Next Article in Journal
Simulation Study and Optimization Strategies for Vacuum Infusion of GFRP Hoses Based on Resin Time-Viscosity Variables
Next Article in Special Issue
A Novel Method to Characterize the Damping Capacity of EPDM/CIIR Blends Using Vibrating Rubber Balls
Previous Article in Journal
Preparation of a Molecularly Imprinted Polymer on Polyethylene Terephthalate Platform Using Reversible Addition-Fragmentation Chain Transfer Polymerization for Tartrazine Analysis via Smartphone
Previous Article in Special Issue
A Systematic Investigation of the Kinetic Models Applied to the Transport Behaviors of Aromatic Solvents in Unfilled Hydrogenated Nitrile Rubber/Ethylene Propylene Diene Monomer Composites
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chitosan-2D Nanomaterial-Based Scaffolds for Biomedical Applications

by
Atanu Naskar
,
Sreenivasulu Kilari
and
Sanjay Misra
*
Vascular and Interventional Radiology Translational Laboratory, Division of Vascular and Interventional Radiology, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
*
Author to whom correspondence should be addressed.
Polymers 2024, 16(10), 1327; https://doi.org/10.3390/polym16101327
Submission received: 11 April 2024 / Revised: 3 May 2024 / Accepted: 7 May 2024 / Published: 8 May 2024

Abstract

:
Chitosan (CS) and two-dimensional nanomaterial (2D nanomaterials)-based scaffolds have received widespread attention in recent times in biomedical applications due to their excellent synergistic potential. CS has garnered much attention as a biomedical scaffold material either alone or in combination with some other material due to its favorable physiochemical properties. The emerging 2D nanomaterials, such as black phosphorus (BP), molybdenum disulfide (MoS2), etc., have taken huge steps towards varying biomedical applications. However, the implementation of a CS-2D nanomaterial-based scaffold for clinical applications remains challenging for different reasons such as toxicity, stability, etc. Here, we reviewed different types of CS scaffold materials and discussed their advantages in biomedical applications. In addition, a different CS nanostructure, instead of a scaffold, has been described. After that, the importance of 2D nanomaterials has been elaborated on in terms of physiochemical properties. In the next section, the biomedical applications of CS with different 2D nanomaterial scaffolds have been highlighted. Finally, we highlighted the existing challenges and future perspectives of using CS-2D nanomaterial scaffolds for biomedical applications. We hope that this review will encourage a more synergistic biomedical application of the CS-2D nanomaterial scaffolds and their utilization clinical applications.

1. Introduction

Chitosan (CS) is a cationic natural linear polysaccharide of β-(1→4)-linked D-glucosamine and N-acetyl-D-glucosamine derived from the alkaline hydrolysis of chitin. CS is the second most naturally abundant biopolymer after cellulose and is one of the most vastly used natural materials in the medicine, agriculture, and food processing industries due its biocompatibility, biodegradability, and physiochemical properties [1,2]. Therefore, CS has been classified as a generally recognized as safe (GRAS) compound by the Food and Drug Administration (FDA) [3]. This is why numerous researchers have extensively followed CS or CS-based scaffolds for biomedical applications, which can be confirmed by the abundant published work based on CS [4,5,6].
Because of the structural similarity of CS to glycosaminoglycans, as one of the components in extracellular matrix (ECM) [7], CS scaffolds are used in biomedical research and the development of therapeutics. Due to its structural advantages, CS scaffolds are also utilized for various vascular regeneration applications [8,9]. Although CS preparation is cost-effective, poor solubility and porosity are limiting factors for their usage in a wide range of biomedical applications [10,11]. However, CS can combine with other polymers, metal, metal oxide, and 2D nanomaterials to augment those properties and lead to scaffold formation [12]. In this respect, 2D nanomaterials, such as graphene, black phosphorus, and MoS2, when combined with CS scaffold have shown synergistic properties [13,14,15]. Further, due to the large surface area of 2D nanomaterials, the chance of cell interaction with 2D materials is higher. However, the nanomaterials have their own toxicity issue which can be overcome by CS capping [16]. Hence, not only can 2D nanomaterials be utilized with CS, but it can be synergistically applied for nanomaterial applications.
Here, we discuss the advantages of CS biomaterial scaffolds and the functionalized synergistic applications of the CS scaffolds in combination with 2D materials. Finally, the prospects and challenges of CS-based 2D materials scaffold in clinical applications are discussed.

2. Advantages of Chitosan for Biomedical Applications

The biomedical applications of a biomaterial are dependent on their favorable physiochemical properties such as porosity, solubility, biodegradability, biocompatibility, etc. In this section, we discuss the chitosan properties that make CS an excellent scaffold biomaterial in biomedical applications.

2.1. Biocompatibility

The biocompatibility of a material can be determined by its compatibility with the biological system with minimal or no adverse effects, including immunogenicity in vivo. The CS materials are relatively non-toxic excellent biocompatible materials which are derivatives of chitin. A highly conserved extracellular matrix appeared across the animals from invertebrates to higher mammals. Moreover, upon degradation, CS releases its constitutive ingredients, D-glucosamine and N-acetyl-D-glucosamine, which are natural components that can be utilized for tissue regeneration and in the healing process. [10]. Although, CS has been extensively investigated as a nanobiomaterial due to its non-toxicity, biodegradability, and biocompatibility and granted FDA Generally Recognized As Safe (GRAS) status (GRN n° 73, 170, 397 and 443), some studies showed toxic effects after using CS in the cell lines of zebrafish [17,18].

2.2. Porosity

Porosity is one of the important features for any scaffold to determine its biocompatibility. Cell adhesion to the scaffold material is dependent on the pore size of the scaffold, which, if too small, results limited cell permeability, whereas too large pores result in a limited surface area and reduced ligand density for the cell to bind. Therefore, it is important to maintain optimal pore size for cell adhesion and growth [19]. Further, different pore size scaffolds are required for certain types of applications in biomedical research. For example, a scaffold with greater than 20–100 µm is a good fit for cell infiltration [20], while more than 100 µm is well recommended for neovascularization studies [21]. Similarly, a scaffold up to 300 µm pore size is ideal for endochondral ossification, whereas pores above 300 µm in scaffolds showed in osteogenesis studies [22]. Therefore, it is necessary to select the appropriate pore size for specific applications.
The pore size of the scaffold can be controlled by regulating the temperature and water content in the scaffolds. For example, the lower the temperature, the greater the water content and the smaller the pore size. Further, the thermal-induced phase separation (TIPS) method is used to synthesize different structures with different pores [23].
Further, the pore size of the CS scaffold also depends on various parameters, such as crosslinkers, freezing temperature, the concentration of polymer, and the addition of other compounds such as drug, nanoparticles, etc. For example, Shavandi et al. [24] demonstrated that the CS scaffold with hydroxyapatite and beta-tricalcium phosphate, prepared at −80 °C and −20 °C vary in their pore size. The CS scaffolds that were prepared at −80 °C, showed elongated pores with an irregularity in shape, whereas the scaffold of −20 °C showed highly layered pores with more irregularities. Similarly, the addition of hydroxyapatite nanoparticles to CS-silk fibroin (SF) scaffold has a reduced porosity compared to the CS-silk fibroin (SF) scaffold [25].

2.3. Molecular Weight

CS is a polysaccharide of D-glucosamine and N-acetyl-D-glucosamine units, and the molecular weight varies with the number of D-glucosamine and N-acetyl-D-glucosamine units. The physiochemical and biological properties, including solubility and the viscosity of CS changes with increases in the molecular weight [26]. Depending on the source and preparation process, the molecular weight of CS ranges from ~300 to 1000 kDa [27]. CS with a higher molecular weight becomes more viscous, less soluble, and consequently less permeable, which is not desirable for various biomedical applications. Hence, low molecular weight CS is commonly used in CS scaffold preparation for biomedical applications, due to its excellent solubility and stability [28].

2.4. Water Retention Ability

The water retention ability of any scaffold material can be described as the ability to swell and hold certain volumes of water after being placed in a liquid medium [29]. The scaffolds materials water absorption resulted in increased pore size and swelling. The content of aminosugars in CS determine its swelling ability [30]. The cationic CS materials have an electrostatic interaction with anionic polymers, resulting in polymeric complexation and decreased swelling. In this context, the inclusion of silicon dioxide and zirconia nano particles significantly reduced the swelling behavior of the CS scaffold [31]. The addition of bioactive glass ceramic nanoparticles (nBGC) to the CS–gelatin scaffold have shown a significant reduction in the swelling ability of the CS scaffold [32]. Together, these reports indicate that the swelling ability of CS scaffolds can be modified as needed.

2.5. Biodegradability

The process of degradation and he longevity of a scaffold material in the biological system are key factors in selecting the biomaterial therapeutics [33]. The process of degradation can be hydrolysis and or enzymatic, and the resulting degraded products should be non-immunogenic and non-toxic and are incorporated into metabolic pathways or excreted [34]. As previously mentioned, the CS materials are derived from the chitin one of the extracellular components in the biological systems, which is highly conserved across the species and, therefore, CS materials are non-toxic and minimal immunogenic. The CS is hydrolyzed to acetylated and amino sugars which may be re-cycled or excreted [35]. The rate of CS degradation also depends on degree of deacetylation and hydrolysis by lysozyme [36]. However, due to its high degradation rates, the usage of CS scaffolds in vivo for long term application is limited. The addition of nanoparticles or other polymers into the CS scaffold seemed to affect the degradation rate. For example, Saravanan et al. [37] showed that the addition of nano-hydroxyapatite (nHAp) into the CS scaffold seemed to accelerate the derogation rate, while the opposite result can be seen after the addition of nano silver (nAg) in the CS matrix. Similarly, the addition of bioactive glass ceramic nanoparticles (nBGC) to the CS–gelatin scaffold considerably reduced their degradation rate [32].

3. Types of Chitosan Scaffold

CS materials, such as hydrogel, sponges nanofiber membrane, etc., have been used in various biomedical applications, including wound healing and tissue engineering. In this section, we discuss the different types of CS scaffold used in tissue engineering.

3.1. Hydrogel Scaffold

Hydrogels are cross-linked and a polymeric network of hydrophilic units and the gelation can be initiated via physical and or chemical reactions [38]. Hydrogel-based scaffolds are supporting materials that have the potential to mimic the extracellular matrix, which provides cell–cell communications with the sustained release of water and other biomolecules for tissue regeneration and the healing process [39]. The hydrophilic structure of the hydrogel scaffold gives it the capability to maintain considerable amounts of water or other biological fluids, which helps in nutrient diffusion. It is worthy to note that a proper hydrogel should be able to regenerate specific tissues, while achieving the minimum requirements for vascularization, cell growth, proliferation, and concurrent degradation during the healing process, along with its biocompatible and non-toxic properties [40]. Superior physical and mechanical stability, high biodegradability, and high durability are some of the other characteristics of a proper hydrogel scaffold. The advantages of a CS hydrogel scaffold are its excellent inherent biodegradability, biocompatibility, and hydrophilic surface. However, its extreme viscosity, combined with its mechanical weakness, are some of the limitations which are yet to be resolved [40].
In recent years, the urgency to develop smart injectable hydrogels has increased due to its minimal invasive approach. Smart injectable hydrogels are liquid at room temperature but form a gel when injected into a fractured location, which has the potential for scar size reduction, less post-operative pain, the rapid recovery of patients, and obvious cost-effectiveness [41]. Naturally occurring polysaccharides are especially relevant to hydrogel preparation as they mimics many features of the extracellular matrix. Chitosan, a naturally occurring polysaccharide and a pH-responsive polymer is significant in this scenario [42]. The anionic nature of most human tissues can perfectly adhere to the cationic character of chitosan and the subsequent adherence of CS hydrogels to tissue sites [43]. Additionally, the polycationic nature of chitosan enabled the preparation of cross-linked hydrogels without any use of cross-linking agents, which might be toxic.

3.2. Sponges

The primary advantage of chitosan sponges is that its micro-porous structure enables it to absorb high amounts of fluids. In some cases, this amount of absorbed fluid is 20 times more than its dry weight, without compromising its flexibility and texture [44]. With respect to wound healing applications, CS sponges prevent contamination in wound and dehydration due to its porous structure [45]. For example, CS/tricalcium phosphate [46], CS/collagen sponges [47] are used as scaffolds in bone regeneration. Du et al. [48] showed the excellent wound healing potential of micro-channeled alkylated chitosan sponge, which are able to guide in situ tissue regeneration for noncompressible hemorrhages. In another example, Wu et al. [49] prepared ampicillin-grafted chitosan sponges as an antibacterial material against Staphylococcus aureus, Candida albicans, and Escherichia coli and showed its potential as a wound dressing material. In a similar experiment, Al-Mofty et al. [50] showed the antibacterial and hemostatic activity of PVA/chitosan sponges loaded with hydroxyapatite and ciprofloxacin. It is worthy to note that CS sponges also have some disadvantages such as poor mechanical properties and rapid degradation prepared in acidic conditions, which hindered its growth in application processes [51].

3.3. Fiber Scaffolds

Fiber scaffolds were generally utilized to disperse the bioactive agents within the fibrous matrix. The bioactive agents either can also be adsorbed on the surface of the fibers or blended into the electrospinning polymer solution to produce fiber scaffolds [52]. The release of bioactive molecules from the fiber scaffold is straightforward where the fiber scaffolds usually burst release due to the dissolution of bioactive agents [53]. However, despite the simplicity of the process, the release rate of the bioactive agents directly depends on the degradation rate of the polymer matrix. Moreover, the solvents in the electrospinning solution utilized to disperse the bioactive agents can also hinder the activity of the molecules. Fiber-based chitosan scaffolds were also utilized to resolve the high viscosity problem of chitosan [54]. In this case, a nanofiber diameter of 140 nm can be achieved with chitosan that is hydrolyzed for 48 h. Additionally, electrospinning conditions and the solvent concentration also affected the fiber diameter.

3.4. Microspheres Scaffolds

CS microsphere scaffolds have been used for controlled drug release and increased bioavailability [55]. The preparation of the CS microsphere was enabled after reacting chitosan with controlled amounts of multivalent anion, which, in turn, resulted in cross-linking between the chitosan molecules [55]. Precipitation, cross-linking with anions, modified emulsification, thermal cross-linking, etc., are some the techniques utilized to prepare the CS microsphere [56]. The nature of drug molecule which needs to be incorporated into CS microsphere decides the selection of preparation process.
Hu et al. [57] utilized a combination of biodegradable poly-(lactic acid-co-trimethylene carbonate) and chitosan microspheres for bone tissue engineering. The porosity, pore size, and mechanical properties of these CS microsphere scaffolds can be controlled through the preparation methods and parameters. Moreover, this CS microsphere-based scaffolds possessed shape-memory effects, i.e., it can recover to its initial shape when heated to 37 °C within 300 s. The scaffold has the potential for bone regeneration applications. In another example, Budhiraja et al. [58] exploited the formulation of mupirocin-loaded chitosan microspheres embedded in Piper betle extract containing a collagen scaffold for the purpose of wound healing activity. Similarly, Fan et al. [59] showed the effectiveness of covalent and injectable chitosan-chondroitin sulfate hydrogels embedded with chitosan microspheres as an injectable drug and cell delivery system in cartilage tissue engineering. The porosity, pore size, and mechanical properties of these CS microsphere scaffolds can be tuned through the preparation methods. Moreover, these CS microsphere scaffolds regain their shape upon heating to 37 °C within 300 s. Such a shape memory effect is favorable for spatial implantation applications.

4. Types of Chitosan Nanostructures

Chitosan is used as hydrogel scaffolding material [8]. However, the poor mechanical properties of CS hydrogel or CS films have hindered their application in scaffolds, despite their having excellent biomedical properties [60]. Hence, efforts have been made to incorporate different forms of CS, such as CS nanoparticles, CS nanosphere, CS nanosheets.

4.1. Chitosan Nanoparticles (CS NPs)

Chitosan NPs have been successfully utilized because of their mucoadhesive capacity, enhanced bioavailability, non-toxic, and biocompatibility, etc. [61]. Additionally, CS NPs have a large surface-to-volume ratio which, in turn, enables it to provide a great binding capacity for biological macromolecules in various biomedical applications [62]. Moreover, the growth factors and signaling molecules can be easily loaded into the scaffolding materials through the incorporation of CS NPs [63].
Further, the addition of CS NPs to the scaffolding materials have showed an enhanced biocompatibility and accelerated hydrolytic degradation for potential in tissue engineering applications as listed in Table 1.

4.2. Chitosan Nanospheres (CS NSs)

Chitosan nanospheres (CS NSs) is another nanomaterial which is used for drug delivery application mainly because of its high surface area, excellent porosity, effective chemical stability, and stable geometric structure [71,72,73].
There are various examples of CS NSs as a nanocomposite material or as a scaffold material in biomedical applications. For example, Yang et al. [71] synthesized an injectable carboxymethyl chitosan/nanosphere-based hydrogel for drug release and lubrication in ameliorating from arthritis. The average size of the NP utilized in this hydrogel was in the range of 47.7 nm to 52.1 nm. Moreover, CS NSs are also used for the delivery of the anticancer drug 5-fluorouracil [72]. The mean diameter of CS NSs was ~200 nm. However, despite its potential for excellent biomedical applications, its particle size and morphology are not fully controllable, which limits its potential in biomedical applications.

4.3. Chitosan Nanosheets (CS NTs)

Chitosan nanosheets are another nanostructure which have shown excellent potential for biomedical applications. There are limited studies using CS NTs in wound healing activities with lower inflammatory cells infiltration, along with new epithelium thickness [74].

5. The Advantages of 2D Nanomaterials for Biomedical Applications

Two-dimensional nanomaterials such as graphene, black phosphorus, metal carbides, and nitrides (MXenes), etc., have shown excellent potential as biomaterials in various biomedical applications [75,76]. Recent studies on the utilization of 2D nanomaterials in biomedical research can be attributed to their excellent physiochemical properties, [77,78] which makes them attractive candidates for biosensing, bioimaging, drug delivery, and regenerative medicine. Another advantage of various 2D nanomaterials is that they can be utilized with CS or some other polymer material for synergistic biomedical applications, i.e., the 2D material-based CS nanocomposite would show much improved biomedical properties than individual samples. There are several advantages of 2D nano materials such as:
  • High surface-to-volume ratio and tunable interfacial chemistry are some of the most important characteristics of 2D nanomaterials, which are generally required for biomedical applications.
  • 2D nanomaterials showed a rippling or wrinkling effect in the case of out-of-plane bending or folding, which allows cells to strongly attach and spread freely over the underlying substrate [79]. This process of nanocomposite formation helped in biomedical applications as strong cell attachment to the substrate is one of the desired criteria for biomedical applications.
  • Mechanical strain gradients allow electrical polarization, which can regenerate electrically active tissues such as bone, neurons, and cardiac tissue [80].
  • Two-dimensional nanomaterials can interact with cellular membrane in penetration mode as well as attachment mode [79,81]. Hydrophobic attraction drives the penetration mode interaction between the lipid layer of cellular membrane and the 2D nanomaterials, whereas the hydrophilic interaction works for the interaction in attachment mode.
  • The lateral size of the 2D nanomaterials also determine the interaction mode between the cellular membrane and 2D nanomaterials [79,81]. For example, nanomaterials with similar dimensions to plasma membrane implement attachment mode, whereas larger dimension nanomaterials utilize penetration mode.

6. Chitosan-2D Nanomaterial Scaffolds for Biomedical Applications

In recent years, 2D nanomaterials such as graphene, black phosphorus (BP), MoS2, were increasingly utilized for various biomedical applications. A combination of chitosan with 2D nanomaterials used for synergistic biomedical applications are listed in Table 2.

6.1. Chitosan-Graphene

Graphene is a derivative of graphite, a thin 2D nanomaterial with high tensile strength and electrical conductivity. Although there are promising results, the biocompatibility of graphene is under debate. The addition of graphene 2D material to CS scaffold has shown synergistic effects, tissue regeneration, and cardiac repair [13,84]. Hermenean et al. [82] exploited CS-graphene oxide (GO) 3D scaffolds for bone tissue regeneration in critical-size mouse calvarial defects. When combined with GO, CS scaffolds showed the synergistic increment of alkaline phosphatase activity both in vitro and in vivo experiments, along with an increased expression of bone morphogenetic protein (BMP) and Runx-2, and showed its bone tissue regeneration ability. In a similar approach, Dinescu et al. [83] used the GO with a CS-based 3D scaffold, which showed the formation of ordered morphologies and a higher total porosity, combined with a greater surface availability for cell attachment.
CS-based graphene nanocomposites were successfully experimented on as antibacterial scaffolds in hemorrhage control and wound-healing applications [13]. The nanobiocomposite scaffolds were fabricated by the incorporation of graphene-silver-polycationic peptide (GAP) nanocomposite into CS (Figure 1). One of the CS-GAP scaffolds showed excellent antibacterial activity against E. coli and S. aureus, along with excellent porosity, fluid absorption, and mechanical strength. Saravanan et al. [84] also showed the importance of GO and Au nanosheet-based CS scaffolds for the improvement of ventricular contractility and function into Infarcted Heart. The particle size of Au NPs was ~8 nm utilized in the nanocomposite. In another experiment, Sivashankari et al. [85] exploited agarose/CS/graphene composite scaffolds for its potential application in bone and osteochondral tissue engineering. The functional recovery of injured spinal cord in rats was successfully achieved through using GO composite-based CS scaffolds [86]. The various examples of cartilage tissue engineering [87] and cardiac tissue engineering [88] was also achieved through using synergistic GO-based CS scaffolds.

6.2. Chitosan-Black Phosphorus

Black phosphorus (BP) has risen up with huge potential due to its favorable physiochemical properties [97,98,99]. Unlike graphene, BP has its own biocompatible and biodegradable properties. It showed excellent photo active properties, as well as photothermal anticancer or antibacterial applications [99,100]. It is worthy to note that the degradation products of BP are safe PO43− and are capable of enhancing the osteogenesis process. However, the instability of BP hindered its usage in biomedical applications [77,99]. Zhao et al. [14] used chitosan/hydroxyapatite/black phosphorus (CS/HC/HA/BP) hybrid photothermal scaffold (Figure 2) to solve bone tumor-related complications. CS not only stabilized the BP-based scaffold but also synergistically act for simultaneous antitumor/antibacterial properties under the photothermal stimulation of <50 °C. In another work, BP nanosheets were combined with platelet-rich plasma (PRP)-chitosan thermo responsive hydrogel for the preparation of a therapeutic platform for the phototherapy treatment of rheumatoid arthritis [89]. This injectable CS thermo-responsive hydrogel was able to control the degradation products of the BP nanosheets, which were simultaneously used as raw materials for osteanagenesis. Moreover, this hydrogel could protect articular cartilage by reducing the friction on the surrounding tissue. Similarly, He et al. [90] prepared layer-by-layer assembled BP/CS composite coating for a multi-functional bone scaffold for osteosarcoma management and bone repair. Th size of BP was ~200 nm. The biocompatible polyetheretherketone (PEEK) scaffold provided similar mechanical properties and architecture compared to that of the natural bone.

6.3. Chitosan-MoS2

Among the 2D-layered transition metal dichalcogenides, molybdenum disulfide (MoS2) in particular, has shown promising results for applications in environmental and biomedical fields [101,102]. In this regard, Mutalik et al. [103] showed the potential of the phase dependent. The MoS2-based hydrogels showed excellent mechanical properties, along with intrinsic NIR region absorption for useful photothermal conversion efficiency [104]. However, the negative charge surface of MoS2 limits its interaction with cells [91]. Therefore, coating of MoS2 with a cationic biocompatible agent such as CS seemed to be an excellent strategy for more cellular interaction with synergistic nanocomposites. Additionally, the poor hydrophilic property of MoS2 can be modified with CS coating. Therefore, the combination of CS with MoS2 seems to have lots of synergistic potential for various biomedical applications.
Yan et al. [15] used a quaternized chitosan (QCS)-coated MoS2/poly(vinyl alcohol) hydrogel (Figure 3) for NIR-responsive photothermal antibacterial activity against S. aureus and E. coli. The incorporation of QCS- MoS2 seemed to increase the mechanical properties of the hydrogel. Similarly, Shen et al. [91], developed in situ grown bacterial cellulose/MoS2-chitosan nanocomposite (BC/MoS2-CS) for excellent photodynamic and photothermal antibacterial activities against E. coli and S. aureus under visible-light illumination. The cationic CS coating enabled the nanocomposite for more bacteria interaction, which eventually led to bacteria cell killing. Moreover, CS also seemed to potentiate the antibacterial activity of the nanocomposite by bacterial membrane disruption and/or permeability.
MoS2-based CS hydrogels were also utilized for colon cancer treatment [92]. In this experiment, MoS2 nanoflower was doped into CS/oxidized dextran hydrogels and then used for sequential delivery of methotrexate (MTX) and 5-Fluorouracil (5-FU). The NIR irradiation onto the nanocomposite generated hyperthermia due to the presence of MoS2, which led to the consequent release of 5-FU encapsulated. In other experiments, Xu et al. [93] and Mukheem et al. [94] showed photothermal antibacterial activity of CS-based MoS2 hydrogels.

6.4. Chitosan-MXene

Transition metal carbide (MXene) is another 2D material which has shown excellent photothermal properties and biocompatibility, which can be utilized for various biomedical applications [105]. However, it also tends to aggregate in the physiological environment, which limits its usage in biological applications. CS, in combination with MXene, has been shown to be a stable nanocomposite [95,105]. Further, CS and MXene combination shows higher porosity, which is an essential criterion for any functional hydrogel.
Dong et al. [95] prepared Ti3C2Tx MXene-loaded chitosan (MX-CS) hydrogel for photothermal synergistic activity against methicillin-resistant S. aureus. The MX-CS hydrogel not only adsorb MRSA cells via CS-MRSA interactions, but it can also kill the bacteria by NIR-irradiated photothermal hyperthermia. In another study, the porosity of CS-hyaluronate matrix hydrogel nanocomposites was controlled by the addition of 2D Ti3C2Tx MXene [11]. Due to the large porosity of the nanocomposite, a small amount of MXene (1–5 wt.%) in CS-based hydrogel was effective against E. coli, S. aureus, and Bacillus sp. bacteria. In a different application, Liu et al. [96] utilized a MXene@CS based conductive polyacrylamide hydrogel for highly stretchable and sensitive wearable skin.

7. Conclusions and Future Perspectives

Chitosan-based materials were utilized for many biomedical applications, such as antibacterial activity, wound healing, anticancer activity, and tissue engineering (bone, cartilage, cardiac, dental, skin, etc.) applications, etc. However, there are certain limitations of CS alone as a scaffolding material in biomedical applications which include: (1) lacking sufficient mechanical strength, (2) porosity, and (3) solubility. However, the current research shows that the combination of CS with other 2D nanomaterials not only overcomes these limitations but also seems to complement and elicit synergistic effects, including the mechanical strength and porosity. In short, it is valid to say that significant progress has been made for CS-based scaffolds and their biomedical applications. Two-dimensional nanomaterial-based CS scaffolds also showed great promise in clinical application. Additionally, CS-2D nanomaterials scaffolds can also be experimented for wrapping around the vessels in vascular surgery procedures, as previously performed through CorMatrix in our lab [106]. However, significant challenges, such as the ineffective delivery of growth and large scale reproducibility, still need to be overcome as the translation from the lab scale into clinical trials has been limited due to the industrial-scale production quantities and quality. Hence, a different approach for large scale production, such as cross flow filtration (CFF) [107], or some other process to enhance the large-scale production of CS should be executed. Overall, it is fair to conclude that, despite having some limitations to deal with, CS is an excellent base material, whereas other nanomaterials, such as 2D nanomaterials, can be utilized for more direction- and application-oriented research work.

Author Contributions

Conceptualization and writing, A.N., S.K. and S.M.; funding acquisition, S.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by HL98967 and DK135407 to S.M. S.K is partly supported by a Career development award, Mayo foundation.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that there are no conflicts of interest.

References

  1. Harugade, A.; Sherje, A.P.; Pethe, A. Chitosan: A review on properties, biological activities and recent progress in biomedical applications. React. Funct. Polym. 2023, 191, 105634. [Google Scholar] [CrossRef]
  2. Periayah, M.H.; Halim, A.S.; Saad, A.Z. Chitosan: A Promising Marine Polysaccharide for Biomedical Research. Pharmacogn. Rev. 2016, 10, 39–42. [Google Scholar] [CrossRef] [PubMed]
  3. Leonida, M.; Ispas-Szabo, P.; Mateescu, M.A. Self-stabilized chitosan and its complexes with carboxymethyl starch as excipients in drug delivery. Bioact. Mater. 2018, 3, 334–340. [Google Scholar] [CrossRef] [PubMed]
  4. Almajidi, Y.Q.; Ponnusankar, S.; Chaitanya, M.; Marisetti, A.L.; Hsu, C.Y.; Dhiaa, A.M.; Saadh, M.J.; Pal, Y.; Thabit, R.; Adhab, A.H.; et al. Chitosan-based nanofibrous scaffolds for biomedical and pharmaceutical applications: A comprehensive review. Int. J. Biol. Macromol. 2024, 264, 130683. [Google Scholar] [CrossRef] [PubMed]
  5. Rasool, A.; Rizwan, M.; Islam, A.; Abdullah, H.; Shafqat, S.S.; Azeem, M.K.; Rasheed, T.; Bilal, M. Chitosan-Based Smart Polymeric Hydrogels and Their Prospective Applications in Biomedicine. Starch-Starke 2024, 76, 2100150. [Google Scholar] [CrossRef]
  6. Guo, Y.B.; Qiao, D.L.; Zhao, S.M.; Liu, P.; Xie, F.W.; Zhang, B.J. Biofunctional chitosan-biopolymer composites for biomedical applications. Mater. Sci. Eng. R. 2024, 159, 100775. [Google Scholar] [CrossRef]
  7. Del Bakhshayesh, A.R.; Asadi, N.; Alihemmati, A.; Nasrabadi, H.T.; Montaseri, A.; Davaran, S.; Saghati, S.; Akbarzadeh, A.; Abedelahi, A. An overview of advanced biocompatible and biomimetic materials for creation of replacement structures in the musculoskeletal systems: Focusing on cartilage tissue engineering. J. Biol. Eng. 2019, 13, 85. [Google Scholar] [CrossRef]
  8. Wang, Q.L.; Wang, X.Y.; Feng, Y.K. Chitosan Hydrogel as Tissue Engineering Scaffolds for Vascular Regeneration Applications. Gels 2023, 9, 373. [Google Scholar] [CrossRef]
  9. Thottappillil, N.; Nair, P.D. Scaffolds in vascular regeneration: Current status. Vasc. Health Risk Manag. 2015, 11, 79–91. [Google Scholar] [CrossRef]
  10. Aranaz, I.; Alcántara, A.R.; Civera, M.C.; Arias, C.; Elorza, B.; Caballero, A.H.; Acosta, N. Chitosan: An Overview of Its Properties and Applications. Polymers 2021, 13, 3256. [Google Scholar] [CrossRef]
  11. Rozmyslowska-Wojciechowska, A.; Karwowska, E.; Gloc, M.; Wozniak, J.; Petrus, M.; Przybyszewski, B.; Wojciechowski, T.; Jastrzebska, A.M. Controlling the Porosity and Biocidal Properties of the Chitosan-Hyaluronate Matrix Hydrogel Nanocomposites by the Addition of 2D TiCT MXene. Materials 2020, 13, 4587. [Google Scholar] [CrossRef] [PubMed]
  12. Murugesan, S.; Scheibel, T. Chitosan-based nanocomposites for medical applications. J. Polym. Sci. 2021, 59, 1610–1642. [Google Scholar] [CrossRef]
  13. Choudhary, P.; Ramalingam, B.; Das, S.K. Fabrication of Chitosan-Reinforced Multifunctional Graphene Nanocomposite as Antibacterial Scaffolds for Hemorrhage Control and Wound-Healing Application. ACS Biomater. Sci. Eng. 2020, 6, 5911–5929. [Google Scholar] [CrossRef] [PubMed]
  14. Zhao, Y.; Peng, X.; Xu, X.Y.; Wu, M.Z.; Sun, F.; Xin, Q.W.; Zhang, H.B.; Zuo, L.R.; Cao, Y.L.; Xia, Y.H.; et al. Chitosan based photothermal scaffold fighting against bone tumor-related complications: Recurrence, infection, and defects. Carbohydr. Polym. 2023, 300, 120264. [Google Scholar] [CrossRef] [PubMed]
  15. Yan, P.F.; Li, M.Y.; Liu, J.; Song, L.F.; Tang, K.Y. Near-infrared responsive quaternized chitosan-coated MoS2/poly(vinyl alcohol) hydrogel with improved mechanical and rapid antibacterial properties. Eur. Polym. J. 2022, 180, 111593. [Google Scholar] [CrossRef]
  16. Rizeq, B.R.; Younes, N.N.; Rasool, K.; Nasrallah, G.K. Synthesis, Bioapplications, and Toxicity Evaluation of Chitosan-Based Nanoparticles. Int. J. Mol. Sci. 2019, 20, 5776. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, Y.B.; Zhou, J.R.; Liu, L.; Huang, C.J.; Zhou, D.Q.; Fu, L.L. Characterization and toxicology evaluation of chitosan nanoparticles on the embryonic development of zebrafish. Carbohydr. Polym. 2016, 141, 204–210. [Google Scholar] [CrossRef] [PubMed]
  18. Hu, Y.L.; Qi, W.; Han, F.; Shao, J.Z.; Gao, J.Q. Toxicity evaluation of biodegradable chitosan nanoparticles using a zebrafish embryo model. Int. J. Nanomed. 2011, 6, 3351–3359. [Google Scholar] [CrossRef]
  19. Lutzweiler, G.; Halili, A.N.; Vrana, N.E. The Overview of Porous, Bioactive Scaffolds as Instructive Biomaterials for Tissue Regeneration and Their Clinical Translation. Pharmaceutics 2020, 12, 602. [Google Scholar] [CrossRef]
  20. Ameer, J.M.; Kumar, P.R.A.; Kasoju, N. Strategies to Tune Electrospun Scaffold Porosity for Effective Cell Response in Tissue Engineering. J. Funct. Biomater. 2019, 10, 30. [Google Scholar] [CrossRef]
  21. Choi, S.W.; Zhang, Y.; MacEwan, M.R.; Xia, Y.N. Neovascularization in Biodegradable Inverse Opal Scaffolds with Uniform and Precisely Controlled Pore Sizes. Adv. Healthc. Mater. 2013, 2, 145–154. [Google Scholar] [CrossRef] [PubMed]
  22. Gupte, M.J.; Swanson, W.B.; Hu, J.; Jin, X.B.; Ma, H.Y.; Zhang, Z.P.; Liu, Z.N.; Feng, K.; Feng, G.J.; Xiao, G.Y.; et al. Pore size directs bone marrow stromal cell fate and tissue regeneration in nanofibrous macroporous scaffolds by mediating vascularization. Acta Biomater. 2018, 82, 1–11. [Google Scholar] [CrossRef] [PubMed]
  23. Conoscenti, G.; Schneider, T.; Stoelzel, K.; Pavia, F.C.; Brucato, V.; Goegele, C.; La Carrubba, V.; Schulze-Tanzil, G. PLLA scaffolds produced by thermally induced phase separation (TIPS) allow human chondrocyte growth and extracellular matrix formation dependent on pore size. Mater. Sci. Eng. C-Mater. 2017, 80, 449–459. [Google Scholar] [CrossRef] [PubMed]
  24. Shavandi, A.; Bekhit, A.E.D.A.; Ali, M.A.; Sun, Z. Bio-mimetic composite scaffold from mussel shells, squid pen and crab chitosan for bone tissue engineering. Int. J. Biol. Macromol. 2015, 80, 445–454. [Google Scholar] [CrossRef] [PubMed]
  25. Lima, P.A.L.; Resende, C.X.; Soares, G.D.D.; Anselme, K.; Almeida, L.E. Preparation, characterization and biological test of 3D-scaffolds based on chitosan, fibroin and hydroxyapatite for bone tissue engineering. Mater. Sci. Eng. C-Mater. 2013, 33, 3389–3395. [Google Scholar] [CrossRef] [PubMed]
  26. Tangsadthakun, C.; Kanokpanont, S.; Sanchavanakit, N.; Pichyangkura, R.; Banaprasert, T.; Tabata, Y.; Damrongsakkul, S. The influence of molecular weight of chitosan on the physical and biological properties of collagen/chitosan scaffolds. J. Biomater. Sci.-Polym. Ed. 2007, 18, 147–163. [Google Scholar] [CrossRef]
  27. Saravanan, S.; Leena, R.S.; Selvamurugan, N. Chitosan based biocomposite scaffolds for bone tissue engineering. Int. J. Biol. Macromol. 2016, 93, 1354–1365. [Google Scholar] [CrossRef] [PubMed]
  28. Levengood, S.K.L.; Zhang, M.Q. Chitosan-based scaffolds for bone tissue engineering. J. Mater. Chem. B 2014, 2, 3161–3184. [Google Scholar] [CrossRef] [PubMed]
  29. Wu, Z.Y.; Li, Q.; Pan, Y.K.; Yao, Y.; Tang, S.C.; Su, J.C.; Shin, J.W.; Wei, J.; Zhao, J. Nanoporosity improved water absorption, in vitro degradability, mineralization, osteoblast responses and drug release of poly(butylene succinate)-based composite scaffolds containing nanoporous magnesium silicate compared with magnesium silicate. Int. J. Nanomed. 2017, 12, 3637–3651. [Google Scholar] [CrossRef]
  30. Jiménez-Gómez, C.P.; Cecilia, J.A. Chitosan: A Natural Biopolymer with a Wide and Varied Range of Applications. Molecules 2020, 25, 3981. [Google Scholar] [CrossRef]
  31. Pattnaik, S.; Nethala, S.; Tripathi, A.; Saravanan, S.; Moorthi, A.; Selvamurugan, N. Chitosan scaffolds containing silicon dioxide and zirconia nano particles for bone tissue engineering. Int. J. Biol. Macromol. 2011, 49, 1167–1172. [Google Scholar] [CrossRef] [PubMed]
  32. Peter, M.; Binulal, N.S.; Nair, S.V.; Selvamurugan, N.; Tamura, H.; Jayakumar, R. Novel biodegradable chitosan-gelatin/nano-bioactive glass ceramic composite scaffolds for alveolar bone tissue engineering. Chem. Eng. J. 2010, 158, 353–361. [Google Scholar] [CrossRef]
  33. Modrák, M.; Trebunová, M.; Balogová, A.F.; Hudák, R.; Zivcák, J. Biodegradable Materials for Tissue Engineering: Development, Classification and Current Applications. J. Funct. Biomater. 2023, 14, 159. [Google Scholar] [CrossRef] [PubMed]
  34. Tajvar, S.; Hadjizadeh, A.; Samandari, S.S. Scaffold degradation in bone tissue engineering: An overview. Int. Biodeterior. Biodegrad. 2023, 180, 105599. [Google Scholar] [CrossRef]
  35. Kim, S.; Cui, Z.K.; Koo, B.; Zheng, J.W.; Aghaloo, T.; Lee, M. Chitosan Lysozyme Conjugates for Enzyme-Triggered Hydrogel Degradation in Tissue Engineering Applications. ACS Appl. Mater. Inter. 2018, 10, 41138–41145. [Google Scholar] [CrossRef] [PubMed]
  36. Mathaba, M.; Daramola, M.O. Effect of Chitosan’s Degree of Deacetylation on the Performance of PES Membrane Infused with Chitosan during AMD Treatment. Membranes 2020, 10, 52. [Google Scholar] [CrossRef] [PubMed]
  37. Saravanan, S.; Nethala, S.; Pattnaik, S.; Tripathi, A.; Moorthi, A.; Selvamurugan, N. Preparation, characterization and antimicrobial activity of a bio-composite scaffold containing chitosan/nano-hydroxyapatite/nano-silver for bone tissue engineering. Int. J. Biol. Macromol. 2011, 49, 188–193. [Google Scholar] [CrossRef] [PubMed]
  38. Bashir, S.; Hina, M.; Iqbal, J.; Rajpar, A.H.; Mujtaba, M.A.; Alghamdi, N.A.; Wageh, S.; Ramesh, K.; Ramesh, S. Fundamental Concepts of Hydrogels: Synthesis, Properties, and Their Applications. Polymers 2020, 12, 2702. [Google Scholar] [CrossRef] [PubMed]
  39. Mantha, S.; Pillai, S.; Khayambashi, P.; Upadhyay, A.; Zhang, Y.L.; Tao, O.; Pham, H.M.; Tran, S.D. Smart Hydrogels in Tissue Engineering and Regenerative Medicine. Materials 2019, 12, 3323. [Google Scholar] [CrossRef]
  40. Radulescu, D.M.; Neacsu, I.A.; Grumezescu, A.M.; Andronescu, E. New Insights of Scaffolds Based on Hydrogels in Tissue Engineering. Polymers 2022, 14, 799. [Google Scholar] [CrossRef]
  41. Almawash, S.; Osman, S.K.; Mustafa, G.; El Hamd, M.A. Current and Future Prospective of Injectable Hydrogels-Design Challenges and Limitations. Pharmaceuticals 2022, 15, 371. [Google Scholar] [CrossRef] [PubMed]
  42. Tang, G.K.; Tan, Z.H.; Zeng, W.S.; Wang, X.; Shi, C.G.; Liu, Y.; He, H.L.; Chen, R.; Ye, X.J. Recent Advances of Chitosan-Based Injectable Hydrogels for Bone and Dental Tissue Regeneration. Front. Bioeng. Biotech. 2020, 8, 587658. [Google Scholar] [CrossRef] [PubMed]
  43. Jiang, Y.; Fu, C.H.; Wu, S.H.; Liu, G.H.; Guo, J.; Su, Z.Q. Determination of the Deacetylation Degree of Chitooligosaccharides. Mar. Drugs 2017, 15, 332. [Google Scholar] [CrossRef] [PubMed]
  44. Jayakumar, R.; Prabaharan, M.; Kumar, P.T.S.; Nair, S.V.; Tamura, H. Biomaterials based on chitin and chitosan in wound dressing applications. Biotechnol. Adv. 2011, 29, 322–337. [Google Scholar] [CrossRef] [PubMed]
  45. Mahmoud, A.A.; Salama, A.H. Norfloxacin-loaded collagen/chitosan scaffolds for skin reconstruction: Preparation, evaluation and wound healing assessment. Eur. J. Pharm. Sci. 2016, 83, 155–165. [Google Scholar] [CrossRef] [PubMed]
  46. Lee, Y.M.; Park, Y.J.; Lee, S.J.; Ku, Y.; Han, S.B.; Choi, S.M.; Klokkevold, P.R.; Chung, C.P. Tissue engineered bone formation using chitosan/tricalcium phosphate sponges. J. Periodontol. 2000, 71, 410–417. [Google Scholar] [CrossRef] [PubMed]
  47. Arpornmaeklong, P.; Pripatnanont, P.; Suwatwirote, N. Properties of chitosan-collagen sponges and osteogenic differentiation of rat-bone-marrow stromal cells. Int. J. Oral Maxillofac. Surg. 2008, 37, 357–366. [Google Scholar] [CrossRef] [PubMed]
  48. Du, X.C.; Wu, L.; Yan, H.Y.; Jiang, Z.Y.; Li, S.L.; Li, W.; Bai, Y.L.; Wang, H.J.; Cheng, Z.J.; Kong, D.L.; et al. Microchannelled alkylated chitosan sponge to treat noncompressible hemorrhages and facilitate wound healing. Nat. Commun. 2021, 12, 4733. [Google Scholar] [CrossRef] [PubMed]
  49. Wu, J.M.; Su, C.; Jiang, L.; Ye, S.; Liu, X.F.; Shao, W. Green and Facile Preparation of Chitosan Sponges as Potential Wound Dressings. ACS Sustain. Chem. Eng. 2018, 6, 9145–9152. [Google Scholar] [CrossRef]
  50. Al-Mofty, S.E.; Karaly, A.H.; Sarhan, W.A.; Azzazy, H.M.E. Multifunctional Hemostatic PVA/Chitosan Sponges Loaded with Hydroxyapatite and Ciprofloxacin. ACS Omega 2022, 7, 13210–13220. [Google Scholar] [CrossRef]
  51. Cao, S.J.; Xu, G.; Li, Q.J.; Zhang, S.K.; Yang, Y.F.; Chen, J.D. Double crosslinking chitosan sponge with antibacterial and hemostatic properties for accelerating wound repair. Compos. Part B-Eng. 2022, 234, 109746. [Google Scholar] [CrossRef]
  52. Afsharian, Y.P.; Rahimnejad, M. Bioactive electrospun scaffolds for wound healing applications: A comprehensive review. Polym. Test. 2021, 93, 106952. [Google Scholar] [CrossRef]
  53. Zhang, Z.P.; Hu, J.; Ma, P.X. Nanofiber-based delivery of bioactive agents and stem cells to bone sites. Adv. Drug Deliv. Rev. 2012, 64, 1129–1141. [Google Scholar] [CrossRef] [PubMed]
  54. Homayoni, H.; Ravandi, S.A.H.; Valizadeh, M. Electrospinning of chitosan nanofibers: Processing optimization. Carbohydr. Polym. 2009, 77, 656–661. [Google Scholar] [CrossRef]
  55. Mitra, A.; Dey, B. Chitosan microspheres in novel drug delivery systems. Indian. J. Pharm. Sci. 2011, 73, 355–366. [Google Scholar] [CrossRef]
  56. Chi, H.G.; Qiu, Y.Q.; Ye, X.Q.; Shi, J.L.; Li, Z.Y. Preparation strategy of hydrogel microsphere and its application in skin repair. Front. Bioeng. Biotech. 2023, 11, 1239183. [Google Scholar] [CrossRef] [PubMed]
  57. Hu, X.L.; He, J.; Yong, X.; Lu, J.L.; Xiao, J.P.; Liao, Y.J.; Li, Q.; Xiong, C.D. Biodegradable poly (lactic acid-co-trimethylene carbonate)/chitosan microsphere scaffold with shape-memory effect for bone tissue engineering. Colloids Surf. B 2020, 195, 111218. [Google Scholar] [CrossRef] [PubMed]
  58. Budhiraja, M.; Zafar, S.; Akhter, S.; Alrobaian, M.; Rashid, M.A.; Barkat, M.A.; Beg, S.; Ahmad, F.J. Mupirocin-Loaded Chitosan Microspheres Embedded in Extract Containing Collagen Scaffold Accelerate Wound Healing Activity. AAPS PharmSciTech 2022, 23, 77. [Google Scholar] [CrossRef]
  59. Fan, M.; Ma, Y.; Tan, H.P.; Jia, Y.; Zou, S.Y.; Guo, S.X.; Zhao, M.; Huang, H.; Ling, Z.H.; Chen, Y.; et al. Covalent and injectable chitosan-chondroitin sulfate hydrogels embedded with chitosan microspheres for drug delivery and tissue engineering. Mater. Sci. Eng. C-Mater. 2017, 71, 67–74. [Google Scholar] [CrossRef]
  60. Thakhiew, W.; Champahom, M.; Devahastin, S.; Soponronnarit, S. Improvement of mechanical properties of chitosan-based films via physical treatment of film-forming solution. J. Food Eng. 2015, 158, 66–72. [Google Scholar] [CrossRef]
  61. Shoueir, K.R.; El-Desouky, N.; Rashad, M.M.; Ahmed, M.K.; Janowska, I.; El-Kemary, M. Chitosan based-nanoparticles and nanocapsules: Overview, physicochemical features, applications of a nanofibrous scaffold, and bioprinting. Int. J. Biol. Macromol. 2021, 167, 1176–1197. [Google Scholar] [CrossRef] [PubMed]
  62. Grenha, A. Chitosan nanoparticles: A survey of preparation methods. J. Drug Target. 2012, 20, 291–300. [Google Scholar] [CrossRef] [PubMed]
  63. Tayebi, T.; Baradaran-Rafii, A.; Hajifathali, A.; Rahimpour, A.; Zali, H.; Shaabani, A.; Niknejad, H. Biofabrication of chitosan/chitosan nanoparticles/polycaprolactone transparent membrane for corneal endothelial tissue engineering. Sci. Rep. 2021, 11, 7060. [Google Scholar] [CrossRef] [PubMed]
  64. Azizian, S.; Hadjizadeh, A.; Niknejad, H. Chitosan-gelatin porous scaffold incorporated with Chitosan nanoparticles for growth factor delivery in tissue engineering. Carbohydr. Polym. 2018, 202, 315–322. [Google Scholar] [CrossRef] [PubMed]
  65. Karri, V.V.S.R.; Kuppusamy, G.; Talluri, S.V.; Mannemala, S.S.; Kollipara, R.; Wadhwani, A.D.; Mulukutla, S.; Raju, K.R.S.; Malayandi, R. Curcumin loaded chitosan nanoparticles impregnated into collagen-alginate scaffolds for diabetic wound healing. Int. J. Biol. Macromol. 2016, 93, 1519–1529. [Google Scholar] [CrossRef]
  66. Modaresifar, K.; Hadjizadeh, A.; Niknejad, H. Design and fabrication of GelMA/chitosan nanoparticles composite hydrogel for angiogenic growth factor delivery. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1799–1808. [Google Scholar] [CrossRef]
  67. Seddighian, A.; Ganji, F.; Baghaban-Eslaminejad, M.; Bagheri, F. Electrospun PCL scaffold modified with chitosan nanoparticles for enhanced bone regeneration. Prog. Biomater. 2021, 10, 65–76. [Google Scholar] [CrossRef]
  68. Delan, W.K.; Ali, I.H.; Zakaria, M.; Elsaadany, B.; Fares, A.R.; ElMeshad, A.N.; Mamdouh, W. Investigating the bone regeneration activity of PVA nanofibers scaffolds loaded with simvastatin/chitosan nanoparticles in an induced bone defect rabbit model. Int. J. Biol. Macromol. 2022, 222, 2399–2413. [Google Scholar] [CrossRef]
  69. Kaparekar, P.S.; Pathmanapan, S.; Anandasadagopan, S.K. Polymeric scaffold of Gallic acid loaded chitosan nanoparticles infused with collagen-fibrin for wound dressing application. Int. J. Biol. Macromol. 2020, 165, 930–947. [Google Scholar] [CrossRef]
  70. Amnieh, Y.A.; Ghadirian, S.; Mohammadi, N.; Shadkhast, M.; Karbasi, S. Evaluation of the effects of chitosan nanoparticles on polyhydroxy butyrate electrospun scaffolds for cartilage tissue engineering applications. Int. J. Biol. Macromol. 2023, 249, 126064. [Google Scholar] [CrossRef]
  71. Yang, L.M.; Zhao, X.D.; Kong, Y.S.; Li, R.J.; Li, T.; Wang, R.; Ma, Z.F.; Liang, Y.M.; Ma, S.H.; Zhou, F. Injectable carboxymethyl chitosan/nanosphere-based hydrogel with dynamic crosslinking network for efficient lubrication and sustained drug release. Int. J. Biol. Macromol. 2023, 229, 814–824. [Google Scholar] [CrossRef] [PubMed]
  72. Cavalli, R.; Leone, F.; Minelli, R.; Fantozzi, R.; Dianzani, C. New Chitosan Nanospheres for the Delivery of 5-Fluorouracil: Preparation, Characterization and Studies. Curr. Drug Deliv. 2014, 11, 270–278. [Google Scholar] [CrossRef]
  73. Xing, L.; Zhao, Q.; Zheng, X.Y.; Hui, M.W.; Peng, Y.L.; Zhu, X.; Hu, L.; Yao, W.L.; Yan, Z.Q. Porous Ag-Chitosan Nanospheres Bridged by Cysteine Residues for Colorimetric Sensing of Trace Hg. ACS Appl. Nano Mater. 2021, 4, 3639–3646. [Google Scholar] [CrossRef]
  74. Askari, M.; Afshar, M.; Khorashadizadeh, M.; Zardast, M.; Naghizadeh, A. Wound Healing Effects of Chitosan Nanosheets/Honey Compounds in Male BALB/c Mice. Int. J. Low. Extrem. Wounds 2022. [Google Scholar] [CrossRef] [PubMed]
  75. Derakhshi, M.; Daemi, S.; Shahini, P.; Habibzadeh, A.; Mostafavi, E.; Ashkarran, A.A. Two-Dimensional Nanomaterials beyond Graphene for Biomedical Applications. J. Funct. Biomater. 2022, 13, 27. [Google Scholar] [CrossRef]
  76. Jiang, M.J.; Zhu, Y.N.; Li, Q.S.; Liu, W.X.; Dong, A.; Zhang, L. 2D nanomaterial-based 3D network hydrogels for anti-infection therapy. J. Mater. Chem. B 2024, 12, 916–951. [Google Scholar] [CrossRef] [PubMed]
  77. Naskar, A.; Kim, K.S. Black phosphorus nanomaterials as multi-potent and emerging platforms against bacterial infections. Microb. Pathog. 2019, 137, 103800. [Google Scholar] [CrossRef] [PubMed]
  78. Naskar, A.; Khan, H.; Sarkar, R.; Kumar, S.; Halder, D.; Jana, S. Anti-biofilm activity and food packaging application of room temperature solution process based polyethylene glycol capped Ag-ZnO-graphene nanocomposite. Mater. Sci. Eng. C-Mater. 2018, 91, 743–753. [Google Scholar] [CrossRef]
  79. Murali, A.; Lokhande, G.; Deo, K.A.; Brokesh, A.; Gaharwar, A.K. Emerging 2D nanomaterials for biomedical applications. Mater. Today 2021, 50, 276–302. [Google Scholar] [CrossRef]
  80. Cho, C.; Zhang, Z.C.; Kim, J.M.; Ma, P.J.; Haque, M.F.; Snapp, P.; Nam, S. Spatial Tuning of Light-Matter Interaction via Strain-Gradient-Induced Polarization in Freestanding Wrinkled 2D Materials. Nano Lett. 2023, 23, 9340–9346. [Google Scholar] [CrossRef]
  81. Baig, N. Two-dimensional nanomaterials: A critical review of recent progress, properties, applications, and future directions. Compos. Part A Appl. Sci. Manuf. 2023, 165, 107362. [Google Scholar] [CrossRef]
  82. Hermenean, A.; Codreanu, A.; Herman, H.; Balta, C.; Rosu, M.; Mihali, C.V.; Ivan, A.; Dinescu, S.; Ionita, M.; Costache, M. Chitosan-Graphene Oxide 3D scaffolds as Promising Tools for Bone Regeneration in Critical-Size Mouse Calvarial Defects. Sci. Rep. 2017, 7, 16641. [Google Scholar] [CrossRef] [PubMed]
  83. Dinescu, S.; Ionita, M.; Ignat, S.R.; Costache, M.; Hermenean, A. Graphene Oxide Enhances Chitosan-Based 3D Scaffold Properties for Bone Tissue Engineering. Int. J. Mol. Sci. 2019, 20, 5077. [Google Scholar] [CrossRef] [PubMed]
  84. Saravanan, S.; Sareen, N.; Abu-El-Rub, E.; Ashour, H.; Sequiera, G.L.; Ammar, H.I.; Gopinath, V.; Shamaa, A.A.; Saved, S.S.E.; Moudgil, M.; et al. Graphene Oxide-Gold Nanosheets Containing Chitosan Scaffold Improves Ventricular Contractility and Function After Implantation into Infarcted Heart. Sci. Rep. 2018, 8, 15069. [Google Scholar] [CrossRef] [PubMed]
  85. Sivashankari, P.R.; Prabaharan, M. Three-dimensional porous scaffolds based on agarose/chitosan/graphene oxide composite for tissue engineering. Int. J. Biol. Macromol. 2020, 146, 222–231. [Google Scholar] [CrossRef]
  86. Yang, B.; Wang, P.B.; Mu, N.; Ma, K.; Wang, S.; Yang, C.Y.; Huang, Z.B.; Lai, Y.; Feng, H.; Yin, G.F.; et al. Graphene oxide-composited chitosan scaffold contributes to functional recovery of injured spinal cord in rats. Neural Regen. Res. 2021, 16, 1829–1835. [Google Scholar] [CrossRef] [PubMed]
  87. Shamekhi, M.A.; Mirzadeh, H.; Mahdavi, H.; Rabiee, A.; Mohebbi-Kalhori, D.; Eslaminejad, M.B. Graphene oxide containing chitosan scaffolds for cartilage tissue engineering. Int. J. Biol. Macromol. 2019, 127, 396–405. [Google Scholar] [CrossRef]
  88. Jiang, L.L.; Chen, D.Y.; Wang, Z.; Zhang, Z.M.; Xia, Y.L.; Xue, H.Y.; Liu, Y. Preparation of an Electrically Conductive Graphene Oxide/Chitosan Scaffold for Cardiac Tissue Engineering. Appl. Biochem. Biotech. 2019, 188, 952–964. [Google Scholar] [CrossRef] [PubMed]
  89. Pan, W.Z.; Dai, C.B.; Li, Y.; Yin, Y.M.; Gong, L.; Machuki, J.O.; Yang, Y.; Qiu, S.; Guo, K.J.; Gao, F.L. PRP-chitosan thermoresponsive hydrogel combined with black phosphorus nanosheets as injectable biomaterial for biotherapy and phototherapy treatment of rheumatoid arthritis. Biomaterials 2020, 239, 119851. [Google Scholar] [CrossRef]
  90. He, M.M.; Zhu, C.; Sun, D.; Liu, Z.; Du, M.X.; Huang, Y.; Huang, L.Z.; Wang, J.H.; Liu, L.M.; Li, Y.B.; et al. Layer-by-layer assembled black phosphorus/chitosan composite coating for multi-functional PEEK bone scaffold. Compos. Part. B-Eng. 2022, 246, 110266. [Google Scholar] [CrossRef]
  91. Shen, H.Y.; Jiang, C.Y.; Li, W.; Wei, Q.F.; Ghiladi, R.A.; Wang, Q.Q. Synergistic Photodynamic and Photothermal Antibacterial Activity of In Situ Grown Bacterial Cellulose/MoS-Chitosan Nanocomposite Materials with Visible Light Illumination. ACS Appl. Mater. Interfaces 2021, 13, 31193–31205. [Google Scholar] [CrossRef] [PubMed]
  92. Liang, Z.Y.; Gao, J.; Yin, Z.Z.; Li, J.Y.; Cai, W.R.; Kong, Y. A sequential delivery system based on MoS nanoflower doped chitosan/oxidized dextran hydrogels for colon cancer treatment. Int. J. Biol. Macromol. 2023, 233, 123616. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, Q.L.; Zhang, L.; Liu, Y.H.; Cai, L.; Zhou, L.Z.; Jiang, H.J.; Chen, J. Encapsulating MoS-nanoflowers conjugated with chitosan oligosaccharide into electrospun nanofibrous scaffolds for photothermal inactivation of bacteria. J. Nanostructure Chem. 2022, 14, 137–151. [Google Scholar] [CrossRef]
  94. Mukheem, A.; Shahabuddin, S.; Akbar, N.; Anwar, A.; Sarih, N.M.; Sudesh, K.; Khan, N.A.; Sridewi, N. Fabrication of biopolymer polyhydroxyalkanoate/chitosan and 2D molybdenum disulfide-doped scaffolds for antibacterial and biomedical applications. Appl. Microbiol. Biotechnol. 2020, 104, 3121–3131. [Google Scholar] [CrossRef] [PubMed]
  95. Dong, Y.H.; Liu, J.H.; Chen, Y.; Zhu, T.; Li, Y.H.; Zhang, C.L.; Zeng, X.; Chen, Q.M.; Peng, Q. Photothermal and natural activity-based synergistic antibacterial effects of Ti3C2Tx MXene-loaded chitosan hydrogel against methicillin-resistant Staphylococcus aureus. Int. J. Biol. Macromol. 2023, 240, 124482. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, Y.Q.; Xu, D.R.; Ding, Y.; Lv, X.X.; Huang, T.T.; Yuan, B.L.; Jiang, L.; Sun, X.Y.; Yao, Y.Q.; Tang, J. A conductive polyacrylamide hydrogel enabled by dispersion-enhanced MXene@chitosan assembly for highly stretchable and sensitive wearable skin. J. Mater. Chem. B 2021, 9, 8862–8870. [Google Scholar] [CrossRef]
  97. Naskar, A.; Lee, S.; Kim, K.S. Au-ZnO Conjugated Black Phosphorus as a Near-Infrared Light-Triggering and Recurrence-Suppressing Nanoantibiotic Platform against. Pharmaceutics 2021, 13, 52. [Google Scholar] [CrossRef]
  98. Cho, H.; Naskar, A.; Lee, S.; Kim, S.; Kim, K.S. A New Surface Charge Neutralizing Nano-Adjuvant to Potentiate Polymyxins in Killing Mcr-1 Mediated Drug-Resistant. Pharmaceutics 2021, 13, 250. [Google Scholar] [CrossRef]
  99. Luo, M.M.; Fan, T.J.; Zhou, Y.; Zhang, H.; Mei, L. 2D Black Phosphorus-Based Biomedical Applications. Adv. Funct. Mater. 2019, 29, 1808306. [Google Scholar] [CrossRef]
  100. Naskar, A.; Cho, H.; Kim, K.S. Black phosphorus-based CuS nanoplatform: Near-infrared-responsive and reactive oxygen species-generating agent against environmental bacterial pathogens. J. Environ. Chem. Eng. 2022, 10, 108226. [Google Scholar] [CrossRef]
  101. Naskar, A.; Shin, J.; Kim, K.S. A MoS based silver-doped ZnO nanocomposite and its antibacterial activity against β-lactamase expressing. RSC Adv. 2022, 12, 7268–7275. [Google Scholar] [CrossRef] [PubMed]
  102. Yadav, V.; Roy, S.; Singh, P.; Khan, Z.; Jaiswal, A. 2D MoS-Based Nanomaterials for Therapeutic, Bioimaging, and Biosensing Applications. Small 2019, 15, 1803706. [Google Scholar] [CrossRef] [PubMed]
  103. Mutalik, C.; Okoro, G.; Chou, H.L.; Lin, I.H.; Yougbaré, S.; Chang, C.C.; Kuo, T.R. Phase-Dependent 1T/2H-MoS Nanosheets for Effective Photothermal Killing of Bacteria. ACS Sustain. Chem. Eng. 2022, 10, 8949–8957. [Google Scholar] [CrossRef]
  104. Shi, J.P.; Li, J.; Wang, Y.; Cheng, J.J.; Zhang, C.Y. Recent advances in MoS-based photothermal therapy for cancer and infectious disease treatment. J. Mater. Chem. B 2020, 8, 5793–5807. [Google Scholar] [CrossRef] [PubMed]
  105. Chen, L.; Dai, X.Y.; Feng, W.; Chen, Y. Biomedical Applications of MXenes: From Nanomedicine to Biomaterials. Acc. Mater. Res. 2022, 3, 785–798. [Google Scholar] [CrossRef]
  106. Yang, B.X.; Kilari, S.; Brahmbhatt, A.; McCall, D.L.; Torres, E.N.; Leof, E.B.; Mukhopadhyay, D.; Misra, S. CorMatrix Wrapped Around the Adventitia of the Arteriovenous Fistula Outflow Vein Attenuates Venous Neointimal Hyperplasia. Sci. Rep. 2017, 7, 14298. [Google Scholar] [CrossRef]
  107. Almeida, A.; Günday-Türeli, N.; Sarmento, B. A scale-up strategy for the synthesis of chitosan derivatives used in micellar nanomedicines. Int. J. Pharm. 2021, 609, 121151. [Google Scholar] [CrossRef]
Figure 1. Schematic representation for the preparation of CS-GAP nanobiocomposite film and sponge. Reproduced with permission from Ref. [13] Copyright 2020, American Chemical Society.
Figure 1. Schematic representation for the preparation of CS-GAP nanobiocomposite film and sponge. Reproduced with permission from Ref. [13] Copyright 2020, American Chemical Society.
Polymers 16 01327 g001
Figure 2. Schematic representation of the Biomineralized CS/HC/HA/BP scaffold formation for the photothermal treatment of bacterial elimination, tumor ablation, and subsequent osteogenesis. Reproduced with permission from Ref. [14] Copyright 2023, Elsevier.
Figure 2. Schematic representation of the Biomineralized CS/HC/HA/BP scaffold formation for the photothermal treatment of bacterial elimination, tumor ablation, and subsequent osteogenesis. Reproduced with permission from Ref. [14] Copyright 2023, Elsevier.
Polymers 16 01327 g002
Figure 3. Schematic representation of the QCS-MoS2/PVA hydrogel formation for photothermal treatment of bacterial elimination. Reproduced with permission from Ref. [15]. Copyright 2022, Elsevier.
Figure 3. Schematic representation of the QCS-MoS2/PVA hydrogel formation for photothermal treatment of bacterial elimination. Reproduced with permission from Ref. [15]. Copyright 2022, Elsevier.
Polymers 16 01327 g003
Table 1. Chitosan nanoparticle (CS NP)-based scaffolds and their applications.
Table 1. Chitosan nanoparticle (CS NP)-based scaffolds and their applications.
MaterialEffectNP Size Ref.
CS NPs-BSA-bFGFSignificantly affected the physical properties of chitosan-gelatin scaffold∼266 nm[64]
CUR-CS NPsImproved stability and solubility for better tissue regeneration applications∼197 nm[65]
GelMA/CS NPs-bFGFProvide a sustained release of growth factors∼267 nm[66]
CS NPs-PCL-DEXEnhanced osteogenic differentiation of the mesenchymal stem cells∼285 nm[67]
PVA NF with SIM/CS NPsControlled drug delivery for bone regeneration application∼110–140 nm[68]
GA-CSNPsWound healing∼96–357 nm[69]
CS NPs-PHBCartilage tissue engineering∼255 nm[70]
Abbreviations: CS NPs: Chitosan nanoparticles, BSA: bovine serum albumin, CUR: curcumin, GelMA: Gelatin methacryloyl, PCL: poly-ε-caprolacton, DEX: dexamethasone, PVA NF: Polyvinyl alcohol nanofiber, SIM: Simvastatin, GA: Gallic acid, PHB: polyhydroxy butyrate.
Table 2. Chitosan (CS)-2D nanomaterial-based scaffolds and their applications.
Table 2. Chitosan (CS)-2D nanomaterial-based scaffolds and their applications.
MaterialEffectRef.
CS-GO-1Bone tissue regeneration in critical-size mouse calvarial defects[82]
CS-GO-2Ability to support stem cell differentiation processes for bone tissue engineering[83]
CS-GAPAntibacterial scaffolds for hemorrhage control and wound-healing application[13]
CS-GO-AuImprovement of the ventricular contractility and function into infarcted heart in rat model.[84]
Agarose/CS/GOPotential application in bone and osteochondral tissue engineering[85]
GO-composited CSFunctional recovery of injured spinal cord in rats[86]
CS-GO-3Cartilage tissue engineering[87]
GO/CSCardiac tissue engineering[88]
CS/HC/HA/BPPhotothermal scaffold for bone tumor-related application[14]
BP/CS/PRPPhotothermal treatment of rheumatoid arthritis[89]
BP/CS compositeThe biocompatible polyetheretherketone (PEEK) scaffold provided similar mechanical properties and architecture compared to that of the natural bone.[90]
QCS-MoS2-PVAPhotothermal antibacterial activity against S. aureus and E. coli.[15]
BC/MoS2-CSPhotodynamic and photothermal antibacterial activities against E. coli and S. aureus[91]
MoS2 doped CS/OD hydrogelsPhotothermal colon cancer treatment[92]
MoS2-LA-COSPhotothermal antibacterial activity against S. aureus and E. coli.[93]
PHA-CS/MoS2Antibacterial activity against multi-drug-resistant E. coli K1 and methicillin-resistant S. aureus (MRSA)[94]
MX-CSSynergistic photothermal antibacterial activity against MRSA[95]
MX-CS-hyaluronateAntibacterial activity against E. coli, S. aureus, and Bacillus sp.[11]
MXene@CSHighly stretchable and sensitive wearable skin[96]
Abbreviations: CS: Chitosan, GO: Graphene oxide, GAP: graphene-silver-polycationic peptide, HC: hydroxypropyltrimethyl ammonium chloride chitosan, HA: hydroxyapatite, BP: black phosphorus, PRP: platelet-rich plasma, QCS: quaternized chitosan, BC: Bacterial cellulose, OD: oxidized dextran, LA: α-lipoic acid, COS: chitosan oligosaccharide, PHA: polyhydroxyalkanoate, MX: Ti3C2Tx MXene.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Naskar, A.; Kilari, S.; Misra, S. Chitosan-2D Nanomaterial-Based Scaffolds for Biomedical Applications. Polymers 2024, 16, 1327. https://doi.org/10.3390/polym16101327

AMA Style

Naskar A, Kilari S, Misra S. Chitosan-2D Nanomaterial-Based Scaffolds for Biomedical Applications. Polymers. 2024; 16(10):1327. https://doi.org/10.3390/polym16101327

Chicago/Turabian Style

Naskar, Atanu, Sreenivasulu Kilari, and Sanjay Misra. 2024. "Chitosan-2D Nanomaterial-Based Scaffolds for Biomedical Applications" Polymers 16, no. 10: 1327. https://doi.org/10.3390/polym16101327

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