Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3387 word(s) 3387 2022-03-16 08:57:34 |
2 formating Meta information modification 3387 2022-03-17 04:42:18 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Reyes, L. Polymeric Platforms and Delivery Systems of Probiotics. Encyclopedia. Available online: https://encyclopedia.pub/entry/20653 (accessed on 15 September 2024).
Reyes L. Polymeric Platforms and Delivery Systems of Probiotics. Encyclopedia. Available at: https://encyclopedia.pub/entry/20653. Accessed September 15, 2024.
Reyes, Luis. "Polymeric Platforms and Delivery Systems of Probiotics" Encyclopedia, https://encyclopedia.pub/entry/20653 (accessed September 15, 2024).
Reyes, L. (2022, March 16). Polymeric Platforms and Delivery Systems of Probiotics. In Encyclopedia. https://encyclopedia.pub/entry/20653
Reyes, Luis. "Polymeric Platforms and Delivery Systems of Probiotics." Encyclopedia. Web. 16 March, 2022.
Polymeric Platforms and Delivery Systems of Probiotics
Edit

The selection of optimal material for probiotic encapsulation and the appropriate processing route are key parameters to ensure an efficient delivery strategy, where early degradation by GIT stimuli and harsh conditions are largely avoided. Nevertheless, selecting the materials for superior performance in complex physiological environments such as the GIT is a task made challenging not only by the obstacles to be overcome to reach the target site but also by the need to maintain high biological activities and positive responses to the changing surroundings. The fast-growing notion that encapsulates can be composed of active biomaterials should be driven by matching the material’s properties with expected responses through the GIT. Particularly, polymers exhibit versatile molecular moieties that have been widely exploited to fabricate chemical and physical delivery platforms with properties that can be finely tuned by adjusting interchain interactions. Chemical polymeric scaffolds are formed by covalent bonds between adjacent chains, while the physical ones are maintained together by charged polyvalent surfactants or ion interactions. Moreover, their versatile processability schemes facilitate obtaining different morphologies, functionalities, and the possibility to form composites with nanostructured materials in search of an enhanced response when subjected to a stimulus. Typical polymeric encapsulates comprise microparticles, microspheres, microcapsules, hydrogels, and, more recently, nanocomposite 3D matrices.

Nanostructure Hydrogels Polymeric probiotic

1. Introduction

The term “probiotic” has been complimented since its first appearance in the 1960s. It was initially defined as a substance secreted by microorganisms that has beneficial effects on the human body [1][2]. Then, in 1980, some specific characteristics were added to this definition. Additional claims stated that probiotics are “strains that have a beneficial impact, non-toxic, non-allergic, and nonpathogenic, available in large quantities as viable cells, suitable for the environment of the gut, and storable as well as stable” [1]. The Food and Agriculture Organization (FAO) and the World Health Organization (WHO) defined probiotics as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host” [1][2][3][4]. Additionally, both organizations have classified products containing live organisms into four categories: (a) live or active cultures; (b) probiotics in food or supplements without a health claim; (c) probiotics in food or supplements with a specific health claim; and (d) probiotic drugs [2].
Probiotic strains generally belong to the following genera: Lactobacillus, Bifidobacterium, Saccharomyces, Pediococcus, Streptococcus, and Leuconostoc [1]. Lactobacillus and Bifidobacterium are the most common strains. Different aspects need to be considered when selecting probiotic strains, and these include stomach pH and bile tolerance, adherence to epithelial surfaces, capacity for immunostimulation, antagonistic activity against pathogens, and antimutagenic and anticarcinogenic properties [5]. Probiotics have been successfully employed in manufacturing a wide variety of fermented products for daily consumption, including yogurt, kefir, sour pickles, milk, miso soup, and several soft cheeses [6]. The average probiotic consumption for a single person varies from 107 to 109 CFU/mg/day, whereas the significant benefit probiotic content in food must be of the order of 106 CFU/g [1][4]. Major health benefits have been attributed to these microorganisms, which nowadays can be used for the prevention and treatment of ailments such as liver disorders, cardiovascular diseases, dental caries, gastrointestinal inflammation, diarrhea, diabetes, obesity, and irritable bowel syndrome [1][3][6][7].
Probiotics can produce essential metabolites, including enzymes, vitamins, amino acids, peptides, exopolysaccharides, antioxidants, and anti-inflammatory agents. For example, some Bifidobacterium strains can produce B6 vitamin, while L. reuteri can produce cobalamin [8]. Some studies performed on pediatric patients who suffered from some kind of food allergy showed, with moderate certainty, that consumption of probiotics such as L. rhamnosius GG, LC705, L. casei LOCK 0900 and LOCK 0908, L. paracasei LOCK 0919, B. breve Bbi99, Propionibacterium freudenreichii ssp, or Shermanii JS could alleviate the symptoms caused by bovine lactose intolerance. This is because probiotics are thought to induce the production of β-galactosidase and lactase enzymes, helping to metabolize lactose quite effectively [8][9].
However, it is important to keep in mind that first, the strain must reach its site of action, usually the gut, and thus survive the physiological stress met during its ingestion, i.e., acid stomach and gut pH and the presence of biliary salts. Furthermore, its ingestion must not lead to any major risks for the host, maintaining its characteristics and remaining stable during the manufacturing process where it is usually incorporated into a delivery matrix [6]. A few strains can maintain viability after 1 h at a pH of 1, and most of them lose viability after 3 h at a pH of 3. The human stomach pH varies from 1 when fasting to 4.5 after eating. The process can take more than 3 h [10].
Different strategies exist to protect probiotics and their viability by edible carriers such as cheese, drinks, and bread [11][12]. Also, polymeric matrices have attracted significant attention for encapsulation, protection, and probiotic release [13]. Probiotics have evolved in sophistication from the first to the fourth generation. First-generation probiotics are either fresh or lyophilized cells without any coating. This has led to a low survival rate between 7% and 30%. Second-generation probiotics are incorporated into polymeric capsules or tablets with fillers. Usually, these strains show higher survival rates, but low performance due to rapid metabolite degradation. Third-generation probiotics are those encapsulated in natural, semi-synthetic, or synthetic polymers. The microcapsules are designed to be consumed gradually, which helps maintain the metabolites’ activity. Their structure can be a 3D matrix, a crosslinked construct, or have an external coating. Fourth-generation probiotics are those incorporated into biofilms, which improve protection when transiting the gastrointestinal tract [14][15].
Encapsulation techniques are widely used for varied applications in the food industry, including masking and design of flavors, colors, and odors, improving the shelf life of products, protecting some components against nutritional loss, and regulating undesirable oxidative reactions. With probiotics, encapsulation provides protection from media effects and enhanced viability and allows controlled dosing and handling of cells [16]. One of the biggest challenges when encapsulating is selecting effective and safe materials for the capsules’ manufacturing, an efficient release system, and proper production techniques (e.g., extrusion, emulsion, spray-drying, etc.). A much more comprehensive discussion of different methods used in the design of encapsulation microgels is given by McClements [13]. Additionally, it is vital to consider economic, regulatory, and consumption factors to assure scalability and successful market penetration [13][16].
Materials used for capsule manufacturing have diverse origins. The most common ones are derivates from cellulose, proteins, polysaccharides, carrageenan, gelatin, pectin, and alginate [16]. Materials are chosen depending on the characteristic physicochemical and structural features of the capsules, and generally, polysaccharides and proteins are selected due to their versatility. In this regard, natural polymers such as alginate, xanthan gum with divalent cations, casein gels, or gelatins have been chosen due to their ease of crosslinking, and the possibility of combining them to achieve different levels of mechanical resistance. Chitosan, lysine, or whey protein are used for the external coating of structures [15]. Some of the most important physicochemical properties of the materials to consider for a rational design are solubility, gelation mechanisms, degradability, and electric properties [13]. There are different methods to perform encapsulation processes such as injection (e.g., extrusion, atomization, and microfluidic), template techniques (e.g., emulsions), biopolymer phase separation, precipitation, reduction, drying, and more recently, biofilm formation to promote colonization and enhance the permanence of probiotics in the host intestinal mucosa [13][14].
The gastrointestinal tract comprises the mouth, esophagus, stomach, gut, and colon. Its microbiota concentration varies over the tract due to changes in pH and the presence of bile and enzymes. For example, in the stomach, such concentration is low (101 bacteria/g), increasing through the duodenum (103 bacteria/g), the jejunum (104 bacteria/g), and the ileum (107 bacteria/g). The largest concentration of microorganisms is found in the gut and colon, rounding 1011 to 1012 bacteria/g [17]. Such microbiota presents a great phylogenetic diversity, allowing the required metabolic performance. The present microbiota is mainly composed of Bacteroidetes, Firmicutes, and Proteobacteria, and to a lesser extent by Actinobacteria, Clostridium, Enterobacter, Verrumicrobia, bacteriophages, viruses, and several Aspergillus, Candida, Cryptococcus, and Penicillium fungi genres [7][18][19].
The digestion and nutrient absorption processes are carried out by the small intestine, where there is an intestinal barrier composed of a mucosae layer and a cell component (intestinal epithelium and underlying lamina propria) that acts as a physical barrier to the microorganisms present in the gut [3]. Mucosae is composed of an outer loosely adhering layer and a dense inner layer. This last one is the first effective defense mechanism because of its high density, preventing most bacteria from penetrating and adhering [17]. The intestinal epithelium creates a separation between the gut lumen and the lamina propria, and comprises enterocytes, goblet, Paneth, and endoenterocrine cells. In contrast, the lamina propria is formed by dendritic cells, macrophages, and plasma cells that can engulf pathogens and eliminate apoptotic cells and waste [3].
The microbiota existing along the intestine have an immunological vigilance function that allows the detection of pathogens and stimulates the immune system to respond adequately. The pathogen control mechanism comprises four major steps: first, production of bacteriocin and other inhibitors; second, the competitive exclusion by the binding sites; third, stimulation of the immune response; and last, the inhibition of virulent genes or expression of proteins in pathogens [7]. Intestinal homeostasis occurs when the immune system establishes an equilibrium between commensal, mutualistic, and opportunistic bacteria. This happens when the microbiota communicates effectively with the immune system through a healthy intestinal barrier [3].

2. Polymeric Materials in Probiotic Encapsulation

In protected-delivery technologies, a suitable polymeric material should be able to preserve its core from adverse environmental conditions (e.g., reduce the acid-induced degradation of probiotics by gastric fluids in the stomach), exhibit inertness with the encapsulated materials, promote a controlled release of the encapsulate, achieve higher encapsulation efficiencies of bioactive compounds on a per mass basis and, ultimately, favor high levels of absorption into the targeted organs (i.e., the overall efficacy of the compounds) [20][21][22]. All the selected materials must also be biodegradable and biocompatible since they will be in direct contact with various types of cells [20][23].
Among the natural polymers, alginate, a heteropolysaccharide, has been applied successfully as a pH-sensitive material for the encapsulation of probiotic bacteria [24]. Alginate, extracted from algae, is composed of two monosaccharide units: α-L-guluronic acid and β-D-mannuronic acid, linked together by a β (1–4) glycosidic bond. Due to its toxicity, inexpensiveness, ease of processing, and biocompatibility, calcium alginate has been extensively employed in the encapsulation of probiotics [21][25][26][27][28][29]. Yet, calcium alginate encapsulates are chemically susceptible to disintegration in the presence of excess monovalent ions, Ca2+-chelating agents such as phosphate and citrate, and harsh chemical conditions (e.g., low pH) [30][31][32]. To increase the stability of alginate and decrease the loss of encapsulated material, alginate is usually coated with polycationic polymers such as chitosan and poly-L-lysine [20][23][32][33].
Chitosan is a very abundant polysaccharide obtained from chitin and is composed of (1,4)-linked 2-amino-deoxy-b-d-glucan [34]. It also shows high biocompatibility and biodegradability under physiological conditions. For these reasons, it has enabled several encapsulation applications in the food and pharmaceutical industries, including liposome coating, chitosan–alginate coating, controlled delivery of small molecules and biologicals, and the release of bioactive metabolites (e.g., essential oils, probiotics, vitamins, antioxidants, and flavors). The unique cationic character of chitosan allows forming multi-layer systems with anionic alginate for probiotic encapsulation, which can bring efficient protection to cargoes, reduced porosity, stability at various pH ranges, and reduced leakage of the encapsulated probiotic. In experiments where the gastric environment was simulated, chitosan coating was more efficient than poly-l-lysine and alginate coatings in protecting probiotics, which represents a possible route to overcome the challenges of oral delivery [34]. Furthermore, chitosan coating with drying processes can prolong the long-term storage of some probiotics at different temperatures [35].
Another frequently studied material for encapsulation is gelatin, a commercially available denatured protein obtained by the hydrolysis of collagen from the skin and bones of bovines or fish. This protein might be positively or negatively charged, depending on whether an acidic or alkaline method was used for its extraction, which can be exploited to design multifunctional controlled release systems [36]. For example, gelatin-coated alginate capsules and microspheres can protect labile drugs from the stomach’s acidic environment, enabling their release in the target intestinal area, whose environment has a basic pH [37]. Other studies have reported that the addition of fish gelatin significantly raised polymer matrix density and improved the physical integrity of alginate capsules because of a more stable and ordered 3D structure. Some other aspects, such as the survival rate of microorganisms during the GIT’s passage, are enhanced compared to non-encapsulated cells [38].
Poly-l-lysine is a cationic, non-ribosomal, non-toxic, biocompatible, biodegradable, and antimicrobial homopolymer produced by modified strains of Streptomyces albulus. Lysine is frequently used as a preservative and food additive [39], and studied as an alginate bead coating. The marked antimicrobial activity of most cationic polymers poses a major challenge because they tend to inhibit the growth of some microorganism strains, depending on pH and incubation times. That is the case not only of lysine but also of chitosan and polyethyleneimines [40]. However, different in vitro studies that have implemented mixed manufacturing techniques, such as freeze-frying, have shown that poly-l-lysine coatings for alginate capsules are well-suited to maintain cells’ growth and proliferation at low pH values and viability for a storage period of up to 16 weeks at 4 °C [41].
The materials mentioned above correspond to the most popular and studied polymers for survival enhancement and protection of probiotics in hostile environments, such as those found in the compartments of the GIT. Many other materials might be suitable for producing capsules and coatings for this application, including polyethyleneimines, poly(2-dimethyl(aminoethyl)methacrylate), dextran, pectin, Arabic gum, starch, sodium caseinate, polyvinyl alcohol, polyethylene glycol, polyacrylic acid, and succinylated or acylated carrageenan [41][39][42][43].

3. Hydrogels

Hydrogels have drawn particular attention among encapsulation alternatives for probiotics, given their ease of processing, the wide range of materials available for their fabrication, and their capability to form three-dimensional networks that protect the probiotic’s integrity [44]. Remarkably, the inertness of hydrogels in environments with high water activity makes them suitable to entrap molecules and microorganisms, and ensure their integrity and viability in physiological environments [45]. However, one of the main concerns about implementing hydrogels is the proper tuning of mechanical performance, along with optimal porosity to enable microorganism survival while maintaining sufficient cell entrapment and a considerable degree of swelling [46]. This has been addressed by chemically modified polymers through different routes, including the addition of ionizable functional groups, functionalized backbones, and combined polymeric blending [47][48][49].
Hydrogels can be classified mainly according to their (i) composition (homo or copolymers), (ii) network size (macrogels, microgels, nanogels), (iii) electrical charge (non-ionic, cationic, anionic, amphoteric or zwitterionic), and (iv) crosslinking method (physical or chemical) [50][51]. Moreover, recent studies have demonstrated the fabrication of chemically crosslinked platforms oriented toward the encapsulation of probiotics into hydrogel beads crosslinked with the aid of glutaraldehyde, Genipin, calcium chloride (CaCl2), and ferrous sulfate (FeSO4) [52][53][54][55]. Alginate, gelatin, and chitosan are the three main polymers of choice for hydrogel synthesis as they have proven to be effective in protecting probiotic cells from harsh environmental conditions [51][56].
One of the most intensive research areas is the development of strategies for tuning degradation rates and matrix porosities to improve their performance in protecting encapsulated living organisms [57]. This has been achieved by the supramolecular design of monomeric structures and by controlling polymerization reactions with carefully applied light and thermal stimuli [58][59]. Depending on the polymerization scheme selected (i.e., bulk polymerization, solution polymerization, suspension polymerization, emulsion polymerization, and graft polymerization), macroscopic and microscopic properties such as porosity and polymeric mesh size might change significantly [60]. Notably, the capacity of engineering the tortuosity and interconnectivity of hydrogels’ mesh has been reported as critical for the smart release of the encapsulated cargoes [61]. Further control over such a process can be achieved by chemical modifications with hygroscopic polymers such as polyethyleneglycol, which leads to the enhanced mucoadhesiveness of the encapsulates by both physical entanglement and hydrogen bonding with the base encapsulating polymer [62][63]. Therefore, the following section will discuss the fast-growing area of stimuli-responsive hydrogels, emphasizing how they can be activated, de-activated, or re-activated for a particular delivery purpose [64].

4. Microencapsulates

Microparticles, microcapsules, and microspheres usually made of food-grade polymers, such as alginate, chitosan, carboxymethyl cellulose, cellulose acetate phthalate, xanthan gum, starch, carrageenan, gelatin, and pectin [25][65], have demonstrated to be protective barriers of high performance against the GIT’s environmental conditions [66][67][68][69][70]. These microencapsulates’ dimensions usually range between 1 and 1000 µm [71]. According to recent studies, an effective microencapsulation system should maintain the stability of the probiotics during storage, protect them from the harsh conditions of the upper GIT, release them in the colon, and finally, promote their ability to colonize the mucosal surfaces [71][72][73][74].
Microparticles typically consist of a core composed of one to several ingredients surrounded by a wall or barrier of uniform or non-uniform thickness, either single-layered or multi-layered. The design of microencapsulated ingredients requires knowledge of (1) the core, (2) the materials for encapsulation, (3) the interactions between the core, matrix, and the environment, (4) the stability of the microencapsulated ingredients under storage conditions and when incorporated into food matrices, and (5) the mechanisms that control the release of the core [22][25][65]. Matrix degradation, and consequently, the release of its contents, can be controlled to occur at different times. Larger particles generally release encapsulated compounds more slowly and over more extended periods, while particle size reduction favors adhesiveness and therefore prolonged GI transit time, leading to a higher drug bioavailability [22][25][65][75].
Typical technologies employed for probiotic encapsulation include emulsification [76][77][78], emulsification and enzymatic gelation [79][80][81][82], atomization (e.g., spray drying [77][78][83], spray freeze drying [77][78][83][84][85]), coating and agglomeration [78][86][87][88], and extrusion [89][90].

5. Nanostructured Platforms

One of the leading strategies to improve microencapsulates’ tolerance to different GIT environments and ensure their efficient delivery is physical and chemical blending with stimuli-responsive and high mechanical performance nanomaterials [91][92][93]. This approach allows superior control over probiotic delivery due to the unique properties attainable by forming nanocomposites and the possibility of providing multimodal delivery platforms (i.e., including more than one encapsulated component) where survival of probiotics is increased substantially [94][95]. Accordingly, several nanostructured materials have been explored in the fabrication of next-generation delivery platforms, including polymeric and iron oxide-based nanoparticles, nanosheets (e.g., graphene oxide—GO and phyllosilicate clays), nanoliposomes, micelles, and nanoparticles derived from naturally occurring polymers such as nanocellulose and starch nanocrystals [96][97][98].
The dispersion of nanoparticles (NPs) into polymeric arrangements (to form nanocomposites) is attractive mainly due to their intrinsic capacity to fill out pores, therefore avoiding the diffusion of molecules such as hydrogen ions, bile salts, or digestive enzymes that may lead to the undesirable degradation of the encapsulated probiotics [99]. For instance, magnesium oxide (MgO) NPs have been incorporated into alginate–gelatin microgels, which resulted in a more stable encapsulation of probiotics when compared to unmodified microgels. The MgO NPs help neutralize the hydrogen ions present in the gastric fluids, which diminishes the acid-induced degradation of probiotics and maintains a neutral pH inside the microgels [20]. Alternatively, the use of NPs can help improve some physicochemical properties of the encapsulates, such as hardness, compressibility, cohesiveness, and adhesiveness. This is the case of chitosan NPs, which have been reported to enhance the mucoadhesive properties of hydrogels [100]. Through this approach, the interaction with the intestinal mucus is improved by the electrostatic interaction and physical entanglements of the chitosan-containing matrices facilitated by the positive charge of chitosan.
Another approach suggests that combining polymeric platforms with nanocrystals derived from polysaccharides (e.g., cellulose and starch) enhances mechanical stability and shelling properties [101], and increases the surface area for target delivery [102]. For instance, when cellulose nanocrystals (NCs) are combined with alginate during the microencapsulation process, the dissolution time increases while porosity is reduced significantly [103]. Moreover, ionic interaction between the material and the nanocellulose filler reduces the infiltration of gastric fluids, preventing the degradation of probiotics [104]. When starch NCs are implemented as fill-in alginate-based delivery platforms, thicker barrier protection against gastric and intestinal juices provides a stable mechanism to decrease probiotics mortality [96].
Remarkably, novel developments in the fabrication of nanocarbon-based materials (e.g., reduced graphene oxide, graphene quantum dots, graphene nanoribbons), silica-based nanocarriers, and inorganic nanoparticles have enabled the fabrication of emerging nanocomposites with improved mechanical strengths, high drug loading, and reduced toxicity [91][105][106]. These features have been reported to not only favor the controlled release of on-cargo molecules but also increase the survival of encapsulated living organisms (e.g., probiotics and mammalian cells) [107][108]. Similarly, clay mineral silicates have gained popularity given their unique cationic exchange properties that can be exploited to fabricate water barriers due to hydrogen bonding [109][110]. Kim and colleagues reported superior shape integrity for bentonite/alginate-based encapsulates during gastric fluid exposure and appropriate disintegration in the intestinal area upon oral administration in mice [105].

References

  1. Ranjha, M.M.A.N.; Shafique, B.; Batool, M.; Kowalczewski, P.; Shehzad, Q.; Usman, M.; Manzoor, M.F.; Zahra, S.M.; Yaqub, S.; Aadil, R.M. Nutritional and Health Potential of Probiotics: A Review. Appl. Sci. 2021, 11, 11204.
  2. Quigley, E.M. Prebiotics and Probiotics in Digestive Health. Clin. Gastroenterol. Hepatol. 2018, 17, 333–344.
  3. La Fata, G.; Weber, P.; Mohajeri, M.H. Probiotics and the Gut Immune System: Indirect Regulation. Probiot. Antimicrob. Proteins 2018, 10, 11–21.
  4. Oberoi, K.; Tolun, A.; Altintas, Z.; Sharma, S. Effect of Alginate-Microencapsulated Hydrogels on the Survival of Lactobacillus rhamnosus under Simulated Gastrointestinal Conditions. Foods 2021, 10, 1999.
  5. Mattila-Sandholm, T.; Myllärinen, P.; Crittenden, R.; Mogensen, G.; Fondén, R.; Saarela, M. Technological challenges for future probiotic foods. Int. Dairy J. 2002, 12, 173–182.
  6. Butel, M.-J. Probiotics, gut microbiota and health. Med. Mal. Infect. 2014, 44, 1–8.
  7. Vitetta, L.; Briskey, D.; Alford, H.; Hall, S.; Coulson, S. Probiotics, prebiotics and the gastrointestinal tract in health and disease. Inflammopharmacology 2014, 22, 135–154.
  8. Chugh, B.; Kamal-Eldin, A. Bioactive compounds produced by probiotics in food products. Curr. Opin. Food Sci. 2020, 32, 76–82.
  9. Tan-Lim, C.S.C.; Esteban-Ipac, N.A.R. Probiotics as treatment for food allergies among pediatric patients: A meta-analysis. World Allergy Organ. J. 2018, 11, 25.
  10. Maragkoudakis, P.A.; Zoumpopoulou, G.; Miaris, C.; Kalantzopoulos, G.; Pot, B.; Tsakalidou, E. Probiotic potential of Lactobacillus strains isolated from dairy products. Int. Dairy J. 2006, 16, 189–199.
  11. Rolim, F.R.; Neto, O.C.F.; Oliveira, M.E.G.; Oliveira, C.J.; Queiroga, R.C. Cheeses as food matrixes for probiotics: In vitro and in vivo tests. Trends Food Sci. Technol. 2020, 100, 138–154.
  12. Soares, M.B.; Martinez, R.C.; Pereira, E.P.; Balthazar, C.F.; Cruz, A.G.; Ranadheera, C.S.; Sant’Ana, A.S. The resistance of Bacillus, Bifidobacterium, and Lactobacillus strains with claimed probiotic properties in different food matrices exposed to simulated gastrointestinal tract conditions. Food Res. Int. 2019, 125, 108542.
  13. McClements, D.J. Designing biopolymer microgels to encapsulate, protect and deliver bioactive components: Physicochemical aspects. Adv. Colloid Interface Sci. 2016, 240, 31–59.
  14. Salas-Jara, M.J.; Ilabaca, A.; Vega, M.; García, A. Biofilm Forming Lactobacillus: New Challenges for the Development of Probiotics. Microorganisms 2016, 4, 35.
  15. Trush, E.; Poluektova, E.A.; Beniashvilli, A.G.; Shifrin, O.S.; Poluektov, Y.M.; Ivashkin, V.T. The Evolution of Human Probiotics: Challenges and Prospects. Probiot. Antimicrob. Proteins 2020, 12, 1291–1299.
  16. Huq, T.; Khan, A.; Khan, R.A.; Riedl, B.; Lacroix, M. Encapsulation of Probiotic Bacteria in Biopolymeric System. Crit. Rev. Food Sci. Nutr. 2013, 53, 909–916.
  17. Dieterich, W.; Schink, M.; Zopf, Y. Microbiota in the Gastrointestinal Tract. Med. Sci. 2018, 6, 116.
  18. Fanaro, S.; Chierici, R.; Guerrini, P.; Vigi, V. Intestinal microflora in early infancy: Composition and development. Acta Paediatr. 2007, 92, 48–55.
  19. Tannock, G.W. Molecular assessment of intestinal microflora. Am. J. Clin. Nutr. 2001, 73, 410s–414s.
  20. Yao, M.; Li, B.; Ye, H.; Huang, W.; Luo, Q.; Xiao, H.; McClements, D.J.; Li, L. Enhanced viability of probiotics (Pediococcus pentosaceus Li05) by encapsulation in microgels doped with inorganic nanoparticles. Food Hydrocoll. 2018, 83, 246–252.
  21. Safi, M.A.E. Materials and Techniques for Microencapsulation of Probiotics. Biosci. Biotechnol. Res. Commun. 2021, 14, 922–928.
  22. da Silva, B.V.; Barreira, J.C.; Oliveira, M.B.P. Natural phytochemicals and probiotics as bioactive ingredients for functional foods: Extraction, biochemistry and protected-delivery technologies. Trends Food Sci. Technol. 2016, 50, 144–158.
  23. Survival of Lactobacillus Acidophilus as Probiotic Bacteria using Chitosan Nanoparticles. Int. J. Eng. 2017, 30, 456–463.
  24. Allan-Wojtas, P.; Hansen, L.T.; Paulson, A. Microstructural studies of probiotic bacteria-loaded alginate microcapsules using standard electron microscopy techniques and anhydrous fixation. LWT 2008, 41, 101–108.
  25. Shori, A.B. Microencapsulation Improved Probiotics Survival During Gastric Transit. HAYATI J. Biosci. 2017, 24, 1–5.
  26. Hansen, L.T.; Allan-Wojtas, P.; Jin, Y.-L.; Paulson, A. Survival of Ca-alginate microencapsulated Bifidobacterium spp. in milk and simulated gastrointestinal conditions. Food Microbiol. 2002, 19, 35–45.
  27. Etchepare, M.D.A.; Barin, J.S.; Cichoski, A.J.; Jacob-Lopes, E.; Wagner, R.; Fries, L.L.M.; De Menezes, C.R. Microencapsulation of probiotics using sodium alginate. Ciência Rural 2015, 45, 1319–1326.
  28. Mathews, S. Microencapsulation of Probiotics by Calcium Alginate and Gelatin and Evaluation of its Survival in Simulated Human Gastro-Intestinal Condition. Int. J. Curr. Microbiol. Appl. Sci. 2017, 6, 2080–2087.
  29. Khosravi Zanjani, M.A.; Tarzi, B.G.; Sharifan, A.; Mohammadi, N. Microencapsulation of probiotics by calcium alginate-gelatinized starch with chitosan coating and evaluation of survival in simulated human gastro-intestinal condition. Iran. J. Pharm. Res. 2014, 13, 843–852.
  30. Krasaekoopt, W.; Bhandari, B.; Deeth, H. The influence of coating materials on some properties of alginate beads and survivability of microencapsulated probiotic bacteria. Int. Dairy J. 2004, 14, 737–743.
  31. Shu, X.; Zhu, K. The release behavior of brilliant blue from calcium–alginate gel beads coated by chitosan: The preparation method effect. Eur. J. Pharm. Biopharm. 2002, 53, 193–201.
  32. Lee, J.S.; Cha, D.S.; Park, H.J. Survival of Freeze-Dried Lactobacillus bulgaricus KFRI 673 in Chitosan-Coated Calcium Alginate Microparticles. J. Agric. Food Chem. 2004, 52, 7300–7305.
  33. Koo, S.M.; Cho, Y.H.; Huh, C.S.; Baek, Y.J.; Park, J.Y. Improvement of the Stability of Lactobacillus casei YIT 9018 by Microencapsulation Using Alginate and Chitosan. J. Microbiol. Biotechnol. 2001, 11, 376–383.
  34. Călinoiu, L.-F.; Ştefănescu, B.E.; Pop, I.D.; Muntean, L.; Vodnar, D.C. Chitosan Coating Applications in Probiotic Microencapsulation. Coatings 2019, 9, 194.
  35. Santos, M.A.S.; Machado, M.T.C. Coated alginate–chitosan particles to improve the stability of probiotic yeast. Int. J. Food Sci. Technol. 2020, 56, 2122–2131.
  36. Albadran, H.A.; Monteagudo-Mera, A.; Khutoryanskiy, V.V.; Charalampopoulos, D. Development of chitosan-coated agar-gelatin particles for probiotic delivery and targeted release in the gastrointestinal tract. Appl. Microbiol. Biotechnol. 2020, 104, 5749–5757.
  37. Annan, N.; Borza, A.; Hansen, L.T. Encapsulation in alginate-coated gelatin microspheres improves survival of the probiotic Bifidobacterium adolescentis 15703T during exposure to simulated gastro-intestinal conditions. Food Res. Int. 2008, 41, 184–193.
  38. Liu, J.; Liu, F.; Ren, T.; Wang, J.; Yang, M.; Yao, Y.; Chen, H. Fabrication of fish gelatin/sodium alginate double network gels for encapsulation of probiotics. J. Sci. Food Agric. 2021, 101, 4398–4408.
  39. Ramos, P.E.; Cerqueira, M.A.; Teixeira, J.A.; Vicente, A.A. Physiological protection of probiotic microcapsules by coatings. Crit. Rev. Food Sci. Nutr. 2018, 58, 1864–1877.
  40. Hlaing, S.P.; Kim, J.; Lee, J.; Kwak, D.; Kim, H.; Yoo, J.-W. Enhanced Viability of Probiotics Against Gastric Acid by One-Step Coating Process with Poly-L-Lysine: In Vitro and In Vivo Evaluation. Pharmaceutics 2020, 12, 662.
  41. Asgari, S.; Pourjavadi, A.; Licht, T.R.; Boisen, A.; Ajalloueian, F. Polymeric carriers for enhanced delivery of probiotics. Adv. Drug Deliv. Rev. 2020, 161–162, 1–21.
  42. Misra, S.; Pandey, P.; Mishra, H.N. Novel approaches for co-encapsulation of probiotic bacteria with bioactive compounds, their health benefits and functional food product development: A review. Trends Food Sci. Technol. 2021, 109, 340–351.
  43. Zhu, Y.; Wang, Z.; Bai, L.; Deng, J.; Zhou, Q. Biomaterial-based encapsulated probiotics for biomedical applications: Current status and future perspectives. Mater. Des. 2021, 210, 110018.
  44. Xiao, Y.; Lu, C.; Liu, Y.; Kong, L.; Bai, H.; Mu, H.; Li, Z.; Geng, H.; Duan, J. Encapsulation of Lactobacillus rhamnosus in Hyaluronic Acid-Based Hydrogel for Pathogen-Targeted Delivery to Ameliorate Enteritis. ACS Appl. Mater. Interfaces 2020, 12, 36967–36977.
  45. Mosqueda, I.S.; Bousquets, A.L.; Montiel-Sosa, J.F.; Corona, L.; Álvarez, Z.G.; Gochi, L.C. Encapsulation of Lactobacillus plantarum ATCC 8014 and Pediococcus acidilactici ATCC 8042 in a freeze-dried alginate-gum arabic system and its in vitro testing under gastrointestinal conditions. J. Microencapsul. 2019, 36, 591–602.
  46. Li, R.; Zhang, Y.; Polk, D.B.; Tomasula, P.M.; Yan, F.; Liu, L. Preserving viability of Lactobacillus rhamnosus GG in vitro and in vivo by a new encapsulation system. J. Control. Release 2016, 230, 79–87.
  47. Abad, I.; Conesa, C.; Sánchez, L. Development of Encapsulation Strategies and Composite Edible Films to Maintain Lactoferrin Bioactivity: A Review. Materials 2021, 14, 7358.
  48. Su, J.; Cai, Y.; Zhi, Z.; Guo, Q.; Mao, L.; Gao, Y.; Yuan, F.; Van der Meeren, P. Assembly of propylene glycol alginate/β-lactoglobulin composite hydrogels induced by ethanol for co-delivery of probiotics and curcumin. Carbohydr. Polym. 2020, 254, 117446.
  49. Dafe, A.; Etemadi, H.; Dilmaghani, A.; Mahdavinia, G.R. Investigation of pectin/starch hydrogel as a carrier for oral delivery of probiotic bacteria. Int. J. Biol. Macromol. 2017, 97, 536–543.
  50. El-Husseiny, H.M.; Mady, E.A.; Hamabe, L.; Abugomaa, A.; Shimada, K.; Yoshida, T.; Tanaka, T.; Yokoi, A.; Elbadawy, M.; Tanaka, R. Smart/stimuli-responsive hydrogels: Cutting-edge platforms for tissue engineering and other biomedical applications. Mater. Today Bio 2021, 13, 100186.
  51. Sharpe, L.A.; Daily, A.M.; Horava, S.D.; A Peppas, N. Therapeutic applications of hydrogels in oral drug delivery. Expert Opin. Drug Deliv. 2014, 11, 901–915.
  52. Qi, X.; Simsek, S.; Chen, B.; Rao, J. Alginate-based double-network hydrogel improves the viability of encapsulated probiotics during simulated sequential gastrointestinal digestion: Effect of biopolymer type and concentrations. Int. J. Biol. Macromol. 2020, 165, 1675–1685.
  53. Patarroyo, J.L.; Florez-Rojas, J.S.; Pradilla, D.; Valderrama-Rincón, J.D.; Cruz, J.C.; Reyes, L.H. Formulation and Characterization of Gelatin-Based Hydrogels for the Encapsulation of Kluyveromyces lactis—Applications in Packed-Bed Reactors and Probiotics Delivery in Humans. Polymers 2020, 12, 1287.
  54. Ghibaudo, F.; Gerbino, E.; Orto, V.C.D.; Gomez-Zavaglia, A. Pectin-iron capsules: Novel system to stabilise and deliver lactic acid bacteria. J. Funct. Foods 2017, 39, 299–305.
  55. Falco, C.Y.; Falkman, P.; Risbo, J.; Cárdenas, M.; Medronho, B. Chitosan-dextran sulfate hydrogels as a potential carrier for probiotics. Carbohydr. Polym. 2017, 172, 175–183.
  56. Patarroyo, J.; Fonseca, E.; Cifuentes, J.; Salcedo, F.; Cruz, J.; Reyes, L. Gelatin-Graphene Oxide Nanocomposite Hydrogels for Kluyveromyces lactis Encapsulation: Potential Applications in Probiotics and Bioreactor Packings. Biomolecules 2021, 11, 922.
  57. Badeau, B.A.; Deforest, C.A. Programming Stimuli-Responsive Behavior into Biomaterials. Annu. Rev. Biomed. Eng. 2019, 21, 241–265.
  58. Dutta, S.; Samanta, P.; Dhara, D. Temperature, pH and redox responsive cellulose based hydrogels for protein delivery. Int. J. Biol. Macromol. 2016, 87, 92–100.
  59. Noirbent, G.; Dumur, F. Photoinitiators of polymerization with reduced environmental impact: Nature as an unlimited and renewable source of dyes. Eur. Polym. J. 2020, 142, 110109.
  60. Ahmed, E.M. Hydrogel: Preparation, characterization, and applications: A review. J. Adv. Res. 2015, 6, 105–121.
  61. Samal, S.K.; Dash, M.; Dubruel, P.; Van Vlierberghe, S. Smart polymer hydrogels: Properties, synthesis and applications. In Smart Polymers and their Applications, 1st ed.; Aguilar, M.R., Román, J.S., Eds.; Woodhead Publishing Limited: Cambridge, UK, 2014; pp. 237–270. ISBN 978-085-709-702-6.
  62. Liu, L.; Yao, W.; Rao, Y.; Lu, X.; Gao, J. pH-Responsive carriers for oral drug delivery: Challenges and opportunities of current platforms. Drug Deliv. 2017, 24, 569–581.
  63. Peppas, N.A.; Huang, Y. Nanoscale technology of mucoadhesive interactions. Adv. Drug Deliv. Rev. 2004, 56, 1675–1687.
  64. Tang, Y.; Heaysman, C.L.; Willis, S.; Lewis, A.L. Physical hydrogels with self-assembled nanostructures as drug delivery systems. Expert Opin. Drug Deliv. 2011, 8, 1141–1159.
  65. Sultana, K.; Godward, G.; Reynolds, N.; Arumugaswamy, R.; Peiris, P.; Kailasapathy, K. Encapsulation of probiotic bacteria with alginate–starch and evaluation of survival in simulated gastrointestinal conditions and in yoghurt. Int. J. Food Microbiol. 2000, 62, 47–55.
  66. Holkem, A.T.; Favaro-Trindade, C.S. Potential of solid lipid microparticles covered by the protein-polysaccharide complex for protection of probiotics and proanthocyanidin-rich cinnamon extract. Food Res. Int. 2020, 136, 109520.
  67. González-Ferrero, C.; Irache, J.; González-Navarro, C. Soybean protein-based microparticles for oral delivery of probiotics with improved stability during storage and gut resistance. Food Chem. 2018, 239, 879–888.
  68. Harshitha, K.; Kulkarni, P.; Vaghela, R.; Varma, V.K.; Deshpande, D.; Hani, U. Probiotic and Prebiotic-probiotic PEC Microparticles for Sustaining and Enhancing Intestinal Probiotic Growth. Curr. Drug Deliv. 2015, 12, 299–307.
  69. Haffner, F.B.; Girardon, M.; Fontanay, S.; Canilho, N.; Duval, R.E.; Mierzwa, M.; Etienne, M.; Diab, R.; Pasc, A. Core–shell microparticles encapsulating probiotics. J. Mater. Chem. B 2016, 4, 7929–7935.
  70. Di Natale, C.; Lagreca, E.; Panzetta, V.; Gallo, M.; Passannanti, F.; Vitale, M.; Fusco, S.; Vecchione, R.; Nigro, R.; Netti, P. Morphological and Rheological Guided Design for the Microencapsulation Process of Lactobacillus paracasei CBA L74 in Calcium Alginate Microspheres. Front. Bioeng. Biotechnol. 2021, 9, 660691.
  71. Yao, M.; Xie, J.; Du, H.; McClements, D.J.; Xiao, H.; Li, L. Progress in microencapsulation of probiotics: A review. Compr. Rev. Food Sci. Food Saf. 2020, 19, 857–874.
  72. Anselmo, A.C.; McHugh, K.J.; Webster, J.; Langer, R.; Jaklenec, A. Layer-by-Layer Encapsulation of Probiotics for Delivery to the Microbiome. Adv. Mater. 2016, 28, 9486–9490.
  73. Fakhrullin, R.F.; Lvov, Y.M. “Face-Lifting” and “Make-Up” for Microorganisms: Layer-by-Layer Polyelectrolyte Nanocoating. ACS Nano 2012, 6, 4557–4564.
  74. Tripathi, P.; Beaussart, A.; Alsteens, D.; Dupres, V.; Claes, I.; von Ossowski, I.E.; De Vos, W.M.; Palva, A.; Lebeer, S.; Vanderleyden, J.; et al. Adhesion and Nanomechanics of Pili from the Probiotic Lactobacillus rhamnosus GG. ACS Nano 2013, 7, 3685–3697.
  75. Pothakamury, U.R.; Barbosa-Cánovas, G.V. Fundamental aspects of controlled release in foods. Trends Food Sci. Technol. 1995, 6, 397–406.
  76. Chen, M.-J.; Chen, K.-N. Applications of Probiotic Encapsulation in Dairy Products. In Encapsulation and Controlled Release Technologies in Food Systems; Blackwell Publishing: Ames, IA, USA, 2007; pp. 83–112.
  77. Kailasapathy, K. Encapsulation technologies for functional foods and nutraceutical product development. CAB Rev. Perspect. Agric. Veter. Sci. Nutr. Nat. Resour. 2009, 4.
  78. de Vos, P.; Faas, M.M.; Spasojevic, M.; Sikkema, J. Encapsulation for preservation of functionality and targeted delivery of bioactive food components. Int. Dairy J. 2010, 20, 292–302.
  79. Picot, A.; Lacroix, C. Encapsulation of bifidobacteria in whey protein-based microcapsules and survival in simulated gastrointestinal conditions and in yoghurt. Int. Dairy J. 2004, 14, 505–515.
  80. Heidebach, T.; Först, P.; Kulozik, U. Microencapsulation of probiotic cells by means of rennet-gelation of milk proteins. Food Hydrocoll. 2009, 23, 1670–1677.
  81. Heidebach, T.; Först, P.; Kulozik, U. Transglutaminase-induced caseinate gelation for the microencapsulation of probiotic cells. Int. Dairy J. 2009, 19, 77–84.
  82. Li, X.Y.; Chen, X.G.; Cha, D.S.; Park, H.J.; Liu, C. Microencapsulation of a probiotic bacteria with alginate–gelatin and its properties. J. Microencapsul. 2008, 26, 315–324.
  83. Semyonov, D.; Ramon, O.; Kaplun, Z.; Levin-Brener, L.; Gurevich, N.; Shimoni, E. Microencapsulation of Lactobacillus paracasei by spray freeze drying. Food Res. Int. 2010, 43, 193–202.
  84. Wang, Z.; Finlay, W.; Peppler, M.; Sweeney, L. Powder formation by atmospheric spray-freeze-drying. Powder Technol. 2006, 170, 45–52.
  85. Zuidam, N.J.; Shimoni, E. Overview of Microencapsulates for Use in Food Products or Processes and Methods to Make Them. In Encapsulation Technologies for Active Food Ingredients and Food Processing; Zuidam, N.J., Nedovic, V., Eds.; Springer: New York, NY, USA, 2010; Volume 1, pp. 3–29.
  86. Champagne, C.; Fustier, P. Microencapsulation for delivery of probiotics and other ingredients in functional dairy products. In Functional Dairy Products; Elsevier: Amsterdam, The Netherlands, 2007; pp. 404–426.
  87. Liu, H.; Cui, S.W.; Chen, M.; Li, Y.; Liang, R.; Xu, F.; Zhong, F. Protective approaches and mechanisms of microencapsulation to the survival of probiotic bacteria during processing, storage and gastrointestinal digestion: A review. Crit. Rev. Food Sci. Nutr. 2019, 59, 2863–2878.
  88. Zaghari, L.; Basiri, A.; Rahimi, S. Preparation and characterization of double-coated probiotic bacteria via a fluid-bed process: A case study on Lactobacillus reuteri. Int. J. Food Eng. 2020, 16.
  89. Krasaekoopt, W.; Bhandari, B.; Deeth, H. Evaluation of encapsulation techniques of probiotics for yoghurt. Int. Dairy J. 2003, 13, 3–13.
  90. Lee, Y.; Ji, Y.R.; Lee, S.; Choi, M.-J.; Cho, Y. Microencapsulation of Probiotic Lactobacillus acidophilus KBL409 by Extrusion Technology to Enhance Survival under Simulated Intestinal and Freeze-Drying Conditions. J. Microbiol. Biotechnol. 2019, 29, 721–730.
  91. Reddy, S.; He, L.; Ramakrishana, S.; Luo, H. Graphene nanomaterials for regulating stem cell fate in neurogenesis and their biocompatibility. Curr. Opin. Biomed. Eng. 2019, 10, 69–78.
  92. Shi, H.; Liu, W.; Xie, Y.; Yang, M.; Liu, C.; Zhang, F.; Wang, S.; Liang, L.; Pi, K. Synthesis of carboxymethyl chitosan-functionalized graphene nanomaterial for anticorrosive reinforcement of waterborne epoxy coating. Carbohydr. Polym. 2020, 252, 117249.
  93. Gholamali, I.; Yadollahi, M. Bio-nanocomposite Polymer Hydrogels Containing Nanoparticles for Drug Delivery: A Review. Regen. Eng. Transl. Med. 2021, 7, 129–146.
  94. Lattuada, E.; Leo, M.; Caprara, D.; Salvatori, L.; Stoppacciaro, A.; Sciortino, F.; Filetici, P. DNA-GEL, Novel Nanomaterial for Biomedical Applications and Delivery of Bioactive Molecules. Front. Pharmacol. 2020, 11.
  95. Rabiee, N.; Bagherzadeh, M.; Ghadiri, A.M.; Fatahi, Y.; Baheiraei, N.; Safarkhani, M.; Aldhaher, A.; Dinarvand, R. Bio-multifunctional noncovalent porphyrin functionalized carbon-based nanocomposite. Sci. Rep. 2021, 11, 6604.
  96. Thangrongthong, S.; Puttarat, N.; Ladda, B.; Itthisoponkul, T.; Pinket, W.; Kasemwong, K.; Taweechotipatr, M. Microencapsulation of probiotic Lactobacillus brevis ST-69 producing GABA using alginate supplemented with nanocrystalline starch. Food Sci. Biotechnol. 2020, 29, 1475–1482.
  97. Yuan, L.; Wei, H.; Yang, X.-Y.; Geng, W.; Peterson, B.W.; van der Mei, H.C.; Busscher, H.J. Escherichia coli Colonization of Intestinal Epithelial Layers In Vitro in the Presence of Encapsulated Bifidobacterium breve for Its Protection against Gastrointestinal Fluids and Antibiotics. ACS Appl. Mater. Interfaces 2021, 13, 15973–15982.
  98. Duarte, I.F.B.; Mergulhão, N.L.O.N.; Silva, V.D.C.; de Bulhões, L.C.G.; Júnior, I.D.B.; Silva, A.C. Natural Probiotics and Nanomaterials: A New Functional Food. In Prebiotics and Probiotics—From Food to Health; IntechOpen: London, UK, 2021.
  99. Yu, H.; Park, J.-Y.; Kwon, C.W.; Hong, S.-C.; Park, K.-M.; Chang, P.-S. An Overview of Nanotechnology in Food Science: Preparative Methods, Practical Applications, and Safety. J. Chem. 2018, 2018, 5427978.
  100. Razavi, S.; Janfaza, S.; Tasnim, N.; Gibson, D.L.; Hoorfar, M. Nanomaterial-based encapsulation for controlled gastrointestinal delivery of viable probiotic bacteria. Nanoscale Adv. 2021, 3, 2699–2709.
  101. Mettu, S.; Hathi, Z.; Athukoralalage, S.; Priya, A.; Lam, T.N.; Ong, K.L.; Choudhury, N.R.; Dutta, N.K.; Curvello, R.; Garnier, G.; et al. Perspective on Constructing Cellulose-Hydrogel-Based Gut-Like Bioreactors for Growth and Delivery of Multiple-Strain Probiotic Bacteria. J. Agric. Food Chem. 2021, 69, 4946–4959.
  102. Hasan, N.; Rahman, L.; Kim, S.-H.; Cao, J.; Arjuna, A.; Lallo, S.; Jhun, B.H.; Yoo, J.-W. Recent advances of nanocellulose in drug delivery systems. J. Pharm. Investig. 2020, 50, 553–572.
  103. Park, M.; Lee, D.; Hyun, J. Nanocellulose-alginate hydrogel for cell encapsulation. Carbohydr. Polym. 2015, 116, 223–228.
  104. Huq, T.; Fraschini, C.; Khan, A.; Riedl, B.; Bouchard, J.; Lacroix, M. Alginate based nanocomposite for microencapsulation of probiotic: Effect of cellulose nanocrystal (CNC) and lecithin. Carbohydr. Polym. 2017, 168, 61–69.
  105. Liu, Y.; Lyu, Y.; Hu, Y.; An, J.; Chen, R.; Chen, M.; Du, J.; Hou, C. Novel Graphene Oxide Nanohybrid Doped Methacrylic Acid Hydrogels for Enhanced Swelling Capability and Cationic Adsorbability. Polymers 2021, 13, 1112.
  106. Phan, L.; Vo, T.; Hoang, T.; Cho, S. Graphene Integrated Hydrogels Based Biomaterials in Photothermal Biomedicine. Nanomaterials 2021, 11, 906.
  107. Wang, X.; Guo, W.; Li, L.; Yu, F.; Li, J.; Liu, L.; Fang, B.; Xia, L. Photothermally triggered biomimetic drug delivery of Teriparatide via reduced graphene oxide loaded chitosan hydrogel for osteoporotic bone regeneration. Chem. Eng. J. 2020, 413, 127413.
  108. Lu, Y.-J.; Lan, Y.-H.; Chuang, C.-C.; Lu, W.-T.; Chan, L.-Y.; Hsu, P.-W.; Chen, J.-P. Injectable Thermo-Sensitive Chitosan Hydrogel Containing CPT-11-Loaded EGFR-Targeted Graphene Oxide and SLP2 shRNA for Localized Drug/Gene Delivery in Glioblastoma Therapy. Int. J. Mol. Sci. 2020, 21, 7111.
  109. Kim, J.; Hlaing, S.P.; Lee, J.; Saparbayeva, A.; Kim, S.; Hwang, D.S.; Lee, E.H.; Yoon, I.-S.; Yun, H.; Kim, M.-S.; et al. Exfoliated bentonite/alginate nanocomposite hydrogel enhances intestinal delivery of probiotics by resistance to gastric pH and on-demand disintegration. Carbohydr. Polym. 2021, 272, 118462.
  110. Batista, D.P.C.; de Oliveira, I.N.; Ribeiro, A.R.B.; Fonseca, E.J.S.; Santos-Magalhães, N.S.; de Sena-Filho, J.G.; Teodoro, A.V.; Grillo, L.A.M.; de Almeida, R.S.; Dornelas, C.B. Encapsulation and release of Beauveria bassiana from alginate–bentonite nanocomposite. RSC Adv. 2017, 7, 26468–26477.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 856
Revisions: 2 times (View History)
Update Date: 17 Mar 2022
1000/1000
ScholarVision Creations