Next Article in Journal
Mechanism for the Intercalation of Aniline Cations into the Interlayers of Graphite
Previous Article in Journal
Stable Dried Catalase Particles Prepared by Electrospraying
Previous Article in Special Issue
Liposomal Form of 2,4-Dinitrophenol Lipophilic Derivatives as a Promising Therapeutic Agent for ATP Synthesis Inhibition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomimetic Nanomaterials: Diversity, Technology, and Biomedical Applications

1
Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia
2
Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
3
SciBear OU, Tartu mnt 67/1-13b, Kesklinna Linnaosa, 10115 Tallinn, Estonia
4
Research Center of Biotechnology of the Russian Academy of Sciences, Institute of Bioengineering, 119071 Moscow, Russia
5
Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
6
Center of Translational Cancer Research (TranslaTUM), Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
7
Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
8
National Center for Neurosurgery, Nur-Sultan 010000, Kazakhstan
*
Authors to whom correspondence should be addressed.
Nanomaterials 2022, 12(14), 2485; https://doi.org/10.3390/nano12142485
Submission received: 23 June 2022 / Revised: 15 July 2022 / Accepted: 18 July 2022 / Published: 20 July 2022
(This article belongs to the Special Issue Biomimetic and Biogenic Multifunctional Nanomaterials)

Abstract

:
Biomimetic nanomaterials (BNMs) are functional materials containing nanoscale components and having structural and technological similarities to natural (biogenic) prototypes. Despite the fact that biomimetic approaches in materials technology have been used since the second half of the 20th century, BNMs are still at the forefront of materials science. This review considered a general classification of such nanomaterials according to the characteristic features of natural analogues that are reproduced in the preparation of BNMs, including biomimetic structure, biomimetic synthesis, and the inclusion of biogenic components. BNMs containing magnetic, metal, or metal oxide organic and ceramic structural elements (including their various combinations) were considered separately. The BNMs under consideration were analyzed according to the declared areas of application, which included tooth and bone reconstruction, magnetic and infrared hyperthermia, chemo- and immunotherapy, the development of new drugs for targeted therapy, antibacterial and anti-inflammatory therapy, and bioimaging. In conclusion, the authors’ point of view is given about the prospects for the development of this scientific area associated with the use of native, genetically modified, or completely artificial phospholipid membranes, which allow combining the physicochemical and biological properties of biogenic prototypes with high biocompatibility, economic availability, and scalability of fully synthetic nanomaterials.

1. Introduction

“Biomimetics” is a concept introduced in 1957 by Otto Schmitt [1,2] that, generally, means the imitation of structures, characteristics, models, and compositions of natural objects to solve various problems [3]. The word “biomimetic” comes from the Greek word “bios” (life) and the suffix “mimetic” (mimicry) [4]. This concept also includes approaches and processes mimicking biological or natural ones such as, for example, the synthesis of nanomaterials or nanostructures that reproduce the physicochemical, mechanical, and biological properties of natural (biogenic) materials at the nano- or macroscale [5,6]. Similar to the formation of biogenic materials, the technology of biomimetic materials includes the processes of self-assembly and interfacial molecular recognition [7,8]. Other important directions of development for such materials are biopolymers [9] and biocomposites [10], which are receiving immense consideration in biomedical and other fields.
At the same time, the process of biomimetic material formation can differ greatly from the natural analog process in terms of parameters (for example, high energies during plasma deposition [11] or electric currents during electrospinning [12]). The transfer of an idea or mechanism from living systems to nonliving ones is not trivial. The direct copying of a biological prototype may not be successful, even if it is feasible with modern technology [13]. Currently, biomimetic materials play an important role in medical science in the development of drug delivery systems and theranostics [14,15,16,17,18,19,20,21,22,23,24,25,26], tissue engineering [27,28,29], and combined solutions to these problems [30]. Chronologically, the first attempts of delivery-system-mimicking were directed towards the development of “biologically inert” nanoparticles (NPs), demonstrating reduced interaction with immune cells. The next generation of delivery system had to provide immune inertness and specific binding to target pathogenic cells, such as cells present in pathological lesions in neurodegenerative disorders, inflammatory endothelial cells, etc. These tasks required the development of “targeted” NPs functionalized with monoclonal antibodies or other types of ligands. The next step was the development of biomimetic NPs partially complementary to target cells that reproduced the surface characteristics of native cells, which further improved the effectiveness of delivery systems [31]. A schematic diagram illustrating the evolution of NP technology for biomedicine is shown in Figure 1a.
An example of the technological evolution of NPs is the synthesis of biomimetic nanomaterials (BNMs) that specifically accumulate in the inflammatory zone. Over the past thirty years, the development of such NPs has largely relied on three strategies [16]:
(1)
Synthetic NPs modified with targeting ligands that mimic cell surface proteins;
(2)
NPs covered with a native cell membrane;
(3)
Liposomes formed using cell membrane proteins (Figure 1b).
Biomimetic NPs take advantage of both cell membranes and synthetic NPs and are distinguished by three main features: prolonged circulation in the bloodstream, specific binding, and reduced toxicity [16]. BNMs intended for the targeted therapy of atherosclerosis and inflammatory diseases can be coated with cell membranes derived from various types of cells, such as erythrocytes, macrophages, and platelets. The methods for obtaining cell-membrane-coated NPs can be summarized as a three-step process. At the first stage, the cell membrane is isolated from the original cells (cells can be lysed using various methods, after which centrifugation is used to separate fragments of the cell membrane). Then, the prepared cell wall fragments are repeatedly pressed through a polycarbonate membrane with a pore diameter of ~200–400 nm to obtain microcapsules from the cell membrane, and an NP core with a dissolved drug is also prepared. In the last step, NPs are encapsulated into microcapsules made of the cell membrane (see Figure 1c) [20].
Based on the above, we propose a general classification of BNMs based on the purposes of reproduced BNMs and defined as materials with biomimetic structural elements (for example, liposomes incorporating cell wall proteins [16]), materials produced using biomimetic methods (for example, NPs grown using magnetosome-associated proteins [32]), and materials containing biogenic components, i.e., so-called nanobiohydrides [9] (e.g., polymeric NPs coated with erythrocyte cell membranes [33]). Due to the wide variety of proposed structures and compositions of BNMs, the use of magnetic, metal, and metal oxide materials, as well as organic, ceramic, and hybrid (multicomponent) structural elements, are considered separately. The proposed classification with some examples from the literature, including information on biomimetic structure [16,34,35,36,37,38], biomimetic synthesis [32,39,40,41,42,43], biogenic components [31,33,44,45,46,47], magnetic BNMs [48,49,50,51,52,53,54], metal and metal oxide BNMs [55,56,57,58,59,60,61], organic, ceramic and hybrid BNMs [62,63,64,65,66,67], is shown in Figure 2.
Thus, in this review, BNMs are understood as functional materials containing nanosized components and having structural and technological similarities to natural (biogenic) analogues.

2. Interaction between Biomimetic Nanomaterials and Biological Tissue

The shapes, sizes, and surface characteristics of BNMs provide outstanding advantages that enable them to deliver water-insoluble drugs, prevent premature drug release in vivo, enhance biodistribution, control drug release, improve pharmacokinetics, and increase intracellular penetration. Due to their small sizes, these particles are able to pass through the finest blood capillaries to enable passive targeting of tumors, as enhanced permeability and retention can affect tumor vasculature [68]. Untargeted theranostic agents reduce therapeutic efficiency by nonspecific accumulation in other tissues. There are two possible mechanisms to improve the tumor selectivity of nanoparticles: (i) passive targeting through enhanced permeability retention and (ii) active targeting based on a specific cell-surface receptor with a ligand on the nanoparticles. Integrins, heterodimeric cell adhesion proteins involved in many mechanisms, including cell attachment, angiogenesis, and the metastasis of solid tumors, have been identified for the active targeting of tumor tissue in clinical trials. The authors of [69] synthesized chitosan-oligosaccharide-coated biocompatible palladium nanoparticles for photo-based imaging and therapy with effective accumulation in breast cancer cells. Further, the NIR-based photothermal ablation and PAT imaging efficiency of the formulated particles were examined using in vitro and in vivo models, as shown in Figure 3.
Conventional silica NPs are widely applied in drug delivery systems and as optical contrast agents for imaging due to their water solubility, chemical and thermal stability, low toxicity, and good biocompatibility; however, the functionalization of silica NPs is often limited by their surface properties. Biotemplated mesoporous silica NPs exhibit a higher surface area and a tunable pore volume, allowing for a higher therapeutic drug-loading capacity. A variety of methods have been established to prepare mesoporous silica NPs. Although the silica wall is amorphous, the interior of the material possesses an extremely ordered framework with uniform mesopores. It was proposed that cationic surfactant molecules could self-organize into a hexagonal structure to serve as a template, followed by the co-condensation of silica precursors and cylindrical micelles to form mesoporous silica nanoparticles (MSNs) with a pore size range of 2–50 nm [70]. Based on recent developments in nanomaterials research, silicon-based nanomaterials, along with combinations of silicon-based nanomaterials and other nanomaterials, have gained attention also in noninvasive disease diagnosis. MSNs can be suitable platforms for imaging agents because of their highly specific surface areas and functionalized surfaces [22]. Yang et al. [71] proposed a more complex schematic illustration of the composition and architecture of engineered silica-based nanoparticles with diverse appealing properties as nanocarriers, including cellular uptake, DNA transfection, adhesion and transcytosis, and biomodulators, including dendritic cell maturation, cancer cell immunogenicity, macrophage polarization, and tumor microenvironment, for biomedical applications. Cage-like polyhedral oligomeric silsesquioxanes, owing to their biocompatibility and ability to incorporate with different polymers, were shown to offer high potential for several biomedical applications such as drug delivery, dental composites, biosensors, biomedical devices, and tissue engineering [72].
It was shown in [73] that organic–inorganic hybrid polydopamine-chitosan-coated AgNPs obtained via an mussel-inspired electropolymerization strategy could be used for the noninvasive electrochemical detecting of a malondialdehyde biomarker. This result confirmed the perspectives of BNM uses both in vivo and in vitro. Another approach for understanding BNM and tissue interaction was proposed in [74], where the authors considered some typical examples of human bovine serum albumin, ferritin, and human transferrin-based nanoplatforms loaded with noble metals, oxides, semiconductors, and polymers for multifunctional applications, including chemodynamic therapy, chemotherapy, X-ray computed tomography, fluorescent imaging, magnetic resonance imaging, photoacoustic therapy, photodynamic therapy, positron emission computed tomography, photothermal therapy, and radiotherapy.
Imaging methods using upconversion nanoparticles (UCNPs) have been proposed as prospective diagnosis tools due to their ability to absorb near-infrared radiation and transform it into visible light through an upconversion process of multiple-photon absorption. Sharipov et al. [75] developed novel UCNP-loaded phosphate micelles that could be cleaved by the secreted phospholipase A2 (sPLA-2) enzyme, allowing the release and delivery of UCNPs directly to prostate cancer cells. The ability to release UCNPs in a precise location provided several advantages, including the efficient delivery of UCNPs in low concentrations, an increase in dispersion, and a remarkably high selectivity to prostate cancer cells. Correspondingly, these benefits could reduce biological side effects that other delivery systems have faced. Nanoparticles were prepared with a hydrothermal method, and a delivery system was developed in the form of phosphate micelles synthesized from biocompatible materials (see Figure 4).
Renu et al. [76] considered green-chemistry (plant extract)-mediated synthesized metal nanoparticles (silver nanoparticles) and incorporated biodegradable polymeric nanocomposite preparation, characterization, and its mechanism of antimicrobial-based wound-healing activity. Their examination of the literature displayed that there are two key types of nanocomposites of metals with polymers: (1) inorganic, metal-core nanoparticles enclosed in a polymer shell and (2) inorganic metal nanoparticles inserted into a polymer matrix. Transition-metal-based nanomaterials have shown great potential in cancer therapy due to their intensive near-infrared absorption, excellent photothermal conversion efficiency, strong X-ray attenuation, and magnetic properties. Functional polymers are usually introduced via one-step or multistep methods to further endow these nanomaterials with great biocompatibility and physiological stability. Polymer-decorated transition metal BNMs can be used in multimodal imaging diagnosis and cancer therapy [77]. Magnetic NPs with properly functionalized surfaces can be physically and chemically stable, biocompatible, and environmentally safe. Surface-coating strategies are required to facilitate the application of magnetic NPs in nanomedicine. In the biomedical applications of synthetic nanocomposites, one of the most important properties these materials should possess is high biocompatibility (or low toxicity) when exposed to cells, tissues, or organisms [78]. The authors of [79] considered a new general approach for devising multifunctional magnetic NPs containing different layers, i.e., core-magnetic NPs, with a middle layer containing different therapeutic and imaging agents and an outer layer containing different functional groups (peptides, antibodies, and aptamers) for targeting tumor tissues and for achieving simultaneous sensing, imaging, and therapy. Poon et al. [80] synthesized hybrid metal oxide–peptide amphiphile micelles consisting of an iron oxide or manganese oxide core and a fibrin-binding peptide self-assembling into 20–30 nm spherical NPs. These hybrid NPs were found to be biocompatible with human aortic endothelial cells in vitro, and they bound to human clots three to five times more efficiently than their nontargeted counterparts. Lee et al. [81] revealed four main routes of the cytotoxic mechanism of AgNPs (Figure 5).
Next, we dwell in more detail on the consideration of three groups of BNMs that differ in their compositions of functional elements (fillers).

3. Magnetic Biomimetic Nanomaterials

The biomimetic synthesis approach has been successfully used in the synthesis of magnetic materials ranging from ferrimagnetic Fe3O4 (found in magnetotactic bacteria (MTB)) to hard magnetic alloys (such as FePt and CoPt). The use of protein cages for the synthesis of magnetic BNMs provides a number of unique advantages since such structures are of biological origin and, at the same time, are suitable for scalable production and can also be modified using chemical or genetic engineering approaches to give them a specific chemical or structural functionality [82,83]. Ten years ago [64], the overwhelming majority of publications on the synthesis of magnetic NPs were devoted to physical and chemical methods, while only a small part of the works was related to biological methods (using protein constructs, bacteria, or fungi). In recent years, the development of the technology of magnetic BNMs has made it possible to reproduce chains of magnetic NPs similar to those formed in MTBs in a laboratory. Various biomimetic approaches to the synthesis of such chains with optimal properties for biomedical use have been demonstrated [84].
One of the studied types of magnetic BNMs is polymer-coated and nonpolymer-coated magnetite NPs obtained using magnetosome-associated MTB proteins [32]. When implementing this method, the biomineralization of Fe3O4 is carried out in an oxygen-free aqueous solution containing the recombinant magnetosome protein, usually for 30 days. Such magnetic NPs can be used for photothermia or magnetic hyperthermia, chemotherapy, and enzyme immobilization [32,48,85,86,87]. It was experimentally established in [32] that, when two magnetosome proteins (MamC and Mms6) were used for biosynthesis at once, it was possible to obtain magnetic NPs that were close in shape and size to magnetosome crystals [88]. Electron microscopic images of magnetic NPs obtained chemically and using the MamC and Mms6 proteins are shown in Figure 6 [32].
Another option for magnetic BNMs is the use of engineered structures based on bacterial magnetosomes. In this case, magnetosomes are isolated from MTB cells and subjected to purification, followed by biotinylation or encapsulation in an inorganic shell. Such magnetic NPs can be used for contrast enhancement in magnetic resonance imaging, magnetic particle imaging, and magnetic hyperthermia [44,89].
The construction of magnetic BNMs based on magnetite NPs coated with hydroxyapatite (HAP) is described. The preparation of such constructs is usually carried out by introducing a liquid HAP precursor into a solution containing preliminarily formed magnetite cores. The biomedical applications of such magnetic NPs can be magnetic hyperthermia, the creation of magnetic scaffolds for bone tissue restoration, and the delivery of genetic material [35,51], as shown in Figure 7.
Integration in a single biomimetic matrix (for example, SiO2) of magnetic NPs and the active substance is possible. Such BNMs can be obtained by the hydrolysis of tetramethyl orthosilicate in the presence of preliminarily obtained magnetite NPs and an enzyme solution. A possible area of application is the development of biocatalysts for targeted enzyme prodrug therapy [37].
A summary of the literature data on the structure, synthesis, and applications of magnetic BNMs is given in Table 1.
An analysis of the methods for the synthesis of magnetic BNMs allowed us to draw the following conclusions. All the methods considered in Table 1 could be conditionally divided into two groups: methods based on the use of previously obtained or commercial magnetic NPs and methods that included the process of obtaining magnetic NPs. The first group of methods included the introduction of precursors of the HAP biomimetic component into a colloidal solution of magnetic NPs [35,51], acrylamide and ethylene glycol dimethacrylate copolymerization in the presence of S-naproxen on silica-coated Fe3O4 NPs [90], preliminarily prepared Fe3O4 NP incubation in an alkaline dopamine solution [93], and the self-assembly of Fe3O4 NPs coated with a hydrophilic polymer into 1D nanochains in water [95].
Methods of the second group, from our point of view, were more promising due to the ability to create a material structure similar to natural analogues at the level of individual atoms. They included the biosynthesis of magnetic NPs from oxygen-free solutions containing recombinant MamC in anaerobic conditions [32,48,85,86,87], magnetite biomineralization using PEGylated human ferritin NPs [52], and the silica encapsulation or biotinylation of isolated bacterial magnetosomes [44,89]. Thus, the methods of the second group made it possible to achieve a combination of the advantages of biogenic magnetic NPs (magnetosomes) and their synthetic counterparts, minimizing the disadvantages of both.
Despite the long history of the development of magnetic BNM technology [68], this class of materials is not among the most widespread group of biomimetics.

4. Metal and Metal Oxide Biomimetic Nanomaterials

The physical and chemical properties of metal and metal oxide NPs can be controlled by choosing their micro- or nanoenvironments during synthesis. Some of the best-known platforms currently in use, in particular for the biomimetic synthesis of metal oxides, include ferritin, viral capsids, or bacterial cells. These biotemplates provide strictly defined conditions for the formation of NPs and, thus, their narrow distribution in shape and size [96]. In addition, such synthetic approaches are of interest due to the development of environmentally friendly NP technology. One of the most discussed approaches is the synthesis of metal NPs using organisms, of which plants are considered the best candidates and are suitable for the scalable production. The growing advantages of using plants and herbs for the biomimetic synthesis of metal NPs has prompted scientists to search for the mechanisms of metal ion biological reduction and to study in more depth the mechanisms of metal NP synthesis in plants [97]. Obtaining Ag NPs for the purpose of therapy for multiresistant pathogenic microorganisms can be carried out using plant components. Thus, the preparation of such NPs by the reduction of silver nitrate in an aqueous solution with the addition of extract of Musa balbisiana or Phlomis bracteosa seedlings, as well as Saraca indica leaves, as a reducing agent, has been described [55,98,99].
The development of biomimetic methods for the synthesis of metallic nanomaterials plays an important role in dentistry. In recent years, various approaches have been used to modify the topographical or chemical properties of traditional implant surfaces in order to improve the adhesion of the implant material to bone cells. Changing the surface of a titanium implant makes it possible to stimulate the bone tissue, minimizing the period of osseointegration and ensuring the good transfer of occlusal mechanical loads from the implant to the bone [38] (Figure 8).
A titanium base with a nanostructured calcium phosphate coating obtained by introducing HAP ceramic particles into a plasma jet directed at the treated Ti surface can be used in dentures [100].
One of the areas of application of metallic BNMs is the detection and elimination of natural and synthetic pollutants. Gold, silver, and bimetallic Ag-Au NPs for the purpose of creating sensors for biological substances and research in the field of nanotoxicology were obtained using a biomimetic method in an aqueous solution of gelatin with the successive addition of silver nitrate or hydrogen tetrachloroaurate [39].
The new, proposed biomimetic methods for obtaining Au NPs and Ag NPs include the use of “cubosomes”, model cubic nanostructures based on lipid membranes. Such BNMs are obtained through incubating preformed Au or Ag NPs with cubosomes in an aqueous solution [34] (Figure 9). The purpose of BNMs based on cubosomes is the targeted delivery of drugs.
Another important direction in the development of BNM technology for use in sensors of biological substances and oncology is the production of metal–organic frameworks (MOFs) [58,101]. Pt NPs could be synthesized using MOFs prepared using Fe(III)tetra(4-carboxyphenyl)porphine chloride as a template [101]. More complex structures, porphyrin Zr-MOFs coated with a cell membrane for the purposes of antiangiogenesis and photodynamic therapy in oncology, were obtained using solution chemistry followed by the deposition of manganese oxide from KMnO4 and camouflaging with a cell membrane [58]. Composite sponges of stearic acid do not exhibit good adsorption capacity as compared to MOFs and carbonaceous-material-based sponges. Moreover, they have complex synthesis procedures and difficult oil recovery methods. Azam et al. [102] fabricated a composite sponge with hydrophobicity as high as that of stearic acid sponges, as high an oil absorption capacity as that of MOF-based sponges, and excellent reusability of up to 10 cycles utilizing the inherited properties of MOFs (i.e., high surface area, controllable pore size, and chemical functionality at the molecular level), the nonwetting and superhydrophobic property of stearic acid, and the regular 3D skeleton of a highly porous polyurethane sponge.
The authors of [103] explored the use of MOFs formed using the self-assembly of metal ions and organic building blocks to safeguard collagen integrity in the functional dentin matrix. They demonstrated that collagen fibrils (from demineralized human dentin) could induce the biomimetic growth of MOF crystals as protective coatings to strengthen and stabilize the fibrils. Chen et al. [104] focused on the recent progress in biomimetic MOF catalysts. The authors summarized principles and strategies for the design and synthesis of biomimetic MOF catalysts, discussed structure-related catalytic properties, and particularly addressed important factors, including (1) active sites, (2) microenvironment, (3) transmission channels, and (4) co-catalytic sites, on the distinct catalytic properties of biomimetic MOFs. In addition, some examples have been given to illustrate the synergistic catalysis between multiple factors that are closely related to enzymatic catalysis. It is highly desirable, yet remains challenging, to achieve the synergy of specific functions between MOF host and guest species. Cheng et al. [105] obtained a novel MOF composite biomimetic structure inspired by a natural multienzyme system and based on the co-encapsulation of glucose oxidase and L-arginine into Cu-MOFs with Fenton-like catalytic activity to achieve a synergistic antibacterial effect. A facile strategy to prepare a biomimetic cascade reaction system by combining the advantages of enzyme immobilization and biomimetic catalysis in a one-pot reaction system based on a hierarchically porous metal–organic framework was reported [106].
MOFs are ideal candidates for building biomimetic systems because their uniform cavities can generate a high density of biomimetic active centers. In addition, thermally and chemically stable MOFs can be built with a variety of metal clusters. Furthermore, the channels of MOFs provide confined pockets, which protect the catalytic centers and enhance substrate specificity [107]. Liang et al. [108] reported that a wide range of biomacromolecules, including proteins, DNA and enzymes, could efficiently induce MOF formation and control the morphology of the resultant porous crystal via a biomimetic mineralization process under physiological conditions. The authors demonstrated that the biomimetic mineralization of MOFs formed a nanoporous shell, which encapsulated the biomacromolecules and afforded unprecedented protection from biological, thermal, and chemical degradation with the maintenance of bioactivity (Figure 10).
Singh et al. [109] proposed a protective framework offering thermal stability especially for vaccines wherein additives and stabilizers alone may not be sufficient. The authors applied an MOF biomimetic mineralization technique for live viral vaccine encapsulation. This is a ubiquitous stabilization technology that is a simple, rapid, and one-step scalable approach using cost-effective ingredients and ambient aqueous conditions. Water-stable Fe(III)-based MOFs, which exhibit intrinsic peroxidase-like activities, catalyzing the oxidation of 3,3′,5,5′-tetramethylbenzidine and o-phenylenediamin when H2O2 served as the oxidant, were constructed and characterized [110]. Their catalytic performances strongly relied on pH value, temperature, catalyst dosage, and H2O2 concentration. Compared to natural enzymes, peroxidase mimetics had the advantages of low cost, ease of preparation and storage, greater resistance to biodegradation, and less vulnerability to denaturation. The presented results could provide a possibility of building MOF-based platforms as enzymatic mimic catalysts and could facilitate their utilization in immunoassays and biotechnology. With a reactivity that mimicked the activity of an enzyme, the MOF-based material also catalyzed the isotopic exchange of oxygen between water and carbon dioxide. Wright et al. [111] provided convincing evidence that the metal nodes in MOFs had high structural fidelity with respect to the active sites of enzymes.
Ma et al. [112] designed a renewable, inexpensive, and easy-to-assemble filter prepared by growing an MOF on wood, followed by high-temperature carbonization. The proposed filter demonstrated a superior dye removal performance and was expected to replace commercial activated carbon and to be successfully applied in practical wastewater treatment fields. The effect of a peptide or protein sequence on the biomimetic mineralization of zeolite imidazolate frameworks (ZIFs), as well as a general approach to make peptide@ZIFs via a biomimetic mineralization process, was investigated [113]. The reaction rate of peptide@ZIF formation was compared for different types of peptides with different isoelectric points. Wang et al. [114] developed a new MOF–poly(amidoxime) composite membrane with a permeable macroporous structure, good mechanical properties, and high efficiency for uranium extraction from seawater. Cancer-cell-membrane-coated, triphenylphosphonium-decorated nano-MOF constructs were synthesized to achieve homologous mitochondria-targeted sonodynamic therapy efficacy in combination with simultaneous delivery of the Toll-like receptor agonist R837 as an immune adjuvant [115] (see Figure 11).
The above, as well as other examples of BNMs based on metal particles and metal oxides, are summarized in Table 2.
Comparing the summary data in Table 2 with those presented earlier in Table 1, one can draw attention to the predominantly different synthesis techniques for obtaining BNMs of the two considered groups, despite the wide variety in the approaches to synthesis proposed to date. In the case of BNMs based on magnetic particles, the biomineralization of magnetite and the use of preformed magnetic NPs are most commonly used.
For classical, well-developed BNMs based on metal particles and metal oxides, reduction from precursor solutions is most often used as a method. Nevertheless, in recent years, a newer class of BNMs including metal components has been developed rapidly in the form of MOFs. The wide perspectives of this class of materials are mainly due to its multifunctionality, including antiangiogenesis and photodynamic therapy [58]; biosensing [101]; sonodynamic therapy and immune checkpoint blockade immunotherapy [115]; protection from biological, thermal, and chemical degradation with the maintenance of bioactivity [108]; antibacterial therapy; and biomimetic catalysis [106]. Nevertheless, the technology of MOFs and, especially, composite BNMs containing MOFs is still challenging [104].
This may be due, from our point of view, not only to the chemical natures of the oxide and metal components of BNMs, but to the differences in the main claimed applications of these materials, as shown later in Section 5.

5. Organic, Ceramic, and Hybrid Biomimetic Nanomaterials

BNMs based on organic and ceramic components are by far the most widely represented compared to the two groups of BNMs described above. When obtaining BNMs based on organic components, molecular mechanisms for constructing proteins and peptides can be used, which control the synthesis of nanosized objects and the self-assembly of systems of multifunctional materials of a higher order. For this, solid-binding peptides are used, and the formation, assembly, and organization of functional nanoobjects are controlled [119]. Protein- and peptide-based biomimetic mineralization has been shown to be an efficient and promising strategy for the synthesis of magnetic and ceramic NPs. Proteins or peptides can bind metal ions through various metal-binding sites, such as N-terminal amines or multi-metal binding sites, and thus, their unique structures allow them to be used as NP growth templates [120]. A new class of biomimetic polymers is peptoids (or poly-N-substituted glycines), which are perceived due to highly efficient synthesis, high chemical stability, resistance to enzymatic hydrolysis, and biocompatibility. Due to their properties, amphiphilic peptoids are considered as customizable building blocks for obtaining BNMs with hierarchical structures and specified functionalities [121].
The development of ceramic BNM technology is largely aimed at creating scaffolds that mimic natural tissue and reproduce its properties, such as composition and microstructure. In particular, for bone tissue engineering, this is most often achieved by fabricating highly porous ceramic structures that resemble cancellous bone. These scaffolds possess many important properties, including biological activity, space for cells, and ingrowth of new tissues, as well as acceptable mechanical strength. However, when using such scaffolds, the new tissue is often disorganized and has poor mechanical properties [122]. The formation of three-dimensional tissue in the laboratory is highly dependent on the biomimetic environment, engineered extracellular matrix, and cell type, as well as biologically active components. Scaffolds should resemble the structural and biochemical features of the natural extracellular environment in order to support and control cell migration and growth [123]. In addition to being used in solving tissue-engineering problems, biomimetic ceramic scaffolds are often loaded with various therapeutic molecules to increase their biological effectiveness [124]. The peptide-mediated remineralization of artificially induced tooth lesions and the characteristics of orientation, formation, and composition of a newly formed enamel-like apatite layer were experimentally evaluated. Amelogenin was used as a model peptide [41,125].
A biomimetic design of light, high-temperature ceramics based on functionally differentiated structures was proposed. Such structures can be found in nature, in particular, on cross-sections of the bones, teeth, and stems of many plants, such as bamboo. Originally conceived for spacecraft heat shields, functionally differentiated structures are finding more and more applications in other areas, including biomedicine [126]. Researchers are attracted by the hierarchical structure of nacre, which contains at least six levels of hierarchy in the size range from a few millimeters to ~100 nm. In addition, the biomineralization of nacre is carried out by the self-assembly of elementary units under mild conditions at ambient temperature and either neutral or physiological pH, always in an aquatic environment [127]. For the production of bioactive orthopedic biomaterials using biomimetic nanocrystalline apatites, several approaches have been used, among which are in situ formation, which allows obtaining a highly reactive structure, and low-temperature molding [128]. Biomimetic ceramics of complex shapes can be obtained on the basis of self-forming, two-layer microstructures based on aluminum oxide. Such a material is produced by the repeated coagulation of a suspension of Al2O3 NPs with a ferrofluid under the action of a magnetic field, followed by high-temperature annealing [36].
A promising object of research is biomimetic catalysts based on chiral nanomaterials, which can cause the same biological effects as natural catalysts and has demonstrated high efficiency in biological applications [129]. The biomimetic synthesis of materials and the template growth of inorganic or hybrid networks using self-assembling, hybrid, organic–inorganic interfaces are also of considerable interest to researchers since artificially obtained hybrid materials lie at the junction of the organic, inorganic, and biological worlds [130].
Fatima et al. [9] obtained a composite based on bacterial cellulose waste modified with bioactive plant extract. The material showed high bactericidal activity against S. aureus and produced a clear inhibition zone, whereas negligible activity was observed against E. coli, indicating its bactericidal activity mainly against Gram-positive bacteria. Overall, their study illustrated that there is a huge potential for developing valuable biomaterials from food wastes and utilizing their liquid-holding capabilities for value-added applications in the medical and pharmaceutical fields. Chen et al. [131] reported a bamboo–nigrosine–poly(N-isopropylacrylamide) composite hydrogel actuator capable of complex deformations and near-infrared light response based on bamboo sheets. The simple and rapid ultraviolet in situ polymerization method closely connected bamboo and hydrogel so that the composite hydrogel actuator was difficult to peel off during the actuation process. In addition, the composite hydrogel actuator exhibited diverse shape deformations because of the natural anisotropy of the bamboo sheets. A composite thermoresponsive actuator with multifunctional actuation upon an alternative magnetic field was fabricated [132]. Fe3O4-NP-embedded thermoplastic polyurethane film and a photo-crosslinkable electrospun fibrous poly(N-isopropylacrylamide) mat formed the bilayer actuator.
The removal of systemically administered bacterial toxins could be accomplished using anisotropic polymeric NPs camouflaged with an erythrocyte membrane. Spherical particles of poly(lactic acid-glycolic acid) (PLGA) were immobilized on a film of polyvinyl alcohol and glycerol mechanically stretched to change the shape of the NPs from isotropic to anisotropic, and then erythrocyte membranes were deposited with an ultrasonic treatment [33] (Figure 12).
Important areas of application for polymeric NPs camouflaged by cell membranes are bioimaging, phototheranostics, and the development of nanovaccines. NPs of PLGA with tumor cell membrane envelopes could be obtained by depositing the drug in PLGA using precipitation from a solution, followed by coating with a cell membrane [45].
As well as metal NPs (see Section 4), more complex NPs can be obtained biomimically using plant extracts as sources of natural reducing agents and stabilizers. Thus, Ag–TiO2 NPs were obtained through sonochemical synthesis using Origanum majorana leaf extract as a restoring and stabilizing agent, which could be used for antibacterial and antioxidant therapy [40] (Figure 13).
Much attention from BNM researchers has been paid to works in the field of dentistry, primarily to the restoration of demineralized tooth enamel. One of proposed approaches is amelogenin-containing chitosan hydrogel [133,134]. The preparation of this material is carried out by the mixing of a solution of chitosan, calcium chloride, and recombinant porcine amelogenin, followed by mechanical agitation. Li et al. used HAP crystallized with a polyamidoamine (PAMAM) dendrimer template [135,136,137]. The treated tooth enamel was placed in a solution containing CaCl2, KH2PO4, and PAMAM dendrimers modified with carboxyl groups.
An important task in the preparation of BNMs based on NPs of different natures (oxide, metal, polymer, and semiconductor) is the possibility of experimental evaluation of the efficiency of coating particles with a cell membrane obtained from tumor cells or normal human cells (macrophages, platelets, or erythrocytes).
The use of a fluorescence-quenching assay to assess the integrity of cell membranes covering BNMs has been proposed. Using this approach, the authors of [138] measured and compared the degree of membrane coverage of BNMs and found that the approach used today did not provide adequate integrity of the cell membrane. It was demonstrated that such partially coated NPs were able to enter tumor cells through a cooperative mechanism based on NP aggregation.
Another large group of problems solved through the use of BMNs and, in particular, materials based on a natural or artificial phospholipid membrane encapsulating a drug agent is the therapy of inflammatory diseases, including atherosclerosis. The potential use for anti-inflammatory therapy of leukocyte-based biomimetic NPs obtained by combining phospholipids and leukocyte membrane proteins followed by incubation with binding antibodies has been described. Moreover, the process can be carried out both in a batch [109] and in a continuous flow (microfluidic) [67] reactor. Such biomimetic nanovesicles are called leukosomes [67,139].
In addition to leukocytes, macrophages can serve as a basis for constructing BNMs intended for the treatment of atherosclerosis. The authors of [46] proposed a construct based on a macrophage membrane in which polymeric NPs capable of responding to reactive oxygen species (ROS) were camouflaged. The synthesis was carried out by applying a shell of cell membranes to preliminarily obtained polymeric NPs by extrusion through a polycarbonate membrane with a pore size of 400 nm. The resulting ROS-responsive biomimetic NPs had a small hydrodynamic size with a negative surface charge, retained functional proteins from macrophage membranes, and demonstrated ROS-responsive drug release. Due to cell membrane camouflage, the NPs were able to effectively avoid capture by macrophages and target inflammatory endothelial cells. In addition, such BNMs have demonstrated the ability to inhibit the proliferation of macrophages and smooth muscle cells in vitro without significant cytotoxicity.
A summary of the above and additional examples of BNMs based on polymer, ceramic, and mixed components is shown in Table 3.
As one of the most intensively developing classes of nanomaterials, hybrid BNMs have attracted considerable attention, especially toward the development of technologies for their formation. By analyzing the data presented in Table 3, it was concluded that there were two main groups of hybrid BNMs: the first one included BNMs based on biomimetic nanoceramics, and the second one was based on the use of biogenic or artificial cell membranes with organic (polymeric) or inorganic functional cores. We believe that both groups of BNMs can be considered as equally prospective for biomedical use.
Biomimetic nanoceramics have been obtained using various methods, including Al2O3 NP repeated coagulation with ferrofluid under a magnetic field following sintering [36], nonstoichiometric silicon nitride (SixNy) deposition on both sides of a silicon wafer using low-pressure chemical vapor deposition [142], self-assembly of a layered chitosan–maleic acid matrix followed by monetite mineralization and transformation to HAP [147], and alumina powder addition to a chitosan solution followed by alginate dissolution and genipin (cross-linking agent) addition. The main proposed application of these biomimetic nanoceramics has been tissue engineering for orthopedic and dentistry purposes.
Membrane-camouflaged NPs have usually been synthesized, for example, via drug-encapsulating PLGA prepared via nanoprecipitation with a consequent coating with cancer cell membranes [45], RBC hypotonic treatment and extrusion followed by mixing with PLGA NPs via extrusion through a porous membrane [66], stretching of spherical PLGA NPs immobilized on a PVA-glycerol film followed by sonication-assisted coating with ultrasound-derived RBC membranes [33], and biodegradable NPs conjugated with (3-aminopropyl)triethoxysilane followed by incubation with proteolipid solution [144]. Such BNMs have been mainly proposed for the development of drug delivery carriers.

6. Summary

The scientific literature reviewed was summarized based on the application areas of BNMs of the three considered main groups. The distribution of the applications of BNMs indicated in Table 1, Table 2 and Table 3 is shown in Figure 14. The diagram shows that biomedical applications of such materials are currently predominant.
The main applications included tasks related to the use of BNMs inside the human body: the restoration of damaged tooth enamel and the restoration of bone tissue; magnetic and infrared hyperthermia; chemotherapy and immunotherapy for oncological diseases; the development of new drugs for targeted therapy; and antibacterial and anti-inflammatory therapy and bioimaging. It can be assumed that, due to the high technological complexity and production costs of BNMs, their practical uses are justified, first of all, to treat socially significant infectious and noninfectious diseases.
From our point of view, BNM technologies based on the uses of natural (native), genetically modified, or completely artificial phospholipid membranes are currently of the highest demand in biomedical applications. Such constructs are able to combine the physicochemical and biological properties of biogenic prototypes (for example, bacterial magnetosomes) with high biocompatibility (due to the absence of microbial toxins), economic availability (expensive biotechnological synthesis is not required), and scalability for the production of synthetic nanomaterials. The most frequently used inorganic bases (cores) for obtaining such biomimetic nanostructures include (i) magnetite, (ii) hydroxyapatite, (iii) Au and Ag, and their combinations due to in-demand applications in the fields of drug delivery, tissue engineering, and antibacterial therapy. The most frequently obtained organic and hybrid cores of BNMs include (i) metal–organic frameworks, (ii) poly(lactic acid-glycolic acid), and (iii) polyamidoamine dendrimers due to applications in nanovaccines, anti-inflammatory therapy, and biocatalysis.
Finally, by analyzing the most recent literary sources, it was possible to list the main challenges that stand in the way of the development of BNM technology: (i) there remains much room for the improvement of BNMs in terms of their stability and biocompatibility since BNMs have been used only in animal studies, but they have not been widely used in clinical practice [22]; (ii) understanding the energetic contributions that rule interactions at the nano–bio interface (i.e., where NPs meet biological barriers, specifically cell membranes) is very complex due to the high compositional heterogeneity of biomembranes and the intrinsic variability of the biological environment [34]; (iii) the clinical application of BNMs encounters complex fabrication processes, unsuitable large-scale production, low yields, and difficult preservation [24]; (iv) the utilization of biosynthesis (e.g., engineered bacteria) requires scaled-up manufacturing, dose determination, and potential biosafety studies [23]; (v) the processing of biomaterials (e.g., bioprinting) that incorporate living cells is still very challenging, especially considering that the production of mechanically rigid and insoluble substrates usually requires nonbiocompatible processes, such as chemical cross-linking or sintering [65].

Author Contributions

K.G.G., Conceptualization and Writing—Original Draft; D.S.G., Writing—Review and Editing; V.V.K., Writing—Review and Editing; N.O.S., Writing—Review and Editing; T.M.Z., Writing—Review and Editing; H.G. and M.S., Funding Acquisition and Writing—Review and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Russian Science Foundation (Grant Number 22-15-00240). H.G. was supported by the National Natural Science Foundation of China (81961138009).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zan, G.; Wu, Q. Biomimetic and Bioinspired Synthesis of Nanomaterials/Nanostructures. Adv. Mater. 2016, 28, 2099–2147. [Google Scholar] [CrossRef] [PubMed]
  2. Sheikhpour, M.; Barani, L.; Kasaeian, A. Biomimetics in drug delivery systems: A critical review. J. Control. Release 2017, 253, 97–109. [Google Scholar] [CrossRef] [PubMed]
  3. Das, D.; Noh, I. Biomimetic Medical Materials: From Nanotechnology to 3D Bioprinting; Springer: Berlin/Heidelberg, Germany, 2018; ISBN 9789811304446. [Google Scholar]
  4. Ruys, A.J. Biomimetic Biomaterials; Elsevier: Amsterdam, The Netherlands, 2013; ISBN 9780857092335. [Google Scholar]
  5. Poupon, E.; Nay, B. Biomimetic Organic Synthesis; Wiley: Hoboken, NJ, USA, 2011; Volume 1–2, ISBN 9783527325801. [Google Scholar]
  6. Moreira, F.T.C.; Guerreiro, J.R.L.; Brandão, L.; Sales, M.G.F. Synthesis of molecular biomimetics. In Biomimetic Technologies: Principles and Applications; Elsevier: Amsterdam, The Netherlands, 2015; pp. 3–31. ISBN 9780081002605. [Google Scholar]
  7. Takai, O. Biomimetic nanotechnology. Ann. N. Y. Acad. Sci. 2006, 1093, 84–97. [Google Scholar] [CrossRef] [PubMed]
  8. Garg, P.; Ghatmale, P.; Tarwadi, K.; Chavan, S. Influence of nanotechnology and the role of nanostructures in biomimetic studies and their potential applications. Biomimetics 2017, 2, 7. [Google Scholar] [CrossRef] [Green Version]
  9. Fatima, A.; Yasir, S.; Khan, M.S.; Manan, S.; Ullah, M.W.; Ul-Islam, M. Plant extract-loaded bacterial cellulose composite membrane for potential biomedical applications. J. Bioresour. Bioprod. 2021, 6, 26–32. [Google Scholar] [CrossRef]
  10. Saad, E.M.; Elshaarawy, R.F.; Mahmoud, S.A.; El-Moselhy, K.M. New Ulva lactuca Algae Based Chitosan Bio-composites for Bioremediation of Cd(II) Ions. J. Bioresour. Bioprod. 2021, 6, 223–242. [Google Scholar] [CrossRef]
  11. Chattopadhyay, S. Biomimetic Architectures by Plasma Processing: Fabrication and Applications; CRC Press: Boca Raton, FL, USA, 2014; ISBN 9789814463959. [Google Scholar]
  12. Ke, P.; Jiao, X.N.; Ge, X.H.; Xiao, W.M.; Yu, B. From macro to micro: Structural biomimetic materials by electrospinning. RSC Adv. 2014, 4, 39704–39724. [Google Scholar] [CrossRef]
  13. Vincent, J.F.V. Biomimetic materials. J. Mater. Res. 2008, 23, 3140–3147. [Google Scholar] [CrossRef]
  14. Gagliardi, M. Biomimetic and bioinspired nanoparticles for targeted drug delivery. Ther. Deliv. 2017, 8, 289–299. [Google Scholar] [CrossRef]
  15. Parodi, A.; Molinaro, R.; Sushnitha, M.; Evangelopoulos, M.; Martinez, J.O.; Arrighetti, N.; Corbo, C.; Tasciotti, E. Bio-inspired engineering of cell- and virus-like nanoparticles for drug delivery. Biomaterials 2017, 147, 155–168. [Google Scholar] [CrossRef]
  16. Jin, K.; Luo, Z.; Zhang, B.; Pang, Z. Biomimetic nanoparticles for inflammation targeting. Acta Pharm. Sin. B 2018, 8, 23–33. [Google Scholar] [CrossRef]
  17. Unnithan, A.R.; Sasikala, A.R.K.; Park, C.H.; Kim, C.S. Biomimetic Nanoengineered Materials for Advanced Drug Delivery; Elsevier: Amsterdam, The Netherlands, 2019; ISBN 9780128149447. [Google Scholar]
  18. Akagi, T.; Cabral, H.; Mi, P. Bio-inspired nanomaterials for biomedical innovation. Sci. Technol. Adv. Mater. 2020, 21, 420–421. [Google Scholar] [CrossRef]
  19. Wang, H.; Liu, Y.; He, R.; Xu, D.; Zang, J.; Weeranoppanant, N.; Dong, H.; Li, Y. Cell membrane biomimetic nanoparticles for inflammation and cancer targeting in drug delivery. Biomater. Sci. 2020, 8, 552–568. [Google Scholar] [CrossRef]
  20. Yang, L.; Zang, G.; Li, J.; Li, X.; Li, Y.; Zhao, Y. Cell-derived biomimetic nanoparticles as a novel drug delivery system for atherosclerosis: Predecessors and perspectives. Regen. Biomater. 2020, 7, 349–358. [Google Scholar] [CrossRef]
  21. Zhao, Y.; Li, A.; Jiang, L.; Gu, Y.; Liu, J. Hybrid Membrane-Coated Biomimetic Nanoparticles (HM@BNPs): A Multifunctional Nanomaterial for Biomedical Applications. Biomacromolecules 2021, 22, 3149–3167. [Google Scholar] [CrossRef]
  22. Feng, Z.; Fan, H.; Cheng, L.; Zhang, H.; Fan, H.; Liu, J. Advanced Biomimetic Nanomaterials for Non-invasive Disease Diagnosis. Front. Mater. 2021, 8, 664795. [Google Scholar] [CrossRef]
  23. Zhu, L.; Zhong, Y.; Wu, S.; Yan, M.; Cao, Y.; Mou, N.; Wang, G.; Sun, D.; Wu, W. Cell membrane camouflaged biomimetic nanoparticles: Focusing on tumor theranostics. Mater. Today Bio. 2022, 14, 100228. [Google Scholar] [CrossRef]
  24. Zhao, X.; Yan, C. Research Progress of Cell Membrane Biomimetic Nanoparticles for Tumor Therapy. Nanoscale Res. Lett. 2022, 17, 36. [Google Scholar] [CrossRef]
  25. Liu, R.; An, Y.; Jia, W.; Wang, Y.; Wu, Y.; Zhen, Y.; Cao, J.; Gao, H. Macrophage-mimic shape changeable nanomedicine retained in tumor for multimodal therapy of breast cancer. J. Control. Release 2020, 321, 589–601. [Google Scholar] [CrossRef]
  26. Yu, W.; He, X.; Yang, Z.; Yang, X.; Xiao, W.; Liu, R.; Xie, R.; Qin, L.; Gao, H. Sequentially responsive biomimetic nanoparticles with optimal size in combination with checkpoint blockade for cascade synergetic treatment of breast cancer and lung metastasis. Biomaterials 2019, 217, 119309. [Google Scholar] [CrossRef]
  27. Stratakis, E.; Ranella, A.; Fotakis, C. Biomimetic micro/nanostructured functional surfaces for microfluidic and tissue engineering applications. Biomicrofluidics 2011, 5, 013411. [Google Scholar] [CrossRef] [Green Version]
  28. Mano, J.F. Biomimetic Approaches for Biomaterials Development; Wiley: Hoboken, NJ, USA, 2012; ISBN 9783527329168. [Google Scholar]
  29. Pasha, M.; Muhammad, N.; Shahnawaz, S.; Najmi, Y.; Shahroz, N.; Liaqat, S. Ceramic Nanomaterials in Dental Applications. In Nanoengineering of Biomaterials; Wiley: Hoboken, NJ, USA, 2022; pp. 123–144. [Google Scholar] [CrossRef]
  30. Narayan, R.J.; Kumta, P.N.; Wagner, W.R. Advances in Biomedical and Biomimetic Materials; Wiley: Hoboken, NJ, USA, 2008; ISBN 9780470538357. [Google Scholar]
  31. Sushnitha, M.; Evangelopoulos, M.; Tasciotti, E.; Taraballi, F. Cell Membrane-Based Biomimetic Nanoparticles and the Immune System: Immunomodulatory Interactions to Therapeutic Applications. Front. Bioeng. Biotechnol. 2020, 8, 627. [Google Scholar] [CrossRef]
  32. Peigneux, A.; Jabalera, Y.; Vivas, M.A.F.; Casares, S.; Azuaga, A.I.; Jimenez-Lopez, C. Tuning properties of biomimetic magnetic nanoparticles by combining magnetosome associated proteins. Sci. Rep. 2019, 9, 8804. [Google Scholar] [CrossRef]
  33. Ben-Akiva, E.; Meyer, R.A.; Yu, H.; Smith, J.T.; Pardoll, D.M.; Green, J.J. Biomimetic anisotropic polymeric nanoparticles coated with red blood cell membranes for enhanced circulation and toxin removal. Sci. Adv. 2020, 6, eaay9035. [Google Scholar] [CrossRef]
  34. Cardellini, J.; Montis, C.; Barbero, F.; De Santis, I.; Caselli, L.; Berti, D. Interaction of Metallic Nanoparticles with Biomimetic Lipid Liquid Crystalline Cubic Interfaces. Front. Bioeng. Biotechnol. 2022, 10, 848687. [Google Scholar] [CrossRef]
  35. Mondal, S.; Manivasagan, P.; Bharathiraja, S.; Moorthy, M.S.; Kim, H.H.; Seo, H.; Lee, K.D.; Oh, J. Magnetic hydroxyapatite: A promising multifunctional platform for nanomedicine application. Int. J. Nanomed. 2017, 12, 8389–8410. [Google Scholar] [CrossRef] [Green Version]
  36. Bargardi, F.L.; Le Ferrand, H.; Libanori, R.; Studart, A.R. Bio-inspired self-shaping ceramics. Nat. Commun. 2016, 7, 13912. [Google Scholar] [CrossRef]
  37. Correa, S.; Puertas, S.; Gutiérrez, L.; Asín, L.; De La Fuente, J.M.; Grazú, V.; Betancor, L. Design of stable magnetic hybrid nanoparticles of Si-entrapped HRP. PLoS ONE 2019, 14, e0214004. [Google Scholar] [CrossRef] [PubMed]
  38. Al-Zubaidi, S.M.; Madfa, A.A.; Mufadhal, A.A.; Aldawla, M.A.; Hameed, O.S.; Yue, X.G. Improvements in Clinical Durability from Functional Biomimetic Metallic Dental Implants. Front. Mater. 2020, 7, 106. [Google Scholar] [CrossRef]
  39. Liu, Y.; Liu, X.; Wang, X. Biomimetic Synthesis of Gelatin Polypeptide-Assisted Noble-Metal Nanoparticles and Their Interaction Study. Nanoscale Res. Lett. 2011, 6, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Bhardwaj, D.; Singh, R. Green biomimetic synthesis of Ag–TiO2 nanocomposite using Origanum majorana leaf extract under sonication and their biological activities. Bioresour. Bioprocess. 2021, 8, 1. [Google Scholar] [CrossRef]
  41. Mukherjee, K.; Ruan, Q.; Nutt, S.; Tao, J.; De Yoreo, J.J.; Moradian-Oldak, J. Peptide-Based Bioinspired Approach to Regrowing Multilayered Aprismatic Enamel. ACS Omega 2018, 3, 2546–2557. [Google Scholar] [CrossRef] [Green Version]
  42. Wen, X.; Xiao, Z.; Jiang, T.; Li, J.; Zhang, W.; Zhang, L.; Shao, H. Constructing novel fiber reinforced plastic (FRP) composites through a biomimetic approach: Connecting glass fiber with nanosized boron nitride by polydopamine coating. J. Nanomater. 2013, 2013, 470583. [Google Scholar] [CrossRef]
  43. Yu, H.; Li, J.; Li, J.; Chen, Y.; Hou, X.; Chen, S.; Yang, H. Biomimetic preparation of a ceramic combined with sea urchin stereom structure and nacre mineral bridge structure. Mater. Des. 2019, 178, 107844. [Google Scholar] [CrossRef]
  44. Mickoleit, F. Generation of nanomagnetic biocomposites by genetic engineering of bacterial magnetosomes. Bioinspired, Biomim. Nanobiomaterials 2019, 8, 86–98. [Google Scholar] [CrossRef] [Green Version]
  45. Jin, J.; Bhujwalla, Z.M. Biomimetic Nanoparticles Camouflaged in Cancer Cell Membranes and Their Applications in Cancer Theranostics. Front. Oncol. 2020, 9, 1560. [Google Scholar] [CrossRef] [Green Version]
  46. Tang, D.; Wang, Y.; Wijaya, A.; Liu, B.; Maruf, A.; Wang, J.; Xu, J.; Liao, X.; Wu, W.; Wang, G. ROS-responsive biomimetic nanoparticles for potential application in targeted anti-atherosclerosis. Regen. Biomater. 2021, 8, rbab033. [Google Scholar] [CrossRef]
  47. Wang, D.; Chen, L.; Li, C.; Long, Q.; Yang, Q.; Huang, A.; Tang, H. CRISPR/Cas9 delivery by NIR-responsive biomimetic nanoparticles for targeted HBV therapy. J. Nanobiotechnol. 2022, 20, 27. [Google Scholar] [CrossRef]
  48. Peigneux, A.; Glitscher, E.A.; Charbaji, R.; Weise, C.; Wedepohl, S.; Calderón, M.; Jimenez-Lopez, C.; Hedtrich, S. Protein corona formation and its influence on biomimetic magnetite nanoparticles. J. Mater. Chem. B 2020, 8, 4870–4882. [Google Scholar] [CrossRef]
  49. Abdeen, M.; Sabry, S.; Ghozlan, H.; El-Gendy, A.A.; Carpenter, E.E. Microbial-Physical Synthesis of Fe and Fe3O4 Magnetic Nanoparticles Using Aspergillus Niger YESM1 and Supercritical Condition of Ethanol. J. Nanomater. 2016, 2016, 9174891. [Google Scholar] [CrossRef] [Green Version]
  50. Santana, S.D.F.; Dhadge, V.L.; Roque, A.C.A. Dextran-coated magnetic supports modified with a biomimetic ligand for IgG purification. ACS Appl. Mater. Interfaces 2012, 4, 5907–5914. [Google Scholar] [CrossRef]
  51. Del Bianco, L.; Lesci, I.G.; Fracasso, G.; Barucca, G.; Spizzo, F.; Tamisari, M.; Scotti, R.; Ciocca, L. Synthesis of nanogranular Fe3O4/biomimetic hydroxyapatite for potential applications in nanomedicine: Structural and magnetic characterization. Mater. Res. Express 2015, 2, 65002. [Google Scholar] [CrossRef]
  52. Yang, C.; Cao, C.; Cai, Y.; Xu, H.; Zhang, T.; Pan, Y. Effects of PEGylation on biomimetic synthesis of magnetoferritin nanoparticles. J. Nanopart. Res. 2017, 19, 101. [Google Scholar] [CrossRef]
  53. Samal, S.K.; Dash, M.; Shelyakova, T.; Declercq, H.A.; Uhlarz, M.; Bañobre-López, M.; Dubruel, P.; Cornelissen, M.; Herrmannsdörfer, T.; Rivas, J.; et al. Biomimetic magnetic silk scaffolds. ACS Appl. Mater. Interfaces 2015, 7, 6282–6292. [Google Scholar] [CrossRef]
  54. Jiang, Q.; Wang, K.; Zhang, X.; Ouyang, B.; Liu, H.; Pang, Z.; Yang, W. Platelet Membrane-Camouflaged Magnetic Nanoparticles for Ferroptosis-Enhanced Cancer Immunotherapy. Small 2020, 16, e2001704. [Google Scholar] [CrossRef]
  55. Singh, M.; Renu; Kumar, V.; Upadhyay, S.K.; Singh, R.; Yadav, M.; Seema; Kumari, S.; Sharma, A.K.; Manikanadan, S. Biomimetic synthesis of silver nanoparticles from aqueous extract of saraca indica and its profound antibacterial activity. Biointerface Res. Appl. Chem. 2021, 11, 8110–8120. [Google Scholar] [CrossRef]
  56. Sarker, N.H.; Barnaby, S.N.; Dowdell, A.P.; Nakatsuka, N.; Banerjee, I.A. Biomimetic formation of Pd and Au-Pd nanocomposites and their catalytic applications. Soft Mater. 2013, 11, 403–413. [Google Scholar] [CrossRef]
  57. Slocik, J.M.; Stone, M.O.; Naik, R.R. Synthesis of gold nanoparticles using multifunctional peptides. Small 2005, 1, 1048–1052. [Google Scholar] [CrossRef] [PubMed]
  58. Min, H.; Wang, J.; Qi, Y.; Zhang, Y.; Han, X.; Xu, Y.; Xu, J.; Li, Y.; Chen, L.; Cheng, K.; et al. Biomimetic Metal–Organic Framework Nanoparticles for Cooperative Combination of Antiangiogenesis and Photodynamic Therapy for Enhanced Efficacy. Adv. Mater. 2019, 31, e1808200. [Google Scholar] [CrossRef] [PubMed]
  59. Xie, J.; Lee, J.Y.; Wang, D.I.C.; Ting, Y.P. Identification of active biomolecules in the high-yield synthesis of single-crystalline gold nanoplates in algal solutions. Small 2007, 3, 672–682. [Google Scholar] [CrossRef] [PubMed]
  60. Mallampati, R.; Valiyaveettil, S. Biomimetic metal oxides for the extraction of nanoparticles from water. Nanoscale 2013, 5, 3395–3399. [Google Scholar] [CrossRef]
  61. Gruber, S.; Zollfrank, C. Noble metal nanoparticles on biotemplated nanowires. Bioinspired Biomim. Nanobiomater. 2012, 1, 95–100. [Google Scholar] [CrossRef]
  62. Banerjee, P.; Das, J. Biomimetic synthesis of nanocrystalline hydroxyapatite from sharkskin collagen. Bioinspired Biomim. Nanobiomater. 2017, 7, 27–36. [Google Scholar] [CrossRef]
  63. Chung, H.Y.; Li, C.C. Microstructure and nanomechanical properties of enamel remineralized with asparagine-serine-serine peptide. Mater. Sci. Eng. C 2013, 33, 969–973. [Google Scholar] [CrossRef]
  64. Fu, C.; Ravindra, N.M. Magnetic iron oxide nanoparticles: Synthesis and applications. Bioinspired Biomim. Nanobiomater. 2012, 1, 229–244. [Google Scholar] [CrossRef]
  65. Mainardi, J.C.; Rezwan, K.; Maas, M. Genipin-crosslinked chitosan/alginate/alumina nanocomposite gels for 3D bioprinting. Bioprocess Biosyst. Eng. 2022, 45, 171–185. [Google Scholar] [CrossRef]
  66. Hu, C.M.J.; Zhang, L.; Aryal, S.; Cheung, C.; Fang, R.H.; Zhang, L. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc. Natl. Acad. Sci. USA 2011, 108, 10980–10985. [Google Scholar] [CrossRef] [Green Version]
  67. Molinaro, R.; Evangelopoulos, M.; Hoffman, J.R.; Corbo, C.; Taraballi, F.; Martinez, J.O.; Hartman, K.A.; Cosco, D.; Costa, G.; Romeo, I.; et al. Design and Development of Biomimetic Nanovesicles Using a Microfluidic Approach. Adv. Mater. 2018, 30, e1702749. [Google Scholar] [CrossRef]
  68. Singh, D.; Singh, D.; Zo, S.; Han, S.S. Nano-biomimetics for nano/micro tissue regeneration. J. Biomed. Nanotechnol. 2014, 10, 3141–3161. [Google Scholar] [CrossRef]
  69. Bharathiraja, S.; Bui, N.Q.; Manivasagan, P.; Moorthy, M.S.; Mondal, S.; Seo, H.; Phuoc, N.T.; Vy Phan, T.T.; Kim, H.; Lee, K.D.; et al. Multimodal tumor-homing chitosan oligosaccharide-coated biocompatible palladium nanoparticles for photo-based imaging and therapy. Sci. Rep. 2018, 8, 500. [Google Scholar] [CrossRef]
  70. Albert, K.; Huang, X.C.; Hsu, H.Y. Bio-templated silica composites for next-generation biomedical applications. Adv. Colloid Interface Sci. 2017, 249, 272–289. [Google Scholar] [CrossRef]
  71. Yang, Y.; Zhang, M.; Song, H.; Yu, C. Silica-Based Nanoparticles for Biomedical Applications: From Nanocarriers to Biomodulators. Acc. Chem. Res. 2020, 53, 1545–1556. [Google Scholar] [CrossRef]
  72. Ghanbari, H.; de Mel, A.; Seifalian, A.M. Cardiovascular application of polyhedral oligomeric silsesquioxane nanomaterials: A glimpse into prospective horizons. Int. J. Nanomed. 2011, 6, 775–786. [Google Scholar] [CrossRef] [Green Version]
  73. Hasanzadeh, M.; Babaie, P.; Jouyban-Gharamaleki, V.; Jouyban, A. The use of chitosan as a bioactive polysaccharide in non-invasive detection of malondialdehyde biomarker in human exhaled breath condensate: A new platform towards diagnosis of some lung disease. Int. J. Biol. Macromol. 2018, 120, 2482–2492. [Google Scholar] [CrossRef]
  74. Liang, K.; Chen, H. Protein-based nanoplatforms for tumor imaging and therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnology 2020, 12, e1616. [Google Scholar] [CrossRef]
  75. Sharipov, M.; Tawfik, S.M.; Gerelkhuu, Z.; Huy, B.T.; Lee, Y.I. Phospholipase A2-Responsive Phosphate Micelle-Loaded UCNPs for Bioimaging of Prostate Cancer Cells. Sci. Rep. 2017, 7, 16073. [Google Scholar] [CrossRef] [Green Version]
  76. Renu, S.; Shivashangari, K.S.; Ravikumar, V. Incorporated plant extract fabricated silver/poly-D,L-lactide-co-glycolide nanocomposites for antimicrobial based wound healing. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 228, 117673. [Google Scholar] [CrossRef]
  77. Zhao, W.; Li, A.; Zhang, A.; Zheng, Y.; Liu, J. Recent Advances in Functional-Polymer-Decorated Transition-Metal Nanomaterials for Bioimaging and Cancer Therapy. ChemMedChem 2018, 13, 2134–2149. [Google Scholar] [CrossRef] [PubMed]
  78. Wu, K.; Su, D.; Liu, J.; Saha, R.; Wang, J.P. Magnetic nanoparticles in nanomedicine: A review of recent advances. Nanotechnology 2019, 30, 502003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Knežević, N.; Gadjanski, I.; Durand, J.O. Magnetic nanoarchitectures for cancer sensing, imaging and therapy. J. Mater. Chem. B 2019, 7, 9–23. [Google Scholar] [CrossRef] [PubMed]
  80. Poon, C.; Gallo, J.; Joo, J.; Chang, T.; Bañobre-López, M.; Chung, E.J. Hybrid, metal oxide-peptide amphiphile micelles for molecular magnetic resonance imaging of atherosclerosis. J. Nanobiotechnol. 2018, 16, 92. [Google Scholar] [CrossRef] [Green Version]
  81. Lee, S.H.; Jun, B.H. Silver nanoparticles: Synthesis and application for nanomedicine. Int. J. Mol. Sci. 2019, 20, 865. [Google Scholar] [CrossRef] [Green Version]
  82. Klem, M.T.; Young, M.; Douglas, T. Biomimetic magnetic nanoparticles. Mater. Today 2005, 8, 28–37. [Google Scholar] [CrossRef]
  83. Altan, C.L. Biomimetic synthesis, Magnetic Properties and Applications of Magnetite Nanoparticles; Technische Universiteit Eindhoven: Eindhoven, The Netherlands, 2014; ISBN 978-94-6108-843-7. [Google Scholar]
  84. Kralj, S.; Marchesan, S. Bioinspired Magnetic Nanochains for Medicine. Pharmaceutics 2021, 13, 1262. [Google Scholar] [CrossRef]
  85. Vurro, F.; Jabalera, Y.; Mannucci, S.; Glorani, G.; Sola-Leyva, A.; Gerosa, M.; Romeo, A.; Romanelli, M.G.; Malatesta, M.; Calderan, L.; et al. Improving the cellular uptake of biomimetic magnetic nanoparticles. Nanomaterials 2021, 11, 766. [Google Scholar] [CrossRef]
  86. Jabalera, Y.; Sola-Leyva, A.; Carrasco-Jiménez, M.P.; Iglesias, G.R.; Jimenez-Lopez, C. Synergistic photothermal-chemotherapy based on the use of biomimetic magnetic nanoparticles. Pharmaceutics 2021, 13, 625. [Google Scholar] [CrossRef]
  87. Salem, K.; Jabalera, Y.; Puentes-Pardo, J.D.; Vilchez-Garcia, J.; Sayari, A.; Hmida-Sayari, A.; Jimenez-Lopez, C.; Perduca, M. Enzyme Storage and Recycling: Nanoassemblies of α-Amylase and Xylanase Immobilized on Biomimetic Magnetic Nanoparticles. ACS Sustain. Chem. Eng. 2021, 9, 4054–4063. [Google Scholar] [CrossRef]
  88. Gareev, K.G.; Grouzdev, D.S.; Kharitonskii, P.V.; Kosterov, A.; Koziaeva, V.V.; Sergienko, E.S.; Shevtsov, M.A. Magnetotactic bacteria and magnetosomes: Basic properties and applications. Magnetochemistry 2021, 7, 86. [Google Scholar] [CrossRef]
  89. Taher, Z.; Legge, C.; Winder, N.; Lysyganicz, P.; Rawlings, A.; Bryant, H.; Muthana, M.; Staniland, S. Magnetosomes and magnetosome mimics: Preparation, cancer cell uptake and functionalization for future cancer therapies. Pharmaceutics 2021, 13, 367. [Google Scholar] [CrossRef]
  90. Goyal, G.; Bhakta, S.; Mishra, P. Surface Molecularly Imprinted Biomimetic Magnetic Nanoparticles for Enantioseparation. ACS Appl. Nano Mater. 2019, 2, 6747–6756. [Google Scholar] [CrossRef]
  91. Lenders, J.J.M.; Bawazer, L.A.; Green, D.C.; Zope, H.R.; Bomans, P.H.H.; de With, G.; Kros, A.; Meldrum, F.C.; Sommerdijk, N.A.J.M. Combinatorial Evolution of Biomimetic Magnetite Nanoparticles. Adv. Funct. Mater. 2017, 27, 1604863. [Google Scholar] [CrossRef] [Green Version]
  92. Bu, Y.; Hu, Q.; Ke, R.; Sui, Y.; Xie, X.; Wang, S. Cell membrane camouflaged magnetic nanoparticles as a biomimetic drug discovery platform. Chem. Commun. 2018, 54, 13427–13430. [Google Scholar] [CrossRef]
  93. Ren, Y.; Rivera, J.G.; He, L.; Kulkarni, H.; Lee, D.K.; Messersmith, P.B. Facile, high efficiency immobilization of lipase enzyme on magnetic iron oxide nanoparticles via a biomimetic coating. BMC Biotechnol. 2011, 11, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Tengood, J.E.; Alferiev, I.S.; Zhang, K.; Fishbein, I.; Levy, R.J.; Chorny, M. Real-time analysis of composite magnetic nanoparticle disassembly in vascular cells and biomimetic media. Proc. Natl. Acad. Sci. USA 2014, 111, 4245–4250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Zhang, T.; Li, F.; Xu, Q.; Wang, Q.; Jiang, X.; Liang, Z.; Liao, H.; Kong, X.; Liu, J.; Wu, H.; et al. Ferrimagnetic Nanochains-Based Mesenchymal Stem Cell Engineering for Highly Efficient Post-Stroke Recovery. Adv. Funct. Mater. 2019, 29, 1900603. [Google Scholar] [CrossRef]
  96. Deravi, L.F.; Swartz, J.D.; Wright, D.W. The Biomimetic Synthesis of Metal Oxide Nanomaterials; Wiley: Hoboken, NJ, USA, 2010; Volume 2, ISBN 9783527321520. [Google Scholar]
  97. Prasad, R.; Kumar, M.; Kumar, V. Nanotechnology: An Agricultural Paradigm; Springer: Berlin/Heidelberg, Germany, 2017; ISBN 9789811045738. [Google Scholar]
  98. Singh, S.; Abbasi, B.H. Biomimetic synthesis of antimicrobial silver nanoparticles using in vitro-propagated plantlets of a medicinally important endangered species: Phlomis bracteosa. Int. J. Nanomed. 2016, 11, 1663–1675. [Google Scholar] [CrossRef] [Green Version]
  99. Bag, S.S.; Bora, A.; Golder, A.K. Biomimetic Synthesis of Silver Nanoparticles Using Bhimkol (Musa balbisiana) Peel Extract as Biological Waste: Its Antibacterial Activity and Role of Ripen Stage of the Peel. Curr. Nanomater. 2020, 5, 47–65. [Google Scholar] [CrossRef]
  100. Le Guéhennec, L.; Soueidan, A.; Layrolle, P.; Amouriq, Y. Surface treatments of titanium dental implants for rapid osseointegration. Dent. Mater. 2007, 23, 844–854. [Google Scholar] [CrossRef]
  101. Chen, H.; Qiu, Q.; Sharif, S.; Ying, S.; Wang, Y.; Ying, Y. Solution-Phase Synthesis of Platinum Nanoparticle-Decorated Metal-Organic Framework Hybrid Nanomaterials as Biomimetic Nanoenzymes for Biosensing Applications. ACS Appl. Mater. Interfaces 2018, 10, 24108–24115. [Google Scholar] [CrossRef]
  102. Azam, T.; Pervaiz, E.; Farrukh, S.; Noor, T. Biomimetic highly hydrophobic stearic acid functionalized MOF sponge for efficient oil/water separation. Mater. Res. Express 2021, 8, 015019. [Google Scholar] [CrossRef]
  103. Bim-Junior, O.; Alania, Y.; Tabatabaei, F.S.; Frem, R.; Bedran-Russo, A.K.; Lisboa-Filho, P.N. Biomimetic Growth of Metal–Organic Frameworks for the Stabilization of the Dentin Matrix and Control of Collagenolysis. Langmuir 2022, 38, 1600–1610. [Google Scholar] [CrossRef]
  104. Chen, K.; Wu, C. De Designed fabrication of biomimetic metal–organic frameworks for catalytic applications. Coord. Chem. Rev. 2019, 378, 445–465. [Google Scholar] [CrossRef]
  105. Cheng, X.; Zhang, S.; Liu, H.; Chen, H.; Zhou, J.; Chen, Z.; Zhou, X.; Xie, Z.; Kuang, Q.; Zheng, L. Biomimetic Metal-Organic Framework Composite-Mediated Cascade Catalysis for Synergistic Bacteria Killing. ACS Appl. Mater. Interfaces 2020, 12, 36996–37005. [Google Scholar] [CrossRef]
  106. Li, S.F.; Chen, Y.; Wang, Y.S.; Mo, H.L.; Zang, S.Q. Integration of enzyme immobilization and biomimetic catalysis in hierarchically porous metal-organic frameworks for multi-enzymatic cascade reactions. Sci. China Chem. 2022, 65, 1122–1128. [Google Scholar] [CrossRef]
  107. Gu, Z.Y.; Park, J.; Raiff, A.; Wei, Z.; Zhou, H.C. Metal-organic frameworks as biomimetic catalysts. ChemCatChem 2014, 6, 67–75. [Google Scholar] [CrossRef]
  108. Liang, K.; Ricco, R.; Doherty, C.M.; Styles, M.J.; Bell, S.; Kirby, N.; Mudie, S.; Haylock, D.; Hill, A.J.; Doonan, C.J.; et al. Biomimetic mineralization of metal-organic frameworks as protective coatings for biomacromolecules. Nat. Commun. 2015, 6, 4–11. [Google Scholar] [CrossRef] [Green Version]
  109. Singh, R.; White, J.F.; de Vries, M.; Beddome, G.; Dai, M.; Bean, A.G.; Mulet, X.; Layton, D.; Doherty, C.M. Biomimetic metal-organic frameworks as protective scaffolds for live-virus encapsulation and vaccine stabilization. Acta Biomater. 2022, 142, 320–331. [Google Scholar] [CrossRef]
  110. Wang, X.N.; Zhao, Y.; Li, J.L.; Pang, J.D.; Wang, Q.; Li, B.; Zhou, H.C. Biomimetic catalysts of iron-based metal-organic frameworks with high peroxidase-mimicking activity for colorimetric biosensing. Dalt. Trans. 2021, 50, 3854–3861. [Google Scholar] [CrossRef]
  111. Wright, A.M.; Wu, Z.; Zhang, G.; Mancuso, J.L.; Comito, R.J.; Day, R.W.; Hendon, C.H.; Miller, J.T.; Dincă, M. A Structural Mimic of Carbonic Anhydrase in a Metal-Organic Framework. Chem 2018, 4, 2894–2901. [Google Scholar] [CrossRef] [Green Version]
  112. Ma, X.; Zhao, S.; Tian, Z.; Duan, G.; Pan, H.; Yue, Y.; Li, S.; Jian, S.; Yang, W.; Liu, K.; et al. MOFs meet wood: Reusable magnetic hydrophilic composites toward efficient water treatment with super-high dye adsorption capacity at high dye concentration. Chem. Eng. J. 2022, 446, 136851. [Google Scholar] [CrossRef]
  113. Zou, D.; Yu, L.; Sun, Q.; Hui, Y.; Tengjisi; Liu, Y.; Yang, G.; Wibowo, D.; Zhao, C.X. A general approach for biomimetic mineralization of MOF particles using biomolecules. Colloids Surf. B Biointerfaces 2020, 193, 111108. [Google Scholar] [CrossRef]
  114. Wang, J.; Sun, Y.; Zhao, X.; Chen, L.; Peng, S.; Ma, C.; Duan, G.; Liu, Z.; Wang, H.; Yuan, Y.; et al. A poly(amidoxime)-modified MOF macroporous membrane for high-efficient uranium extraction from seawater. e-Polymers 2022, 22, 399–410. [Google Scholar] [CrossRef]
  115. Luo, J.; Wang, X.; Shi, Z.; Zeng, Y.; He, L.; Cao, J.; Sun, Y.; Zhang, T.; Huang, P. Enhancement of antitumor immunotherapy using mitochondria-targeted cancer cell membrane-biomimetic MOF-mediated sonodynamic therapy and checkpoint blockade immunotherapy. J. Nanobiotechnol. 2022, 20, 228. [Google Scholar] [CrossRef]
  116. Aghoutane, Y.; Bari, N.E.; Laghrari, Z.; Bouchikhi, B. Electrochemical Detection of Fenthion Insecticide in Olive Oils by a Sensitive Non-Enzymatic Biomimetic Sensor Enhanced with Metal Nanoparticles. Chem. Proc. 2021, 5, 64. [Google Scholar] [CrossRef]
  117. Yao, Y.; Lan, L.; Liu, X.; Ying, Y.; Ping, J. Spontaneous growth and regulation of noble metal nanoparticles on flexible biomimetic MXene paper for bioelectronics. Biosens. Bioelectron. 2020, 148, 111799. [Google Scholar] [CrossRef]
  118. Wu, K.; Yang, Y.; Zhang, Y.; Deng, J.; Lin, C. Antimicrobial activity and cytocompatibility of silver nanoparticles coated catheters via a biomimetic surface functionalization strategy. Int. J. Nanomed. 2015, 10, 7241–7252. [Google Scholar] [CrossRef] [Green Version]
  119. Tamerler, C.; Sarikaya, M. Molecular biomimetics: Nanotechnology and bionanotechnology using genetically engineered peptides. Philos. Trans. R. Soc. A Math. Phys. Eng. Sci. 2009, 367, 1705–1726. [Google Scholar] [CrossRef] [PubMed]
  120. Li, B.; Wang, F.; Gui, L.; He, Q.; Yao, Y.; Chen, H. The potential of biomimetic nanoparticles for tumor-targeted drug delivery. Nanomedicine 2018, 13, 2099–2118. [Google Scholar] [CrossRef] [PubMed]
  121. Liu, J.; Cai, B.; Cui, L.; Chen, C.L. Peptoid-based hierarchically-structured biomimetic nanomaterials: Synthesis, characterization and applications. Sci. China Mater. 2020, 63, 1099–1112. [Google Scholar] [CrossRef] [Green Version]
  122. Katsanevakis, E. Engineering a Biomimetic Structure for Human Long Bone Regeneration. Ph.D. Thesis, Clemson University, Clemson, SC, USA, 2013; 172p. [Google Scholar]
  123. Taale, M.; Schütt, F.; Carey, T.; Marx, J.; Mishra, Y.K.; Stock, N.; Fiedler, B.; Torrisi, F.; Adelung, R.; Selhuber-Unkel, C. Biomimetic Carbon Fiber Systems Engineering: A Modular Design Strategy to Generate Biofunctional Composites from Graphene and Carbon Nanofibers. ACS Appl. Mater. Interfaces 2019, 11, 5325–5335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Diaz-Rodriguez, P.; Sánchez, M.; Landin, M. Drug-loaded biomimetic ceramics for tissue engineering. Pharmaceutics 2018, 10, 272. [Google Scholar] [CrossRef] [Green Version]
  125. Papagerakis, P. Odontogenesis; Humana Press Inc.: Totowa, NJ, USA, 2019; ISBN 9781493990115. [Google Scholar]
  126. Chan, C.M.; Ruys, A. Biomimetic Design of Haute-Temperature Lightweight Ceramics: A Review. J. Biomimetics, Biomater. Tissue Eng. 2009, 2, 73–93. [Google Scholar] [CrossRef]
  127. Luz, G.M.; Mano, J.F. Biomimetic design of materials and biomaterials inspired by the structure of nacre. Philos. Trans. R. Soc. A Math. Phys. Eng. Sci. 2009, 367, 1587–1605. [Google Scholar] [CrossRef]
  128. Rey, C.; Combes, C.; Drouet, C.; Cazalbou, S.; Grossin, D.; Brouillet, F.; Sarda, S. Surface properties of biomimetic nanocrystalline apatites; Applications in biomaterials. Prog. Cryst. Growth Charact. Mater. 2014, 60, 63–73. [Google Scholar] [CrossRef] [Green Version]
  129. Zhang, H.; Li, S.; Qu, A.; Hao, C.; Sun, M.; Xu, L.; Xu, C.; Kuang, H. Engineering of chiral nanomaterials for biomimetic catalysis. Chem. Sci. 2020, 11, 12937–12954. [Google Scholar] [CrossRef]
  130. Hussain, C.M.; Thomas, S. Handbook of Polymer and Ceramic Nanotechnology; Springer: Berlin/Heidelberg, Germany, 2021; ISBN 9783030405120. [Google Scholar]
  131. Chen, L.; Wei, X.; Sun, Y.; Xue, Y.; Wang, J.; Wu, Q.; Ma, C.; Yang, X.; Duan, G.; Wang, F.; et al. A bamboo/PNIPAM composite hydrogel assembly for both programmable and remotely-controlled light-responsive biomimetic actuations. Chem. Eng. J. 2022, 446, 137072. [Google Scholar] [CrossRef]
  132. Liu, L.; Song, W.; Jiang, S.; Duan, G.; Qin, X. Giving Penetrable Remote-Control Ability to Thermoresponsive Fibrous Composite Actuator with Fast Response Induced by Alternative Magnetic Field. Nanomaterials 2022, 12, 53. [Google Scholar] [CrossRef]
  133. Ruan, Q.; Zhang, Y.; Yang, X.; Nutt, S.; Moradian-Oldak, J. An amelogenin-chitosan matrix promotes assembly of an enamel-like layer with a dense interface. Acta Biomater. 2013, 9, 7289–7297. [Google Scholar] [CrossRef] [Green Version]
  134. Prajapati, S.; Ruan, Q.; Mukherjee, K.; Nutt, S.; Moradian-Oldak, J. The Presence of MMP-20 Reinforces Biomimetic Enamel Regrowth. J. Dent. Res. 2018, 97, 84–90. [Google Scholar] [CrossRef]
  135. Li, J.; Chen, L.; Liang, K.; Li, J.; Wu, D.; Zhou, X. Regeneration of biomimetic hydroxyapatite on etched human enamel by anionic PAMAM template in vitro. Arch. Oral Biol. 2013, 58, 975–980. [Google Scholar] [CrossRef]
  136. Liang, K.; Weir, M.D.; Xie, X.; Wang, L.; Reynolds, M.A.; Li, J.; Xu, H.H.K. Dentin remineralization in acid challenge environment via PAMAM and calcium phosphate composite. Dent. Mater. 2016, 32, 1429–1440. [Google Scholar] [CrossRef] [PubMed]
  137. Li, J.; Yang, J.J.; Li, J.; Chen, L.; Liang, K.; Wu, W.; Chen, X.; Li, J. Bioinspired intrafibrillar mineralization of human dentine by PAMAM dendrimer. Biomaterials 2013, 34, 6738–6747. [Google Scholar] [CrossRef] [PubMed]
  138. Liu, L.; Bai, X.; Martikainen, M.V.; Kårlund, A.; Roponen, M.; Xu, W.; Hu, G.; Tasciotti, E.; Lehto, V.P. Cell membrane coating integrity affects the internalization mechanism of biomimetic nanoparticles. Nat. Commun. 2021, 12, 5726. [Google Scholar] [CrossRef] [PubMed]
  139. Martinez, J.O.; Molinaro, R.; Hartman, K.A.; Boada, C.; Sukhovershin, R.; De Rosa, E.; Kirui, D.; Zhang, S.; Evangelopoulos, M.; Carter, A.M.; et al. Biomimetic nanoparticles with enhanced affinity towards activated endothelium as versatile tools for theranostic drug delivery. Theranostics 2018, 8, 1131–1145. [Google Scholar] [CrossRef]
  140. Engelmann, T.; Desante, G.; Labude, N.; Rütten, S.; Telle, R.; Neuss, S.; Schickle, K. Coatings based on organic/non-organic composites on bioinert ceramics by using biomimetic co-precipitation. Ceramics 2019, 2, 260–270. [Google Scholar] [CrossRef] [Green Version]
  141. Jin, P.; Wang, L.; Sha, R.; Liu, L.; Qian, J.; Ishimwe, N.; Zhang, W.; Qian, J.; Zhang, Y.; Wen, L. A blood circulation-prolonging peptide anchored biomimetic phage-platelet hybrid nanoparticle system for prolonged blood circulation and optimized anti-bacterial performance. Theranostics 2021, 11, 2278–2296. [Google Scholar] [CrossRef]
  142. Jud, C.; Ahmed, S.; Müller, L.; Kinnear, C.; Vanhecke, D.; Umehara, Y.; Frey, S.; Liley, M.; Angeloni, S.; Petri-Fink, A.; et al. Ultrathin Ceramic Membranes as Scaffolds for Functional Cell Coculture Models on a Biomimetic Scale. Biores. Open Access 2015, 4, 457–468. [Google Scholar] [CrossRef]
  143. Li, Z.; Zhang, X.; Liu, C.; Peng, Q.; Wu, Y.; Wen, Y.; Zheng, R.; Yan, Q.; Ma, J. Macrophage-Biomimetic Nanoparticles Ameliorate Ulcerative Colitis through Reducing Inflammatory Factors Expression. J. Innate Immun. 2021, 14, 380–392. [Google Scholar] [CrossRef]
  144. Evangelopoulos, M.; Parodi, A.; Martinez, J.O.; Yazdi, I.K.; Cevenini, A.; van de Ven, A.L.; Quattrocchi, N.; Boada, C.; Taghipour, N.; Corbo, C.; et al. Cell source determines the immunological impact of biomimetic nanoparticles. Biomaterials 2016, 82, 168–177. [Google Scholar] [CrossRef] [Green Version]
  145. Zhang, H.; Yang, J.; Liang, K.; Li, J.; He, L.; Yang, X.; Peng, S.; Chen, X.; Ding, C.; Li, J. Effective dentin restorative material based on phosphate-terminated dendrimer as artificial protein. Colloids Surfaces B Biointerfaces 2015, 128, 304–314. [Google Scholar] [CrossRef]
  146. Ma, Z.; Li, J.; Cao, F.; Yang, J.; Liu, R.; Zhao, D. Porous silicon carbide coated with tantalum as potential material for bone implants. Regen. Biomater. 2020, 7, 453–459. [Google Scholar] [CrossRef]
  147. Ruan, Q.; Liberman, D.; Zhang, Y.; Ren, D.; Zhang, Y.; Nutt, S.; Moradian-Oldak, J. Assembly of Layered Monetite-Chitosan Nanocomposite and Its Transition to Organized Hydroxyapatite. ACS Biomater. Sci. Eng. 2016, 2, 1049–1058. [Google Scholar] [CrossRef] [Green Version]
  148. Pullar, R.C.; Novais, R.M. Ecoceramics. Mater. Today 2017, 20, 45–46. [Google Scholar] [CrossRef]
  149. Pooyan, P.; Brewster, L.P.; Tannenbaum, R.; Garmestani, H. Biomimetic synthesis of two different types of renewable cellulosic nanomaterials for scaffolding in tissue engineering. Green Process. Synth. 2018, 7, 181–190. [Google Scholar] [CrossRef]
  150. Mainardi, J.C.; Demarchi, C.B.; Mirdrikvand, M.; Karim, M.N.; Dreher, W.; Rezwan, K.; Maas, M. 3D bioprinting of hydrogel/ceramic composites with hierarchical porosity. J. Mater. Sci. 2022, 57, 3662–3677. [Google Scholar] [CrossRef]
  151. Aksay, I.A.; Trau, M.; Manne, S.; Honma, I.; Yao, N.; Zhou, L.; Fenter, P.; Eisenberger, P.M.; Gruner, S.M. Biomimetic pathways for assembling inorganic thin films. Science 1996, 273, 892–898. [Google Scholar] [CrossRef]
  152. Ebrahimi, M.; Botelho, M.; Lu, W.; Monmaturapoj, N. Synthesis and characterization of biomimetic bioceramic nanoparticles with optimized physicochemical properties for bone tissue engineering. J. Biomed. Mater. Res. Part A 2019, 107, 1654–1666. [Google Scholar] [CrossRef]
  153. Fu, L.; Xie, L.; Fu, W.; Hu, S.; Zhang, Z.; Leifer, K.; Engqvist, H.; Xia, W. Ultrastrong Translucent Glass Ceramic with Nanocrystalline, Biomimetic Structure. Nano Lett. 2018, 18, 7146–7154. [Google Scholar] [CrossRef]
  154. Ling, S.; Jin, K.; Qin, Z.; Li, C.; Zheng, K.; Zhao, Y.; Wang, Q.; Kaplan, D.L.; Buehler, M.J. Combining In Silico Design and Biomimetic Assembly: A New Approach for Developing High-Performance Dynamic Responsive Bio-Nanomaterials. Adv. Mater. 2018, 30, 1802306. [Google Scholar] [CrossRef]
Figure 1. Biomimetic nanoparticles for biomedical applications. (a) Technology evolution: Early generations of particles were biologically inert and covered with nonfouling coatings, preventing their nonspecific interactions with the cells they encountered in vivo. From here, the next generation of nanoparticles became active, targeting molecules, which enabled the nanoparticles to reach the disease site and engage with the local environment. Taking inspiration from nature, the third generation of cell-membrane-based biomimetic nanoparticles mimicked the surface features of native cells by utilizing the whole cell membrane or membrane protein functionalization onto synthetic carriers (Reprinted from [31], license CC BY 4.0.) (b) Schematic presentation of different strategies of inflammation-targeting biomimetic nanoparticles. Orange and red spheres represent drug-encapsulated synthetic nanoparticles (grey) and liposomes (green), respectively. (Reprinted from [16], license CC BY 4.0.) (c) An example of modern BNM concept implementation: cell-membrane-coated NPs designed for atherosclerosis and inflammation therapy. The membranes of RBCs, platelets, and macrophages are extracted and used to coat different kinds of NPs depending on the site of inflammation and atherosclerosis. Each cell membrane has its own unique surface proteins, such as CD47 on RBC, integrin a4b1 on macrophages, and GPIIb/IIa on platelets, modifying the therapeutic effects. (Reprinted from [20], license CC BY 4.0.)
Figure 1. Biomimetic nanoparticles for biomedical applications. (a) Technology evolution: Early generations of particles were biologically inert and covered with nonfouling coatings, preventing their nonspecific interactions with the cells they encountered in vivo. From here, the next generation of nanoparticles became active, targeting molecules, which enabled the nanoparticles to reach the disease site and engage with the local environment. Taking inspiration from nature, the third generation of cell-membrane-based biomimetic nanoparticles mimicked the surface features of native cells by utilizing the whole cell membrane or membrane protein functionalization onto synthetic carriers (Reprinted from [31], license CC BY 4.0.) (b) Schematic presentation of different strategies of inflammation-targeting biomimetic nanoparticles. Orange and red spheres represent drug-encapsulated synthetic nanoparticles (grey) and liposomes (green), respectively. (Reprinted from [16], license CC BY 4.0.) (c) An example of modern BNM concept implementation: cell-membrane-coated NPs designed for atherosclerosis and inflammation therapy. The membranes of RBCs, platelets, and macrophages are extracted and used to coat different kinds of NPs depending on the site of inflammation and atherosclerosis. Each cell membrane has its own unique surface proteins, such as CD47 on RBC, integrin a4b1 on macrophages, and GPIIb/IIa on platelets, modifying the therapeutic effects. (Reprinted from [20], license CC BY 4.0.)
Nanomaterials 12 02485 g001aNanomaterials 12 02485 g001b
Figure 2. Classification of biomimetic nanomaterials (BNMs) based on the literature data, including information on biomimetic structure [16,34,35,36,37,38], biomimetic synthesis [32,39,40,41,42,43], biogenic components [31,33,44,45,46,47], magnetic BNMs [48,49,50,51,52,53,54], metal and metal oxide BNMs [55,56,57,58,59,60,61], organic, ceramic and hybrid BNMs [62,63,64,65,66,67].
Figure 2. Classification of biomimetic nanomaterials (BNMs) based on the literature data, including information on biomimetic structure [16,34,35,36,37,38], biomimetic synthesis [32,39,40,41,42,43], biogenic components [31,33,44,45,46,47], magnetic BNMs [48,49,50,51,52,53,54], metal and metal oxide BNMs [55,56,57,58,59,60,61], organic, ceramic and hybrid BNMs [62,63,64,65,66,67].
Nanomaterials 12 02485 g002
Figure 3. Chitosan-oligosaccharide-coated biocompatible palladium nanoparticles (Pd@COS NPs) for photo-based imaging and therapy. (a) A scheme showing the preparation of Pd NPs, further surface coating with thiolated chitosan oligosaccharide (Pd@COS NPs), and finally, functionalization using an RGD peptide (Pd@COS-RGD). (b) A systematic illustration showing the photothermal ablation and photoacoustic imaging of tumor tissue using Pd@COS-RGD. (Reprinted from [69], license CC BY 4.0.)
Figure 3. Chitosan-oligosaccharide-coated biocompatible palladium nanoparticles (Pd@COS NPs) for photo-based imaging and therapy. (a) A scheme showing the preparation of Pd NPs, further surface coating with thiolated chitosan oligosaccharide (Pd@COS NPs), and finally, functionalization using an RGD peptide (Pd@COS-RGD). (b) A systematic illustration showing the photothermal ablation and photoacoustic imaging of tumor tissue using Pd@COS-RGD. (Reprinted from [69], license CC BY 4.0.)
Nanomaterials 12 02485 g003
Figure 4. Schematic illustration of encapsulation procedure and release mechanism of biocompatible upconversion nanoparticles (UCNP). (a) Encapsulation of UCNPs in a novel, synthesized phosphate surfactant through sonication at rt. (b) Release of UCNPs after a specific cleavage of phosphate surfactant by the sPLA-2 enzyme. (Reprinted from [75], license CC BY 4.0.)
Figure 4. Schematic illustration of encapsulation procedure and release mechanism of biocompatible upconversion nanoparticles (UCNP). (a) Encapsulation of UCNPs in a novel, synthesized phosphate surfactant through sonication at rt. (b) Release of UCNPs after a specific cleavage of phosphate surfactant by the sPLA-2 enzyme. (Reprinted from [75], license CC BY 4.0.)
Nanomaterials 12 02485 g004
Figure 5. The four main routes of the cytotoxic mechanism of AgNPs. 1: AgNPs adhere to the surface of a cell, damaging its membrane and altering the transport activity; 2: AgNPs and Ag ions penetrate inside the cell and interact with numerous cellular organelles and biomolecules, which can affect corresponding cellular functions; 3: AgNPs and Ag ions participate in the generation of reactive oxygen species (ROS) inside the cell, leading to cell damage; and 4: AgNPs and Ag ions induce the genotoxicity. (Reprinted from [81], license CC BY 4.0.)
Figure 5. The four main routes of the cytotoxic mechanism of AgNPs. 1: AgNPs adhere to the surface of a cell, damaging its membrane and altering the transport activity; 2: AgNPs and Ag ions penetrate inside the cell and interact with numerous cellular organelles and biomolecules, which can affect corresponding cellular functions; 3: AgNPs and Ag ions participate in the generation of reactive oxygen species (ROS) inside the cell, leading to cell damage; and 4: AgNPs and Ag ions induce the genotoxicity. (Reprinted from [81], license CC BY 4.0.)
Nanomaterials 12 02485 g005
Figure 6. HRTEM images of different NPs: (a,b) inorganic magnetite NPs, (ce) MamC magnetite NPs, (fh) Mms6 magnetite NPs, and (ik) Mms6-MamC-mediated NPs. Selected areas of electron diffraction are shown for each sample. (Reprinted from [32], license CC BY 4.0.)
Figure 6. HRTEM images of different NPs: (a,b) inorganic magnetite NPs, (ce) MamC magnetite NPs, (fh) Mms6 magnetite NPs, and (ik) Mms6-MamC-mediated NPs. Selected areas of electron diffraction are shown for each sample. (Reprinted from [32], license CC BY 4.0.)
Nanomaterials 12 02485 g006
Figure 7. Magnetofection for gene delivery: (A) schematic representation of the process and (B) schematic illustration of DNA loading into lamellar magnetic hydroxyapatite (MHAp) nanoparticles for nucleic acid delivery. (Reprinted from [35], license CC BY 3.0.)
Figure 7. Magnetofection for gene delivery: (A) schematic representation of the process and (B) schematic illustration of DNA loading into lamellar magnetic hydroxyapatite (MHAp) nanoparticles for nucleic acid delivery. (Reprinted from [35], license CC BY 3.0.)
Nanomaterials 12 02485 g007
Figure 8. Surface features and scanning electron micrographs of a TiUnite dental implant surface. (Reprinted from [38], license CC BY 4.0.)
Figure 8. Surface features and scanning electron micrographs of a TiUnite dental implant surface. (Reprinted from [38], license CC BY 4.0.)
Nanomaterials 12 02485 g008
Figure 9. Schematic representation of the mechanism and final outcomes of the interaction of Au NPs and Ag NPs with a water dispersion of cubosomes and solid-supported films of cubosomes. (Reprinted from [34], license CC BY 4.0.)
Figure 9. Schematic representation of the mechanism and final outcomes of the interaction of Au NPs and Ag NPs with a water dispersion of cubosomes and solid-supported films of cubosomes. (Reprinted from [34], license CC BY 4.0.)
Nanomaterials 12 02485 g009
Figure 10. Schematic illustration of biomimetically mineralized metal–organic framework (MOF). (a) Schematic of a sea urchin, a hard, porous, protective shell that is biomineralized by soft biological tissue. (b) Schematic of an MOF biocomposite showing a biomacromolecule (for example, protein, enzyme, or DNA) encapsulated within a porous, crystalline shell. (Reprinted from [108], license CC BY 4.0.)
Figure 10. Schematic illustration of biomimetically mineralized metal–organic framework (MOF). (a) Schematic of a sea urchin, a hard, porous, protective shell that is biomineralized by soft biological tissue. (b) Schematic of an MOF biocomposite showing a biomacromolecule (for example, protein, enzyme, or DNA) encapsulated within a porous, crystalline shell. (Reprinted from [108], license CC BY 4.0.)
Nanomaterials 12 02485 g010
Figure 11. Schematic illustration of mechanism of mitochondria-targeted cancer cell membrane biomimetic metal–organic framework mediated sonodynamic therapy and immune checkpoint blockade immunotherapy. (Reprinted from [115], license CC BY 4.0.)
Figure 11. Schematic illustration of mechanism of mitochondria-targeted cancer cell membrane biomimetic metal–organic framework mediated sonodynamic therapy and immune checkpoint blockade immunotherapy. (Reprinted from [115], license CC BY 4.0.)
Nanomaterials 12 02485 g011
Figure 12. Schematic of anisotropic nanoparticle fabrication and RBC membrane coating. (A) Spherical PLGA nanoparticles (NPs) were synthesized and cast onto a thin plastic film of 10% polyvinyl alcohol (PVA) and 2% glycerol. Particles were then stretched under heat in one and two dimensions (2D) to generate prolate and oblate ellipsoidal particles, respectively. (B) RBCs underwent hypotonic lysis and were then sonicated to generate sub—200 nm vesicles. RBC-derived vesicles were then coated on PLGA nanoparticles of all shapes under sonication. (Reprinted from [33], license CC BY 4.0.)
Figure 12. Schematic of anisotropic nanoparticle fabrication and RBC membrane coating. (A) Spherical PLGA nanoparticles (NPs) were synthesized and cast onto a thin plastic film of 10% polyvinyl alcohol (PVA) and 2% glycerol. Particles were then stretched under heat in one and two dimensions (2D) to generate prolate and oblate ellipsoidal particles, respectively. (B) RBCs underwent hypotonic lysis and were then sonicated to generate sub—200 nm vesicles. RBC-derived vesicles were then coated on PLGA nanoparticles of all shapes under sonication. (Reprinted from [33], license CC BY 4.0.)
Nanomaterials 12 02485 g012
Figure 13. Plausible mechanism for the formation of Ag–TiO2 NCs using Origanum majorana leaf extract. (Reprinted from [40], license CC BY 4.0).
Figure 13. Plausible mechanism for the formation of Ag–TiO2 NCs using Origanum majorana leaf extract. (Reprinted from [40], license CC BY 4.0).
Nanomaterials 12 02485 g013
Figure 14. Main applications of biomimetic nanomaterials.
Figure 14. Main applications of biomimetic nanomaterials.
Nanomaterials 12 02485 g014
Table 1. Magnetic biomimetic nanomaterials: structure, synthesis, and applications.
Table 1. Magnetic biomimetic nanomaterials: structure, synthesis, and applications.
CompositionSynthesis TechniqueDeclared ApplicationsRefs.
HAP 1-coated magnetite NPsHAP precursors added into a solution containing iron oxide NPsMagnetic hyperthermia, magnetic scaffold for bone tissue regeneration[35,51]
Dextran-coated magnetite NPs modified with a protein and a mimetic ligandLigand directly synthesized on
dextran-coated particles
Magnetic separation of biomolecules[50]
MamC-mediated biomimetic Fe3O4 NPs with or without polymer coatingBiosynthesis from
oxygen-free solutions containing recombinant MamC in anaerobic conditions for 30 days
Photothermia,
chemotherapy,
magnetic hyperthermia, immobilization of enzymes
[32,48,85,86,87]
Fe and Fe3O4 NPsMicrobial preparation of FeS and Fe2O3 using Aspergillus niger YESM 1, followed by physical process at supercritical conditionsMagnetic resonance imaging, magnetic
hyperthermia
[49]
PEGylated magnetoferritin NPs with magnetite coreMagnetite biomineralization using PEGylated human ferritin NPsMagnetic resonance imaging[52]
Acrylamide-based
biomimetic
magnetic NPs
Acrylamide and ethylene glycol
dimethacrylate copolymerized in the presence of S-naproxen on silica-coated Fe3O4 NPs
Separation of chiral drugs[90]
Biomimetic silica entrapping Fe3O4 NPs and horseradish peroxidaseTetramethyl orthosilicate hydrolysis in the presence of Fe3O4 NP suspension and horseradish peroxidase solutionBiocatalyst for direct enzyme prodrug therapy[37]
Fe3O4 NPsCopolypeptide-promoted Fe3O4 NP biomimetic mineralizationSeparation technology, magnetic resonance imaging[91]
Cell-membrane-camouflaged Fe3O4 NPsCell membrane adsorption onto silica-coated Fe3O4 or onto drug-loaded mesoporous Fe3O4 NPsDrug targeting, cancer immunotherapy[54,92]
Biomimetic magnetic silk scaffoldsMagnetic NP diffusion into silk fibroin protein via dip-coatingTissue engineering, magnetic
hyperthermia
[53]
Engineered bacterial magnetosomesSilica encapsulation or biotinylation of isolated bacterial magnetosomesMagnetic particle imaging, magnetic resonance imaging, magnetic
hyperthermia
[44,89]
Polydopamine-coated Fe3O4 NPsPreliminarily prepared Fe3O4 NP incubation in an alkaline dopamine solutionImmobilization of enzymes[93]
Biodegradable polylactide-based Fe3O4 NPsModified emulsification–solvent evaporation methodDegradation pattern study of NP formulations[94]
Magnetosome-like ferrimagnetic iron oxide nanochainsSelf-assembly of Fe3O4 NPs
coated with hydrophilic polymer
into 1D nanochains in water
Post-stroke recovery[95]
1 Hydroxyapatite.
Table 2. Biomimetic nanomaterials based on particles of metals and metal oxides and areas of their application.
Table 2. Biomimetic nanomaterials based on particles of metals and metal oxides and areas of their application.
CompositionSynthesis TechniqueDeclared ApplicationsRefs.
Au, Ag, and Ag–Au bimetallic NPsBiomimetic synthesis in aqueous gelatin solution with consequent addition of AgNO3 and HAuCl4Biosensing, nanotoxicology[39]
Au and Ag NPs attached to model lipid cubic phase membranesIncubation of preliminarily obtained Au or Ag NPs and cubosomes in aqueous dispersions Cubosome-based targeted drug delivery[34]
Electrochemical sensor based on Au-NP-imprinted polymerSurface modification of metal electrode with 2-aminothiophenol and preliminarily obtained Au NPs, followed by electropolymerizationOrganic pollutant detection[116]
Ag NPsBiological reduction of AgNO3 in aqueous solution with Musa balbisiana or Phlomis bracteosa plantlets or Saraca indica leaf extracts as reducing agentsMultidrug-resistant bacteria treatment[55,98,99]
Au–Ag NPs attached on silica nanowire supportSilica nanowire formation using cellulose nanocrystals as biotemplates, followed by Au–Ag NP attachment via wet chemical processNetwork substrate in surface-enhanced Raman scattering[61]
Au, Pd, and Pt NPs on biomimetic MXene paperSpontaneous growth of metal NPs from aqueous precursor solution on Ti3C2Tx paper obtained using vacuum filtrationFlexible bioelectronics[117]
Au–Pd NPs in amide conjugate structureFormation of Au–Pd NPs from HAuCl4 to PdCl2 self-assembled gallic acid amid conjugatesCatalytic degradation of organic pollutants[56]
Porphyrinic
Zr–MOF 1 NPs cloaked with cell membrane
Wet chemistry synthesis of Zr–MOFs, followed by MnO2-coating in KMnO4 solution and cell-membrane-cloakingAntiangiogenesis
and photodynamic therapy
[58]
Peptide-coated Au NPsReduction of HAuCl4 in aqueous solution of multifunctional peptidesBiosensing, targeting NPs into
cells and organelles
[57]
Au nanoplatesReduction of HAuCl4 in aqueous solution using Chlorella vulgaris extractNear-infrared range
hyperthermia
[59]
ZnO, NiO, CuO, Co3O4, and CeO2Eggshell membrane immersion in metal salt solutions, followed by drying at room temperature and calcination at 750 °CRemoval of NPs from an aqueous environment[60]
Polydopamine-Ag NP membraneTreatment of catheter surface with a dopamine solution, followed by AgNO3 solution immersion and vacuum-dryingCentral venous catheter coating[118]
Pt-NP-decorated metal–organic frameworkSynthesis of Pt NPs templated with MOFs obtained using Fe(III) tetra(4-carboxyphenyl)porphine chlorideBiosensing[101]
Nanostructured calcium-phosphate-coated TiHAP 2 ceramic particle injection into a plasma torch and projection on the surface of titaniumDental implants[38,100]
1 Metal–organic framework; 2 Hydroxyapatite.
Table 3. Polymeric, ceramic, and hybrid biomimetic nanomaterials and their applications.
Table 3. Polymeric, ceramic, and hybrid biomimetic nanomaterials and their applications.
CompositionSynthesis TechniqueDeclared ApplicationsRefs.
Microstructured Al2O3 self-shaped bilayersAl2O3 NP repeated coagulation with ferrofluid under magnetic field following sinteringBiomimetic complex-shaped ceramics[36]
Cancer-cell-membrane-coated polymeric NPsDrug-encapsulating PLGA prepared via nanoprecipitation consequently coated with cancer cell membraneBioimaging, phototheranostics, nanovaccines[45]
ZrO2 coated with HAP 1–bovine serum albumin compositeZrO2 substrate soaked in albumin and simulated body fluid solution, followed by calcium phosphate nanocrystal precipitationOrthopedic and dentistry[140]
Phage–platelet hybrid NPsBinding of a blood-circulation-prolonging, peptide-modified bacteriophage to platelet membrane NPs derived via a repeated freeze–thaw procedureBlood-retention-time-prolonging, antibacterial phage therapy[141]
Ultrathin silicon nitride microporous membranesNonstoichiometric silicon nitride (SixNy) deposition on both sides of a silicon wafer by low-pressure chemical vapor depositionScaffolds for epithelial tissue cell models[142]
PLGA 2 NPs wrapped with MMs 3Mixing of drug-containing PLGA NPs with purified macrophage membranes and following extrusion using a 200 nm polycarbonate membraneUlcerative colitis treatment[143]
Fe3O4, ZIF 4, Au, PLGA, and porous Si coated with cell membraneSonication or extrusion coating of various NPs with HeLa, macrophages, platelets, and RBC 5 cell membranesCancer
nanomedicine
[138]
Leukocyte-based biomimetic NPsCombination of phospholipids and membrane proteins from leukocytes, followed by incubation with specific antibodies in batch or microfluidic processesAnti-inflammatory therapy[67,139]
Aprismatic, enamel-like, nanostructured HAP layersHAP mineralization from CaCl2·2H2O and KH2PO4 in the presence of synthetic peptide solutionDevelopment of enamel-like biomaterials[41]
MM-camouflaged ROS 6-responsive biomimetic NPsCamouflaging of ROS-responsive polymer NPs with MMs extruded through a 400 nm polycarbonate porous membraneAtherosclerosis therapy[46]
Lanthanide NPs-Cas9 7 complex coated with hepatoblastoma cell membraneSynthesis of NaYF4:Yb/Tm/Ca@NaYF4:Yb/Nd core–shell NPs from LnCl3 aqueous solution, followed by Cas9 binding and coating with hepatoblastoma cell membranesHBV 8-targeted
therapy
[47]
NPs functionalized with leukocyte cellular membraneBiodegradable NPs conjugated with (3-aminopropyl)triethoxysilane, followed by incubation with proteolipid solutionDevelopment of
drug delivery carriers
[144]
Anisotropic polymeric NPs coated
with RBC membranes
Stretching of spherical PLGA NPs immobilized on a PVA 9-glycerol film, followed by sonication-assisted coating with ultrasound-derived RBC membranesDetoxification
of systemically administered bacterial toxin
[33]
BN NP-polydopamine-coated glass fiber-epoxy resin nanocompositeFacile, water-assisted dopamine
coating of glass fiber, followed by addition of BN NPs and epoxy resin components
Development of fiber-reinforced plastic composites[42]
Al2TiO5–Al2O3 ceramics with sea urchin and nacre structure elementsBall-milling of Al2O3, SiO2, MgO, and TiO2, followed by vacuum-drying and pressureless-sintering in air atmosphereComposite ceramics,
catalyst carriers, and sound absorbers
[43]
Ag–TiO2 NPsSonochemical synthesis of NPs using leaf extract of Origanum majorana as a bioreductant and a stabilizing agentAntibacterial and antioxidant therapy[40]
Phosphate-terminated polyamidoamine dendrimerG4 PAMAM 10 modification with dimethylhydrogenophosphonate, followed by treatment with bromotrimethylsilaneBone and teeth restoration[145]
Porous SiC coated with TaBioactive metal (Ta) chemical vapor deposition on porous SiC scaffoldsPotential material for bone substitutes[146]
HAP with multi-scale, hierarchically ordered structureSelf-assembly of layered chitosan–maleic acid matrix, followed by monetite mineralization and transformation to HAPDeveloping bone substitute materials[147]
Amelogenin-containing chitosan hydrogel (modified with enamel proteinase)Mixing of chitosan solution, CaCl2, and recombinant full-length porcine amelogenin, followed by stirring overnight (and addition of enamel proteinase)Enamel repair[133,134]
Ceramic biomimetic
3-DOM 11 foam
Cork powder pyrolysis to carbon, followed by infiltration with precursor salt solution and calcination to form the oxide ceramicEnvironmental and energy applications[148]
Cellulose nanowhiskers in biopolymer matricesMicrocrystalline cellulose sulfuric
acid hydrolysis and centrifugation
Scaffolding in tissue engineering[149]
Genipin-crosslinked chitosan, alginate, and alumina nanocomposite gelsAlumina powder added to chitosan solution, followed by alginate dissolution and genipin (cross-linking agent) addition3D bioprinting[65,150]
Ceramic–organic nanocomposite filmsTemplated supramolecular surfactant self-assembly on a mica surfaceLow-temperature thin-film processing[151]
Nanometer-sized HAP–collagen compositeIncubation of Tris-buffered CaCl2 with sharkskin collagen suspensionOrthopedic implants[62]
PAMAM 12-dendrimer-templated HAP crystallizationEnamel immersion in a solution of CaCl2, KH2PO4, and PAMAM dendrimers modified with carboxylic acid groupsEnamel repair[135,136,137]
HAP–tricalcium
phosphate biphasic NPs
Wet-milling of CaHPO4 and CaCO3 powders, followed by double-sieving and high-temperature calcinationBone tissue engineering[152]
HAP NPs obtained using asparagine–serine–serine peptideEnamel exposure to triplet repeats of asparagine–serine–serine solution, followed by soaking in artificial salivaEnamel repair[63]
Erythrocyte-membrane-camouflaged polymeric NPsRBC hypotonic treatment and extrusion, followed by mixing with PLGA NPs via extrusion through a porous membraneTargeted drug delivery[66]
Monocrystalline ZrO2
NPs embedded in an amorphous SiO2 matrix
Spark-plasma-sintering of ZrO2 NPs and amorphous SiO2 powder with a molar ratio of 65% ZrO2/35% SiO2 at 1200 °CHigh-strength
translucent glass ceramic materials
[153]
Nacre-like composite of silk nanofibrils, HAP, and chitin nanofibrilsSelf-assembly of silk nanofibrils, followed by HAP biomineralization, mixing with chitin nanofibril solution, and nacre-like membrane vacuum-assisted deposition“Grab-and-release” actuators[154]
1 Hydroxyapatite. 2 Poly(lactic acid-glycolic acid). 3 Macrophage membrane. 4 Zeolitic imidazolate framework. 5 Red blood cell. 6 Reactive oxygen species. 7 CRISPR-associated protein 9 (CRISPR is the clustered regularly inter-spaced palindromic repeat gene-editing therapy). 8 Hepatitis B virus. 9 Polyvinyl alcohol. 10 Generation 4 of polyamidoamine dendrimer. 11 Three-dimensionally ordered material. 12 Poly(amido amine).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gareev, K.G.; Grouzdev, D.S.; Koziaeva, V.V.; Sitkov, N.O.; Gao, H.; Zimina, T.M.; Shevtsov, M. Biomimetic Nanomaterials: Diversity, Technology, and Biomedical Applications. Nanomaterials 2022, 12, 2485. https://doi.org/10.3390/nano12142485

AMA Style

Gareev KG, Grouzdev DS, Koziaeva VV, Sitkov NO, Gao H, Zimina TM, Shevtsov M. Biomimetic Nanomaterials: Diversity, Technology, and Biomedical Applications. Nanomaterials. 2022; 12(14):2485. https://doi.org/10.3390/nano12142485

Chicago/Turabian Style

Gareev, Kamil G., Denis S. Grouzdev, Veronika V. Koziaeva, Nikita O. Sitkov, Huile Gao, Tatiana M. Zimina, and Maxim Shevtsov. 2022. "Biomimetic Nanomaterials: Diversity, Technology, and Biomedical Applications" Nanomaterials 12, no. 14: 2485. https://doi.org/10.3390/nano12142485

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