Next Article in Journal
The Expression of Selected Factors Related to T Lymphocyte Activity in Canine Mammary Tumors
Next Article in Special Issue
Cannabidiol and Other Non-Psychoactive Cannabinoids for Prevention and Treatment of Gastrointestinal Disorders: Useful Nutraceuticals?
Previous Article in Journal
Nutrition and Cardiovascular Health
Previous Article in Special Issue
A Brief Review of Nutraceutical Ingredients in Gastrointestinal Disorders: Evidence and Suggestions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Updated Overview on Nanonutraceuticals: Focus on Nanoprebiotics and Nanoprobiotics

by
Alessandra Durazzo
1,
Amirhossein Nazhand
2,
Massimo Lucarini
1,
Atanas G. Atanasov
3,4,5,6,
Eliana B. Souto
7,8,
Ettore Novellino
9,
Raffaele Capasso
10,* and
Antonello Santini
9,*
1
CREA—Research Centre for Food and Nutrition; Via Ardeatina 546, 00178 Rome, Italy
2
Biotechnology Department, Sari University of Agricultural Sciences and Natural Resources, 9th km of Farah Abad Road, Mazandaran, 48181 68984 Sari, Iran
3
Institute of Neurobiology, Bulgarian Academy of Sciences, 23 Acad. G. Bonchev str., 1113 Sofia, Bulgaria
4
Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland
5
Department of Pharmacognosy, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
6
Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
7
Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, 3000-548 Coimbra, Portugal
8
CEB-Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
9
Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
10
Department of Agricultural Sciences, University of Napoli Federico II, Via Università 100, 80055 Portici (Napoli), Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(7), 2285; https://doi.org/10.3390/ijms21072285
Submission received: 17 January 2020 / Revised: 12 March 2020 / Accepted: 23 March 2020 / Published: 26 March 2020
(This article belongs to the Special Issue Role of Nutraceuticals in Metabolic and Gastrointestinal Disorders)

Abstract

:
Over the last few years, the application of nanotechnology to nutraceuticals has been rapidly growing due to its ability to enhance the bioavailability of the loaded active ingredients, resulting in improved therapeutic/nutraceutical outcomes. The focus of this work is nanoprebiotics and nanoprobiotics, terms which stand for the loading of a set of compounds (e.g., prebiotics, probiotics, and synbiotics) in nanoparticles that work as absorption enhancers in the gastrointestinal tract. In this manuscript, the main features of prebiotics and probiotics are highlighted, together with the discussion of emerging applications of nanotechnologies in their formulation. Current research strategies are also discussed, in particular the promising use of nanofibers for the delivery of probiotics. Synbiotic-based nanoparticles represent an innovative trend within this area of interest. As only few experimental studies on nanoprebiotics and nanoprobiotics are available in the scientific literature, research on this prominent field is needed, covering effectiveness, bioavailability, and safety aspects.

Graphical Abstract

1. Nanonutraceuticals

1.1. Nutraceuticals

Beside the emerging need for natural origin alternatives to pharmaceuticals, the interest is focusing more and more on possible applications of food derived products that can be used as tools to prevent (and in some cases also cure) or delay the onset of a health issue [1,2,3]. Nutraceuticals, are a novel toolbox not completely explored so far for its full potential in medicine [4,5,6,7]. Nutraceuticals, a portmanteau of the words ‘nutrition’ and ‘pharmaceutical’ [2], have been defined as “the phytocomplex if they derive from a food of vegetal origin, and as the pool of the secondary metabolites if they derive from a food of animal origin, concentrated and administered in the more suitable pharmaceutical form” [8]. Examples of substances that have nutritional and nutraceutical interest are antioxidants, vitamins, polyunsaturated fatty acids, dietary fibres, prebiotics, and probiotics [9]. Nutraceuticals reside nowadays in a gray area between pharmaceuticals and food; their safety and efficacy in health conditions and safety must be substantiated by clinical data; moreover, there is lack of a shared regulatory system for them [7,10].

1.2. From Nanopharmaceuticals to Nanonutraceuticals

1.2.1. Characteristics of Nanoparticles and General Classification

Within the different definitions of nanomaterials, these can be described as the products of nanotechnology, characterized by at least one dimension within the size range below 100 nanometers [11,12,13]. Due to their remarkable properties and versatility, nanomaterials are being exploited in different fields, e.g., agriculture, health, electronics, cosmetics [14,15,16,17,18], representing a great challenge, in particular, in food science and technology, environment, and human health [19]. The progress in pharmaceutical nanotechnology has led to a new class of products, the so-called nanopharmaceuticals [20,21], defined as pharmaceutical drug molecules formulated in nanomaterials. Different types of nanoformulations are being exploited for the treatment of neurodegenerative diseases, cancer, infectious diseases, and others [22,23,24,25,26]. Besides, nanomaterials are also succeeding in offering new advanced tools for imaging and diagnosis [27] which, combined with therapy, have been proposed as nanotheranostics. These formulations are also being tailored for personalized medicine.
Nanoparticles can be produced from natural (e.g., proteins, polysaccharides, lipids) and from synthetic (e.g., polymers) sources. Ideally, materials should be biocompatible, biodegradable, and biotolerable, namely the way by which designed materials are tolerated by the body, and of generally recognized as safe (GRAS) status, in order to be used in pharmaceutical and nutraceutical products. Among the available options, and if the nanoparticles are intended for oral administration (as happens with nanonutraceuticals), lipid nanoparticles are of special interest [28,29,30,31]. Lipids are known for their role as absorption enhancers in the gut, which contribute to improving the oral bioavailability of several drugs and biomolecules. Besides this, the loading of poorly soluble drugs into lipid nanoparticles overcome the limitations encountered in their formulation into final products. Lipid nanoparticles can be produced from well-known lipids existing both in the human body and in foodstuff (e.g., fatty acids, triglycerides, phospholipids, waxes, cholesterol) thereby enhancing their biodegradability, and biocompatibility profiles [32].
Among polysaccharides, chitosan [33,34,35,36,37] and alginate [33,38,39], have been frequently used in the production of nanoparticles for oral delivery. Being a mucoadhesive polysaccharide, chitosan is able to increase cellular permeability and improves the bioavailability of orally administered drugs and proteins. Moreover, the molecule itself exhibits antimicrobial properties, and has a low toxicity. The molecule has chemical functional groups that can be modified for site specific targeting. Alginate is also a versatile mucoadhesive natural polymer with very low toxicity in vivo. Alginate nanoparticles have a hydrophilic character with improved loading capacity for hydrophilic drugs, being able to modify their release profile. Alginate nanoparticles are reported as adjuvants in vaccinations and can be produced conjugated with dextran to modify the release profile of proteins and other macromolecules intended for oral administration [40].
Nanopharmaceuticals and nanonutraceuticals are obtained, respectively, when a pharmaceutical or a nutraceutical is formulated in nanoparticles. The rationale for their development is mainly addressed to improve the physicochemical properties (e.g., solubility) and pharmacokinetic parameters (tmax, Cmax, area under the plasma drug concentration–time curve (AUC)), with the ultimate aim to reduce the dose required to observe the therapeutic/nutraceutical outcome and thus the possible risk of toxicity [41,42,43]. Parameters, such as efficiency, quality, and safety should therefore be considered. Nevertheless, regulatory issues related to nanopharmaceuticals still need further developments [44].

1.2.2. Emerging Area of Applications

Nanopharmaceuticals and the great change of the pharmaceutical industry have a great impact also on nutraceuticals. The recent work of Agarwal et al. [45] gives the patented and approval scenario of nanopharmaceuticals with regards to biomedical application, manufacturing procedure, and safety aspects.
Wu et al. [46] highlighted how nanotherapeutics and nanopharmaceuticals could lead to a more precise individual diagnosis, improve targeted therapies, reduce side effects, and enhance therapeutic monitoring. The same review also underlines that the field of nanomedicine is at its early stage and that further efforts to translate their potential into clinical trials and medical practice are still needed.
A growing number of studies are addressed towards the application of nanotechnologies to nutraceuticals [47,48,49,50] in order to obtain improved bioavailability, delivery, and effect. This leads to the development of an emerging area of innovative products: the nanonutraceuticals [51,52,53].
Nanotechnology can be used to improve absorption, bioavailability, stability, and controlled release of nutrients and nutraceuticals, thereby increasing health benefits; some examples of potential advantages of applications of nanotechnology on the nutraceuticals are (i) efficient encapsulation; (ii) smart delivery and release from a nanoformulation. For example, research on encapsulation of nutraceuticals into biodegradable, environmentally friendly nanocarriers, is ongoing to increase their absorption and their therapeutic potential.
The nanonutraceutical formulations represent a valuable and promising strategy to maintain nutraceutical health beneficial properties at a nano level, to guarantee safety and efficacy, when used in managing health conditions, particularly for patients who are not eligible for a conventional pharmacological therapy. Follow-up studies, as reported by recent works [54,55,56,57], and communication strategies [58], are needed for both the nanopharmaceuticals and nanonutraceuticals [59,60], in view of expanding the area of interest to different health conditions. For instance, Aditya et al. [61] describe the current status of the various delivery systems that are used for the delivery of hydrophilic bioactive compounds and discuss future prospects to be explored for the delivery of hydrophilic bioactive compoundse.g., niosomes, bilosomes, cubosomes.

2. Focus on Nanotechnologies Applied to Prebiotics, Probiotics, and Synbiotics

Focus of this perspective is the application of nanotechnologies to food supplements containing prebiotics, probiotics, and synbiotics. This section consists of (i) shot on prebiotics, probiotics, and synbiotics; (ii) definition and delineation of nano-prebiotics, nano-probiotics, and nano-synbiotics.

2.1. An Overview on Prebiotics, Probiotics, and Synbiotics

2.1.1. Prebiotics

Prebiotics [62,63,64,65,66] are a special form of dietary fiber with health benefits, which invoke alterations in the host microbial ecosystem, not only in the gut, via their selective administration by live host microbes [67]. Food ingredients like prebiotics are classified on the basis of some principles, such as resistance to digestion in upper alimentary tract, selective stimulation of probiotic growth, beneficial health effects in the host, stability in different conditions of food/feed processing, and fermentation process through intestinal microbiota. They are found in various sources, including some non-digestible oligosaccharides, non-digestible carbohydrates, yacon, unrefined wheat, unrefined barley, soybeans, raw oats, breast milk, and inulin sources (e.g., chicory roots and Jerusalem artichoke) [68]. Some compounds found in prebiotics are soya-oligosaccharide, xylo-oligosaccharide, pyrodextrins, gluco-oligosaccharide, lactulose, malto-oligosaccharide, galactans (galacto-oligosaccharide (GOS)), oligofructose, isomalto-oligosaccharide (IOS), fructans (FOS and inulin), mannan-oligosaccharide (MOS), lactitol, and non-starch polysaccharides (NSP). Figure 1 gives an overview of prebiotics.
Several mechanisms are involved in the bioactivity of prebiotics and probiotics [69,70], as described in Figure 2.
The metabolic products of such microorganisms can drop the gastrointestinal (GI) pH by carbohydrate fermentation via Bifidobacteria and Lactobacillus thereby influencing mineral uptake, growth, and spread of gut microbiota, epithelial integrity, and hormonal regulation. They also are able to enhance the absorption of trace elements and especially of iron and act on the regulation of body immune function. The prebiotics can use the short-chain fatty acids (SCFAs) as an energy source.

2.1.2. Probiotics

The FAO (Food and Agriculture Organization) and WHO (World Health Organization) have defined probiotics as non-pathogenic living microorganisms that ensure host health if used properly in foods or as dietary supplements [71,72]. Probiotics come from different sources, such as various natural environments, human gut microbiota, and foods. The main properties of probiotics like the ability to survive through the gastrointestinal tract, the resistance against bile and gastric acidity, and the stimulation of the activity of bile salt hydrolase, promote health benefits to the host [68,73,74,75,76,77,78,79,80,81]. The count of probiotic bacteria (colony-forming units (CFU)/g) in probiotic-containing products differ among the countries; for example, 107 CFU/g in the USA and 109 CFU/g in Canada. The effective dose generally contains >106–108 CFU/g or >108–1010 CFU/d of live probiotic bacteria [82,83]. Most probiotics are found in Gram-positive bacteria, including Streptococcus, Bacillus, Lactobacillus, Enterococcus, and Pediococcus. The probiotics can also include fungal and yeast species such as Saccharomyces cerevisiae and Kluyveromyces. Only some microorganisms such as Lactobacillus spp., Bifidobacterium spp., and Lactococcus are known as generally recognized as safe (GRAS) despite the existence of diverse microorganisms which can act as probiotics with health benefits [84,85,86]. Figure 3 gives an overview of probiotics.
The reported key mechanisms of action of probiotics [87] have been mentioned as follows (see Figure 2): enhancement of epithelial barrier, modulation of insulin-sensitive tissues, synthesis of antimicrobial substances, multi-pathogen competition, and induction of mucin secretion. The probiotics are able to adhere to epithelium, resulting in microbial elimination. They also modulate the immune function via the stimulation of signaling pathways to upregulate anti-inflammatory cytokines and growth factors, to differentiate T-regulatory cells (Tregs), and to interact with the gut-brain axis (GBA) by endocrine regulation and neurologic functions.

2.1.3. Synbiotics

The synbiotic agents are a combination of prebiotics and probiotics with beneficial effects on host through the enhancement of activity and survival of beneficial microorganisms in the gastrointestinal tract, so that they can selectively provoke the growth and stimulate the metabolism of one or more health-promoting bacteria, thereby enhancing the host welfare [88,89,90,91,92,93,94,95,96,97]. The most important issue in the design of synbiotics resides in the prebiotic and probiotic selection criteria and requirements, which should be clearly described.

2.1.4. Health Promoting Effect of Prebiotics, Probiotics, and Synbiotics

The International Scientific Association for Probiotics and Prebiotics (ISAPP) introduced a wide range of products containing the probiotics with health promoting effects, including non-edible products (e.g., vaginal preparations), baby formulas (e.g., first milk), drugs, therapeutic supplements (e.g., for enteral nutrition), and foods (e.g., fermented milk with reportedly health beneficial effects) [98].
Some of the reported beneficial effects of probiotics in human health include anticancer [99,100,101,102,103,104,105,106,107,108,109,110,111], anti-allergic [112,113], anti-diabetic [114,115,116], anti-obesity [117,118,119,120], anti-pathogenic [121,122], immunomodulatory [123], and anti-inflammatory [124,125,126,127] activities [128], as reported in Table 1. In an in vitro study, Sequential Window Acquisition of All Theoretical Mass Spectra (SWATH-MS) as a quantitative analysis technique was applied to evaluate the proteomic profile of colon cancer cells in Lactobacillus kefiri SGL 13, and the results indicated antiproliferative and pro-apoptotic activities for this strain on human colon adenocarcinoma cell line HT29 [99]. In another study, the airway hyper reactivity was suppressed in ovalbumin-sensitized samples by Lactobacillus spp. (such as Lactobacillus and Pediococcus) via a reduction in the level of Th2 cytokines, OVA-specific IgE and IgG1 as well as an increase in the level of IgG2a [112]. Lactobacillus fermentum cell-free supernatant (LCFS) caused cancer cell death in 3D HCT-116 conditions through the induction of apoptosis in the colon cancer cell line and the antiproliferative activity by the inhibition of NF-κB signaling [129]. The use of lactoferrin and Bifidobacterium longum BB536 managed the enteropathy caused by diclofenac in rat samples by modulating the proinflammatory pathway of TLR-2/-4/NF-kB [130]. Othman et al. [131], studied the effect of inactivated Bifidobacterium longum intake on obese diabetes affected mice. They reported a significant decrease of body weight gain, adipose tissue mass and blood glucose levels, as well as a significant reduction in blood glucose after a 5 weeks treatment. The treatment also resulted in reduced levels of cholesterol and triglycerides [131].
The administration of three strains of Bifidobacteria in the adult rats improved neuronal plasticity and cognitive behavior [132].
Prebiotics have been reported to have different activities; for example, generation of bacteriocins, maintenance of gut health [133], possibility to be used as food additive and starter culture, clearance of cholesterol [134,135], potentiation of immune defense [136], inhibition of constipation and risk of obesity [137,138], inhibition of colitis [139], protection of colon and other organs against cancer [140,141,142], reduction of cardiovascular disease risk factors, antioxidant activity [143,144], over-bioavailability [145]. According to scientific published data, the administration of oligofructose-enriched inulin (OEI) promotes malondialdehyde content, lipid profile, glycemic indices, and antioxidant level in female patients suffering from type II diabetes [146]. The supplementation of inulin in shaken cultures was found to increase the growth rate of L. plantarum ST16 [147]. Based on the findings from Ramos et al. [148], the administration of fructooligosaccharides (FOS) was tolerated and decreased the total and free p-cresyl sulfate (PCS) in the serum samples of patients with non-diabetic chronic kidney disease (NDD-CKD).
The therapeutic potential of synbiotics has been comprehensively discussed in a recent review published by Flesch et al. [149]. According to their findings, the patients with irritable bowel syndrome (IBS) when receiving B. longum BB536 and L. rhamnosus HN001 plus vitamin B6 showed restoration of intestinal permeability and gut microbiota, as well as amelioration of the disease symptoms [150]. In the research of Mohan et al., the synbiotic AMFTM 15+ manuka honey yogurt showed antibacterial properties, followed by increasing probiotic bacteria and producing lactic and propionic acids [151]. A study reported gut health enhancement following the administration of seaweed-based synbiotic of Gracilaria coronopifolia which caused the reduction of inflammation, the generation of reactive oxygen species (ROS), and diminution of the oxidative stress-induced cell damage [152]. According to Sarwar et al., the textural properties, such as adhesiveness, cohesiveness, and hardness, were enhanced following the co-administration of inulin and Saccharomyces boulardii [153]. In Table 1 an updated overview of in vitro and in vivo studies on prebiotic, probiotic, and synbiotic products is given.

2.2. Nano-Prebiotics, Nano-Probiotics, and Nano-Synbiotics

Recently, emerging applications of nanotechnologies in prebiotics and probiotics have been developed and carried out as reported in Table 2 [154,155,156,157,158,159,160,161,162,163,164,165,166,167].
Caneus et al. [168] remarked how nanomedicine, together with the known practices of prebiotics, probiotics, and synbiotics, represents a valuable approach in creating an optimal environment within the gastrointestinal tract.
Exploring the nanonization strategies of probiotics and the utility of nanoprobiotics in the delivery of encapsulated bacteria is being carried out. For encapsulation of probiotic have been used mainly nanoparticles i.e., with of selenium and gold particles of a size in the range 10–1000 nm; nanolayers, consisting of at least three layers of a charged polyelectrolyte, a polymeric layer, and a functionalized polysaccharide or polyether; nanoemulsions consisting of a liquid phase dispersion in another liquid phase with droplet size less 200 nm; nanobeads (nanosized bacteria-enabled autonomous delivery system) and emerging product of nanofibers [169]. The best technique for probiotics encapsulation was mainly chosen for protecting the cells against an adverse environment in the gastrointestinal tract, in order to allow their release in a viable and metabolically active state in the intestine [170].
Kazmierczak et al. [171] describe an innovative engineering approach to load such nanoparticles onto a biological “mailman” (a novel, nontoxic, therapeutic strain of Salmonella typhimurium engineered to preferentially and precisely seek out, penetrate, and hinder prostate cancer cells as biological delivery system) that will deliver the therapeutics to a target site. Another example of probiotic bacteria encapsulated with nanoparticles was given by Hu et al. [172] that showed how coating live bacterial cells with synthetic nanoparticles represents a promising strategy to engineer efficient and versatile DNA vaccines. Feher et al. [173] have reported the use of nano-sized particles of probiotics for preventing and treating neuroinflammation.
Probiotics are indeed receiving special interest as an alternative to the classical antibiotics to overcome bacterial resistance. As prebiotics enhance the activity of probiotics, Kim et al. [162] proposed the development of a prebiotic formulation composed of Pediococcus acidilactidi loaded in phthalyl dextran nanoparticles by conjugating phthalic anhydride with dextran [162]. The authors evaluated the cellular effects of the produced nanomaterial and checked the antimicrobial properties of the probiotics. The loading of P. acidilactidi into phthalyl dextran nanoparticles was found to enhance the production of antimicrobial peptides by probiotics by a self-defense mechanism, with improved antimicrobial effect against Gram (+) and Gram (−) micro-organisms compared to the probiotics alone. The same authors previously reported that prebiotic phthalyl inulin nanoparticles could also enhance the antimicrobial activities of P. acidilactici [174].
Hong et al. also reported the enhanced antimicrobial activity of phthalyl pullulan nanoparticles treated with L. plantarum against Escherichia coli K99 and Listeria monocytogenes [164]. The nanoparticles were internalized into the L. plantarum by an energy-dependent and galactose transporter-dependent mechanism and a higher amount of plantaricin, a natural antibacterial peptide, was secreted from the developed nanoprobiotic than from probiotic alone.
The use of spores from probiotics have been recently proposed as a delivery system for chemotherapeutic drugs. Song et al. [175] produced deoxycholic acid-modified spores to be loaded with doxorubicin and sorafenib as an approach for autonomous production of nanoparticles in the gastrointestinal tract. Such approach envisions drug protection upon oral administration to improve bioavailability. Besides, the release is based on the disintegrated hydrophobic protein and the hydrophilic deoxycholic acid with enhanced uptake by the epithelial cells via the bile acid pathway, increasing basolateral drug release.
The anticancer activity of silver/Lactobacillus rhamnosus GG nanoparticles was described by Aziz et al. [155]. Using the MTT assay, the authors demonstrated that the viability of HT-29 cell lines has been significantly reduced when applying the highest tested nanoparticle concentration, leading to apoptosis. The method of synthesizing silver/Lactobacillus rhamnosus GG nanoparticles was also found to be cost-effective, offering a viable nanoprobiotic approach for biomedical applications.
It is worth mentioning the work of Fung et al. [176] where, by investigating the agrowaste-based nanofibers as a probiotic encapsulant, has proposed the use of nanofibers for the nanoencapsulation of L. acidophilus using 8% poly(vinyl alcohol) to produce nanofibers by electrospinning technology. The authors suggested how thermal behavior of nanofibers suggested possible thermal protection of probiotics in heat-processed foods. Nagy et al. [177] by investigating the suitability of electrospinning for biodrugs delivery to produce vaginal drug delivery systems, concluded how nanofibers can provide long term stability for huge amounts of living bacteria if they are kept at (or below) 7 °C. The recent work of Zupancic et al. [178], who studied the incorporation of a range of safe lactic acid bacteria into poly(ethylene oxide)-based nanofibers, evidenced that all of the lactic acid bacteria were viable after incorporation into nanofibers, with 0–3 log CFU/mg loss in viability, depending on the species. Moreover, the authors reported that viability can be correlated with the hydrophobicity and to the extreme length of lactic acid bacteria, whereas a horizontal or vertical electrospinning set-up did not have any role. Development of nanofibers via electrospinning has a great potential and use in pharmaceutical and food industry for their properties i.e., sterile nature, biocompatibility, adhesiveness, efficiency, and as vehicle for controlled and sustained release in drug delivery [179,180,181,182]. Electrospinning and electrospraying represent innovative technologies for the delivery of nutraceuticals [183].
An example of nanolayers coated probiotics has been given by Franz et al. [184] who developed layer-by-layer nano self-assembly coating of Allochromatium vinosum with different polyelectrolyte combinations and investigated substrate uptake in bacteria: surface charge neither affected sulfide uptake nor the contact formation between the cells and solid sulfur, whereas increasing layers slowed or inhibited the uptake of sulfide and elemental sulfur.
The recent work of Ebrahimnejad et al. [185] described the use of chitosan for nanoencapsulation of L. acidophilus as probiotic bacteria, by concluding how nanoencapsulation of probiotic bacteria represents a promising strategy in enhancing the viability and survival of them against gastro-intestinal environmental conditions.
Ranjan et al. [186] reported physicochemical characterization and potential prebiotic effect of whey protein isolate/inulin nano complex.
Atia et al. [167] developed an encapsulated oral-symbiotic supplement by studying the effect of adding inulin in alginate beads and observed their ability to protect three probiotic strains, namely, P. acidilactici, L. reuteri, and L. salivarius. The antimicrobial and probiotic properties of bacterial strains were found not to be affected by the encapsulation.
Krithika and Preetha [166] have developed a protein-based inulin incorporated symbiotic nanoemulsion for enhanced stability of probiotic; whey protein concentrate/inulin nano complex can be recommended as a delivery system for various probiotics in food products.
Salmerón et al. [187] reported the development fermented beverages with synbiotic properties, and the incorporation of nanoparticles with unique and specific bioactivity, to improve organoleptic characteristics, absorption, and delivery of nutrients and bioactive compounds which has opened a new horizon in this segment of food created to improve human health and well-being.
Formulation of protein-based inulin incorporated synbiotic nanoemulsion for enhanced stability of probiotic are currently studied extensively.
It is worth mentioning the work of Rezaee et al. [188] that investigated the antimicrobial activity of Ag and TiO2 nano-particles on three species of Lactobacillus i.e., L. casei ATCC 39392, L. plantarum ATCC 8014, and L. fermentum ATCC 9338 in the presence and absence of raffinose, lactulose, and inulin, respectively. The results indicated that silver nanoparticles decreased 85%, 85%, and 71% of L. casei, L. plantarum, and L. fermentum, respectively, after 48 h and decreased percentages of L. casei, L. plantarum, and L. fermentum that were 16%, 64%, and 4% in the presence of the prebiotics. Nano TiO2 particles decreased 59%, 85%, and 61% of L. casei, L. plantarum, and L. fermentum, respectively, after 48 h, and decreased percentages of L. casei, L. plantarum, and L. fermentum which were 16%, 2%, and 4% in the presence of these prebiotics.
The treatment of gastrointestinal disorders (e.g., diarrhea) using nanoprobiotics is also a relatively unexplored field. Khan et al. [189] aimed at quantifying the concentration of nanomaterials commercialized in chocolates and evaluated their effect on a commercial probiotic formulation (containing Bacillus coagulans, Enterococcus faecalis, and Enterococcus faecium) usually used to treat diarrhea in children [189]. The known probiotic activities, such as acid production, biofilm formation, growth, and antibiotic resistance were observed from isolated bacteria, while the isolated titanium oxide nanoparticles from chocolates were shown to inhibit the growth and activity of the probiotic formulation in a concentration range of 125–500 µg/mL in vitro [189]. The outcomes of this study concluded that TiO2 in chocolate discourages the survival of probiotic bacteria in the gastrointestinal tract.
To trace target probiotics in situ and in real-time, Liu et al. [190] developed an in vivo probing strategy using persistent luminescence nanophosphors surface-modified by plasmid-like DNA as optical labelling and background-free fluorescence bioimaging as signal readout. The surface modification with DNA molecules was shown to promote the nanoparticles penetration into the bacteria and facilitated in vivo bioimaging. Such an approach opens new research perspectives in terms of food safety making use of nanotechnologies.

3. Conclusions

Only a few experimental studies are present in literature on nanoprebiotics and nanoprobiotics, while studies on this prominent issue are needed, covering effectiveness and safety aspects as it has been developed for pharmaceuticals. The potential of nanotechnologies in the food area is an emerging challenge as well as the nanonutraceuticals, which are an emerging field of study in the nutraceuticals area. Safety and regulatory aspects should be considered to depict the potentiality of nanoprobiotics and nanoprebiotics. Nanoformulation should be accompanied with regulatory requirements to ensure efficacy, safety, and authorization procedures. As a general guideline, the European Authority for Food Safety (EFSA) [191] has developed an approach for assessing the potential risks arising from the applications of nanoscience and nanotechnologies in the food and feed chain. Regarding prebiotics and probiotics, McClements and Xiao [192] developed a summary of the possible applications of inorganic and organic nanoparticles in foods, a description of the nanoparticle characteristics, and discussed the importance of the food matrix and gastrointestinal tract effects on nanoparticle properties as well as potential possible toxicity mechanisms of different food-grade nanoparticles. The same authors concluded, however, that many of these nanoparticles are unlikely to have adverse-side effects on human health in line with previously reported data [193]. Nonetheless, in order to assess the effective use of food-grade nanoparticles, further studies are expected to exploit and assess safety, improved bioavailability, and efficacy.

Author Contributions

Conceptualization, A.D., A.N., A.S.; Methodology, M.L., E.B.S., R.C., A.S.; Validation, R.C., A.S., E.N., A.G.A., and A.D.; Writing—Review and Editing, A.N., A.D., R.C., M.L., E.B.S., A.S.; Visualization, E.N., M.L., E.B.S.; Supervision, A.D., A.S.; all Authors contributed equally to the manuscript final preparation. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge the support of the research project: Nutraceutica come supporto nutrizionale nel paziente oncologico, CUP: B83D18000140007. E. B. Souto acknowledges the sponsorship of the projects M-ERA-NET-0004/2015-PAIRED and UIDB/04469/2020 (strategic fund), receiving support from the Portuguese Science and Technology Foundation, Ministry of Science and Education (FCT/MEC) through national funds, and co-financed by FEDER, under the Partnership Agreement PT2020.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Santini, A.; Novellino, E. Nutraceuticals: Beyond the diet before the drugs. Curr. Bioact. Comp. 2014, 10, 1–12. [Google Scholar] [CrossRef]
  2. Santini, A.; Novellino, E. To Nutraceuticals and back: Rethinking a concept. Foods 2017, 6, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Abenavoli, L.; Izzo, A.A.; Milić, N.; Cicala, C.; Santini, A.; Capasso, R. Milk thistle (Silybum marianum): A concise overview on its chemistry, pharmacological, and nutraceutical uses in liver diseases. Phytother. Res. 2018, 32, 2202–2213. [Google Scholar] [CrossRef] [PubMed]
  4. Durazzo, A. Extractable and Non-extractable polyphenols: An overview. In Non-Extractable Polyphenols and Carotenoids: Importance in Human Nutrition and Health; Saura-Calixto, F., Pérez-Jiménez, J., Eds.; Royal Society of Chemistry: London, UK, 2018; pp. 1–37. [Google Scholar]
  5. Santini, A.; Novellino, E. Nutraceuticals-shedding light on the grey area between pharmaceuticals and food. Expert Rev. Clin. Pharmacol. 2018, 11, 545–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Durazzo, A.; Lucarini, M. A current shot and re-thinking of antioxidant research strategy. Braz. J. Anal. Chem. 2018, 5, 9–11. [Google Scholar] [CrossRef]
  7. Santini, A.; Cammarata, S.M.; Capone, G.; Ianaro, A.; Tenore, G.C.; Pani, L.; Novellino, E. Nutraceuticals: Opening the debate for a regulatory framework. Br. J. Clin. Pharmacol. 2018, 84, 659–672. [Google Scholar] [CrossRef] [Green Version]
  8. Daliu, P.; Santini, A.; Novellino, E. A decade of nutraceutical patents: Where are we now in 2018? Expert Opin. Ther. Pat. 2018, 28, 875–882. [Google Scholar] [CrossRef]
  9. Durazzo, A.; D’Addezio, L.; Camilli, E.; Piccinelli, R.; Turrini, A.; Marletta, L.; Marconi, S.; Lucarini, M.; Lisciani, S.; Gabrielli, P.; et al. From plant compounds to botanicals and back: A current snapshot. Molecules 2018, 23, 1844. [Google Scholar] [CrossRef] [Green Version]
  10. Santini, A.; Tenore, G.C.; Novellino, E. Nutraceuticals: A paradigm of proactive medicine. Eur. J. Pharm. Sci. 2017, 96, 53–61. [Google Scholar] [CrossRef]
  11. De Jong, W.H.; Borm, P.J.A. Drug delivery and nanoparticles: Applications and hazards. Int. J. Nanomed. 2008, 3, 133–149. [Google Scholar] [CrossRef] [Green Version]
  12. Jeevanandam, J.; Barhoum, A.; Chan, Y.S.; Dufresne, A.; Danquah, M.K. Review on nanoparticles and nanostructured materials: History, sources, toxicity and regulations. Beilstein. J. Nanotechnol. 2018, 9, 1050–1074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Auffan, M.; Rose, J.; Bottero, J.Y.; Lowry, G.V.; Jolivet, J.P.; Wiesner, M.R. Towards a definition of inorganic nanoparticles from an environmental, health and safety perspective. Nat. Nanotechnol. 2009, 4, 634–641. [Google Scholar] [CrossRef] [PubMed]
  14. Chaudhry, Q.; Scotter, M.; Blackburn, J.; Ross, B.; Boxall, A.; Castle, L.; Aitken, R.; Watkins, R. Applications and implications of nanotechnologies for the food sector. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2008, 25, 241–258. [Google Scholar] [CrossRef] [PubMed]
  15. Ljubimova, J.Y.; Holler, E. Biocompatible nanopolymers: The next generation of breast cancer treatment? Nanomedicine 2012, 7, 1467–1470. [Google Scholar] [CrossRef] [Green Version]
  16. Peters, R.J.B.; Bouwmeester, H.; Gottardo, S.; Amenta, V.; Arena, M.; Brandho, P.; Marvin, H.J.P.; Mech, A.; Moniz, F.B.; Pesudo, L.Q.; et al. Nanomaterials for products and application in agriculture, feed and food. Trends Food Sci. Technol. 2016, 54, 155–164. [Google Scholar] [CrossRef]
  17. Dudefoi, W.; Villares, A.; Peyron, S.; Moreau, C.; Ropers, M.-H.; Gontard, N.; Cathala, B. Nanoscience and nanotechnologies for biobased materials, packaging and food applications: New opportunities and concerns. Innov. Food Sci. Emerg. Technol. 2018, 46, 107–121. [Google Scholar] [CrossRef] [Green Version]
  18. He, X.; Deng, H.; Hwang, H.-M. The current application of nanotechnology in food and agriculture. J. Food Drug Anal. 2019, 27, 1–21. [Google Scholar] [CrossRef] [Green Version]
  19. Das, G.; Patra, J.K.; Paramithiotis, S.; Shin, H.S. The sustainability challenge of food and environmental nanotechnology: Current status and imminent perceptions. Int. J. Environ. Res. Public Health 2019, 16, 4848. [Google Scholar] [CrossRef] [Green Version]
  20. Farokhzad, O.C.; Langer, R. Nanomedicine: Developing smarter therapeutic and diagnostic modalities. Adv. Drug Deliv. Rev. 2006, 58, 1456–1459. [Google Scholar] [CrossRef]
  21. Davis, M.E.; Chen, Z.G.; Shin, D.M. Nanoparticle therapeutics: An emerging treatment modality for cancer. Nat. Rev. Drug Discov. 2008, 7, 771–782. [Google Scholar] [CrossRef]
  22. Norouzi, M.; Amerian, M.; Amerian, M.; Atyabi, F. Clinical applications of nanomedicine in cancer therapy. Drug Discov. Today 2019. [Google Scholar] [CrossRef] [PubMed]
  23. Teleanu, D.M.; Chircov, C.; Grumezescu, A.M.; Teleanu, R.I. Neuronanomedicine: An Up-to-Date Overview. Pharmaceutics 2019, 11, 101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Sánchez-López, E.; Guerra, M.; Dias-Ferreira, J.; Lopez-Machado, A.; Ettcheto, M.; Cano, A.; Espina, M.; Camins, A.; Garcia, M.L.; Souto, E.B. Current Applications of Nanoemulsions in Cancer Therapeutics. Nanomaterials 2019, 9, 821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Andreani, T.; Severino, P.; de Hollanda, L.M.; Vazzana, M.; Souto, S.B.; Santini, A.; Silva, A.M.; Souto, E.B. Cancer therapies: Applications, nanomedicines and nanotoxicology. In Nanostructures for Cancer Therapy; Ficai, A., Grumezescu, A.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; Chapter 9; pp. 241–260. [Google Scholar] [CrossRef]
  26. Do Ceu Texeira, M.; Santini, A.; Souto, E.B. Nanocancer therapies: Drug delivery formulation and nanotoxicology. In Nanostructures for Antimicrobial Therapy—Micro and Nano Technologies; Multi-Volume SET I-V: Nanostructures in Therapeutic Medicine Series; Elsevier: Amsterdam, The Netherlands, 2017; Chapter 8; pp. 203–222. [Google Scholar] [CrossRef]
  27. Petros, R.A.; DeSimone, J.M. Strategies in the design of nanoparticles for therapeutic applications. Nat. Rev. Drug Discov. 2010, 9, 615–627. [Google Scholar] [CrossRef] [PubMed]
  28. Doktorovova, S.; Kovacevic, A.B.; Garcia, M.L.; Souto, E.B. Preclinical safety of solid lipid nanoparticles and nanostructured lipid carriers: Current evidence from in vitro and in vivo evaluation. Eur. J. Pharm. Biopharm. 2016, 108, 235–252. [Google Scholar] [CrossRef] [PubMed]
  29. Souto, E.B.; Muller, R.H. Lipid nanoparticles: Effect on bioavailability and pharmacokinetic changes. In Drug Delivery; Springer: Berlin/Heidelberg, Germany, 2010. [Google Scholar] [CrossRef]
  30. Martins, S.; Silva, A.C.; Ferreira, D.C.; Souto, E.B. Improving oral absorption of Salmon calcitonin by trimyristin lipid nanoparticles. J. Biomed. Nanotechnol. 2009, 5, 76–83. [Google Scholar] [CrossRef]
  31. Muller, R.H.; Runge, S.; Ravelli, V.; Mehnert, W.; Thunemann, A.F.; Souto, E.B. Oral bioavailability of cyclosporine: Solid lipid nanoparticles (SLN) versus drug nanocrystals. Int. J. Pharm. 2006, 317, 82–89. [Google Scholar] [CrossRef]
  32. Severino, P.; Pinho, S.C.; Souto, E.B.; Santana, M.H. Polymorphism, crystallinity and hydrophilic-lipophilic balance of stearic acid and stearic acid-capric/caprylic triglyceride matrices for production of stable nanoparticles. Colloids Surf. B Biointerfaces 2011, 86, 125–130. [Google Scholar] [CrossRef]
  33. Andreani, T.; Fangueiro, J.F.; Severino, P.; Souza, A.L.R.; Martins-Gomes, C.; Fernandes, P.M.V.; Calpena, A.C.; Gremiao, M.P.; Souto, E.B.; Silva, A.M. The Influence of Polysaccharide Coating on the Physicochemical Parameters and Cytotoxicity of Silica Nanoparticles for Hydrophilic Biomolecules Delivery. Nanomaterials 2019, 9, 1081. [Google Scholar] [CrossRef] [Green Version]
  34. Ferreira da Silva, C.; Severino, P.; Martins, F.; Santana, M.H.; Souto, E.B. Didanosine-loaded chitosan microspheres optimized by surface-response methodology: A modified “Maximum Likelihood Classification” approach formulation for reverse transcriptase inhibitors. Biomed. Pharmacother. 2015, 70, 46–52. [Google Scholar] [CrossRef]
  35. Severino, P.; Da Silva, C.F.; Dalla Costa, T.C.; Silva, H.; Chaud, M.V.; Santana, M.H.; Souto, E.B. In vivo absorption of didanosine formulated in pellets composed of chitosan microspheres. In Vivo 2014, 28, 1045–1050. [Google Scholar] [PubMed]
  36. Severino, P.; de Oliveira, G.G.G.; Ferraz, H.G.; Souto, E.B.; Santana, M.H.A. Preparation of gastro-resistant pellets containing chitosan microspheres for improvement of oral didanosine bioavailability. J. Pharm. Anal. 2012, 2, 188–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Severino, P.; Souto, E.B.; Pinho, S.C.; Santana, M.H. Hydrophilic coating of mitotane-loaded lipid nanoparticles: Preliminary studies for mucosal adhesion. Pharm. Dev. Technol. 2013, 18, 577–581. [Google Scholar] [CrossRef] [PubMed]
  38. Severino, P.; Chaud, M.V.; Shimojo, A.; Antonini, D.; Lancelloti, M.; Santana, M.H.; Souto, E.B. Sodium alginate-cross-linked polymyxin B sulphate-loaded solid lipid nanoparticles: Antibiotic resistance tests and HaCat and NIH/3T3 cell viability studies. Colloids Surf. B Biointerfaces 2015, 129, 191–197. [Google Scholar] [CrossRef] [PubMed]
  39. Severino, P.; da Silva, C.F.; Andrade, L.N.; de Lima Oliveira, D.; Campos, J.; Souto, E.B. Alginate Nanoparticles for Drug Delivery and Targeting. Curr. Pharm. Des. 2019, 25, 1312–1334. [Google Scholar] [CrossRef]
  40. Sarei, F.; Mohammadpour Dounighi, N.; Zolfagharian, H.; Khaki, P.; Moradi Bidhendi, S. Alginate Nanoparticles as a Promising Adjuvant and Vaccine Delivery System. Indian J. Pharm. Sci. 2013, 75, 442–449. [Google Scholar] [CrossRef] [Green Version]
  41. Havel, H.A. Where are the nanodrugs? An industry perspective on development of drug products containing nanomaterials. AAPS J. 2016, 18, 1351–1353. [Google Scholar] [CrossRef]
  42. Feng, J.; Markwalter, C.E.; Tian, C.; Armstrong, M.; Prud’homme, R.K. Translational formulation of nanoparticle therapeutics from laboratory discovery to clinical scale. J. Transl. Med. 2019, 17, 200. [Google Scholar] [CrossRef]
  43. Öztürk-Atar, K.; Eroğlu, H.; Gürsoy, R.N.; Çaliş, S. Current advances in nanopharmaceuticals. J. Nanosci. Nanotechnol. 2019, 19, 3686–3705. [Google Scholar] [CrossRef]
  44. Souto, E.B.; Silva, G.F.; Dias-Ferreira, J.; Zielinska, A.; Ventura, F.; Durazzo, A.; Lucarini, M.; Novellino, E.; Santini, A. Nanopharmaceutics: Part I-Clinical Trials Legislation and Good Manufacturing Practices (GMP) of Nanotherapeutics in the EU. Pharmaceutics 2020, 12, 146. [Google Scholar] [CrossRef] [Green Version]
  45. Agarwal, V.; Bajpai, M.; Sharma, A. Patented and Approval Scenario of Nanopharmaceuticals with Relevancy to Biomedical Application, Manufacturing Procedure and Safety Aspects. Recent. Pat. Drug Deliv. Formul. 2018, 12, 40–52. [Google Scholar] [CrossRef] [PubMed]
  46. Wu, L.P.; Wang, D.; Li, Z. Grand challenges in nanomedicine. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 106, 110302. [Google Scholar] [CrossRef] [PubMed]
  47. Daliu, P.; Santini, A.; Novellino, E. From pharmaceuticals to nutraceuticals: Bridging disease prevention and management. Expert Rev. Clin. Pharmacol. 2019, 12, 1–7. [Google Scholar] [CrossRef] [PubMed]
  48. Durazzo, A.; Lucarini, M.; Souto, E.B.; Cicala, C.; Caiazzo, E.; Izzo, A.A.; Novellino, E.; Santini, A. Polyphenols: A concise overview on the chemistry, occurrence and human health. Phyt. Res. 2019, 33, 2221–2243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Durazzo, A.; Lucarini, M. Extractable and Non-extractable antioxidants. Molecules 2019, 24, 1933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Durazzo, A.; Lucarini, M. Editorial: The State of Science and Innovation of Bioactive Research and Applications, Health, and Diseases. Front. Nutr. 2019, 6, 178. [Google Scholar] [CrossRef]
  51. Watkins, R.; Wu, L.; Zhang, C.; Davis, R.M.; Xu, B. Natural product-based nanomedicine: Recent advances and issues. Int. J. Nanomed. 2015, 10, 6055–6074. [Google Scholar]
  52. Pimentel-Moral, S.; Teixeira, M.C.; Fernandes, A.R.; Arráez-Román, D.; Martínez-Férez, A.; Segura-Carretero, A.; Souto, E.B. Lipid nanocarriers for the loading of polyphenols—A comprehensive review. Adv. Colloid Interface Sci. 2018, 260, 85–94. [Google Scholar] [CrossRef]
  53. Singh, B. Nanonutraceuticals, 1st ed.; CRC Press: Boca Raton, FL, USA, 2018; 326p. [Google Scholar]
  54. Menditto, E.; Guerriero, F.; Orlando, V.; Crola, C.; Di Somma, C.; Illario, M.; Morisky, D.; Colao, A. Self-Assessment of Adherence to Medication: A Case Study in Campania Region Community-Dwelling Population. J Aging Res. 2015, 2015, 682503. [Google Scholar] [CrossRef]
  55. Putignano, D.; Bruzzese, D.; Orlando, V.; Fiorentino, D.; Tettamanti, A.; Menditto, E. Differences in drug use between men and women: An Italian cross sectional study. BMC Women’s Health 2017, 17, 73. [Google Scholar] [CrossRef] [Green Version]
  56. Menditto, E.; Cahir, C.; Aza-Pascual-Salcedo, M.; Bruzzese, D.; Poblador-Plou, B.; Malo, S.; Costa, E.; González-Rubio, F.; Gimeno-Miguel, A.; Orlando, V.; et al. Adherence to chronic medication in older populations: Application of a common protocol among three European cohorts. Patient Prefer. Adherence 2018, 12, 1975–1987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Iolascon, G.; Gimigliano, F.; Moretti, A.; Riccio, I.; Di Gennaro, M.; Illario, M.; Monetti, V.M.; Orlando, V.; Menditto, E. Rates and reasons for lack of persistence with anti-osteoporotic drugs: Analysis of the Campania region database. Clin. Cases Miner. Bone Metab. 2016, 13, 126–129. [Google Scholar] [CrossRef] [PubMed]
  58. Scala, D.; Menditto, E.; Armellino, M.F.; Manguso, F.; Monetti, V.M.; Orlando, V.; Antonino, A.; Makoul, G.; De Palma, M. Italian translation and cultural adaptation of the communication assessment tool in an outpatient surgical clinic. BMC Health Serv. Res. 2016, 16, 163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Wiwanitkit, V. Delivery of nutraceuticals using nanotechnology. Int. J. Pharm. Investig. 2012, 2, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Bernela, M.; Kaur, P.; Ahuja, M.; Thakur, R. Nano-based Delivery System for Nutraceuticals: The Potential Future. In Advances in Animal Biotechnology and Its Applications; Gahlawat, S., Duhan, J., Salar, R., Siwach, P., Kumar, S., Kaur, P., Eds.; Springer: Singapore, 2018. [Google Scholar]
  61. Aditya, N.P.; Espinosa, Y.G.; Norton, I.T. Encapsulation systems for the delivery of hydrophilic nutraceuticals: Food application. Biotechnol. Adv. 2017, 35, 450–457. [Google Scholar] [CrossRef] [Green Version]
  62. Thammarutwasik, P.; Hongpattarakere, T.; Chantachum, S.; Kijroongrojana, K.; Itharat, A.; Reanmongkol, W.; Tewtrakul, S.; Ooraikul, B. Prebiotics—A Review. Songklanakarin J. Sci. Technol. 2009, 31, 401–408. [Google Scholar]
  63. Patel, S.; Goyal, A. The current trends and future perspectives of prebiotics research: A review. Biotech 2012, 2, 115–125. [Google Scholar] [CrossRef] [Green Version]
  64. Al-Sheraji, S.H.; Ismail, A.; Manap, M.Y.; Mustafa, S.; Yusof, R.M.; Hassan, F.A. Prebiotics as functional foods: A review. J. Funct. Foods 2013, 5, 1542–1553. [Google Scholar] [CrossRef]
  65. Bindels, L.B.; Delzenne, N.M.; Cani, P.D.; Walter, J. Towards a more comprehensive concept for prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 303–310. [Google Scholar] [CrossRef]
  66. Hutkins, R.W.; Krumbeck, J.A.; Bindels, L.B.; Cani, P.D.; Fahey, G., Jr.; Goh, Y.J.; Hamaker, B.; Martens, E.C.; Mills, D.A.; Rastal, R.A.; et al. Prebiotics: Why definitions matter. Curr. Opin. Biotechnol. 2016, 37, 1–7. [Google Scholar] [CrossRef]
  67. Monteagudo-Mera, A.; Rastall, R.A.; Gibson, G.R.; Charalampopoulos, D.; Chatzifragkou, A. Adhesion mechanisms mediated by probiotics and prebiotics and their potential impact on human health. Appl. Microbiol. Biotechnol. 2019, 103, 6463–6472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Pandey, K.R.; Naik, S.R.; Vakil, B.V. Probiotics, prebiotics and synbiotics—A review. J. Food Sci. Technol. 2015, 52, 7577–7587. [Google Scholar] [CrossRef] [PubMed]
  69. Khangwal, I.; Shukla, P. Potential prebiotics and their transmission mechanisms: Recent approaches. J. Food Drug Anal. 2019, 27, 649–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, J.S.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, Types, Sources, Mechanisms, and Clinical Applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef] [Green Version]
  71. FAO. Guidelines for the Evaluation of Probiotics in Food; Report of a Joint FAO/WHO Working Group on Drafting Guidelines for the Evaluation of Probiotics in Food; FAO: London, ON, Canada, 2002. [Google Scholar]
  72. Food and Agriculture Organization. FAO Technical Meeting on Prebiotics: Food Quality and Standards Service (AGNS); FAO Technical Meeting Report; FAO: Rome, Italy, 2007. [Google Scholar]
  73. Chung, W.S.F.; Walker, A.W.; Louis, P.; Parkhill, J.; Vermeiren, J.; Bosscher, D.; Duncan, S.H.; Flint, H.J. Modulation of the human gut microbiota by dietary fibres occurs at the species level. BMC Biol. 2016, 14, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Scavuzzi, B.M.; Henrique, F.C.; Miglioranza, L.H.S.; Simão, A.N.C.; Dichi, I. Impact of prebiotics, probiotics and synbiotics on components of the metabolic syndrome. Ann. Nutr. Disord. Ther. 2014, 1, 1009. [Google Scholar]
  75. Ustundag, G.H.; Altuntas, H.; Soysal, Y.D.; Kokturk, F. The effects of synbiotic Bifidobacterium lactis B94 plus Inulin addition on standard triple therapy of Helicobacter pylori eradication in children. Can. J. Gastroenterol. Hepatol. 2017, 2017, 8130596. [Google Scholar] [CrossRef]
  76. Roškar, I.; Švigelj, K.; Štempelj, M.; Volfand, J.; Štabuc, B.; Malovrh, Š.; Rogelj, I. Effects of a probiotic product containing Bifidobacterium animalis subsp. animalis IM386 and Lactobacillus plantarum MP2026 in lactose intolerant individuals: Randomized, placebo-controlled clinical trial. J. Funct. Foods 2017, 35, 1–8. [Google Scholar] [CrossRef]
  77. Brahe, L.K.; Le Chatelier, E.; Prifti, E.; Pons, N.; Kennedy, S.; Blædel, T.; Håkansson, J.; Dalsgaard, T.K.; Hansen, T.; Pedersen, O. Dietary modulation of the gut microbiota—A randomised controlled trial in obese postmenopausal women. Br. J. Nutr. 2015, 114, 406–417. [Google Scholar] [CrossRef] [Green Version]
  78. Ivey, K.L.; Hodgson, J.M.; Kerr, D.A.; Thompson, P.L.; Stojceski, B.; Prince, R.L. The effect of yoghurt and its probiotics on blood pressure and serum lipid profile; a randomised controlled trial. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 46–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Hariri, M.; Salehi, R.; Feizi, A.; Mirlohi, M.; Ghiasvand, R.; Habibi, N. A randomized, double-blind, placebo-controlled, clinical trial on probiotic soy milk and soy milk: Effects on epigenetics and oxidative stress in patients with type II diabetes. Genes Nutr. 2015, 10, 52. [Google Scholar] [CrossRef] [PubMed]
  80. Tonucci, L.B.; Olbrich Dos Santos, K.M.; Licursi de Oliveira, L.; Rocha Ribeiro, S.M.; Duarte Martino, H.S. Clinical application of probiotics in type 2 diabetes mellitus: Arandomized, double-blind, placebo-controlled study. Clin. Nutr. 2015, 36, 85–92. [Google Scholar] [CrossRef] [PubMed]
  81. Mohamadshahi, M.; Veissi, M.; Haidari, F.; Javid, A.Z.; Mohammadi, F.; Shirbeigi, E. Effects of probiotic yogurt consumption on lipid profile in type 2 diabetic patients: A randomized controlled clinical trial. J. Res. Med. Sci. 2014, 19, 531–536. [Google Scholar] [PubMed]
  82. Champagne, C.P.; Ross, R.P.; Saarela, M.; Hansen, K.F.; Charalampopoulos, D. Recommendations for the viability assessment of probiotics as concentrated cultures and in food matrices. Int. J. Food Microbiol. 2011, 149, 185–193. [Google Scholar] [CrossRef]
  83. Homayoni Rad, A.; Mehrabany, E.V.; Alipoor, B.; Mehrabany, L.V.; Javadi, M. Do probiotics act more efficiently in foods than in supplements? Nutrition 2012, 28, 733–736. [Google Scholar] [CrossRef]
  84. Markowiak, P.; Slizewska, K. Effects of Probiotics, Prebiotics, and Synbiotics on Human Health. Nutrients 2017, 9, 1021. [Google Scholar] [CrossRef]
  85. Sarkar, A.; Mandal, S. Bifidobacteria—Insight into clinical outcomes and mechanisms of its probiotic action. Microbiol. Res. 2016, 192, 159–171. [Google Scholar] [CrossRef]
  86. Turroni, F.; Duranti, S.; Milani, C.; Lugli, A.G.; van Sinderen, D.; Ventura, M. Bifidobacterium bifidum: A Key Member of the Early Human Gut Microbiota. Microorganisms 2019, 7, 544. [Google Scholar] [CrossRef] [Green Version]
  87. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10, S49–S66. [Google Scholar] [CrossRef] [Green Version]
  88. Lee, C.W.; Chen, H.J.; Chien, Y.H.; Hsia, S.M.; Chen, J.H.; Shih, C.K. Synbiotic Combination of Djulis (Chenopodium formosanum) and Lactobacillus acidophilus Inhibits Colon Carcinogenesis in Rats. Nutrients 2019, 12, 103. [Google Scholar] [CrossRef] [Green Version]
  89. Navaei, M.; Haghighat, S.; Janani, L.; Vafa, S.; Saneei Totmaj, A.; Raji Lahiji, M.; Emamat, H.; Salehi, Z.; Amirinejad, A.; Izad, M.; et al. The Effects of Synbiotic Supplementation on Antioxidant Capacity and Arm Volumes in Survivors of Breast Cancer-Related Lymphedema. Nutr. Cancer 2020, 72, 62–73. [Google Scholar] [CrossRef] [PubMed]
  90. Le, B.; Ngoc, A.P.T.; Yang, S.H. Synbiotic fermented soymilk with Weissella cibaria FB069 and xylooligosaccharides prevents proliferation in human colon cancer cells. J. Appl. Microbiol. 2019. [Google Scholar] [CrossRef] [PubMed]
  91. Faraki, A.; Noori, N.; Gandomi, H.; Banuree, S.A.H.; Rahmani, F. Effect of Auricularia auricula aqueous extract on survival of Lactobacillus acidophilus La-5 and Bifidobacterium bifidum Bb-12 and on sensorial and functional properties of synbiotic yogurt. Food Sci. Nutr. 2020, 8, 1254–1263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Athiyyah, A.; Widjaja, N.; Fitri, P.; Setiowati, A.; Darma, A.; Ranuh, R.; Sudarmo, S. Effects of a multispecies synbiotic on intestinal mucosa immune responses. Iran. J. Microbiol. 2019, 11. [Google Scholar] [CrossRef] [Green Version]
  93. Morshedi, M.; Saghafi-Asl, M.; Hosseinifard, E.-S. The potential therapeutic effects of the gut microbiome manipulation by synbiotic containing-Lactobacillus plantarum on neuropsychological performance of diabetic rats. J. Transl. Med. 2020, 18, 1–14. [Google Scholar] [CrossRef] [Green Version]
  94. Ghafouri, A.; Zarrati, M.; Shidfar, F.; Heydari, I.; Shokouhi Shoormasti, R.; Eslami, O. Effect of synbiotic bread containing lactic acid on glycemic indicators, biomarkers of antioxidant status and inflammation in patients with type 2 diabetes: A randomized controlled trial. Diabetol. Metab. Syndr. 2019, 11, 103. [Google Scholar] [CrossRef] [Green Version]
  95. Moser, A.M.; Spindelboeck, W.; Halwachs, B.; Strohmaier, H.; Kump, P.; Gorkiewicz, G.; Hogenauer, C. Effects of an oral synbiotic on the gastrointestinal immune system and microbiota in patients with diarrhea-predominant irritable bowel syndrome. Eur. J. Nutr. 2019, 58, 2767–2778. [Google Scholar] [CrossRef] [Green Version]
  96. Pistol, G.C.; Marin, D.E.; Dragomir, C.; Taranu, I. Synbiotic combination of prebiotic grape pomace extract and probiotic Lactobacillus sp. reduced important intestinal inflammatory markers and in-depth signalling mediators in lipopolysaccharide-treated Caco-2 cells. Br. J. Nutr. 2018, 121, 291–305. [Google Scholar] [CrossRef]
  97. Sengupta, S.; Koley, H.; Dutta, S.; Bhowal, J. Hepatoprotective effects of synbiotic soy yogurt on mice fed a high-cholesterol diet. Nutrition 2019, 63, 36–44. [Google Scholar] [CrossRef]
  98. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  99. Brandi, J.; Di Carlo, C.; Manfredi, M.; Federici, F.; Bazaj, A.; Rizzi, E.; Cornaglia, G.; Manna, L.; Marengo, E.; Cecconi, D. Investigating the Proteomic Profile of HT-29 Colon Cancer Cells after Lactobacillus kefiri SGL 13 Exposure Using the SWATH Method. J. Am. Soc. Mass Spectrom. 2019, 30, 1690–1699. [Google Scholar] [CrossRef] [PubMed]
  100. Ragul, K.; Kandasamy, S.; Devi, P.B.; Shetty, P.H. Evaluation of functional properties of potential probiotic isolates from fermented brine pickle. Food Chem. 2020, 311, 126057. [Google Scholar] [CrossRef] [PubMed]
  101. Ayyash, M.; Abu-Jdayil, B.; Itsaranuwat, P.; Galiwango, E.; Tamiello-Rosa, C.; Abdullah, H.; Esposito, G.; Hunashal, Y.; Obaid, R.S.; Hamed, F. Characterization, bioactivities, and rheological properties of exopolysaccharide produced by novel probiotic Lactobacillus plantarum C70 isolated from camel milk. Int. J. Biol. Macromol. 2020, 144, 938–946. [Google Scholar] [CrossRef] [PubMed]
  102. Rahbar Saadat, Y.; Yari Khosroushahi, A.; Movassaghpour, A.A.; Talebi, M.; Pourghassem Gargari, B. Modulatory role of exopolysaccharides of Kluyveromyces marxianus and Pichia kudriavzevii as probiotic yeasts from dairy products in human colon cancer cells. J. Funct. Foods 2020, 64, 103675. [Google Scholar] [CrossRef]
  103. Justino, P.F.C.; Franco, A.X.; Pontier-Bres, R.; Monteiro, C.E.S.; Barbosa, A.L.R.; Souza, M.H.L.P.; Czerucka, D.; Soares, P.M.G. Modulation of 5-fluorouracil activation of toll-like/MyD88/NF-κB/MAPK pathway by Saccharomyces boulardii CNCM I-745 probiotic. Cytokine 2020, 125, 154791. [Google Scholar] [CrossRef] [PubMed]
  104. Ghanavati, R.; Asadollahi, P.; Shapourabadi, M.B.; Razavi, S.; Talebi, M.; Rohani, M. Inhibitory effects of Lactobacilli cocktail on HT-29 colon carcinoma cells growth and modulation of the Notch and Wnt/β-catenin signaling pathways. Microb. Pathog. 2020, 139, 103829. [Google Scholar] [CrossRef]
  105. Rupasinghe, H.P.V.; Parmar, I.; Neir, S.V. Biotransformation of Cranberry Proanthocyanidins to Probiotic Metabolites by Lactobacillus rhamnosus Enhances Their Anticancer Activity in HepG2 Cells in Vitro. Oxid. Med. Cell. Longev. 2019, 2019, 4750795. [Google Scholar] [CrossRef] [Green Version]
  106. Nozari, S.; Faridvand, Y.; Etesami, A.; Ahmad Khan Beiki, M.; Miresmaeili Mazrakhondi, S.A.; Abdolalizadeh, J. Potential anticancer effects of cell wall protein fractions from Lactobacillus paracasei on human intestinal Caco-2 cell line. Lett. Appl. Microbiol. 2019, 69, 148–154. [Google Scholar] [CrossRef]
  107. Chandel, D.; Sharma, M.; Chawla, V.; Sachdeva, N.; Shukla, G. Isolation, characterization and identification of antigenotoxic and anticancerous indigenous probiotics and their prophylactic potential in experimental colon carcinogenesis. Sci. Rep. 2019, 9, 14769. [Google Scholar] [CrossRef] [Green Version]
  108. Lin, P.-Y.; Li, S.-C.; Lin, H.-P.; Shih, C.-K. Germinated brown rice combined with Lactobacillus acidophilus and Bifidobacterium animalis subsp. lactis inhibits colorectal carcinogenesis in rats. Food Sci. Nutr. 2019, 7, 216–224. [Google Scholar] [CrossRef] [Green Version]
  109. Karimi Ardestani, S.; Tafvizi, F.; Tajabadi Ebrahimi, M. Heat-killed probiotic bacteria induce apoptosis of HT-29 human colon adenocarcinoma cell line via the regulation of Bax/Bcl2 and caspases pathway. Hum. Exp. Toxicol. 2019, 38, 1069–1081. [Google Scholar] [CrossRef] [PubMed]
  110. He, L.; Yang, H.; Tang, J.; Liu, Z.; Chen, Y.; Lu, B.; He, H.; Tang, S.; Sun, Y.; Liu, F.; et al. Intestinal probiotics E. coli Nissle 1917 as a targeted vehicle for delivery of p53 and Tum-5 to solid tumors for cancer therapy. J. Biol. Eng. 2019, 13, 58. [Google Scholar] [CrossRef] [PubMed]
  111. Shi, L.; Sheng, J.; Wang, M.; Luo, H.; Zhu, J.; Zhang, B.; Liu, Z.; Yang, X. Combination Therapy of TGF-beta Blockade and Commensal-derived Probiotics Provides Enhanced Antitumor Immune Response and Tumor Suppression. Theranostics 2019, 9, 4115–4129. [Google Scholar] [CrossRef] [PubMed]
  112. Chen, J.C.; Tsai, C.-C.; Hsieh, C.C.; Lan, A.; Huang, C.C.; Leu, S.F. Multispecies probiotics combination prevents ovalbumin-induced airway hyperreactivity in mice. Allergol. Immunopathol. 2018, 46, 354–360. [Google Scholar] [CrossRef] [PubMed]
  113. Nakamura, S.; Mitsunaga, F. Anti-Allergic Effect of Para-Probiotics from Non-Viable Acetic Acid Bacteria in Ovalbumin-Sensitized Mice. Food Nutr. Sci. 2018, 9, 1376–1385. [Google Scholar] [CrossRef] [Green Version]
  114. Koh, W.Y.; Utra, U.; Ahmad, R.; Rather, I.A.; Park, Y.-H. Evaluation of probiotic potential and anti-hyperglycemic properties of a novel Lactobacillus strain isolated from water kefir grains. Food Sci. Biotechnol. 2018, 27, 1369–1376. [Google Scholar] [CrossRef]
  115. Yadav, R.; Dey, D.K.; Vij, R.; Meena, S.; Kapila, R.; Kapila, S. Evaluation of anti-diabetic attributes of Lactobacillus rhamnosus MTCC: 5957, Lactobacillus rhamnosus MTCC: 5897 and Lactobacillus fermentum MTCC: 5898 in streptozotocin induced diabetic rats. Microb. Pathog. 2018, 125, 454–462. [Google Scholar] [CrossRef]
  116. Miraghajani, M.; Zaghian, N.; dehkohneh, A.; Mirlohi, M.; Ghiasvand, R. Probiotic Soy Milk Consumption and Renal Function among Type 2 Diabetic Patients with Nephropathy: A Randomized Controlled Clinical Trial. Probiotics Antimicrob. Proteins 2019, 11, 124–132. [Google Scholar] [CrossRef]
  117. Choi, W.J.; Dong, H.J.; Jeong, H.U.; Jung, H.H.; Kim, Y.-H.; Kim, T.H. Antiobesity Effects of Lactobacillus plantarum LMT1-48 Accompanied by Inhibition of Enterobacter cloacae in the Intestine of Diet-Induced Obese Mice. J. Med. Food 2019, 22, 560–566. [Google Scholar] [CrossRef] [Green Version]
  118. Legrand, R.; Lucas, N.; Dominique, M.; Azhar, S.; Deroissart, C.; Le Solliec, M.-A.; Rondeaux, J.; Nobis, S.; Guérin, C.; Léon, F.; et al. Commensal Hafnia alvei strain reduces food intake and fat mass in obese mice-a new potential probiotic for appetite and body weight management. Int. J. Obes. 2020. [Google Scholar] [CrossRef] [Green Version]
  119. Kang, D.; Su, M.; Duan, Y.; Huang, Y. Eurotium cristatum, a potential probiotic fungus from Fuzhuan brick tea, alleviated obesity in mice by modulating gut microbiota. Food Funct. 2019, 10, 5032–5045. [Google Scholar] [CrossRef] [PubMed]
  120. Huang, C.H.; Ho, C.Y.; Chen, C.T.; Hsu, H.F.; Lin, Y.H. Probiotic BSH Activity and Anti-Obesity Potential of Lactobacillus plantarum Strain TCI378 Isolated from Korean Kimchi. Prev. Nutr. Food Sci. 2019, 24, 434–441. [Google Scholar] [CrossRef] [PubMed]
  121. Hsu, T.C.; Yi, P.J.; Lee, T.Y.; Liu, J.R. Probiotic characteristics and zearalenone-removal ability of a Bacillus licheniformis strain. PLoS ONE 2018, 13, e0194866. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, J.; Zeng, Y.; Wang, S.; Liu, H.; Zhang, D.; Zhang, W.; Wang, Y.; Ji, H. Swine-Derived Probiotic Lactobacillus plantarum Inhibits Growth and Adhesion of Enterotoxigenic Escherichia coli and Mediates Host Defense. Front. Microbiol. 2018, 9, 1364. [Google Scholar] [CrossRef] [PubMed]
  123. Rocha-Ramírez, M.L.; Hernández-Ochoa, B.; Gómez-Manzo, S.; Marcial-Quino, J.; Cárdenas-Rodríguez, N.; Centeno-Leija, S.; García-Garibay, M. Evaluation of Immunomodulatory Activities of the Heat-Killed Probiotic Strain Lactobacillus casei IMAU60214 on Macrophages in Vitro. Microorganisms 2020, 8, 79. [Google Scholar] [CrossRef] [Green Version]
  124. Beller, A.; Kruglov, A.; Durek, P.; von Goetze, V.; Hoffmann, U.; Maier, R.; Heiking, K.; Siegmund, B.; Heinz, G.; Mashreghi, M.F.; et al. P104 Anaeroplasma, a potential anti-inflammatory probiotic for the treatment of chronic intestinal inflammation. Ann. Rheum. Dis. 2019, 78, A45–A46. [Google Scholar] [CrossRef] [Green Version]
  125. Coqueiro, A.Y.; Raizel, R.; Bonvini, A.; Tirapegui, J.; Rogero, M.M. Probiotics for inflammatory bowel diseases: A promising adjuvant treatment. Int. J. Food Sci. Nutr. 2019, 70, 20–29. [Google Scholar] [CrossRef]
  126. Machado Prado, M.R.; Boller, C. Anti-inflammatory effects of probiotics. In Discovery and Development of Anti-Inflammatory Agents from Natural Products; Brahmachari, G., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; Chapter 9; pp. 259–282. [Google Scholar] [CrossRef]
  127. Chen, Y.; Zhang, L.; Hong, G.; Huang, C.; Qian, W.; Bai, T.; Song, J.; Song, Y.; Hou, X. Probiotic mixtures with aerobic constituent promoted the recovery of multi-barriers in DSS-induced chronic colitis. Life Sci. 2020, 240, 117089. [Google Scholar] [CrossRef]
  128. Zhang, Z.; Lv, J.; Pan, L.; Zhang, Y. Roles and applications of probiotic Lactobacillus strains. Appl. Microbiol. Biotechnol. 2018, 102, 8135–8143. [Google Scholar] [CrossRef]
  129. Lee, J.-E.; Lee, J.; Kim, J.H.; Cho, N.; Lee, S.H.; Park, S.B.; Koh, B.; Kang, D.; Kim, S.; Yoo, H.M. Characterization of the Anti-Cancer Activity of the Probiotic Bacterium Lactobacillus fermentum Using 2D vs. 3D Culture in Colorectal Cancer Cells. Biomolecules 2019, 9, 557. [Google Scholar] [CrossRef] [Green Version]
  130. Fornai, M.; Pellegrini, C.; Benvenuti, L.; Tirotta, E.; Gentile, D.; Natale, G.; Ryskalin, L.; Colucci, R.; Piccoli, E.; Ghelardi, E.; et al. Protective effects of the combination Bifidobacterium longum plus lactoferrin against NSAID-induced enteropathy. Nutrition 2020, 70, 110583. [Google Scholar] [CrossRef] [PubMed]
  131. Ben Othman, M.; Sakamoto, K. Effect of inactivated Bifidobacterium longum intake on obese diabetes model mice (TSOD). Food Res. Int. 2020, 129, 108792. [Google Scholar] [CrossRef] [PubMed]
  132. Talani, G.; Biggio, F.; Mostallino, M.C.; Locci, V.; Porcedda, C.; Boi, L.; Saolini, E.; Piras, R.; Sanna, E.; Biggio, G. Treatment with gut bifidobacteria improves hippocampal plasticity and cognitive behavior in adult healthy rats. Neuropharmacology 2020, 165, 107909. [Google Scholar] [CrossRef] [PubMed]
  133. Khangwal, I.; Shukla, P. Prospecting prebiotics, innovative evaluation methods, and their health applications: A review. Biotech 2019, 9, 187. [Google Scholar] [CrossRef] [PubMed]
  134. Korcz, E.; Kerényi, Z.; Varga, L. Dietary fibers, prebiotics, and exopolysaccharides produced by lactic acid bacteria: Potential health benefits with special regard to cholesterol-lowering effects. Food. Funct. 2018, 9, 3057–3068. [Google Scholar] [CrossRef]
  135. Shehata, M.; El-sahn, M.A.; El-Sohaimy, S.A.; Youssef, M.M. Role and Mechanisms Lowering Cholesterol by Dietary of Probiotics and Prebiotics: A Review. J. Appl. Sci. 2019, 19, 737–746. [Google Scholar] [CrossRef]
  136. Zhu, W.; Zhou, S.; Liu, J.; McLean, R.J.C.; Chu, W. Prebiotic, immuno-stimulating and gut microbiota-modulating effects of Lycium barbarum polysaccharide. Biomed. Pharmacother. 2020, 121, 109591. [Google Scholar] [CrossRef]
  137. Cerdó, T.; García-Santos, A.J.; G. Bermúdez, M.; Campoy, C. The Role of Probiotics and Prebiotics in the Prevention and Treatment of Obesity. Nutrients 2019, 11, 635. [Google Scholar] [CrossRef] [Green Version]
  138. Vyas, N.; Nair, S.; Rao, M.; Miraj, S.S. Chapter 29—Childhood Obesity and Diabetes: Role of Probiotics and Prebiotics. In Global Perspectives on Childhood Obesity, 2nd ed.; Bagchi, D., Ed.; Academic Press: Cambridge, MA, USA, 2019. [Google Scholar]
  139. Laurell, A.; Sjöberg, K. Prebiotics and synbiotics in ulcerative colitis. Scand. J. Gastroenterol. 2017, 52, 477–485. [Google Scholar] [CrossRef]
  140. Rani, A.; Baruah, R.; Goyal, A. Prebiotic Chondroitin Sulfate Disaccharide Isolated from Chicken Keel Bone Exhibiting Anticancer Potential against Human Colon Cancer Cells. Nutr. Cancer 2019, 71, 825–839. [Google Scholar] [CrossRef]
  141. Wen, Y.; Wen, P.; Hu, T.G.; Linhardt, R.J.; Zong, M.H.; Wu, H.; Chen, Z.Y. Encapsulation of phycocyanin by prebiotics and polysaccharides-based electrospun fibers and improved colon cancer prevention effects. Int. J. Biol. Macromol. 2020, 149, 672–681. [Google Scholar] [CrossRef] [PubMed]
  142. Ohara, T.; Mori, T. Antiproliferative Effects of Short-chain Fatty Acids on Human Colorectal Cancer Cells via Gene Expression Inhibition. Anticancer Res. 2019, 39, 4659–4666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Zhou, L.; Xie, M.; Yang, F.; Liu, J. Antioxidant activity of high purity blueberry anthocyanins and the effects on human intestinal microbiota. LWT 2020, 117, 108621. [Google Scholar] [CrossRef]
  144. Li, E.; Yang, S.; Zou, Y.; Cheng, W.; Li, B.; Hu, T.; Li, Q.; Wang, W.; Liao, S.; Pang, D. Purification, Characterization, Prebiotic Preparations and Antioxidant Activity of Oligosaccharides from Mulberries. Molecules 2019, 24, 2329. [Google Scholar] [CrossRef] [Green Version]
  145. Weinborn, V.; Valenzuela, C.; Olivares, M.; Arredondo, M.; Weill, R.; Pizarro, F. Prebiotics increase heme iron bioavailability and do not affect non-heme iron bioavailability in humans. Food. Funct. 2017, 8, 1994–1999. [Google Scholar] [CrossRef] [PubMed]
  146. Aliasgharzadeh, A.; Khalili, M.; Mirtaheri, E.; Pourghassem Gargari, B.; Tavakoli, F.; Abbasalizad Farhangi, M.; Babaei, H.; Dehghan, P. A Combination of Prebiotic Inulin and Oligofructose Improve Some of Cardiovascular Disease Risk Factors in Women with Type 2 Diabetes: A Randomized Controlled Clinical Trial. Adv. Pharm. Bull. 2015, 5, 507–514. [Google Scholar] [CrossRef]
  147. Da Silva Sabo, S.; Converti, A.; Todorov, S.D.; Domínguez, J.M.; de Souza Oliveira, R.P. Effect of inulin on growth and bacteriocin production by Lactobacillus plantarum in stationary and shaken cultures. Int. J. Food Sci. Technol. 2015, 50, 864–870. [Google Scholar] [CrossRef]
  148. Ramos, C.I.; Armani, R.G.; Canziani, M.E.F.; Dalboni, M.A.; Dolenga, C.J.R.; Nakao, L.S.; Campbell, K.L.; Cuppari, L. Effect of prebiotic (fructooligosaccharide) on uremic toxins of chronic kidney disease patients: A randomized controlled trial. Nephrol. Dial. Transpl. 2018, 34, 1876–1884. [Google Scholar] [CrossRef]
  149. Flesch, A.G.; Poziomyck, A.K.; Damin, D.C. The therapeutic use of symbiotics. Arq. Bras. Cir. Dig. 2014, 27, 206–209. [Google Scholar] [CrossRef] [Green Version]
  150. Bonfrate, L.; Palo, D.M.; Celano, G.; Albert, A.; Vitellio, P.; De Angelis, M.; Gobbetti, M.; Portincasa, P. Effects of Bifidobacterium longum BB536 and Lactobacillus rhamnosus HN001 in IBS patients. Eur. J. Clin. Investig. 2020, 50, e13201. [Google Scholar] [CrossRef]
  151. Mohan, A.; Hadi, J.; Gutierrez-Maddox, N.; Li, Y.; Leung, I.K.H.; Gao, Y.; Shu, Q.; Quek, S.Y. Sensory, Microbiological and Physicochemical Characterisation of Functional Manuka Honey Yogurts Containing Probiotic Lactobacillus reuteri DPC16. Foods 2020, 9, 106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Li, P.H.; Lu, W.C.; Chan, Y.J.; Zhao, Y.P.; Nie, X.B.; Jiang, C.X.; Ji, Y.X. Feasibility of Using Seaweed (Gracilaria coronopifolia) Synbiotic as a Bioactive Material for Intestinal Health. Foods 2019, 8, 623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Sarwar, A.; Aziz, T.; Al-Dalali, S.; Zhao, X.; Zhang, J.; ud Din, J.; Chen, C.; Cao, Y.; Yang, Z. Physicochemical and Microbiological Properties of Synbiotic Yogurt Made with Probiotic Yeast Saccharomyces boulardii in Combination with Inulin. Foods 2019, 8, 468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Karimi, M.; Yazdi, F.T.; Mortazavi, S.A.; Shahabi-Ghahfarrokhi, I.; Chamani, J. Development of active antimicrobial poly (l-glutamic) acid-poly (l-lysine) packaging material to protect probiotic bacterium. Polym. Test. 2020, 83, 106338. [Google Scholar] [CrossRef]
  155. Aziz Mousavi, S.M.A.; Mirhosseini, S.A.; Rastegar Shariat Panahi, M.; Mahmoodzadeh Hosseini, H. Characterization of Biosynthesized Silver Nanoparticles Using Lactobacillus rhamnosus GG and its in Vitro Assessment against Colorectal Cancer Cells. Probiotics Antimicrob. Proteins 2019. [Google Scholar] [CrossRef]
  156. Kouhkan, M.; Ahangar, P.; Babaganjeh, L.; Allahyari-Devin, M. Biosynthesis of copper oxide nanoparticles using Lactobacillus casei subsp. casei and its anticancer and antibacterial activities. Curr. Nanosci. 2019, 15. [Google Scholar] [CrossRef]
  157. Xu, C.; Qiao, L.; Guo, Y.; Ma, L.; Cheng, Y. Preparation, characteristics and antioxidant activity of polysaccharides and proteins-capped selenium nanoparticles synthesized by Lactobacillus casei ATCC 393. Carbohydr. Polym. 2018, 195, 576–585. [Google Scholar] [CrossRef]
  158. Xu, C.; Guo, Y.; Qiao, L.; Ma, L.; Cheng, Y.; Roman, A. Biogenic Synthesis of Novel Functionalized Selenium Nanoparticles by Lactobacillus casei ATCC 393 and Its Protective Effects on Intestinal Barrier Dysfunction Caused by Enterotoxigenic Escherichia coli K88. Front. Microbiol. 2018, 9, 1129. [Google Scholar] [CrossRef] [Green Version]
  159. Lee, H.A.; Kim, H.; Lee, K.W.; Park, K.Y. Dead Nano-Sized Lactobacillus plantarum Inhibits Azoxymethane/Dextran Sulfate Sodium-Induced Colon Cancer in Balb/c Mice. J. Med. Food 2015, 18, 1400–1405. [Google Scholar] [CrossRef]
  160. Markus, J.; Mathiyalagan, R.; Kim, Y.J.; Abbai, R.; Singh, P.; Ahn, S.; Perez, Z.E.J.; Hurh, J.; Yang, D.C. Intracellular synthesis of gold nanoparticles with antioxidant activity by probiotic Lactobacillus kimchicus DCY51(T) isolated from Korean kimchi. Enzym. Microb. Technol. 2016, 95, 85–93. [Google Scholar] [CrossRef]
  161. Jimenez-Sanchez, M.; Perez-Morales, R.; Goycoolea, F.M.; Mueller, M.; Praznik, W.; Loeppert, R.; Bermudez-Morales, V.; Zavala-Padilla, G.; Ayala, M.; Olvera, C. Self-assembled high molecular weight inulin nanoparticles: Enzymatic synthesis, physicochemical and biological properties. Carbohydr. Polym. 2019, 215, 160–169. [Google Scholar] [CrossRef]
  162. Kim, W.; Han, G.; Hong, L.; Kang, S.-K.; Shokouhimehr, M.; Choi, Y.-J.; Cho, C. Novel production of natural bacteriocin via internalization of dextran nanoparticles into probiotics. Biomaterials 2019, 218, 119360. [Google Scholar] [CrossRef] [PubMed]
  163. Cui, L.H.; Yan, C.G.; Li, H.S.; Kim, W.S.; Hong, L.; Kang, S.K.; Choi, Y.J.; Cho, C.S. A New Method of Producing a Natural Antibacterial Peptide by Encapsulated Probiotics Internalized with Inulin Nanoparticles as Prebiotics. J. Microbiol. Biotechnol. 2018, 28, 510–519. [Google Scholar] [CrossRef] [PubMed]
  164. Hong, L.; Kim, W.S.; Lee, S.M.; Kang, S.K.; Choi, Y.J.; Cho, C.S. Pullulan Nanoparticles as Prebiotics Enhance the Antibacterial Properties of Lactobacillus plantarum through the Induction of Mild Stress in Probiotics. Front. Microbiol. 2019, 10, 142. [Google Scholar] [CrossRef] [Green Version]
  165. Kaur, K.; Rath, G. Formulation and evaluation of UV protective synbiotic skin care topical formulation. J. Cosmet. Laser Ther. Off. Publ. Eur. Soc. Laser Dermatol. 2019, 21, 332–342. [Google Scholar] [CrossRef] [PubMed]
  166. Krithika, B.; Preetha, R. Formulation of protein based inulin incorporated synbiotic nanoemulsion for enhanced stability of probiotic. Mat. Res. Express 2019, 6, 114003. [Google Scholar] [CrossRef]
  167. Atia, A.; Gomaa, A.; Fliss, I.; Beyssac, E.; Garrait, G.; Subirade, M. A prebiotic matrix for encapsulation of probiotics: Physicochemical and microbiological study. J. Microencapsul. 2016, 33, 89–101. [Google Scholar] [CrossRef]
  168. Caneus, D. Nanotechnology and its Partnership with Synbiotics. J. Nanomed. Res. 2017, 6, 142. [Google Scholar] [CrossRef] [Green Version]
  169. Pathak, K.; Akhtar, N. Nanoprobiotics: Progress and Issues. In Nanonutraceuticals, 1st ed.; Singh., B., Ed.; CRC Press: Boca Raton, FL, USA, 2018; Chapter 9; 326p. [Google Scholar]
  170. Rajendran, K.; Sen, S.; Latha, P. Nanotechnology in probiotics and prebiotics. In Nanotechnology in Nutraceuticals: Production to Consumption, 1st ed.; Sen, S., Pathak, Y., Eds.; CRC Press: Oxfordshire, UK; Taylor & Francis Group: Oxfordshire, UK; Abingdon-on-Thames: Oxfordshire, UK, 2016; Chapter 9. [Google Scholar]
  171. Kazmierczak, R.; Choe, E.; Sinclair, J.; Eisenstark, A. Direct attachment of nanoparticle cargo to Salmonella typhimurium membranes designed for combination bacteriotherapy against tumors. Methods Mol. Biol. 2015, 1225, 151–163. [Google Scholar] [CrossRef]
  172. Hu, Q.; Wu, M.; Fang, C.; Cheng, C.; Zhao, M.; Fang, W.; Chu, P.K.; Ping, Y.; Tang, G. Engineering Nanoparticle-Coated Bacteria as Oral DNA Vaccines for Cancer Immunotherapy. Nano Lett. 2015, 15, 2732–2739. [Google Scholar] [CrossRef]
  173. Feher, F.; Pinter, E.; Helyes, Z.; Szolcsanyi, J. Nano-size particles of probiotics for preventing and treating neuroinflammation. ARVO Annual Meeting. Investig. Ophthalmol. Vis. Sci. 2012, 53, 331. [Google Scholar]
  174. Kim, W.S.; Lee, J.Y.; Singh, B.; Maharjan, S.; Hong, L.; Lee, S.M.; Cui, L.H.; Lee, K.J.; Kim, G.; Yun, C.H.; et al. A new way of producing pediocin in Pediococcus acidilactici through intracellular stimulation by internalized inulin nanoparticles. Sci. Rep. 2018, 8, 5878. [Google Scholar] [CrossRef] [PubMed]
  175. Song, Q.; Zheng, C.; Jia, J.; Zhao, H.; Feng, Q.; Zhang, H.; Wang, L.; Zhang, Z.; Zhang, Y. A Probiotic Spore-Based Oral Autonomous Nanoparticles Generator for Cancer Therapy. Adv. Mater. 2019, 31, e1903793. [Google Scholar] [CrossRef] [PubMed]
  176. Fung, W.Y.; Yuen, K.H.; Liong, M.T. Agrowaste-based nanofibers as a probiotic encapsulant: Fabrication and characterization. J. Agric. Food Chem. 2011, 59, 8140–8147. [Google Scholar] [CrossRef] [PubMed]
  177. Nagy, Z.K.; Wagner, I.; Suhajda, A.; Tobak, T.; Harsztos, A.H.; Vigh, T.; Soti, P.L.; Pataki, K.; Molnar, K.; Marosi, G. Nanofibrous solid dosage form of living bacteria prepared by electrospinning. Express Polym. Lett. 2014, 8, 352–361. [Google Scholar] [CrossRef] [Green Version]
  178. Zupancic, S.; Škrlec, K.; Kocbek, P.; Kristl, J.; Berlec, A. Effects of electrospinning on the viability of ten species of lactic acid bacteria in poly(ethylene oxide) nanofibers. Pharmaceutics 2019, 11, 483. [Google Scholar] [CrossRef] [Green Version]
  179. Wang, A.; Xu, C.; Zhang, C.; Gan, Y.; Wang, B. Experimental Investigation of the Properties of Electrospun Nanofibers for Potential Medical Application. J. Nanomater. 2015, 2015, 418932. [Google Scholar] [CrossRef]
  180. Xue, J.; Xie, J.; Liu, W.; Xia, Y. Electrospun Nanofibers: New Concepts, Materials, and Applications. Acc. Chem. Res. 2017, 50, 1976–1987. [Google Scholar] [CrossRef]
  181. Shahriar, S.M.; Mondal, J.; Hasan, M.N.; Revuri, V.; Lee, D.Y.; Lee, Y.K. Electrospinning Nanofibers for Therapeutics Delivery. Nanomaterials 2019, 9, 532. [Google Scholar] [CrossRef] [Green Version]
  182. Torres-Martínez, E.T.; Cornejo Bravo, J.M.; Serrano Medina, A.; Pérez González, G.L.; Villarreal Gómez, L.J. A summary of electrospun nanofibers as drug delivery system: Drugs loaded and biopolymers used as matrices. Curr. Drug Deliv. 2018, 15, 1360–1374. [Google Scholar] [CrossRef]
  183. Ghorani, B.; Tucker, N. Fundamentals of electrospinning as novel delivery for bioactive compounds in food nanotechnology. Food Hydrocoll. 2015, 51, 227–240. [Google Scholar] [CrossRef]
  184. Franz, B.; Balkundi, S.S.; Dahl, C.; Lvov, Y.M.; Prange, A. Layer-by-layer nano-encapsulation of microbes: Controlled cell surface modification and investigation of substrate uptake in bacteria. Macromol. Biosci. 2010, 11, 164–172. [Google Scholar] [CrossRef] [PubMed]
  185. Ebrahimnejad, P.; Khavarpour, M.; Khalilid, S. Survival of Lactobacillus Acidophilus as probiotic bacteria using chitosan nanoparticles. IJE Trans. A Basics 2017, 30, 456–463. [Google Scholar]
  186. Ranjan, S.; Dasgupta, N.; Chakraborty, A.R.; Melvin Samuel, S.; Ramalingam, C.; Shanker, R.; Kumar, A. Nanoscience and nanotechnologies in food industries: Opportunities and research trends. J. Nanopart. Res. 2014, 16, 2464. [Google Scholar] [CrossRef]
  187. Salmerón, I. Fermented cereal beverages: From probiotic, prebiotic and synbiotic towards Nanoscience designed healthy drinks. Lett. Appl. Microbiol. 2017, 65, 114–124. [Google Scholar] [CrossRef] [Green Version]
  188. Rezaee, P.; Kasra Kermanshahi, R.; Katouli, M. Prebiotics decrease the antibacterial effect of nano silver and nano TiO2 particles against probiotic bacteria of food. Curr. Nutr. Food Sci. 2014, 10. [Google Scholar] [CrossRef]
  189. Khan, S.T.; Saleem, S.; Ahamed, M.; Ahmad, J. Survival of probiotic bacteria in the presence of food grade nanoparticles from chocolates: An in vitro and in vivo study. Appl. Microbiol. Biotechnol. 2019, 103, 6689–6700. [Google Scholar] [CrossRef]
  190. Liu, J.M.; Zhao, N.; Wang, Z.H.; Lv, S.W.; Li, C.Y.; Wang, S. In-Taken Labeling and in Vivo Tracing Foodborne Probiotics via DNA-Encapsulated Persistent Luminescence Nanoprobe Assisted Autofluorescence-Free Bioimaging. J. Agric. Food Chem. 2019, 67, 514–519. [Google Scholar] [CrossRef]
  191. EFSA Scientific Committee. Scientific Opinion: Guidance on the risk assessment of the application of nanoscience and nanotechnologies in the food and feed chain. EFSA J. 2011, 9, 2140. [Google Scholar] [CrossRef]
  192. McClements, D.J.; Xiao, H. Is nano safe in foods? Establishing the factors impacting the gastrointestinal fate and toxicity of organic and inorganic food-grade nanoparticles. NPJ Sci. Food 2017, 1, 6. [Google Scholar] [CrossRef]
  193. Gwinn, M.R.; Vallyathan, V. Nanoparticles: Health Effects—Pros and Cons. Environ. Health Perspect. 2006, 114, 1818–1825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Overview of prebiotics.
Figure 1. Overview of prebiotics.
Ijms 21 02285 g001
Figure 2. Overview of mechanism of action of pre and probiotics.
Figure 2. Overview of mechanism of action of pre and probiotics.
Ijms 21 02285 g002
Figure 3. An overview of probiotics.
Figure 3. An overview of probiotics.
Ijms 21 02285 g003
Table 1. An updated overview of in vitro and in vivo studies on prebiotic, probiotic, and synbiotic products.
Table 1. An updated overview of in vitro and in vivo studies on prebiotic, probiotic, and synbiotic products.
TypeMicroorganisms/PrebioticsActivityStudyReferences
ProbioticBacillus and EnterobacterAnticancer and antioxidant effectThe intracellular cell-free supernatants (CFS) from Bacillus licheniformis KT921419 and the ethyl acetate extracts could control the growth of HT-29, a colon cancer cell line[100]
L. plantarum C70Anticancer effectL. plantarum C70 by releasing the exopolysaccharide caused 73.1% and 88.1% cytotoxic properties against the breast and colon cancers, respectively[101]
Kluyveromyces marxianus and Pichia kudriavzeviiAnticancer effectAccording to analysis of Annexin V/PI and DAPI, an apoptotic induction was observed due to exopolysaccharides released by probiotic yeasts of Kluyveromyces marxianus and Pichia kudriavzevii[102]
Lactobacilli cocktailAnticancer effectHT-29, a human colorectal carcinoma cell line was controlled by Lactobacilli cocktail via the modulation of the Notch and Wnt/β-catenin signaling pathways[104]
L. rhamnosusAnticancer effectThe bioconversion of cranberry proanthocyanidins to Lactobacillus rhamnosus could result in the IC50 values of 20.1 and 47.8 μg/mL[105]
Bifidobacterium infantis, L. acidophilus, Enterococcus faecalis, Bacillus cereusAnti-inflammatory effectA mixture of aerobic probiotics improved the functions of various intestinal barriers and the restoration of lucrative intestinal microbiota in the mouse model of DSS-induced chronic colitis, meaning anti-inflammatory properties[127]
Saccharomyces boulardii CNCM I-745Anti-inflammatory effectThe inflammatory response was modulated in mucositis caused by 5-FU (fluorouracil) via the probiotic Saccharomyces boulardii CNCM I-745 through the control of TLR 2 and 4 as well as the reduction of pro-inflammatory and NF-κB cytokines[103]
L. casei IMAU60214Immunomodulatory effectThe use of L. casei IMAU60214 killed by heat increased the activity of M1-like pro-inflammatory phenotype through the TLR2 signaling pathway[123]
L. plantarumAntimicrobial effectL. plantarum ZLP001 impeded the ETEC adhesion and linked with IPEC-J2 cells via the competition and exclusion[122]
LactobacillusAnti-diabetic effectThe lactobacillus strain alleviated the levels of blood sugar and HbA1c in diabetic rats[115]
L. plantarum LMT1-48Anti-obesity effectThe body weight and abdominal fat content were decreased in mouse models fed a modified diet through the administration of L. plantarum LMT1-48 at a density of 106 CFU/mL[117]
Hafnia alveiAnti-obesity effectFat mass, food intake, and body weights were reduced in the mouse model of obesity and hyperphagia[118]
Eurotium cristatumAnti-obesity effectThe administration of Eurotium cristatum showed anti-obesity activity in mice fed a high-fat diet (HFD) through the modulation of gut microbiota[119]
L. plantarum strain TCI378Anti-obesityThe expression of glucose transporter type 4 (GLUT-4) and adipocyte-specific genes perilipin 1 was suppressed by metabolism derivatives from L. plantarum strain TCI378[120]
PrebioticGalacto-oligosaccharides and phycocyaninAnticancer effectThe prebiotics co-administered by phycocyanin arrested the cell cycle at the G0/G1 phase, resulting in inhibited growth of HCT116 cells[141]
Chondroitin Sulfate DisaccharideAnticancer effectThe growth of HT-29, human colon cancer cell line, was controlled by Chondroitin sulfate (CS)-Keel disaccharide (CSD) generated by chondroitin AC lyase, estimating at 80% antiproliferative activity[140]
Short-chain fatty acidsAntiproliferative effectsThe administration of short-chain fatty acids (SCFAs) prevented the expression of genes involved in human colorectal cancer cells[142]
Blueberry anthocyaninsAntioxidant effectThe density and composition of intestinal microbiota in human models were increased by consumption of high purity blueberry anthocyanins through the increase in the modulatory and prebiotic activities[143]
OligosaccharidesAntioxidant effectThe water-soluble oligosaccharide of EMOS-1a showed 1420% proliferation level[144]
Lycium barbarum polysaccharideImmunomodulation
effect
The administration of polysaccharides derived from Lycium barbarum in mice showed immunomodulatory effects, and enhanced density of beneficial bacteria and gut microbiota[136]
SynbioticDjulis (Chenopodium formosanum) with L. acidophilusAnticancer effectThe co-administration of Djulis (Chenopodium formosanum Koidz.) and Lactobacillus acidophilus inhibited the growth of rat colon cancer cells through the promotion of apoptosis, proliferation, and inflammation[80]
L. casei, acidophilus, rhamnosus, bulgaricus, Bifidobacterium breve, longum and Streptococcus thermophilus with fructo-oligosaccharides.Anticancer and antioxidant effectTen weeks of low-calorie diet program along with synbiotic supplementation enhanced the activity of superoxide dismutase (SOD) and reduced the serum level of malondialdehyde (MDA) in obese patients suffering from breast cancer-related lymphedema[89]
Weissella cibaria FB069 with xylooligosaccharidesAnticancer effectThe use of synbiotic-fermented soymilk (containing xylooligosaccharides and Weissella cibaria FB069) inhibited the proliferation of HCT116 and Caco-2, colorectal cancer cell lines, through the reduction in the transcription of MD2/TLR4/MyD88/NF-κB[90]
Auricularia auricula aqueous with L. acidophilus La-5 and Bifidobacterium bifidum Bb-12Antioxidant effectThe aqueous extract of Auricularia auricula in the presence of L. acidophilus La-5 and Bifidobacterium bifidum Bb-12 significantly elevated the level of phenolic compounds and the activity of antioxidant properties up to 1057.6 mg of Gallic acid/kg and 115.30 of mg BHT eq/kg following 28-day storage[91]
L. bulgaricus PXN 39, L. casei subsp. casei PXN 37, Bifidobacterium breve PXN 25, L. rhamnosus PXN 54, B. infantis PXN 27 Lactobacillus acidophilus PXN 35, Streptococcus thermophilus PXN 66 with fructo-oligosaccharidesImmunomodulation
effect
The use of multispecies symbiotic showed immunoregulatory effects on the expression levels of CD4 and IgA in mice exposed to lipopolysaccharide (LPS)[92]
L. plantarum with inulinNeuropsychological effectConcomitant administration of inulin and L. plantarum in diabetic rats improved CREB/BDNF/TrkB signaling pathway, serotonin secretion, brain parameters, intestinal microbial composition, and oxidative stress, thus leading to improved memory and learning disorders[93]
β-glucan, Bacillus coagulans, and inulin, lactic acidAnti-diabetic effectEight weeks of taking daily synbiotic plus lactic acid improved the levels of GSH-Px, SOD and HbA1c in patients with type II diabetes[94]
Corn starch, maltodextrin, inulin, fructooligosaccharides, potassium chloride, magnesium sulfate, mangan sulfate with L. casei W56, acidophilus W22, paracasei W20, salivarius W24, plantarum W62, Lactococcus lactis W19, Bifidobacterium lactis W51 and W52, and Bifidobacterium bifidum W23Improve symptoms of diarrhea-predominant irritable bowel syndromeIrritable bowel syndrome (IBS) symptoms were improved by synbiotic treatment through an increase in fecal acetate and butyrate, colonic CD4+ T cells, mucosal microbial diversity as well as a decrease in surrogate of intestinal barrier function and fecal zonulin[95]
Grape pomace extract with lactobacilliAnti- inflammatory effectThe co-administration of lactobacilli and prebiotic grape pomace caused a downregulation of inflammatory genes, proteins, signaling molecules through the symbiotic effects[96]
L. acidophilus, L. rhamnosus, B. longum and Bifidobacterium bifidum, Saccharomyces boulardii with fructo-oligosaccharidesHepatoprotective effectsThe administration of synbiotic soy yogurt controlled hypercholesterolemia in mice liver by reducing the levels of low-density lipoprotein cholesterol, triacylglycerols, blood cholesterol, and lipid peroxidation.[97]
Table 2. Emerging applications of nanotechnologies on nanoprobiotics, nanoprebiotics, and nano synbiotics.
Table 2. Emerging applications of nanotechnologies on nanoprobiotics, nanoprebiotics, and nano synbiotics.
TypeActivityStudyReferences
ProbioticAntimicrobial effectThe polylysine-induced poly glutamic acid (PG) films caused protection of probiotics against food-borne pathogens[154]
Anticancer effectThe high levels of synthesized silver/Lactobacillus rhamnosus GG nanoparticles (Ag-LNPs) led to a decline in the rate of HT-29 live cells[155]
Anticancer and antimicrobial effectThe fabrication of copper oxide nanoparticles (CuO-NPs) using L. casei could control the proliferation of HT-29, a human colon carcinoma cell line, and human gastric carcinoma cell line, as well as could eliminate Pseudomonas aeruginosa and Staphylococcus aureus[156]
Anticancer and antioxidant effectThe L. casei capped-SeNPS suppressed the cytotoxicity caused by Diquat and oxidative damage, impeded the cell damage and apoptosis induced by H2O2, and induced the apoptosis mediated by the HepG2 cell line[157]
Anticancer and antioxidant effectThe findings from the administration of L. casei 393-SeNPs were the induction of HepG2 cell line apoptosis, the elevation of oxidative damage caused by Diquat in IECs, and the reduction in gut barrier dysfunction caused by ETEC K88 via the antioxidant functions, the regulation of inflammation, the establishment of gut epithelial barrier integrity, and the balance of gut microflora[158]
Anticancer effectDead nano-scale L. plantarum could impede the proliferation of a colorectal cancer cell line through an increase in the expression level of IgA, an induction of cancer cell cycle arrest and apoptosis, and a suppression of inflammatory response[159]
Anticancer and antioxidant effectThe synthesis gold nanoparticles (AuNps) having antioxidant activity and low cytotoxicity using L. kimchicus DCY51T strain exhibited the activity of a protective protein capping layer[160]
PrebioticImprove drug deliveryHigh molecular weight (HMW) inulin nanoparticles were fabricated to achieve drug delivery system, whose concentration of <200 μg/mL had no toxicity for peripheral blood mononuclear cells (PBMCs)[161]
Antimicrobial effectThe probiotics were internalized by phthalyl dextran nanoparticles (PDNs) to construct pediocin, aiming at the alteration of gut microbiome composition, the suppression of pathogenic intestinal infections, and the elevation of beneficial bacteria species[162]
Antimicrobial effectThe higher pediocin generation following the administration of PIN-internalized probiotics with 0.171 polydispersity index (PDI) with a size of about 203 nm showed the maximum antimicrobial properties[163]
SynbioticAntimicrobial effectThe activity of Listeria monocytogenes and Escherichia coli K99 was inhibited by L. plantarum exposed to phthalylpullulan nanoparticle (PPN) due to production of antimicrobial peptides via intracellular stimulation[164]
The photo protective effectA cream containing L. rhamnosus plus Selenium nanoparticles could heal the side effects induced by sunburn and showed sun protection factor (SPF) of 29.77 in Wistar rat model[165]
Improve delivery systemA new formulation of nano-emulsion containing E. faecium plus inulin could increase probiotic bacterial viability and stability[166]
Improve tolerance of probiotic bacteriaBeads reinforced by inulin (5% w/v) had the highest effect on bacterial protection against bile salts[167]

Share and Cite

MDPI and ACS Style

Durazzo, A.; Nazhand, A.; Lucarini, M.; Atanasov, A.G.; Souto, E.B.; Novellino, E.; Capasso, R.; Santini, A. An Updated Overview on Nanonutraceuticals: Focus on Nanoprebiotics and Nanoprobiotics. Int. J. Mol. Sci. 2020, 21, 2285. https://doi.org/10.3390/ijms21072285

AMA Style

Durazzo A, Nazhand A, Lucarini M, Atanasov AG, Souto EB, Novellino E, Capasso R, Santini A. An Updated Overview on Nanonutraceuticals: Focus on Nanoprebiotics and Nanoprobiotics. International Journal of Molecular Sciences. 2020; 21(7):2285. https://doi.org/10.3390/ijms21072285

Chicago/Turabian Style

Durazzo, Alessandra, Amirhossein Nazhand, Massimo Lucarini, Atanas G. Atanasov, Eliana B. Souto, Ettore Novellino, Raffaele Capasso, and Antonello Santini. 2020. "An Updated Overview on Nanonutraceuticals: Focus on Nanoprebiotics and Nanoprobiotics" International Journal of Molecular Sciences 21, no. 7: 2285. https://doi.org/10.3390/ijms21072285

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

Article Metrics

Back to TopTop