Open access peer-reviewed chapter

Improving Antimicrobial Stewardship in Human Health Using Probiotics and Their Derivatives

Written By

Nesisa Nyathi, Duduzile Ndhlovu, Esma Rabvukwa and Abigarl Ndudzo

Submitted: 29 May 2023 Reviewed: 17 July 2023 Published: 05 January 2024

DOI: 10.5772/intechopen.112497

From the Edited Volume

Antimicrobial Stewardship - New Insights

Edited by Ghulam Mustafa

Chapter metrics overview

55 Chapter Downloads

View Full Metrics

Abstract

Since antimicrobial resistance poses the biggest risks to human health, antimicrobial stewardship implies a strategy of responsible management. To lessen antimicrobial resistance and its impacts, a number of antimicrobial stewardship strategies are being used. One such crucial tactic is the use of probiotics and their derivatives, which directly affect the gut microbiota and have been linked to the development and management of a number of human diseases. Undoubtedly, the gut microbiota has a significant impact on the host immunological response, defense against pathogen overgrowth, biosynthesis, and metabolism. There is a rising need to incorporate strategies for altering the gut microbiota as a means of therapy or infection prevention in routine clinical practice as our understanding of the connections between the gut microbiota and host immunity and infectious illnesses deepens. Probiotics and their derivatives influence the development of various species in the gut microbiome to support the host’s health. This review aims to investigate how the gut microbiota is modulated by probiotics, ghost probiotics, postbiotics, and synbiotics, and what this means for infection prevention and antibiotic stewardship.

Keywords

  • probiotics
  • ghost probiotics
  • synbiotics
  • postbiotics
  • antimicrobial resistance
  • antimicrobial stewardship

1. Introduction

The maintenance of a healthy and diverse gut microbiota plays an important role in the prevention and acquisition of antimicrobial resistance, and strategies that modulate its composition have great potential in impacting human health [1]. By limiting the detrimental effects of antimicrobials on the gut flora, the principles of antimicrobial stewardship are strengthened. Antimicrobial stewardship refers to a logical sequence of steps that confirms the effective therapeutic availability of antimicrobials when required [1]. Antimicrobial stewardship has three objectives. The first objective is to collaborate with medical professionals to ensure that each patient receives the best antibiotic at the right dosage and for the right amount of time. The correct drug, dose, de-escalation to pathogen-directed therapy, and length of therapy are essential components of the best antimicrobial therapy [2]. Preventing misuse, abuse, and overuse of antibiotics is the second objective. Reduction of the emergence of antimicrobial resistance is the third objective [3].

Antimicrobials are the foundation upon which the health system is standing and are regarded as a global public good that has boosted health care, saved lives, and increased economic advantages [4]. Antimicrobial stewardship optimizes patient antimicrobial functioning to enhance outcomes and lowers antimicrobial resistance. In addition to encouraging the use of agents that are less likely to select for resistant microorganisms, it aids in the development of techniques and interventions that aim to reduce the unnecessary use of antimicrobials. Its initiatives lower prices, antimicrobial resistance, and toxicity [5]. According to Shiffen et al. [6], antimicrobial resistance is the modification of bacteria that renders them resistant to an antimicrobial. Antimicrobial resistance is one of the top ten global public health problems now facing the world [7].

In order to maintain antibiotic effectiveness and prevent the emergence and spread of infections that are antibiotic-resistant, antimicrobial stewardship promotes the cautious application of antimicrobials [8]. Antimicrobial treatments can cause microbes including viruses, fungi, and parasites to adapt and develop resistance. For instance, bacteria that are exposed to antibiotics develop a resistance that allows them to proliferate in the presence of antibiotics and pass on their resistance genes to their progeny [9]. As time goes on, highly resistant strains of microbes develop which are difficult or impossible to treat with available antimicrobials [10]. For antimicrobial therapies to be effective, antimicrobial stewardship programs promote the appropriate use of antibiotics, which includes proper prescription of drugs, the right dose for the right duration, and only when it is necessary [11]. As a result, the selective pressure driving the formation of antimicrobial resistance is reduced. It ensures that these drugs successfully treat infections and reduces the risk of microbial resistance, which is connected to morbidity and mortality [12].

Focus should be placed on the antimicrobial potential of probiotics against pathogenic microbes and host immunity when considering the current pandemic scenario. It has been said that the gut microbiota is an underappreciated organ that constantly establishes bi- or multidirectional interactions with other organs [13]. Therefore, the modification of the human immune system, gut microbiota, and treatment of illness by probiotics and their derivatives has a strong positive impact on reducing antimicrobial resistance and thereby increasing antimicrobial stewardship. The capacity of a healthy microbiota to limit the spread of potentially harmful bacteria is known as colonization resistance [1]. Probiotics play a crucial role in antimicrobial stewardship by maintaining or re-establishing colonization resistance by regulating the gut microbiota to prevent infection [6].

Advertisement

2. Probiotics

Probiotics are living organisms that, when consumed in sufficient proportions, help the host’s health, however, the dead bacteria and their components can also exhibit probiotic properties [14]. They help with gastrointestinal health, immune system stimulation, and infection risk reduction [15]. By upsetting the normal equilibrium of gut flora, they reduce antibiotic-associated diarrhea [16]. Additionally, they prevent an infection, which is an objective of antimicrobial stewardship, which reduces the need for antimicrobials. Microbiome therapy aims to modify the gut microbiome by applying native or artificially created bacteria in an additive, subtractive, or modulatory manner. [17]. Probiotic bacteria have the additional capacity to create and release antimicrobial substances like chemical compounds [7].

2.1 Mechanisms of action of probiotics

Probiotics function by:

  • Strengthening the epithelial barrier. The gut barrier plays a crucial role in preventing the entry of harmful substances into the bloodstream. Probiotics can enhance the integrity of the intestinal barrier by strengthening the tight junctions between intestinal cells. This helps to maintain a healthy gut lining and reduces the risk of harmful substances crossing into the body [13],

  • Boosting adherence to the intestinal mucosa,

  • Blocking pathogen adhesion. Probiotics can influence the composition and activity of the gut microbiota. They compete with harmful bacteria for resources and adhesion sites in the gut, effectively crowding out pathogenic microorganisms. Probiotics can also produce antimicrobial substances that inhibit the growth of harmful bacteria [14].

  • Eliminating harmful bacteria through competition, creating anti-microbe compounds, and altering the immune system. The colonization of intestinal microbe communities, competitive exclusion of pathogens [18].

  • Modulating the immune response. They interact with immune cells in the gut-associated lymphoid tissue (GALT) and stimulate the production of beneficial cytokines, such as interleukin-10 (IL-10), which helps to regulate the immune system and reduce inflammation. This immunomodulatory effect may be beneficial in conditions such as allergies, autoimmune diseases, and certain gastrointestinal disorders [19],

  • Production of bioactive compounds: Probiotics can produce bioactive compounds such as vitamins (such as vitamin K and some B vitamins), enzymes, and antimicrobial substances. These compounds can have direct health benefits and contribute to overall production of bacteriocins, modulation of enzymatic activities related to metabolization of specific carcinogens, and production of volatile fatty acids [14]. Probiotics boost the formation of mucin and cell adhesion.

2.2 Effects of probiotics on the gut microbiota

By encouraging the growth of helpful bacteria like Lactobacillus and Bifidobacterium and suppressing the growth of pathogenic bacteria like Clostridium difficile, probiotics can alter the balance of bacteria in the gut [20]. This can help safeguard against certain illnesses while enhancing intestinal health. Depending on the probiotic, harmful bacteria may be killed or its growth may be inhibited [19]. By doing so, the danger of infection can be decreased and pathogenic bacteria in the gut cannot develop [21]. By encouraging the creation of mucus and tight junction proteins, which can help stop harmful bacteria from entering the bloodstream, probiotics can enhance the intestinal barrier [19]. By increasing the production of anti-inflammatory cytokines and decreasing the production of pro-inflammatory cytokines, probiotics can also modify the immune system [20]. This may lessen intestinal inflammation and enhance gut health. A number of infectious and non-infectious diseases can be effectively treated using probiotics and their derivatives.

2.3 Role of probiotics in antimicrobial stewardship

The World Health Organization [11] has come to the conclusion that probiotics are the second-most important immune defense mechanism when frequently advised antimicrobials are unsuccessful against a limited number of pathogens. A number of beneficial intestinal bacteria, including Lactobacillus, Bifidobacterium, and Enterococcus, as shown in Table 1, contribute to the improvement of gut microbiota stability [28]. Probiotics can be found in food, beverages, and dietary supplements. Additionally, consuming fermented foods, which are probiotic bacteria’s natural habitat, is of immense importance which eventually helps in the better treatment of several diseases, including gut-related disorders [28].

ProbioticReported effectsReferences
Lactobacillus rhamnosus GGImproved gut health, reduced diarrhea[22]
Bifidobacterium animalis DN-173010Enhanced immune response - They can boost antibody synthesis, amplify immune cell activity, and fortify the intestinal barrier, lowering the risk of opportunistic infections while using antibiotics; fewer stomach aches and pains[23]
Saccharomyces boulardiiAAD (antibiotic-associated diarrhea) prevention and treatment. The natural balance of benevolent bacteria in the stomach can be upset by antibiotics, which can result in diseases like AAD. When given simultaneously with antibiotics, probiotics can aid in microbial restoration and lower the risk of AAD.[22]
Lactobacillus acidophilus NCFMReduction of lactose intolerance[24]
Streptococcus thermophilusEnhanced digestion of lactose[22]
Lactobacillus plantarum 299vImproved symptoms of irritable bowel syndrome[25]
Bifidobacterium bifidumEnhanced immune function[25]
Lactobacillus
Streptococcus
Treatment Adherence - Probiotics may improve a patient’s treatment adherence with antibiotics. Probiotics have been shown to improve treatment compliance, decrease side effects, and improve patient satisfaction when added to antibiotic regimens.[26]
Lactobacillus
Saccharomyces
Inhibiting the growth and colonization of Clostridium difficile[27]

Table 1.

Roles of probiotics.

If it happens that once in the human body the probiotics die, they can no longer have a positive effect on health therefore scientists are conducting research to formulate technologies that produce dead and broken-down bacteria which are like probiotics or better in terms of immune enhancing properties [29].

Advertisement

3. Ghost probiotics

It has recently been discovered that ghost probiotics, or inactivated or heat-killed probiotic cells, can be used in place of live probiotics. In both animals and people, they have demonstrated effectiveness in modifying anti-inflammatory and pro-inflammatory immune responses [30]. Ghost probiotics, commonly referred to as para-probiotics, are beneficial microorganisms that have been rendered inactive. They can either be whole or broken [8]. Provoking the human immune system, promoting a favorable immunological response, and having anti-inflammatory effects in both people and animals are all impacts of ghost probiotics [31]. They are favored because they have a longer shelf life, which makes it easier to distribute them without refrigeration to various locations. In immune-compromised people, the use of ghost probiotics lowers the likelihood of contamination, microbial translocation, or heightened inflammatory immune responses [8, 32].

3.1 Mechanisms of action of ghost probiotics

Probiotics that have been inactivated may influence immunological responses by interacting with the immune system. A study suggested that the beneficial effects of ghost probiotics on gut microbiota may be due to the release of soluble factors such as cell membrane fragments and metabolites that can modulate immune function and promote the growth of beneficial bacteria [4]. These immuno-modulatory actions might suppress immune-related disorders and control immune response and inflammation. In the stomach, they fight against harmful microbes for adhesion sites and nutrition. They might hinder the attachment and proliferation of harmful bacteria by occupying these locations. In another study, researchers discovered that supplementation with ghost probiotics derived from Lactobacillus plantarum had positive effects on gut microbiota, including an increase in beneficial bacteria such as Bifidobacterium and Lactobacillus and a decrease in potentially harmful bacteria such as Clostridium [17].

They might help keep the intestinal barrier healthy and functioning properly. They are able to interact with gut epithelial cells, enhancing tight junction protein production and preserving the integrity of the gut barrier. Inactivated probiotics can aid in preventing the transfer of hazardous chemicals from the gut into the circulation by enhancing the barrier function [33]. Even in their non-viable state, inactivated probiotics may retain some antimicrobial properties. They may produce antimicrobial compounds such as organic acids, bacteriocins, or antimicrobial peptides that can inhibit the growth of pathogenic bacteria in the gut. These antimicrobial effects contribute to maintaining a balanced gut microbiota [8]. Inactivated probiotics may still have metabolic effects. They can interact with dietary components and release bioactive compounds such as short-chain fatty acids (SCFAs) or other metabolites. SCFAs, for example, can provide energy to colon cells, support the gut barrier function, and have systemic effects on metabolism [32].

3.2 Roles of ghost probiotics in antimicrobial stewardship

In antimicrobial stewardship, ghost probiotics are used as a possible remedy in fighting against antimicrobial resistance [34], as illustrated in Table 2. A study by Thursby and Juge [40], revealed a significant decrease in the number of infections in the treatment group compared to the control group when the efficacy of a ghost probiotic formula in preventing respiratory tract infections in a population of elderly individuals in a nursing home was tested. In 2019, another study investigated the potential use of ghost probiotics as a preventive strategy against Clostridium difficile infections in a mouse model. Researchers found that a ghost probiotic formula of Lactobacillus reuteri DSM 17938 significantly shortens the duration of acute infectious diarrhea and reduces abdominal pain in patients with colitis [19, 41].

Ghost probioticReported effectsReferences
Non-pathogenic Escherichia coli Nissle 1917Competitive exclusion of pathogenic bacteria. They compete with pathogenic bacteria for binding sites and nutrients in the gut. This competitive exclusion mechanism can help prevent the attachment and colonization of harmful bacteria, reducing the risk of infections.[26]
Lactobacillus rhamnosus GGReduced inflammatory response in the gut. When it comes to viral disorders, where inflammation can cause tissue damage, they can help regulate immune responses and minimize excessive inflammation.[35]
Bacillus subtilisEnhanced immune response; inhibition of pathogens[36]
Lactobacillus acidophilusModulation of immune responses. Similar to living probiotics, they influence the immune system by promoting the synthesis of substances that modulate the immune system. They have the power to affect immune cell activity and cytokine synthesis, which can strengthen immunological responses and promote the body’s fight against infections.[37]
BifidobacteriumBoost intestinal health and aid in keeping the intestinal barrier’s integrity. They can strengthen tight connections between cells, increase mucus production, and improve the gut epithelium’s defensive capabilities. This may help lower the possibility of bacterial translocation and ensuing illnesses.[24]
StreptococcusTreat and prevent infections[38]
Lactobacillus
Streptococcus
Bifidobacterium
Reduce use of antibiotics. In some circumstances, they might act as an additional therapy to lessen the requirement for antibiotics. Ghost probiotics may enhance the body’s natural defense mechanisms by boosting the immune system and encouraging a healthy gut flora, potentially lowering the need for antibiotics and limiting the emergence of antibiotic resistance.[39]

Table 2.

Roles of ghost probiotics.

Advertisement

4. Synbiotics

The term synbiotics describes probiotics and prebiotics that have been combined in dietary supplements or food ingredients to create a synergistic effect. According to Wang et al. [42], prebiotics are common foods that strengthen the immune system while encouraging the development of probiotic bacteria in the human stomach. Prebiotics, which are necessary for the development and function of microorganisms, should not be mistaken with probiotics. Supplements and specific foods like yogurt, whole grains, and sauerkraut contain synbiotics [43]. Bifidobacterium, Lactobacillus, Saccharomyces boulardii, Bacillus cougulans, and prebiotics like Inulin and XOS are the most widely utilized synbiotics [44]. The most popular fibers that are typically utilized in conjunction with probiotics are fructans. Synbiotics were created in order to get over potential probiotic survival issues [43]. Synbiotics play a significant role in gut bacterial balance, which benefits the human immune system, metabolism, and gut health [43].

4.1 Effects of synbiotics on gut microbiota

The gut microbiota appears to play a role in the pathogenesis of obesity and associated diseases. Therefore, gut microbiota can also be considered a promising target in the comprehensive dietary approach to the prevention and treatment of obesity, including weight loss and weight maintenance [45]. Synbiotic supplementation increases the abundance of gut bacteria associated with positive health effects, especially Bifidobacterium and Lactobacillus, and it also appears to increase the gut microbiota richness [45].

According to Roy and Dhaneshwar [46], synbiotics can aid in enhancing the integrity of the intestinal barrier, which may lessen intestinal inflammation and enhance immunological function. Short chain fatty acids (SCFAs) are vital energy sources for colonocytes and have anti-inflammatory characteristics. Synbiotics help to boost production of SCFAs [47]. According to Phavichitr et al. [45], synbiotics can determine the abundance of harmful bacteria in the stomach, including Escherichia coli and Clostridium difficile. Additionally, synbiotics may enhance cholesterol, glucose, and inflammation metabolism, which could be advantageous for metabolic health [44].

4.2 How do synbiotics work?

Synbiotics work by combining prebiotics and probiotics to provide a synergistic effect that promotes optimal gut health. The probiotic adds beneficial bacteria that greatly benefit the gut microbiome. The prebiotic fuels the beneficial bacteria that is already present in the gut microbiome. There are two types of synbiotics which are synergistic synobiotics and complementary synbiotics [43]. Synergistic synbiotics are formulated to have live probiotics fueled by the prebiotic substrate that is co-administered in the product, allowing the two elements to work together as a self-contained whole [44]. Complementary synbiotics are formulated so that both elements work independently, with the prebiotic chosen to target resident microorganisms in the gut [44].

Synbiotics help to promote the growth of beneficial bacteria in the gut, leading to improved gut microbiome balance and overall gut health. A study by Palai et al. [43], supported that symbiotic supplements used in clinical trial modulate human gut microbiota by increasing abundance of potentially beneficial microbial species. Synbiotics have been shown to boost immune function by promoting the growth of beneficial bacteria that can help fight off harmful pathogens. A study showed that a synbiotic consisting of Lactobacillus acidophilus and Inulin improved the immune response in elderly individuals by increasing the production of immunoglobulin A and reducing the levels of pro-inflammatory cytokines such as interleukin-6 and tumor necrosis factor-alpha [44].

Synbiotics can improve digestion by promoting the growth of beneficial bacteria that aid in the breakdown and absorption of nutrients. They display positive effects on digestion and enzymatic activity, which may help alleviate many of the symptoms associated with lactose intolerance. For instance, the synbiotic of Lactobacillius acidophilus and Inulin improves the digestion of lactose in lactose-intolerant individuals by increasing the population of lactase-producing bacteria in the gut [47]. Also, probiotics like Bifidobacterium lactis combined together with fructooligosaccharides (FOS) improve the digestion of protein and fat in healthy adults by enhancing the production of pancreatic enzymes. Synbiotics have been shown to have anti-inflammatory effects, which can help to reduce inflammation in the gut and throughout the body. For example, research showed that a synbiotic consisting of Lactobacillus acidophilus and FOS reduces levels of C-reactive protein, a cause of inflammation [48].

4.3 Roles of synbiotics in antimicrobial stewardship

Synbiotics play a valuable role in antimicrobial stewardship by reducing the need for antimicrobial agents and preventing the development of antibiotic resistance. Synbiotics can be used to prevent and treat a number of conditions that are often treated with antimicrobials, such as Clostridium difficile infection, inflammatory bowel disease, and irritable bowel syndrome [49]. Lactobacillus acidophilus and Bifidobacterium lactis with FOS have been shown to reduce the incidence of antibiotic-associated diarrhea and Clostridium difficile infection in hospitalized patients efficiently [47]. Lactobacillus rhamnosus GG and inulin have been shown to reduce the incidence of antibiotic-associated diarrhea in children [47]. Akkermansia muciniphila and prebiotic fibers have been shown to improve gut barrier function and reduce inflammation in obese and overweight adults [49].

Advertisement

5. Postbiotics

Postbiotics are metabolic by-products of probiotic bacteria [34]. These byproducts include short-chain fatty acids, enzymes, peptides, and other compounds that can have various health benefits. Postbiotics play a role in maintaining gut health, regulating the immune system, providing anti-inflammatory effects and potentially reducing the risk of certain diseases [2]. Postbiotics can be naturally produced by the probiotic bacteria that live in our gut and they can also be produced through fermentation of certain foods such as yogurt, kefir, sauerkraut, and kimchi [50].

5.1 Roles of postbiotics in antimicrobial stewardship

Postbiotics, such as peptides and organic acids, have been shown to have beneficial effects on gut microbiota as shown in Table 3. Certain peptides produced by Lactobacillus species have been shown to inhibit the growth of pathogenic bacteria such as Escherichia coli and Salmonella enteritidis [9]. Organic acids such as acetic acid and lactic acid, which are produced by some probiotic bacteria, can help to lower the pH of the gut, creating an environment that is less favorable for the growth of harmful bacteria [2]. Short-chain fatty acids (SCFAs) are produced by the fermentation of dietary fiber by gut bacteria and have been shown to have antimicrobial properties. They can help to inhibit the growth of harmful bacteria such as Clostridium difficile, which is a common cause of antibiotic-associated diarrhea. By promoting the growth of beneficial bacteria and reducing the need for antibiotics, SCFAs can help to reduce the overall use of antimicrobials and limit the development of antimicrobial resistance [50].

PostbioticReported effectsReferences
Short chain fatty acids from gut microbiota fermentationEnhanced gut barrier function; anti-inflammatory effects. They can reduce the production of pro-inflammatory molecules, promote the production of anti-inflammatory substances, and help alleviate inflammation associated with infections.[51]
Lactic acid bacteria metabolitesModulation of immune responses; antimicrobial properties[52]
BacteriocinsInhibition of pathogenic bacteria[53]
Extracellular vesiclesCellular communication; immune regulation[54]
Organic acidspH regulation; antimicrobial effects[55]
Peptidoglycan derived muropeptidesThe intestinal barrier’s integrity and functionality can be supported by immunomodulation. They can improve the protective qualities of the barrier, encourage mucus secretion, and fortify the tight junctions between intestinal cells. This aids in stopping dangerous bacteria and poisons from crossing the intestinal barrier.[51]
They can aid in forming and preserving a balanced makeup of the gut flora. They can support a healthy microbial environment by selectively promoting the development of advantageous bacteria, preventing the growth of diseases, and both. This may improve infection resistance and intestinal health.[56]
Reduction of negative Effects Related to Antibiotic Use - They might help to lessen the negative effects related to antibiotic use. They can aid in reducing antibiotic-associated diarrhea, maintain gut health while taking antibiotics, and hasten recovery by encouraging a balanced gut flora.[35]

Table 3.

Roles of postbiotics.

Bacteriocins are antimicrobial peptides produced by some probiotic bacteria that can help to inhibit the growth of pathogenic bacteria. For example, nisin, a bacteriocin produced by Lactococcus lactis, has been shown to be effective against a range of Gram-positive bacteria, including Staphylococcus aureus and Streptococcus pneumoniae [57]. By using bacteriocins instead of antimicrobials to target specific pathogens, postbiotics can help to reduce the overall use of antimicrobials and limit the development of antimicrobial resistance.

Exopolysaccharides are complex carbohydrates produced by some probiotic bacteria that can have prebiotic effects, meaning they can promote the growth of beneficial bacteria in the gut [50]. Peptidoglycans are complex molecules found in the cell walls of bacteria that can modulate the immune system and have anti-inflammatory effects. Cell surface proteins are proteins found on the surface of probiotic bacteria that can interact with host cells and modulate immune responses [33].

Advertisement

6. Issues surrounding the use of probiotics and their derivatives in antimicrobial stewardship

To be considered a probiotic, a supplement needs to meet all the requirements set forth by the 2002 Food and Agriculture Organization/World Health [57]. The requirements stipulate that a probiotic must be a live microorganism, needs to be administered and should have a health benefit. Additionally, a probiotic must be safe and void of vectors that can transfer resistance to antibiotics [14]. One issue surrounding the use of probiotics in antimicrobial stewardship is the potential for interactions between probiotics and antibiotics. Some studies have suggested that probiotics may reduce the effectiveness of antibiotics by competing with them for absorption in the gut or by producing antibacterial substances that can neutralize antibiotics. This could lead to suboptimal treatment outcomes and the emergence of antibiotic-resistant strains of bacteria [58]. However, other studies have shown that probiotics can enhance the effectiveness of antibiotics by improving the gut microbiota and reducing the risk of antibiotic-associated diarrhea and other side effects. Another issue is the lack of standardization in the production and labeling of probiotics [57]. There is significant variability in the quality and efficacy of probiotic products, and there are currently no widely accepted standards for the selection, testing, and labeling of probiotics. This makes it difficult for healthcare providers to make informed decisions about which probiotics to use and for what indications [59].

Advertisement

7. Conclusion and future perspectives

In the midst of the ongoing antibiotic resistance challenge, using probiotics, postbiotics, ghost probiotics, and synbiotics offers a secure alternative to treating microbial infections. In order to research future potential uses of probiotics in both human and animal diseases, it is vital to update our knowledge in this area. As a result, this work may be used as a reference to understand naturally occurring probiotic compounds and their prospective applications for the treatment and management of numerous human diseases, providing a significant contribution to antimicrobial stewardship. Probiotics and their derivatives that exhibit powerful activity, both alone and in combination, must therefore be researched in the future.

References

  1. 1. Zawistowska-Rojek A, Tyski S. Are probiotic really safe for humans? Polish Journal of Microbiology. 2018;67(3):251-258. DOI: 10.21307/pjm-2018-044
  2. 2. Aguilar-Toala JE, Garcia-Varela R, Garcia HS, Mata- Haro V, Gonzalez-Cordova AF, Vallejo-Cordoba B. Postbiotics: An evolving term within the functional foods field. Trends Food Science and Technology. 2018;75:105-114. DOI: 10.1016/j.tifs.2018.03.009
  3. 3. Jeong K, Kim M, Jeon SA, Kim YH, Lee S. A randomized trial of lactobacillus rhamnosus IDCC 3201 tyndallizate (RHT3201) for treating atopic dermatitis. Pediatric Allergy and Immunology. 2020;31:783-792. DOI: 10.1111/pai.13269
  4. 4. Arboleya S, Merinero MC, Solis G. Ghost probiotics with a combined regimen: A novel approach for the prevention and treatment of neonatal sepsis. Beneficial Microbes. 2021;12(1). DOI: 10.3920/BM2020.0079
  5. 5. Hoarau C, Martin L, Faugaret D, Baron C, Dauba A, Aubert-Jacquin C, et al. Supernatant from Bifidobacterium differentially modulates transduction signaling pathways for biological functions of human dendritic cells. PLoS One. 2018;3(7):E2753. DOI: 10.1371/journal.pone.0002753
  6. 6. Sniffen JC, McFarland LV, Evans CT, Goldstein EJC. Choosing appropriate probiotic product for your patient: An evidence based particular guide. PLoS One. 2018;13(12):e0202205. DOI: 10.1371/journal.pone.0209205
  7. 7. Szczerbiec D, Piechoka J, Glowacki R, Torzewska A. Organic acids secreted by lactobacillus spp. isolated from urine and their antimicrobial activity against Uropathogenic Proteus mirabilis. Molecules. 2022;27:5557. DOI: 10.3390/molecules27175557
  8. 8. Kim WK, Jang YJ, Han DH, Jeon K, Lee C, Han HS. Lactobacillus paracasei KBL382 administration attenuates atopic dermatitis by modulating immune response and gut microbiota. Gut Microbes. 2020;12:1-14. DOI: 10.1080/19490976.2020.1819156
  9. 9. Chang CM, Tsai MH, Liao WC, Yang PH, Li SW, Chu SM, et al. Effects of probiotics on gut microbiomes of extremely preterm infants in the neonatal intensive care unit: A prospective cohort study. Nutrients. 2022;14(15):3239. DOI: 10.3390/nu14153239
  10. 10. Sun M, Luo J, Liu H, Xi Y, Lin Q. Can mixed strains of lactobacillus and Bifidobacterium reduce eczema in infants under three years of age? A meta-analysis. Nutrients. 2021;13. DOI: 10.3390/nu13051461
  11. 11. World Health Organisation. Antimicrobial Stewardship Programmes in Health Care Facilitating Low- and Middle Income Countries: A WHO Practical Toolkit. 2019. Available from: https://apps.who.int/iris/bitstream/handle/10665/329404/9789241515481-eng.pdf [Accessed: 20 May 2023]
  12. 12. Azim-Majumder MA, Rahman S, Cohall D, Bharatha A, Singh K, Haque M, et al. Antimicrobial stewardship: Fighting antimicrobial resistance and protecting global public health. Infection and Drug Resistance. 2020;13:4713-4738. DOI: 10.2147/IDR.S290835
  13. 13. Halluran K, Underwood MA. Probiotics mechanisms of action. Early Human Development. 2019;135:58-65. DOI: 10.1016/j.earlhumdev.2019.05.010
  14. 14. Plaza-Diaz J, Ruiz-Ojeda FJ, Gil-Campos M, Gil A. Mechanisms of action of probiotics. Advanced Nutrition. 2019;10. DOI: 10.1093/advances/nmy063
  15. 15. Reid G, Gadir AA, Dhir R. Probiotics: Reiterating what they are and what they are not. Frontiers in Microbiology. 2019;10. DOI: 10.3389/fmicb.2019.00424
  16. 16. Blaabjerg S, Artzi DM, Aabenhus R. Probiotics for the prevention of antibiotic-associated diarrhea in outpatients- a systematic review and meta-analysis. Antibiotics. 2017;6(4). DOI: 10.3390/antibiotics6040021
  17. 17. Yadav M, Chauhan NS. Microbiome therapeutics: Exploring the present scenario and challenges. Gastroenterology Report. 2021;10:goab046. DOI: 10.1093/gastro/goab046
  18. 18. Li Y, Xia S, Jiang X, Feng C, Gong S, Ma J, et al. Gut microbiota and diarrhea: An updated review. Frontiers in Cellular and Infection Microbiology. 2021;11. DOI: 10.3389/fcimb.2021.625210
  19. 19. Wu J, Zhang Y, Ye L, Wang C. The anti-cancer effects and mechanisms of lactic acid bacteria exopolysaccharides in vitro: A review. Carbohydrate Polymers. 2021;253:117308. DOI: 10.1016/j.carbpol.2020.117308
  20. 20. Divakar D, Poonam SN. The gut microbiota influenced by the intake of probiotics and functional foods with prebiotics can sustain wellness and alleviate certain ailments like gut-inflammation and colon cancer. 2022. DOI: 10.3390/microorganisms10030665
  21. 21. Case I. The Regulation of Probiotics. Degree Candidate for Masters of Science in Regulatory Science. Johns Hopkins University.Available from: https://www.jhsph.edu/research/centers-and-institutes/center-of-excellence-in-regulatory-science-and-innovation/training/Iris; 2021 [Accessed 24 May 2023]
  22. 22. George Kerry R, Patra JK, Gouda S, Park Y, Shin H, Das G. Benefaction of probiotics for human health: A review. Journal of Food and Drug Analysis. 2018;26(3):927-939. DOI: 10.1016/j.jfda.2018.01.002
  23. 23. Chen M, Lin W, Li N, Wang Q , Zhu S, Zeng A, et al. Therapeutic approaches to colorectal cancer via strategies based on modulation of gut microbiota. Frontiers in Microbiology. 2022;13:945533. DOI: 10.3389/fmicb.2022.945533
  24. 24. Bozomitu L, Miron I, Adam Raileanu A, Lupu A, Paduraru G, Marcu FM, et al. The gut microbiome and its implication in the mucosal digestive disorders. Biomedicine. 2022;10(12):3117. DOI: 10.3390/biomedicines10123117
  25. 25. Mazziotta C, Tognon M, Martini F, Torreggiani E, Rotondo JC. Probiotics mechanism of action on immune cells and beneficial effects on human health. Cell. 2023;12(1). DOI: 10.3390/cells12010184
  26. 26. Aponte M, Murru N, Shoukat M. Therapeutic, prophylactic, and functional use of probiotics: A current perspective. Frontiers in Microbiology. 2020;11:562048. DOI: 10.3389/fmicb.2020.562048
  27. 27. Gunaratnam S, Millette M, McFarland LV, DuPont HL, Lacroix M. Potential role of probiotics in reducing Clostridioides difficile virulence: Interference with quorum sensing systems. Microbial Pathogenesis. 2021;153:104798. DOI: 10.1016/j.micpath.2021.104798
  28. 28. Prakoeswa C, Bonita L, Karim A, Herwanto N, Umborowat MA, Setyaningrum T. Beneficial effect of lactobacillus plantarum IS-10506 supplementation in adults with atopic dermatitis: A randomized controlled trial. Journal of Dermatological Treatment. 2022;33:1491-1498. DOI: 10.1080/09546634.2020.1836310
  29. 29. Marco ML, Heeney D, Binda S, Cifelli CJ, Cotter PD, Foligne B, et al. Health benefits of fermented foods: Microbiota and beyond. Current Opinions in Biotechnology. 2017;44:94-102. DOI: 10.1016/j.copbio.2016.11.010
  30. 30. Siciliano RA, Reale A, Mazzeo MF, Morandi S, Silvetti T, Brasca M. Paraprobiotics: A new perspective for functional foods and nutraceuticals. Nutrients. 2021;13(4). DOI: 10.3390/nu13041225
  31. 31. Hasan A, Paray BA, Hussain A, Qadir FA, Attar C, Aziz FM. A review on the cleavage priming of the spike protein on coronavirus by angiotensin-converting enzyme-2 and furin. Journal of Biomolecular Structure and Dynamics. 2021;39(8):3025-3033. DOI: 10.1080/07391102.2020.1754293
  32. 32. Ndudzo A, Ndlovu S, Nyathi N, Makuvise-Sibanda A. Unlocking the potential of ghost probiotics in combating anti-microbial resistance. In: The Global Antimicrobial Resistance Epidemic: Innovative Approaches and Cutting Edge Solutions. London, UK: IntechOpen; 2022. DOI: 10.5772/intechopen.10426
  33. 33. Kim D, Kim YJ, Kim SH, Yang JS. Short-chain fatty acids produced by lactobacillus plantarun strain Lp27 inhibit Escherichia coli 0157:H7 and salmonella Typhimurium in vitro. Journal of Microbiology and Biotechnology. 2019;29(8):1195-1205
  34. 34. Aguilar-Toalá JE, Cuevas-Gonzalez P, Liceaga A. Postbiotics and Parabiotics: From concepts to applications. Food Research International. 2020;136:109502. DOI: 10.1016/j.foodres.2020.109502
  35. 35. Amara AA, Shibl A. Role of probiotics in health improvement, infection control and disease treatment and management. Saudi Pharmaceutical Journal: SPJ. 2015;23(2):107-114. DOI: 10.1016/j.jsps.2013.07.001
  36. 36. Incrocci R, Negris O, McGrath S, Swartzendruber JA. Bacillus subtilis provides long-term protection in a murine model of allergic lung disease by influencing bacterial composition. Allergie. 2023;3(1):1-10. DOI: 10.3390/allergies3010001
  37. 37. Li HY, Zhou DD, Ren Y, Huang S, Zhao C, Shang A, et al. Effects and mechanisms of probiotics, prebiotics, synbiotics and post biotics on metabolic diseases targeting. Gut Microbiota. 2021;13(9):3211. DOI: 10.3390/nu13093211
  38. 38. Kechagia M, Basoulis D, Konstantopoulou S, Dimitriadi D, Gyftopoulou K, Skarmoutsou N, et al. Health benefits of probiotics: A review. ISRN Nutrition. 2013. DOI: 10.5402/2013/481651
  39. 39. Patangia DV, Ryan CA, Dempsey E, Ross RP, Stanton C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiology Open. 2022;11(1). DOI: 10.1002/mbo3.1260
  40. 40. Thursby E, Juge N. Introduction to the human gut microbiota. The Biochemical Journal. 2017;474(11):1823-1836. DOI: 10.1042/BCJ20160510
  41. 41. Saviano A, Brigida M, Migneco A, Gunawardena G, Zanza C, Candelli M, et al. Lactobacillus Reuteri DSM 17938 (Limosilactobacillus reuteri) in diarrhea and constipation: Two sides of the same coin? Mecina (Kaunas). 2021;57(7):643. DOI: 10.3390/medicina57070643
  42. 42. Wang HT, Anvari S, Anagnostou K. The role of probiotics in preventing allergic disease. Children (Basel, Switzerland). 2019;6(2):24. DOI: 10.3390/children6020024
  43. 43. Palai S, Derecho CMP, Kesh SS, Egbuna C, Onyeike PC. Prebiotics, probiotics, synbiotics and its importance in management of diseases. Chapter 10. In: Functional Foods and Neutraceuticals. Springer; 2020. DOI: 10.1007/978-3-030-42319-3_10
  44. 44. Thilagavathi T. Probiotics, prebiotics, synbiotics and its health benefits, International Journal of Current Microbiology and Applied Sciences. 2020 DOI: 10.20546
  45. 45. Phavichitr N, Wang S, Chomoto S, Tantibhaedhyangkul R, Kakourou A, Intarakhao S, et al. Impact of Synbiotics on gut microbiota during early life: A randomized double-blind study. Scientific Reports. 2021;11(1):3534. DOI: 10.1038/s41598-021-83009-2
  46. 46. Roy S, Dhaneshwar S. Role of prebiotics, probiotics, and synbiotics in management of inflammatory bowel disease: Current perspectives. World Journal of Gastroenterology. 2023;29(14):2078-2100. DOI: 10.3748/wjg.v29.i14.2078
  47. 47. Chatchatee P, Way SL, Eugenia C, Kosuwon P, Simaakachorn N, Yavuz Y, et al. Effects of growing up supplemented with prebiotics and LCPUFAS on infectious in young children. 2018. DOI: 10.1097/MPG.0000000000000252
  48. 48. Xie A, Chen A, Chen Y, Luo Z, Jiang S, Chen D, et al. Lactobacillus for the treatment and prevention of atopic dermatitis: Clinical and experimental evidence. Frontiers in Cellular and Infection Microbiology. 2023;13:1137275. DOI: 10.3389/fcimb.2023.1137275
  49. 49. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nature Medicine. 2019;25(7):1096-1103. DOI: 10.1038/s41591-019-0495-2
  50. 50. Zhang L, Chen H, Wang Y, Yu L. Antimicrobial activity of acetic acid produced by lactobacillus plantarum JD-15 against foodborne pathogens. Journal of Agricultural and Food Chemistry. 2019;67(43):12059-12067. DOI: 10.3390%2Ffoods10123131
  51. 51. Delgado S, Sánchez B, Margolles A, Ruas-Madiedo P, Ruiz L. Molecules produced by probiotics and intestinal microorganisms with immunomodulatory activity. Nutrients. 2020;12(2). DOI: 10.3390/nu12020391
  52. 52. Moon A, Sun Y, Wang Y, Huang J, Zafar Khan MU, Qiu H. Lactic acid bacteria as mucosal immunity enhancers and antivirals through Oral delivery. Applied Microbiology. 2022;2(4):837-854. DOI: 10.3390/applmicrobiol2040064
  53. 53. Francisco D, Alexis J, Rubén J. Bacteriocins: An overview of antimicrobial, toxicity, and biosafety assessment by in vivo models. Frontiers in Microbiology. 2021;12:630695. DOI: 10.3389/fmicb.2021.630695
  54. 54. Buzas EI. The roles of extracellular vesicles in the immune system. Nature Reviews Immunology. 2023;23(4):236-250. DOI: 10.1038/s41577-022-00763-8
  55. 55. Ma L, Tu H, Chen T. Postbiotics in human health: A narrative review. Nutrients. 2023;15(2). DOI: 10.3390/nu15020291
  56. 56. Hou K, Wu Z, Chen X, Wang J, Zhang D, Xiao C, et al. Microbiota in health and diseases. Signal Transduction and Targeted Therapy. 2022;7(1):1-28. DOI: 10.1038/s41392-022-00974-4
  57. 57. FDA. 2017. Food Labeling: Revision of the Nutrition and Supplement Facts Label. Available form: https://www.regulations.gov/document/FDA-2012-N-1210-0875 [Accessed 19 May 2023]
  58. 58. de Simone C. The unregulated probiotic market. Clinical Gastroenterology and Hepatology. 2019;17(5):809-817. DOI: 10.1016/j.cgh.2018.01.018
  59. 59. Grebow J. Will FDA Ever Provide a Practical Regulatory Path for Probiotics? Panel Discusses at NPS’s the Big Natural Conference. 2020. Available from: https://www.nutritionaloutlook.com/view/will-fda-ever-provide-a-practical-regulatory-path-forprobiotics-panel-discusses-at-npa-s-the-big-natural-conference [Accessed: 19 May 2023]

Written By

Nesisa Nyathi, Duduzile Ndhlovu, Esma Rabvukwa and Abigarl Ndudzo

Submitted: 29 May 2023 Reviewed: 17 July 2023 Published: 05 January 2024