age < 37 wk and birth weight < 2500 g): probiotics may reduce enteral fungal colonization and invasive fungal sepsis in low-birth-weight neonates (RCT)[75]
L. reuteri DSM 17938 and L. reuteri ATCC PTA 5289Candida spp.215 elderly people (aged 60–102 y): significant reduction of Candida cells in saliva and plaque (RCT)[85]

Table 3.

Probiotics in prevention/treatment of Candida infections.

However, the mechanisms involved in antifungal activity of probiotics against Candida remain unclarified. Strus et al. found that Lactobacillus strains could inhibit the growth of C. albicans to a certain degree and their anticandidal activity related to H2O2 production [81]. Murzyn et al. reported that S. boulardii was able to secrete active compounds, mainly capric acid, reduced the expression of hwp1, ino1, and csh1 genes that encode virulence factors in C. albicans SC5314 cells, and inhibited filamentation of C. albicans and its mycelial development [87]. Therefore, it is likely that the antimicrobial molecules, organic acids, and Н2О2 produced by probiotic are major factors to limit growth of fungal pathogen Candida. This idea was supported by the research of Köhler et al. They demonstrated that low pH environment caused by lactic acid and the H2O2 production of L. rhamnosus GR-1 and L. reuteri RC-14 strains played important role in their inhibition activity to C. albicans SC5314. Moreover, L. rhamnosus GR-1 and L. reuteri RC-14 inhibited genes associated with C. albicans biofilm formation [87]. This result, together with the findings in Murzyn et al. study, shed light on a novel approach for uncovering the molecular mechanisms of the probiotic effect by using gene expression and related technology.

3.2. In vivo evidences: probiotics in prevention/treatment of Candida infections

In vivo studies, especially RCTs, have also been performed to substantiate the antifungal activity of probiotics in humans. These studies mostly focus on the sites of oral cavity, GI tract, and urogenital tract, which are susceptible to Candida infections (Table 3).

The elderly are a group particularly susceptible to oral candidiasis, because of frequent usage of dentures, hyposalivation, and their weakened immune status. Researches by Hatakka et al. and Kraft-Bodi et al. have shown that the daily consumption of food with L. reuteri DSM17938, L. reuteri ATCC PTA 5289, and L. rhamnosus GG ATCC 53103 significantly reduced the high yeast counts in saliva and biofilms in the elderly [76, 85]. The removal of biofilms by the use of probiotics that reduce the oral burden of Candida could play a major role in preventing oral candidiasis in denture wearers.

For the urogenital tract, chronic vulvovaginal candidiasis (VVC) is the most common candidiasis disease and impacts the life quality of thousands of women around the world. Researches on the effect of probiotics in the treatment and prophylaxis of VVC have been performed [82]. Martinez et al., in an RCT involving 55 women, demonstrated that the administration of L. rhamnosusGR-1 and L. reuteri RC-14 significantly reduced the presence of Candida and therefore reduced the vaginal discharge, itching, and/or burning vaginal feeling, dyspareunia, and/or dysuria [82].

For the GI tract, Candida species are common inhabitants of GI tract. Dysbiosis of GI tract may lead to candidal overgrowth and possible invasive infections, especially in infants. Hence, immunocompromised children, especially preterm neonates with low birth weight, have been the target population of a large number of studies to evaluate the prevention or/and treatment potentials of probiotics to Candida infections [28, 75, 83]. Manzoni et al., in an RCT involving 80 very low birth weight (VLBW) neonates, demonstrated that orally administered L. casei subsp. rhamnosus significantly reduced incidence and intensity of enteric colonization by Candida [28]. Another RCT, by Roy et al., found L. acidophilus, B. lactis, B. longum, and B. bifidum reduced enteral fungal colonization and invasive fungal sepsis in 112 preterm neonates (gestational age < 37 wk and birth weight < 2500 g) [75].

Together, both the laboratory studies and clinical studies showed that probiotics could prevent Candida colonization by inhibiting adhesion, filamentation, and biofilm formation, and therefore supplementation of probiotics could be a potential approach for reducing Candida colonization and invasive candidiasis.

Advertisement

4. Safety of probiotics

Although most commercially available probiotic strains are generally regarded as safe and none of the clinical studies mentioned above were reported to have adverse effects directly related to probiotics, there are some concerns regarding the safety of probiotics, including potential of bacteremia and/or endocarditis occurrence, toxicity to the gastrointestinal tract, and transfer of antibiotic resistance [4].

4.1. Potential of bacteremia and/or endocarditis occurrence

Lactic acid bacteria, including Bifidobacterium, have been reported to cause bacteremia as well as endocarditis [88, 89, 90, 91, 92]. Cannon et al. described that L. rhamnosus caused liver abscess, lactobacillemia, and infective endocarditis in a few case studies, and also the occurrence of Lactobacillus sepsis was directly linked with the ingestion of probiotic supplements, especially among immunocompromised patients and those with endocarditis [89]. Kunz et al. found two premature infants with short gut syndrome developed Lactobacillus bacteremia while taking Lactobacillus GG supplements. However, the risk of infection due to Lactobacilli is extremely rare. Statistic data from surveillance in Finland suggest that there was no increase in Lactobacillus bacteremia during 1990–2000, and Lactobacilli were isolated in 0.02% of all blood cultures [93].

4.2. Toxicity to the gastrointestinal tract

The role of probiotics on gastrointestinal physiology suggests a theoretical possibility that the production of metabolites might be undesirable and also might lead to malabsorption due to deconjugation of bile salts. These might increase the risk of colon cancer; however, there is no epidemiologic or clinical evidence to support this hypothesis [94, 95].

4.3. Transfer of antibiotic resistance

Another major safety concern of theoretical importance is genetic transfer of antibiotic resistance from probiotic strains to pathogenic cells in the gastrointestinal tract [96, 97]. Plasmids with antibiotic-resistance genes, including genes encoding resistance to tetracycline, erythromycin, chloramphenicol, and macrolide-lincosamide-streptogramin, have been found in L. plantarum, L. fermentum, L. acidophilus, and L. reuteri strains. L. plantarum 5057 exhibited tetracycline resistance, and L. lactis was with streptomycin, tetracycline, and chloramphenicol resistances [98, 99, 100]. Although the transfer of native Lactobacillus plasmids is quite rare, there are some cases, e.g., the antibiotic-resistance plasmids from Lactococcus species could transfer to Leuconostoc species and Pediococcus species.

With respect to the potential risks of probiotics, it is important to conduct population-based surveillance for safety concern.

Advertisement

5. Conclusions

Probiotics have the ability to restore the imbalance of intestinal microbiota and could act as both prophylactic and adjunctive therapy against candidiasis. Antifungal effect of probiotics is likely due to their interference with Candida biofilm development and hyphal differentiation. Safety may be of concern in application, as probiotic strains may, although quite rarely, cause bacteremia, fungemia, and sepsis. Well-designed RCTs are required to address these issues before the routine use of probiotics is recommended.

Advertisement

Acknowledgments

This project was funded by the National Key Research and Development Program of China (2016YFD0400400), the National Science Foundation of China (31601449), the International Science and Technology Cooperation Program of China (2013DFA32330), the Natural Science Foundation of Zhejiang Province (LY16C200002), and the Food Science and Engineering—the most important discipline of Zhejiang Province (2017SIAR202).

References

  1. 1. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, 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. Nature Reviews Gastroenterology and Hepatology. 2014;11:506-514. DOI: 10.1038/nrgastro.2014.66
  2. 2. Guarner F, Schaafsma GJ. Probiotics. International Journal of Food Microbiology. 1998;39:237-238
  3. 3. Calatayud GA, Suarez JE. A new contribution to the history of probiotics. Beneficial Microbes. 2017;8:323-325. DOI: 10.3920/BM2017.x002
  4. 4. Snydman DR. The safety of probiotics. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2008;46(Suppl 2):S104-S111. discussion S144-151. DOI: 10.1086/523331
  5. 5. Sanders ME. Clinical use of probiotics: What physicians need to know. American Family Physician. 2008;78:1026
  6. 6. Kligler B, Cohrssen A. Probiotics. American Family Physician. 2008;78:1073-1078
  7. 7. El Hage R, Hernandez-Sanabria E, Van de Wiele T. Emerging trends in “smart probiotics”: Functional consideration for the development of novel health and industrial applications. Frontiers in Microbiology. 2017;8:1889. DOI: 10.3389/fmicb.2017.01889
  8. 8. Saarela M, Mogensen G, Fonden R, Matto J, Mattila-Sandholm T. Probiotic bacteria: Safety, functional and technological properties. Journal of Biotechnology. 2000;84:197-215
  9. 9. Hu HJ, Zhang GQ, Zhang Q, Shakya S, Li ZY. Probiotics prevent Candida colonization and invasive fungal sepsis in preterm neonates: A systematic review and meta-analysis of randomized controlled trials. Pediatrics and Neonatology. 2017;58:103-110. DOI: 10.1016/j.pedneo.2016.06.001
  10. 10. Sinclair A, Xie X, Saab L, Dendukuri N. Lactobacillus probiotics in the prevention of diarrhea associated with Clostridium difficile: A systematic review and Bayesian hierarchical meta-analysis. CMAJ Open. 2016;4:E706-E718. DOI: 10.9778/cmajo.20160087
  11. 11. Lievin-Le Moal V. A gastrointestinal anti-infectious biotherapeutic agent: The heat-treated Lactobacillus LB. Therapeutic Advances in Gastroenterology. 2016;9:57-75. DOI: 10.1177/1756283X15602831
  12. 12. DiRienzo DB. Effect of probiotics on biomarkers of cardiovascular disease: Implications for heart-healthy diets. Nutrition Reviews. 2014;72:18-29. DOI: 10.1111/nure.12084
  13. 13. Rodes L, Khan A, Paul A, Coussa-Charley M, Marinescu D, Tomaro-Duchesneau C, et al. Effect of probiotics Lactobacillus and Bifidobacterium on gut-derived lipopolysaccharides and inflammatory cytokines: An in vitro study using a human colonic microbiota model. Journal of Microbiology and Biotechnology. 2013;23:518-526
  14. 14. Messaoudi S, Manai M, Kergourlay G, Prevost H, Connil N, Chobert JM, et al. Lactobacillus salivarius: Bacteriocin and probiotic activity. Food Microbiology. 2013;36:296-304. DOI: 10.1016/j.fm.2013.05.010
  15. 15. Johnston BC, Goldenberg JZ, Parkin PC. Probiotics and the prevention of antibiotic-associated diarrhea in infants and children. JAMA. 2016;316:1484-1485. DOI: 10.1001/jama.2016.11838
  16. 16. Shida K, Nomoto K. Probiotics as efficient immunopotentiators: Translational role in cancer prevention. The Indian Journal of Medical Research. 2013;138:808-814
  17. 17. Ivey KL, Hodgson JM, Kerr DA, Thompson PL, Stojceski B, Prince RL. The effect of yoghurt and its probiotics on blood pressure and serum lipid profile; a randomised controlled trial. Nutrition, Metabolism, and Cardiovascular Diseases. 2015;25:46-51. DOI: 10.1016/j.numecd.2014.07.012
  18. 18. 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. DOI: 10.1161/HYPERTENSIONAHA.114.03469
  19. 19. Yang Y, Xia Y, Chen H, Hong L, Feng J, Yang J, et al. The effect of perioperative probiotics treatment for colorectal cancer: Short-term outcomes of a randomized controlled trial. Oncotarget. 2016;7:8432-8440. DOI: 10.18632/oncotarget.7045
  20. 20. Lopez-Siles M, Khan TM, Duncan SH, Harmsen HJ, Garcia-Gil LJ, Flint HJ. Cultured representatives of two major phylogroups of human colonic Faecalibacterium prausnitzii can utilize pectin, uronic acids, and host-derived substrates for growth. Applied and Environmental Microbiology. 2012;78:420-428. DOI: 10.1128/AEM.06858-11
  21. 21. Bunesova V, Lacroix C, Schwab C. Mucin cross-feeding of infant Bifidobacteria and Eubacterium hallii. Microbial Ecology. 2017. DOI: 10.1007/s00248-017-1037-4
  22. 22. Belzer C, Chia LW, Aalvink S, Chamlagain B, Piironen V, Knol J, et al. Microbial metabolic networks at the mucus layer lead to diet-independent butyrate and vitamin B12 production by intestinal symbionts. mBio. 2017;8(5):e00770-17. DOI: 10.1128/mBio.00770-17
  23. 23. Timmerman HM, Koning CJ, Mulder L, Rombouts FM, Beynen AC. Monostrain, multistrain and multispecies probiotics—A comparison of functionality and efficacy. International Journal of Food Microbiology. 2004;96:219-233. DOI: 10.1016/j.ijfoodmicro.2004.05.012
  24. 24. Timmerman HM, Niers LE, Ridwan BU, Koning CJ, Mulder L, Akkermans LM, et al. Design of a multispecies probiotic mixture to prevent infectious complications in critically ill patients. Clinical Nutrition. 2007;26:450-459. DOI: 10.1016/j.clnu.2007.04.008
  25. 25. Koning CJ, Jonkers DM, Stobberingh EE, Mulder L, Rombouts FM, Stockbrugger RW. The effect of a multispecies probiotic on the intestinal microbiota and bowel movements in healthy volunteers taking the antibiotic amoxicillin. The American Journal of Gastroenterology. 2008;103:178-189. DOI: 10.1111/j.1572-0241.2007.01547.x
  26. 26. Jiang Q, Stamatova I, Kari K, Meurman JH. Inhibitory activity in vitro of probiotic lactobacilli against oral Candida under different fermentation conditions. Beneficial Microbes. 2015;6:361-368. DOI: 10.3920/BM2014.0054
  27. 27. Machairas N, Pistiki A, Droggiti DI, Georgitsi M, Pelekanos N, Damoraki G, et al. Pre-treatment with probiotics prolongs survival after experimental infection by multidrug-resistant Pseudomonas aeruginosa in rodents: An effect on sepsis-induced immunosuppression. International Journal of Antimicrobial Agents. 2015;45:376-384. DOI: 10.1016/j.ijantimicag.2014.11.013
  28. 28. Manzoni P, Mostert M, Leonessa ML, Priolo C, Farina D, Monetti C, et al. Oral supplementation with Lactobacillus casei subspecies rhamnosus prevents enteric colonization by Candida species in preterm neonates: A randomized study. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2006;42:1735-1742. DOI: 10.1086/504324
  29. 29. Mangell P, Lennernas P, Wang M, Olsson C, Ahrne S, Molin G, et al. Adhesive capability of Lactobacillus plantarum 299v is important for preventing bacterial translocation in endotoxemic rats. Acta Pathologica, Microbiologica, et Immunologica Scandinavica. 2006;114:611-618. DOI: 10.1111/j.1600-0463.2006.apm_369.x
  30. 30. Ruan X, Shi H, Xia G, Xiao Y, Dong J, Ming F, et al. Encapsulated Bifidobacteria reduced bacterial translocation in rats following hemorrhagic shock and resuscitation. Nutrition. 2007;23:754-761. DOI: 10.1016/j.nut.2007.07.002
  31. 31. Sanchez E, Nieto JC, Boullosa A, Vidal S, Sancho FJ, Rossi G, et al. VSL#3 probiotic treatment decreases bacterial translocation in rats with carbon tetrachloride-induced cirrhosis. Liver International: Official Journal of the International Association for the Study of the Liver. 2015;35:735-745. DOI: 10.1111/liv.12566
  32. 32. Shimizu K, Ogura H, Goto M, Asahara T, Nomoto K, Morotomi M, et al. Synbiotics decrease the incidence of septic complications in patients with severe SIRS: A preliminary report. Digestive Diseases and Sciences. 2009;54:1071-1078. DOI: 10.1007/s10620-008-0460-2
  33. 33. Hayakawa M, Asahara T, Ishitani T, Okamura A, Nomoto K, Gando S. Synbiotic therapy reduces the pathological Gram-negative rods caused by an increased acetic acid concentration in the gut. Digestive Diseases and Sciences. 2012;57:2642-2649. DOI: 10.1007/s10620-012-2201-9
  34. 34. Jain PK, McNaught CE, Anderson AD, MacFie J, Mitchell CJ. Influence of synbiotic containing Lactobacillus acidophilus La5, Bifidobacterium lactis Bb 12, Streptococcus thermophilus, Lactobacillus bulgaricus and oligofructose on gut barrier function and sepsis in critically ill patients: A randomised controlled trial. Clinical Nutrition. 2004;23:467-475. DOI: 10.1016/j.clnu.2003.12.002
  35. 35. Mohan R, Koebnick C, Schildt J, Schmidt S, Mueller M, Possner M, et al. Effects of Bifidobacterium lactis Bb12 supplementation on intestinal microbiota of preterm infants: A double-blind, placebo-controlled, randomized study. Journal of Clinical Microbiology. 2006;44:4025-4031. DOI: 10.1128/JCM.00767-06
  36. 36. Sanaie S, Ebrahimi-Mameghani M, Hamishehkar H, Mojtahedzadeh M, Mahmoodpoor A. Effect of a multispecies probiotic on inflammatory markers in critically ill patients: A randomized, double-blind, placebo-controlled trial. Journal of Research in Medical Sciences: The Official Journal of Isfahan University of Medical Sciences. 2014;19:827-833
  37. 37. McNaught CE, Woodcock NP, Anderson AD, MacFie J. A prospective randomised trial of probiotics in critically ill patients. Clinical Nutrition. 2005;24:211-219. DOI: 10.1016/j.clnu.2004.08.008
  38. 38. Ebrahimi-Mameghani M, Sanaie S, Mahmoodpoor A, Hamishehkar H. Effect of a probiotic preparation (VSL#3) in critically ill patients: A randomized, double-blind, placebo-controlled trial (pilot study). Pakistan Journal of Medical Sciences. 2013;29:490-494
  39. 39. Atanassova M, Choiset Y, Dalgalarrondo M, Chobert JM, Dousset X, Ivanova I, et al. Isolation and partial biochemical characterization of a proteinaceous anti-bacteria and anti-yeast compound produced by Lactobacillus paracasei subsp. paracasei strain M3. International Journal of Food Microbiology. 2003;87:63-73
  40. 40. Kanmani P, Satish Kumar R, Yuvaraj N, Paari KA, Pattukumar V, Arul V. Probiotics and its functionally valuable products—A review. Critical Reviews in Food Science and Nutrition. 2013;53:641-658. DOI: 10.1080/10408398.2011.553752
  41. 41. Li P, Li X, Gu Q, Lou X, Zhang X, Song D, et al. Comparative genomic analysis of Lactobacillus plantarum ZJ316 reveals its genetic adaptation and potential probiotic profiles. Journal of Zhejiang University: Science B. 2016. DOI: 10.1631/jzus.B1600176
  42. 42. Li P, Gu Q. Complete genome sequence of Lactobacillus plantarum LZ95, a potential probiotic strain producing bacteriocins and B-group vitamin riboflavin. Journal of Biotechnology. 2016;229:1-2. DOI: 10.1016/j.jbiotec.2016.04.048
  43. 43. Li P, Gu Q, Zhou Q. Complete genome sequence of Lactobacillus plantarum LZ206, a potential probiotic strain with antimicrobial activity against food-borne pathogenic microorganisms. Journal of Biotechnology. 2016;238:52-55. DOI: 10.1016/j.jbiotec.2016.09.012
  44. 44. Stoianova LG, Ustiugova EA, Netrusov AI. Antibacterial metabolites of lactic acid bacteria: Their diversity and properties. Prikladnaia Biokhimiia i Mikrobiologiia. 2012;48:259-275
  45. 45. Kailasapathy K, Chin J. Survival and therapeutic potential of probiotic organisms with reference to Lactobacillus acidophilus and Bifidobacterium spp. Immunology and Cell Biology. 2000;78:80-88. DOI: 10.1046/j.1440-1711.2000.00886.x
  46. 46. Atassi F, Servin AL. Individual and co-operative roles of lactic acid and hydrogen peroxide in the killing activity of enteric strain Lactobacillus johnsonii NCC933 and vaginal strain Lactobacillus gasseri KS120.1 against enteric, uropathogenic and vaginosis-associated pathogens. FEMS Microbiology Letters. 2010;304:29-38. DOI: 10.1111/j.1574-6968.2009.01887.x
  47. 47. Sharma A, Srivastava S. Anti-Candida activity of two-peptide bacteriocins, plantaricins (Pln E/F and J/K) and their mode of action. Fungal Biology. 2014;118:264-275. DOI: 10.1016/j.funbio.2013.12.006
  48. 48. Bommarius B, Jenssen H, Elliott M, Kindrachuk J, Pasupuleti M, Gieren H, et al. Cost-effective expression and purification of antimicrobial and host defense peptides in Escherichia coli. Peptides. 2010;31:1957-1965. DOI: 10.1016/j.peptides.2010.08.008
  49. 49. Gillor O, Etzion A, Riley MA. The dual role of bacteriocins as anti- and probiotics. Applied Microbiology and Biotechnology. 2008;81:591-606. DOI: 10.1007/s00253-008-1726-5
  50. 50. Diep DB, Straume D, Kjos M, Torres C, Nes IF. An overview of the mosaic bacteriocin pln loci from Lactobacillus plantarum. Peptides. 2009;30:1562-1574. DOI: 10.1016/j.peptides.2009.05.014
  51. 51. Valenzuela AS, Ruiz GD, Ben Omar N, Abriouel H, Lopez RL, Canamero MM, et al. Inhibition of food poisoning and pathogenic bacteria by Lactobacillus plantarum strain 2.9 isolated from ben saalga, both in a culture medium and in food. Food Control. 2008;19:842-848. DOI: 10.1016/j.foodcont.2007.08.009
  52. 52. Li X, Gu Q, Lou X, Zhang X, Song D, Shen L, et al. Complete genome sequence of the probiotic Lactobacillus plantarum strain ZJ316. Genome Announcements. 2013;1:e0009413. DOI: 10.1128/genomeA.00094-13
  53. 53. Zhu X, Zhao Y, Sun Y, Gu Q. Purification and characterisation of plantaricin ZJ008, a novel bacteriocin against Staphylococcus spp. from Lactobacillus plantarum ZJ008. Food Chemistry. 2014;165:216-223. DOI: 10.1016/j.foodchem.2014.05.034
  54. 54. Song DF, Zhu MY, Gu Q. Purification and characterization of Plantaricin ZJ5, a new bacteriocin produced by Lactobacillus plantarum ZJ5. PLoS One. 2014;9:e105549. DOI: 10.1371/journal.pone.0105549
  55. 55. Francavilla R, Miniello V, Magista AM, De Canio A, Bucci N, Gagliardi F, et al. A randomized controlled trial of Lactobacillus GG in children with functional abdominal pain. Pediatrics. 2010;126:e1445-e1452. DOI: 10.1542/peds.2010-0467
  56. 56. Al-Sadi R, Nighot P, Guo SH, Al-Omari D, Ma TY. Lactobacillus Acidophilus enhancement of intestinal epithelial tight junction barrier is mediated by p38 kinase and toll-like receptor-2 (TLR-2). Gastroenterology. 2016;150:S1007-S1007
  57. 57. Grosse C, Scherer J, Koch D, Otto M, Taudte N, Grass G. A new ferrous iron-uptake transporter, EfeU (YcdN), from Escherichia coli. Molecular Microbiology. 2006;62:120-131. DOI: 10.1111/j.1365-2958.2006.05326.x
  58. 58. Lewis K, Lutgendorff F, Phan V, Soderholm JD, Sherman PM, McKay DM. Enhanced translocation of bacteria across metabolically stressed epithelia is reduced by butyrate. Inflammatory Bowel Diseases. 2010;16:1138-1148. DOI: 10.1002/ibd.21177
  59. 59. Wong JM, Jenkins DJ. Carbohydrate digestibility and metabolic effects. The Journal of Nutrition. 2007;137:2539S-2546S
  60. 60. Rolfe RD. The role of probiotic cultures in the control of gastrointestinal health. The Journal of Nutrition. 2000;130:396S-402S
  61. 61. Engels C, Ruscheweyh HJ, Beerenwinkel N, Lacroix C, Schwab C. The common gut microbe Eubacterium hallii also contributes to intestinal propionate formation. Frontiers in Microbiology. 2016;7:713. DOI: 10.3389/fmicb.2016.00713
  62. 62. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118:229-241. DOI: 10.1016/j.cell.2004.07.002
  63. 63. Zhang Z, Hinrichs DJ, Lu H, Chen H, Zhong W, Kolls JK. After interleukin-12p40, are interleukin-23 and interleukin-17 the next therapeutic targets for inflammatory bowel disease? International Immunopharmacology. 2007;7:409-416. DOI: 10.1016/j.intimp.2006.09.024
  64. 64. Badia R, Zanello G, Chevaleyre C, Lizardo R, Meurens F, Martinez P, et al. Effect of Saccharomyces cerevisiae var. Boulardii and beta-galactomannan oligosaccharide on porcine intestinal epithelial and dendritic cells challenged in vitro with Escherichia coli F4 (K88). Veterinary Research. 2012;43:4. DOI: 10.1186/1297-9716-43-4
  65. 65. Stier H, Bischoff SC. Influence of Saccharomyces boulardii CNCM I-745 on the gut-associated immune system. Clinical and Experimental Gastroenterology. 2016;9:269-279. DOI: 10.2147/CEG.S111003
  66. 66. Smith IM, Christensen JE, Arneborg N, Jespersen L. Yeast modulation of human dendritic cell cytokine secretion: An in vitro study. PLoS One. 2014;9:e96595. DOI: 10.1371/journal.pone.0096595
  67. 67. Ng SC, Hart AL, Kamm MA, Stagg AJ, Knight SC. Mechanisms of action of probiotics: Recent advances. Inflammatory Bowel Diseases. 2009;15:300-310. DOI: 10.1002/ibd.20602
  68. 68. Plaza-Diaz J, Gomez-Llorente C, Fontana L, Gil A. Modulation of immunity and inflammatory gene expression in the gut, in inflammatory diseases of the gut and in the liver by probiotics. World Journal of Gastroenterology. 2014;20:15632-15649. DOI: 10.3748/wjg.v20.i42.15632
  69. 69. Breyner NM, Michon C, de Sousa CS, Boas PBV, Chain F, Azevedo VA, et al. Microbial anti-inflammatory molecule (MAM) from Faecalibacterium prausnitzii shows a protective effect on DNBS and DSS-induced colitis model in mice through inhibition of NF-kappa B pathway. Frontiers in Microbiology. 2017;8:114. DOI: 10.3389/fmicb.2017.00114
  70. 70. Yan F, Polk DB. Probiotic bacterium prevents cytokine-induced apoptosis in intestinal epithelial cells. The Journal of Biological Chemistry. 2002;277:50959-50965. DOI: 10.1074/jbc.M207050200
  71. 71. Wu Q, Liu MC, Yang J, Wang JF, Zhu YH. Lactobacillus rhamnosus GR-1 ameliorates Escherichia coli-induced inflammation and cell damage via attenuation of ASC-independent NLRP3 inflammasome activation. Applied and Environmental Microbiology. 2015;82:1173-1182. DOI: 10.1128/AEM.03044-15
  72. 72. Zaoutis T. Candidemia in children. Current Medical Research and Opinion. 2010;26:1761-1768. DOI: 10.1185/03007995.2010.487796
  73. 73. Steinbach WJ. Epidemiology of invasive fungal infections in neonates and children. Clinical Microbiology and Infection: The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases. 2010;16:1321-1327. DOI: 10.1111/j.1469-0691.2010.03288.x
  74. 74. Hu H, Merenstein DJ, Wang C, Hamilton PR, Blackmon ML, Chen H, et al. Impact of eating probiotic yogurt on colonization by Candida species of the oral and vaginal mucosa in HIV-infected and HIV-uninfected women. Mycopathologia. 2013;176:175-181. DOI: 10.1007/s11046-013-9678-4
  75. 75. Roy A, Chaudhuri J, Sarkar D, Ghosh P, Chakraborty S. Role of enteric supplementation of probiotics on late-onset sepsis by Candida species in preterm low birth weight neonates: A randomized, double blind, placebo-controlled trial. North American Journal of Medical Sciences. 2014;6:50-57. DOI: 10.4103/1947-2714.125870
  76. 76. Hatakka K, Ahola AJ, Yli-Knuuttila H, Richardson M, Poussa T, Meurman JH, et al. Probiotics reduce the prevalence of oral candida in the elderly—A randomized controlled trial. Journal of Dental Research. 2007;86:125-130. DOI: 10.1177/154405910708600204
  77. 77. Kohler GA, Assefa S, Reid G. Probiotic interference of Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14 with the opportunistic fungal pathogen Candida albicans. Infectious Diseases in Obstetrics and Gynecology. 2012;2012:636474. DOI: 10.1155/2012/636474
  78. 78. Vilela SF, Barbosa JO, Rossoni RD, Santos JD, Prata MC, Anbinder AL, et al. Lactobacillus acidophilus ATCC 4356 inhibits biofilm formation by C. albicans and attenuates the experimental candidiasis in Galleria mellonella. Virulence. 2015;6:29-39. DOI: 10.4161/21505594.2014.981486
  79. 79. Coman MM, Verdenelli MC, Cecchini C, Silvi S, Orpianesi C, Boyko N, et al. In vitro evaluation of antimicrobial activity of Lactobacillus rhamnosus IMC 501((R)), Lactobacillus paracasei IMC 502((R)) and SYNBIO((R)) against pathogens. Journal of Applied Microbiology. 2014;117:518-527. DOI: 10.1111/jam.12544
  80. 80. Ujaoney S, Chandra J, Faddoul F, Chane M, Wang J, Taifour L, et al. In vitro effect of over-the-counter probiotics on the ability of Candida albicans to form biofilm on denture strips. Journal of Dental Hygiene. 2014;88:183-189
  81. 81. Strus M, Kucharska A, Kukla G, Brzychczy-Wloch M, Maresz K, Heczko PB. The in vitro activity of vaginal Lactobacillus with probiotic properties against Candida. Infectious Diseases in Obstetrics and Gynecology. 2005;13:69-75. DOI: 10.1080/10647440400028136
  82. 82. Martinez RC, Franceschini SA, Patta MC, Quintana SM, Candido RC, Ferreira JC, et al. Improved treatment of vulvovaginal candidiasis with fluconazole plus probiotic Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14. Letters in Applied Microbiology. 2009;48:269-274. DOI: 10.1111/j.1472-765X.2008.02477.x
  83. 83. Kumar S, Bansal A, Chakrabarti A, Singhi S. Evaluation of efficacy of probiotics in prevention of candida colonization in a PICU-a randomized controlled trial. Critical Care Medicine. 2013;41:565-572. DOI: 10.1097/CCM.0b013e31826a409c
  84. 84. Li D, Li Q, Liu C, Lin M, Li X, Xiao X, et al. Efficacy and safety of probiotics in the treatment of Candida-associated stomatitis. Mycoses. 2014;57:141-146. DOI: 10.1111/myc.12116
  85. 85. Kraft-Bodi E, Jorgensen MR, Keller MK, Kragelund C, Twetman S. Effect of probiotic bacteria on oral Candida in frail elderly. Journal of Dental Research. 2015;94:181S-186S. DOI: 10.1177/0022034515595950
  86. 86. Kheradmand E, Rafii F, Yazdi MH, Sepahi AA, Shahverdi AR, Oveisi MR. The antimicrobial effects of selenium nanoparticle-enriched probiotics and their fermented broth against Candida albicans. Daru: Journal of Faculty of Pharmacy, Tehran University of Medical Sciences. 2014;22:48. DOI: 10.1186/2008-2231-22-48
  87. 87. Murzyn A, Krasowska A, Stefanowicz P, Dziadkowiec D, Lukaszewicz M. Capric acid secreted by S. boulardii inhibits C. albicans filamentous growth, adhesion and biofilm formation. PLoS One. 2010;5:e12050. DOI: 10.1371/journal.pone.0012050
  88. 88. Kunz AN, Noel JM, Fairchok MP. Two cases of Lactobacillus bacteremia during probiotic treatment of short gut syndrome. Journal of Pediatric Gastroenterology and Nutrition. 2004;38:457-458
  89. 89. Cannon JP, Lee TA, Bolanos JT, Danziger LH. Pathogenic relevance of Lactobacillus: A retrospective review of over 200 cases. European Journal of Clinical Microbiology & Infectious Diseases: Official Publication of the European Society of Clinical Microbiology. 2005;24:31-40. DOI: 10.1007/s10096-004-1253-y
  90. 90. Land MH, Rouster-Stevens K, Woods CR, Cannon ML, Cnota J, Shetty AK. Lactobacillus sepsis associated with probiotic therapy. Pediatrics. 2005;115:178-181. DOI: 10.1542/peds.2004-2137
  91. 91. Mackay AD, Taylor MB, Kibbler CC, Hamilton-Miller JM. Lactobacillus endocarditis caused by a probiotic organism. Clinical Microbiology and Infection: The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases. 1999;5:290-292
  92. 92. Meini S, Laureano R, Fani L, Tascini C, Galano A, Antonelli A, et al. Breakthrough Lactobacillus rhamnosus GG bacteremia associated with probiotic use in an adult patient with severe active ulcerative colitis: Case report and review of the literature. Infection. 2015;43:777-781. DOI: 10.1007/s15010-015-0798-2
  93. 93. Salminen MK, Tynkkynen S, Rautelin H, Saxelin M, Vaara M, Ruutu P, et al. Lactobacillus bacteremia during a rapid increase in probiotic use of Lactobacillus rhamnosus GG in Finland. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America. 2002;35:1155-1160. DOI: 10.1086/342912
  94. 94. Gorbach SL, Goldin BR. The intestinal microflora and the colon cancer connection. Reviews of Infectious Diseases. 1990;12(Suppl 2):S252-S261
  95. 95. Lidbeck A, Nord CE, Gustafsson JA, Rafter J. Lactobacilli, anticarcinogenic activities and human intestinal microflora. European Journal of Cancer Prevention: The Official Journal of the European Cancer Prevention Organisation. 1992;1:341-353
  96. 96. Egervarn M, Danielsen M, Roos S, Lindmark H, Lindgren S. Antibiotic susceptibility profiles of Lactobacillus reuteri and Lactobacillus fermentum. Journal of Food Protection. 2007;70:412-418
  97. 97. Egervarn M, Roos S, Lindmark H. Identification and characterization of antibiotic resistance genes in Lactobacillus reuteri and Lactobacillus plantarum. Journal of Applied Microbiology. 2009;107:1658-1668. DOI: 10.1111/j.1365-2672.2009.04352.x
  98. 98. Gevers D, Danielsen M, Huys G, Swings J. Molecular characterization of tet(M) genes in Lactobacillus isolates from different types of fermented dry sausage. Applied and Environmental Microbiology. 2003;69:1270-1275
  99. 99. Lin CF, Fung ZF, Wu CL, Chung TC. Molecular characterization of a plasmid-borne (pTC82) chloramphenicol resistance determinant (cat-TC) from Lactobacillus reuteri G4. Plasmid. 1996;36:116-124. DOI: 10.1006/plas.1996.0039
  100. 100. Tannock GW, Luchansky JB, Miller L, Connell H, Thode-Andersen S, Mercer AA, et al. Molecular characterization of a plasmid-borne (pGT633) erythromycin resistance determinant (ermGT) from Lactobacillus reuteri 100-63. Plasmid. 1994;31:60-71. DOI: 10.1006/plas.1994.1007

Written By

Ping Li and Qing Gu

Submitted: 14 July 2017 Reviewed: 29 November 2017 Published: 04 July 2018