Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2646 2023-01-25 06:11:44 |
2 format correction -2 word(s) 2644 2023-01-29 03:39:39 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Popova, V.;  Dmitrienko, E.;  Chubarov, A. Magnetic Nanocomposites for Biomedical Applications of Nucleic Acids. Encyclopedia. Available online: https://encyclopedia.pub/entry/40494 (accessed on 26 April 2024).
Popova V,  Dmitrienko E,  Chubarov A. Magnetic Nanocomposites for Biomedical Applications of Nucleic Acids. Encyclopedia. Available at: https://encyclopedia.pub/entry/40494. Accessed April 26, 2024.
Popova, Victoriya, Elena Dmitrienko, Alexey Chubarov. "Magnetic Nanocomposites for Biomedical Applications of Nucleic Acids" Encyclopedia, https://encyclopedia.pub/entry/40494 (accessed April 26, 2024).
Popova, V.,  Dmitrienko, E., & Chubarov, A. (2023, January 25). Magnetic Nanocomposites for Biomedical Applications of Nucleic Acids. In Encyclopedia. https://encyclopedia.pub/entry/40494
Popova, Victoriya, et al. "Magnetic Nanocomposites for Biomedical Applications of Nucleic Acids." Encyclopedia. Web. 25 January, 2023.
Magnetic Nanocomposites for Biomedical Applications of Nucleic Acids
Edit

Magnetic nanocomposites (MNCs) combine the features of magnetic nanoparticles and a second material, which provide distinct physical, chemical, and biological properties. The magnetic core for nanocomposite synthesis is extensively used due to its high saturation magnetization, chemical stability, large surface area, and easy functionalization. Moreover, magnetic nanoparticles (MNPs) have great potential for magnetic resonance imaging (MRI), magnetic particle imaging (MPI), hyperthermia, and targeted drug and gene delivery by an external magnetic field. Numerous composing units exist, which leads to the outstanding application of composites. The authors focused on nucleic acid-based bioapplications of MNCs with polymeric, organic, inorganic, biomolecules, and bioinspared surface coating. The unique types of nanocomposites as magnetic molecularly imprinted polymer (MMIP) properties are presented. The authors aim to discuss the features of nucleic acid-based MNC information available to researchers in this field and guide them through some problems in the area, structure variation, and surface functionalization possibilities. The advancements of MNCs and imprinted polymers in nucleic acid-based therapy, diagnostics, theranostics, magnetic separation, biocatalytic, and biosensing are introduced.

magnetic nanoparticles magnetic nanocomposites nucleic acid

1. Toxicity of MNCs

The toxicity of MNPs is an essential factor for future healthcare applications [1][2][3]. However, the studies on MNCs’ creation and safety assessment remained largely divided. While most of the safety studies have been focused on easy nanoparticles on the cell model, the material area goes forward with various smart constructions. However, most works usually present a primary cytotoxicity test, using an MTT compound on cancer cell lines [4][5]. The MTT assay does not show the interaction with blood proteins, tissue media, and delayed toxicity of the degraded product. The cancer cells are highly adapted to ROS levels and unfavorable media conditions and have activated cell growth and survival systems. In this way, the toxicity of the MNCs is still less explored [2][3][6][7][8]. Recently, studies on the toxicity of MNCs on spheroids were highlighted [9][10][11][12]. Such three-dimensional (3D) cell aggregates can mimic the tumor microenvironment. In recent years, significant progress in the development of spheroids for use as a tumor model has been obtained. The cell viability in two-dimensional (2D) cell culture monolayers and spheroids has not shown the same results [10][12][13][14]. Therefore, dose predictions from conventional cell experiments are often misleading for in vivo applications. Spheroids are a successful replacement for expensive and unethical animal experiments. However, extensive further studies are required for the stable manufacturing of various cancer spheroids and better tumor mimicking as sustainable cell growth, proliferating and non-proliferating cells, a hypoxic center, etc. [14][15].
Herein, the possible cytotoxicity and organ-specific toxicity are discussed. A high indestructibility in biological liquids or low toxicity is not required for some purposes. For example, for magnetic separation, MNCs usually do not interact with human organisms and should only have good magnetic properties and the ability to interact with a target. For in vivo studies, MNCs must be non-toxic, stable in biological liquids, and biocompatible. Unstable MNPs may be extremely toxic due to the formation of reactive oxygen species (ROS), injury to the immune system, metabolic disorders, decrease in growth rate, or changes in alterations, inflammation, ulceration, etc. (Figure 1) [2][3][16][17]. A high ROS level leads to mitochondrial membrane damage, harmful cell proliferation, modulating gene transcription, dysregulation of ion channels, RNA destruction, and DNA and lipids oxidation with a subsequent formation of a point mutation. In extreme cases, significant damage leads to cell death (Figure 1) [7][17]. The oxidative stress mechanism can be provided by the release of ferrous ions due to the instability of the MNCs, direct ROS generation on the MNCs’ surface, and altering the mitochondrial function and signaling pathways [7][17].
Figure 1. Possible toxicity mechanism of MNCs, leading cell processes’ dysregulation and triggering cell death (necrosis or apoptosis).
The MNCs’ size, shape, surface charge, coating, and surface modification highly influence biodistribution and toxicity [2]. MNCs with a size less than 10 nm are quickly removed through renal clearance. Nanocomposites greater than 180–200 nm are filtrated by the spleen. Therefore, MNCs in the range from 10 to 150 nm are the most preferred. The surfaces of nanoparticles are rapidly covered in blood by various proteins. Surface chemistry highly influences nanocomposite biodistribution. For example, albumin coating usually prevents non-specific blood interaction and liver accumulation, prolongs circulation time, and moderates particle uptake in cancer tissue [18]. The wrong coating of the nanocomposites may lead to the MNCs’ destabilization, aggregation, and precipitation [19]. Inside the body, MNCs can be absorbed through interactions with proteins (e.g., protein albumin), blood components, and cells [7]. Blood compatibility is essential for any in vivo application of MNCs. Lack of stability in the blood can trigger liver accumulation with further degradation and elimination from the body. In the worst case, the co-coagulation and precipitation of MNCs and the blood component may happen, activating thrombus formation.
The MNCs’ dose, initial concentration, biodistribution, and circulation time should be taken into account [20]. The nanocomposites can be distributed into various organs and further metabolized. The overload of local MNCs may lead to high levels of free iron ion release and ROS generation in the tissue and cause aberrant cellular reactions and organ-specific toxicity [2][3][21]. The in vivo toxicity experiment is an expensive and vast work, which has greatly shut down the progress in this area [3][7]. Almost all organs are influenced by the toxic effects of MNCs. Among those are the heart, lungs, liver, kidney, and nervous and reproductive systems. For example, heart-specific toxicity provides contractile apparatus and endothelial damage, the violation of a conducting system, and ischemia [3]. The MNCs’ organ-specific toxic effects may be associated with ROS generation, leading to changes in glutathione, superoxide dismutase, and coenzyme levels [3]. Afterward, the in vivo interaction of MNCs and the biological system is quite complicated and dynamic. In the future, extended toxicity studies could help to bridge the gap between in vitro research results and successful clinical trials.

2. Drug and Gene Delivery, Therapy, and Diagnostics (Theranostics)

Nanoparticles and MNCs became extremely popular for cancer therapy due to the possible targeted delivery. MNCs provide drug or gene delivery to the cell or tissue by an external magnetic field. Magnetic transfection, or magnetofection, is a method that uses magnetic fields to transport NA-based MNCs to target cells. Magnetofection has been adapted to various NA types, including aptamers, siRNA, miRNA, shRNA, etc. [22][23][24][25][26][27]. Such technology is a possibility to solve the drug resistance problem and the low efficiency of gene delivery through cell membranes [28]. A combination of MNCs and siRNA or antisense oligonucleotides may be successfully used instead of a chemotherapeutic drug, resulting in a therapeutic effect [22][25][29][30][31][32].
As stated above, the magnetic core of MNCs has multimodal advantages, such as possible tracking by magnetic resonance imaging (MRI) or magnetic particle imaging (MPI) and the hyperthermia effect. MRI and MPI are great non-invasive diagnostic techniques [16][33][34][35][36][37][38][39]. MRI provides a high-resolution and easy image contrast manipulation. MNPs are usually known as T2-contrast agents, which lead to a dark zone on the MRI image. However, the previous simple MNPs were withdrawn due to the side effects. There are many successful in vitro experiments and undergoing pre-clinical animal studies for MNCs [33]. MPI is a relatively novel technology that was presented in 2005. MPI detects tracer MNCs selectively, providing the signal is observed without background with a high signal-to-noise ratio [16]. The method possesses potential for tumor, metastases, and cell detection.
The hyperthermia effect is generated by an alternating magnetic field [40][41]. In the presence of MNCs, heat appears in local regions, which damages tumor cells. The method is limited by the MNCs’ quality, size, morphology, and coating. The simultaneous drug, therapeutic NA, and hyperthermia using single-MNCs is a promising anticancer strategy. Recently, various MNCs to be used for multimodal imaging and theranostics have been developed [16][33][42]. The combination of MRI, MPI, and primary used methods, such as single-photon emission computed tomography (SPECT), computed tomography (CT), positron emission tomography (PET), and optical imaging, have become known [33][42][43][44]. However, theranostic synthesis is a complicated problem, which limits progress in the area [45].

3. Magnetic Separation and Biosensors

Solid-phase magnetic separation is a much more efficient protocol for NA isolation than traditional approaches [46][47][48][49][50][51][52][53][54][55][56]. Magnetic separation is a relatively cheap, quick method for pure NA capture with a high yield. It usually requires ~10 min for NA isolation from the mixture using specific MNCs and a rack with magnets (Figure 2). However, the high cost of commercially available MNCs limits their routine applications. Moreover, the specific “bind-release” of NA MNCs is extremely rare. One of the primarily used magnetic beads has an avidin/streptavidin coating and forms a specific complex with biotin-labeled oligonucleotide [57]. Biotinylated oligonucleotide interacts with a targeted NA. Such systems are specific. However, the high binding constant of avidin to biotin hinders the separation of NA from MNPs and their further use. Other commercially available MNCs for NA isolation are not specific and usually bind all the NA in the probe with a certain degree of purity. Such MNCs work on the ionic interaction of NA to the MNC’s surface. Therefore, protein binding is possible. Finally, new cheap, high-capacity sorbents for the capture of NA are required.
Figure 2. Basic principles of NA magnetic separation.
Recently, magnetic cell separation has become a comprehensive technology for targeted cell population separation for various applications [58]. Some MNCs for cell isolation work by using NA-based MNCs bearing aptamers. However, aptamer-based magnetic cell separation faces many obstacles, which makes it difficult to use such an approach in common practice [58]. DNA aptamers can also provide specific recognition capabilities against many targets used for magnetic separation.
Selectively sensing a single NA allows for the discovery of rich information regarding human health [48]. The separation of NA, with further analysis, is a laborious procedure that, in some cases, does not make sense. The new specific, sensitive methods are required for rapid diagnosis [59]. The surface of the specific MNCs for NA detection may be modified by NA-specific molecules, including antibodies, aptamers, NA, proteins, etc. The transducer from “chemical” to “physical” signal may be electrochemical, optical, piezoelectric, etc. [59][60][61] (Figure 3). In some cases, the isolation of NA with subsequent analysis is required. This procedure may be performed using MNCs with further PCR analysis [57]. For instance, MNCs can improve the sensitivity of the PCR with an extreme detection limit. Compared to traditional PCR approaches, an MNC-based PCR shows richness and a high potential [57].
Figure 3. Basic principles of NA-based biosensors.

4. Magnetic Molecularly Imprinted Polymers

MNPs have been extensively developed for their excellent separation and extraction ability. A new innovative approach in this area is the use of MNPs with a molecularly imprinted polymer [62][63][64]. Molecular imprinting is a technique to create molecule (template)-specific cavities in polymer matrices (Figure 4).
Figure 4. Schematic representation of core–shell imprinting for MMIP preparation. The analyte is a template molecule that MIP has reversibly recognized. The number of cycles is proportional to the efficiency of MIPs.
The procedure is similar to the enzymes’ “lock and key” model. The resulting polymer is called molecularly imprinted polymers (MIPs) [64][65]. For the magnetic core, magnetic molecularly imprinted polymers (MMIPs) have been developed [62][66]. In the structure of MIPs, some regions can specifically interact with template molecules or structurally related molecules. Recognition can occur concerning the shape, size, or due to interactions between the functionalities of the template and polymer [65]. Two types of MIPs are known [67]. Among those, the classic version involves the polymerization of functional monomers, templates, and cross-linking agents (Figure 4, top line). The second one is the change in the polymer state from liquid to solid in the presence of a template.
MIPs have many advantages, such as chemical and physical stability, easy synthesis, reusability, and cost-efficient preparation [64][68]. Polymer matrices may consist of organic or inorganic compounds capable of recognizing molecules or ions [62][64][65][66]. The synthesis methods of MIPs can be classified according to the format (two- or three-dimensional) and bonding type between the functional monomers and template (covalent or non-covalent) and by the nature of the functional monomers (organic or inorganic). MIPs are used in many areas, such as synthetic recognition elements, solid-phase extraction, liquid chromatography, electrochromatography, assays, drug delivery, theranostics, and biosensor production [64][65][69][70]. MIPs are primarily prepared by bulk polymerization as monoliths (3D imprinting). The first templates for MIP recognition were low molecular weight biologically active compounds such as vitamins, hormones, toxins, drugs, nucleotides, NA, and their derivatives [71][72][73][74]. However, this method has various drawbacks, such as a low amount of binding sites near the surface, inaccessible recognition sites within the polymer bulk, a wide range of particle sizes, and non-uniform morphology [75][76]. The transition to the imprinting of biomolecules (nucleic acids, peptides, and proteins) requires significant changes in the existing imprinting protocols and the emergence of new ones [64]. Obtaining MIPs for biomolecules remains a formidable challenge due to their large dimensions, low solubility and stability, complex structure, slow mass transfer, and structural flexibility in solution. The bulk polymerization is limited to the macromolecule and biomolecule imprinting, including peptides, proteins, NA, viruses, and bacteria. Nearly 1200 research articles were published annually on MIP-based biosensors, out of which only nearly 10% included the recognition of biomacromolecules [77]. Surface molecular imprinting seems to be an alternative approach that can address some of the shortcomings of the synthesis of a primary MIP. Inorganic materials, such as silica, magnetic, gold, and silver nanoparticles, are especially widely used as a core for MIPs [78][79]. The combination of MIPs and other materials combines features yielding smart core–shell MIP structures which allow for the control of the size and distribution of the synthesis. The hybrid magnetic MIP (MMIP) has the advantages of the technology of MIPs and MNPs.
The magnetic properties of the MMIP allow for magnetic separation, imaging, hyperthermia, and selective template release [62][63][66][80][81][82]. MMIPs have shown a high potential in identifying a broad spectrum of analytes, from small molecule enantiomers to large proteins, NA, and macromolecules [62][68][83][84]. The possibility to automatize this process by using magnetic properties is also an interesting feature for industrialization and mass production. Nowadays, MMIPs are widely used in various fields of biomedicine, such as biosensors, drug and gene delivery, and NA isolation [65][78][84]. MMIPs show a high potential for use in cancer therapy due to targeted delivery by an external magnetic field, hyperthermia effect, and possible simultaneous drug and therapeutic NA delivery [85]. The use of MMIPs for NA-based applications is also being extensively studied [65][68][86][87][88]. NA can act as both templates and complex macromolecular functional monomers, which provide unique properties to the resulting MIPs [89]. Consequently, combining MIPs with NA with magnetic properties provides a new class of smart synthetic NA receptors, i.e., NA-MMIPs [86]. These materials open up new possibilities in this research area. Table 1 summarizes NA-based MNC biomedical applications.
Table 1. Some examples of nucleic acid-based biomedical applications of MNCs.
Application Area MNCs Type NA-Based Application Reference
Biosensing and diagnostics MNP@Ag-amine-modified anti-miR-155 miR-155 detection through resveratrol interaction (electrochemical label) [90]
MNP@Au Ultrasensitive colorimetric and electrochemical miRNA detection [91][92]
MNP@graphene Electrochemical miRNA detection [92]
MNP@SiO2 DNA and RNA extraction from Hepatocellular Carcinoma, virus RNA extraction and detection by RT-PCR, Taq polymerase fixation for long-term enzyme activity for PCR [57][93]
MNP-oleic acid DNA detection by PCR [57]
MNP-NH2 DNA extraction from blood and detection by PCR [93]
MNP-COOH DNA extraction from staphylococcus aureus bacteriophages, mRNA isolation from mammalian cells [93]
MNP-OH/-NH2/-COOH Hybrid NA separation from animal tissue samples [94]
  MMIP DNA detection [86]
  MNP—rabbit antigoat immunoglobulin Immunoglobulin (IgG) detection [95]
Therapy and diagnostics MNP@PEI micro-RNA intracellular delivery for MYCN inhibition in neuroblastoma [96]
MNP-chitosan Gene delivery [97]
MNP-Hyaluronic acid Gene delivery [98]
MNP-lipids siRNA delivery [31]
MNP lipoplex Theranostics, imaging guided (MRI) delivery of NA [98]
Magnetic separation MNP@Ag mRNA extraction [99]
MNP@Au mRNA, dsDNA extraction [99][100]
MNP@graphene dsDNA extraction [100]
MNP@SiO2 DNA/RNA extraction [46][93][99][100]
MNP@SiO2 NA capture from lysed white blood cells, B. subtilis, E. coli, and Rift Valley fever viruses [101]
MNP@SiO2 viral NA extraction from serum [93]
MNP@SiO2-organic halide DNA extraction [46]
MNP@SiO2-NH2 DNA extraction [46]
MNP@polydopamine genomic DNA extraction [102]
MNP-Nylon-6 RNA extraction [103]
MNP-Streptavidin DNA/RNA extraction, aptamer-based cell separation [58][101]
MNP-CD138 (syndecan-1) antibody conjugated Endothelial cells (HUVEC)
separation
[95]
MNP@PEI dsDNA extraction [100]
MNP-thermosensitive polymer, poly(N-isopropylacrylamide-co-2-aminoethyl methacrylate) DNA extraction [52]
MNP-N-isopropylacrylamide and allyl glycidyl ether, 3,5-difluoro-4-formylphenylboronic acid S. aureus and Salmonella spp. separation [104]

References

  1. Petrov, K.D.; Chubarov, A.S. Magnetite Nanoparticles for Biomedical Applications. Encyclopedia 2022, 2, 1811–1828.
  2. Malhotra, N.; Lee, J.S.; Liman, R.A.D.; Ruallo, J.M.S.; Villaflore, O.B.; Ger, T.R.; Hsiao, C. Der Potential toxicity of iron oxide magnetic nanoparticles: A review. Molecules 2020, 25, 3159.
  3. Chrishtop, V.V.; Mironov, V.A.; Prilepskii, A.Y.; Nikonorova, V.G.; Vinogradov, V.V. Organ-specific toxicity of magnetic iron oxide-based nanoparticles. Nanotoxicology 2021, 15, 167–204.
  4. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63.
  5. Präbst, K.; Engelhardt, H.; Ringgeler, S.; Hübner, H. Basic Colorimetric Proliferation Assays: MTT, WST, and Resazurin. In Cell Viability Assays. Methods in Molecular Biology; Springer: Cham, Switzerland, 2017; pp. 1–17.
  6. Attarilar, S.; Yang, J.; Ebrahimi, M.; Wang, Q.; Liu, J.; Tang, Y.; Yang, J. The Toxicity Phenomenon and the Related Occurrence in Metal and Metal Oxide Nanoparticles: A Brief Review From the Biomedical Perspective. Front. Bioeng. Biotechnol. 2020, 8, 822.
  7. Liu, G.; Gao, J.; Ai, H.; Chen, X. Applications and potential toxicity of magnetic iron oxide nanoparticles. Small 2013, 9, 1533–1545.
  8. Abakumov, M.A.; Semkina, A.S.; Skorikov, A.S.; Vishnevskiy, D.A.; Ivanova, A.V.; Mironova, E.; Davydova, G.A.; Majouga, A.G.; Chekhonin, V.P. Toxicity of iron oxide nanoparticles: Size and coating effects. J. Biochem. Mol. Toxicol. 2018, 32, e22225.
  9. Żuk, M.; Podgórski, R.; Ruszczyńska, A.; Ciach, T.; Majkowska-Pilip, A.; Bilewicz, A.; Krysiński, P. Multifunctional Nanoparticles Based on Iron Oxide and Gold-198 Designed for Magnetic Hyperthermia and Radionuclide Therapy as a Potential Tool for Combined HER2-Positive Cancer Treatment. Pharmaceutics 2022, 14, 1680.
  10. De Simone, U.; Roccio, M.; Gribaldo, L.; Spinillo, A.; Caloni, F.; Coccini, T. Human 3D cultures as models for evaluating magnetic nanoparticle CNS cytotoxicity after short- and repeated long-term exposure. Int. J. Mol. Sci. 2018, 19, 1993.
  11. Kappes, M.; Friedrich, B.; Pfister, F.; Huber, C.; Friedrich, R.P.; Stein, R.; Braun, C.; Band, J.; Schreiber, E.; Alexiou, C.; et al. Superparamagnetic Iron Oxide Nanoparticles for Targeted Cell Seeding: Magnetic Patterning and Magnetic 3D Cell Culture. Adv. Funct. Mater. 2022, 32, 2203672.
  12. Nguyen, K.; Nuß, B.; Mühlberger, M.; Unterweger, H.; Friedrich, R.P.; Alexiou, C.; Janko, C. Superparamagnetic iron oxide nanoparticles carrying chemotherapeutics improve drug efficacy in monolayer and spheroid cell culture by enabling active accumulation. Nanomaterials 2020, 10, 1577.
  13. Anisimov, R.A.; Gorin, D.A.; Abalymov, A.A. 3D Cell Spheroids as A Tool for Evaluating the Effectiveness of Carbon Nanotubes as A Drug Delivery and Photothermal Therapy Agents. C 2022, 8, 56.
  14. Henrique, R.B.L.; Lima, R.R.M.; Monteiro, C.A.P.; Oliveira, W.F.; Pereira, G.; Cabral Filho, P.E.; Fontes, A. Advances in the study of spheroids as versatile models to evaluate biological interactions of inorganic nanoparticles. Life Sci. 2022, 302, 120657.
  15. Juarez-Moreno, K.; Chávez-García, D.; Hirata, G.; Vazquez-Duhalt, R. Monolayer (2D) or spheroids (3D) cell cultures for nanotoxicological studies? Comparison of cytotoxicity and cell internalization of nanoparticles. Toxicol. Vitr. 2022, 85, 105461.
  16. Hepel, M. Magnetic nanoparticles for nanomedicine. Magnetochemistry 2020, 6, 3.
  17. Canaparo, R.; Foglietta, F.; Limongi, T.; Serpe, L. Biomedical applications of reactive oxygen species generation by metal nanoparticles. Materials 2021, 14, 53.
  18. Chubarov, A.S. Serum Albumin for Magnetic Nanoparticles Coating. Magnetochemistry 2022, 8, 13.
  19. Issa, B.; Obaidat, I.M.; Albiss, B.A.; Haik, Y. Magnetic nanoparticles: Surface effects and properties related to biomedicine applications. Int. J. Mol. Sci. 2013, 14, 21266–21305.
  20. Shabatina, T.I.; Vernaya, O.I.; Shabatin, V.P.; Melnikov, M.Y. Magnetic nanoparticles for biomedical purposes: Modern trends and prospects. Magnetochemistry 2020, 6, 30.
  21. Nelson, N.; Port, J.; Pandey, M. Use of Superparamagnetic Iron Oxide Nanoparticles (SPIONs) via Multiple Imaging Modalities and Modifications to Reduce Cytotoxicity: An Educational Review. J. Nanotheranostics 2020, 1, 105–135.
  22. Mendes, B.B.; Conniot, J.; Avital, A.; Yao, D.; Jiang, X.; Zhou, X.; Sharf-Pauker, N.; Xiao, Y.; Adir, O.; Liang, H.; et al. Nanodelivery of nucleic acids. Nat. Rev. Methods Prim. 2022, 2, 1–21.
  23. Huang, R.-Y.; Liu, Z.-H.; Weng, W.-H.; Chang, C.-W. Magnetic nanocomplexes for gene delivery applications. J. Mater. Chem. B 2021, 9, 4267–4286.
  24. Sizikov, A.A.; Kharlamova, M.V.; Nikitin, M.P.; Nikitin, P.I.; Kolychev, E.L. Nonviral locally injected magnetic vectors for in vivo gene delivery: A review of studies on magnetofection. Nanomaterials 2021, 11, 1078.
  25. Laurent, N.; Sapet, C.; Gourrierec, L.L.; Bertosio, E.; Zelphati, O. Nucleic acid delivery using magnetic nanoparticles: The MagnetofectionTM technology. Ther. Deliv. 2011, 2, 471–482.
  26. Kami, D.; Takeda, S.; Itakura, Y.; Gojo, S.; Watanabe, M.; Toyoda, M. Application of magnetic nanoparticles to gene delivery. Int. J. Mol. Sci. 2011, 12, 3705–3722.
  27. Bakshi, S.; Zakharchenko, A.; Minko, S.; Kolpashchikov, D.; Katz, E. Towards Nanomaterials for Cancer Theranostics: A System of DNA-Modified Magnetic Nanoparticles for Detection and Suppression of RNA Marker in Cancer Cells. Magnetochemistry 2019, 5, 24.
  28. Kenchegowda, M.; Rahamathulla, M.; Hani, U.; Begum, M.Y.; Guruswamy, S.; Osmani, R.A.M.; Gowrav, M.P.; Alshehri, S.; Ghoneim, M.M.; Alshlowi, A.; et al. Smart Nanocarriers as an Emerging Platform for Cancer Therapy: A Review. Molecules 2022, 27, 146.
  29. Leach, J.C.; Wang, A.; Ye, K.; Jin, S. A RNA-DNA hybrid aptamer for nanoparticle-based prostate tumor targeted drug delivery. Int. J. Mol. Sci. 2016, 17, 380.
  30. Taghavi Pourianazar, N.; Gunduz, U. CpG oligodeoxynucleotide-loaded PAMAM dendrimer-coated magnetic nanoparticles promote apoptosis in breast cancer cells. Biomed. Pharmacother. 2016, 78, 81–91.
  31. Bassetto, M.; Sen, M.; Poulhes, F.; Arango-Gonzalez, B.; Bonvin, E.; Sapet, C.; Ueffing, M.; Zelphati, O. New Method for Efficient siRNA Delivery in Retina Explants: Reverse Magnetofection. Bioconjug. Chem. 2021, 32, 1078–1093.
  32. Gozuacik, D.; Akkoc, Y.; Kosar, A.; Dogan-Ekici, A.I.; Ekici, S. Anticancer Use of Nanoparticles as Nucleic Acid Carriers. J. Biomed. Nanotechnol. 2014, 10, 1751–1783.
  33. Creţu, B.E.B.; Dodi, G.; Shavandi, A.; Gardikiotis, I.; Şerban, I.L.; Balan, V. Imaging constructs: The rise of iron oxide nanoparticles. Molecules 2021, 26, 3437.
  34. Ganapathe, L.S.; Mohamed, M.A.; Yunus, R.M.; Berhanuddin, D.D. Magnetite (Fe3O4) nanoparticles in biomedical application: From synthesis to surface functionalisation. Magnetochemistry 2020, 6, 68.
  35. Baki, A.; Remmo, A.; Löwa, N.; Wiekhorst, F.; Bleul, R. Albumin-coated single-core iron oxide nanoparticles for enhanced molecular magnetic imaging (Mri/mpi). Int. J. Mol. Sci. 2021, 22, 6235.
  36. Kostevšek, N. A review on the optimal design of magnetic nanoparticle-based t2 mri contrast agents. Magnetochemistry 2020, 6, 11.
  37. Wallyn, J.; Anton, N.; Vandamme, T.F. Synthesis, principles, and properties of magnetite nanoparticles for in vivo imaging applications—A review. Pharmaceutics 2019, 11, 601.
  38. Ellis, C.M.; Pellico, J.; Davis, J.J. Magnetic Nanoparticles Supporting Bio-responsive T1/T2 Magnetic Resonance Imaging. Materials 2019, 12, 4096.
  39. Bruno, F.; Granata, V.; Bellisari, F.C.; Sgalambro, F.; Tommasino, E.; Palumbo, P.; Arrigoni, F.; Cozzi, D.; Grassi, F.; Brunese, M.C.; et al. Advanced Magnetic Resonance Imaging (MRI) Techniques: Technical Principles and Applications in Nanomedicine. Cancers 2022, 14, 1626.
  40. Obaidat, I.M.; Narayanaswamy, V.; Alaabed, S.; Sambasivam, S.; Muralee Gopi, C.V.V. Principles of Magnetic Hyperthermia: A Focus on Using Multifunctional Hybrid Magnetic Nanoparticles. Magnetochemistry 2019, 5, 67.
  41. Gavilán, H.; Simeonidis, K.; Myrovali, E.; Mazarío, E.; Chubykalo-Fesenko, O.; Chantrell, R.; Balcells, L.; Angelakeris, M.; Morales, M.P.; Serantes, D. How size, shape and assembly of magnetic nanoparticles give rise to different hyperthermia scenarios. Nanoscale 2021, 13, 15631–15646.
  42. Mittal, A.; Roy, I.; Gandhi, S. Magnetic Nanoparticles: An Overview for Biomedical Applications. Magnetochemistry 2022, 8, 107.
  43. Xie, J.; Chen, K.; Huang, J.; Lee, S.; Wang, J.; Gao, J.; Li, X.; Chen, X. PET/NIRF/MRI triple functional iron oxide nanoparticles. Biomaterials 2010, 31, 3016–3022.
  44. Wang, X.; Tu, M.; Tian, B.; Yi, Y.; Wei, Z.Z.; Wei, F. Synthesis of tumor-targeted folate conjugated fluorescent magnetic albumin nanoparticles for enhanced intracellular dual-modal imaging into human brain tumor cells. Anal. Biochem. 2016, 512, 8–17.
  45. Jiao, W.; Zhang, T.; Peng, M.; Yi, J.; He, Y.; Fan, H. Design of Magnetic Nanoplatforms for Cancer Theranostics. Biosensors 2022, 12, 38.
  46. Li, P.; Li, M.; Yue, D.; Chen, H. Solid-phase extraction methods for nucleic acid separation. A review. J. Sep. Sci. 2022, 45, 172–184.
  47. Wang, L.; He, K.; Sadak, O.; Wang, X.; Wang, Q.; Xu, X. Visual detection of in vitro nucleic acid replication by submicro- and nano-sized materials. Biosens. Bioelectron. 2020, 169, 112602.
  48. Gessner, I.; Fries, J.W.U.; Brune, V.; Mathur, S. Magnetic nanoparticle-based amplification of microRNA detection in body fluids for early disease diagnosis. J. Mater. Chem. B 2021, 9, 9–22.
  49. Pang, Y.; Wang, C.; Wang, J.; Sun, Z.; Xiao, R.; Wang, S. Fe3O4@Ag magnetic nanoparticles for microRNA capture and duplex-specific nuclease signal amplification based SERS detection in cancer cells. Biosens. Bioelectron. 2016, 79, 574–580.
  50. Pivetal, J.; Frénéa-Robin, M.; Haddour, N.; Vézy, C.; Zanini, L.F.; Ciuta, G.; Dempsey, N.M.; Dumas-Bouchiat, F.; Reyne, G.; Bégin-Colin, S.; et al. Development and applications of a DNA labeling method with magnetic nanoparticles to study the role of horizontal gene transfer events between bacteria in soil pollutant bioremediation processes. Environ. Sci. Pollut. Res. 2015, 22, 20322–20327.
  51. Ali, R.S.; Meng, H.; Li, Z. Zinc-Based Metal-Organic Frameworks in Drug Delivery, Cell Imaging, and Sensing. Molecules 2022, 27, 100.
  52. Hossain, S.; Rahman, M.; Nahar, Y.; Rahman, A.; Sharafat, M.K.; Hossain, M.; Ochiai, B.; Elaissari, A.; Ahmad, H. A simple in situ synthesis of iron oxide magnetic nanoparticles embedded in thermosensitive polymer for DNA capture. J. Mater. Res. 2020, 35, 2441–2450.
  53. Damavandi, F.; Wang, W.; Shen, W.Z.; Cetinel, S.; Jordan, T.; Jovel, J.; Montemagno, C.; Wong, G.K.S. Enrichment of low abundance DNA/RNA by oligonucleotide-clicked iron oxide nanoparticles. Sci. Rep. 2021, 11, 1–10.
  54. Li, B.; Mou, X.; Chen, Z.; Chen, H.; Deng, Y.; Li, S.; Su, E.; He, L.; He, N. The development of a rapid high-quality universal nucleic acid extraction kit based on magnetic separation. Sci. China Chem. 2017, 60, 1602–1608.
  55. Pinchon, E.; Leon, F.; Temurok, N.; Morvan, F.; Vasseur, J.J.; Clot, M.; Foulongne, V.; Cantaloube, J.F.; Perre, P.V.; Daynès, A.; et al. Rapid and specific DNA detection by magnetic field-enhanced agglutination assay. Talanta 2020, 219, 121344.
  56. Camacho-Fernández, J.C.; Jin, M.; Liu, X.; Berg, A.V.D.; Zhou, G. Ultrasensitive DNA detection based on two-step quantitative amplification on magnetic nanoparticles. Nanotechnology 2016, 27, 335102.
  57. Yang, Z.; Shen, B.; Yue, L.; Miao, Y.; Hu, Y.; Ouyang, R. Application of Nanomaterials to Enhance Polymerase. Molecules 2022, 27, 8854.
  58. Frenea-Robin, M.; Marchalot, J. Basic Principles and Recent Advances in Magnetic Cell Separation. Magnetochemistry 2022, 8, 11.
  59. Krishnan, S.; Goud, K.Y. Magnetic Particle Bioconjugates: A Versatile Sensor Approach. Magnetochemistry 2019, 5, 64.
  60. Rocha-Santos, T.A.P. Sensors and biosensors based on magnetic nanoparticles. TrAC Trends Anal. Chem. 2014, 62, 28–36.
  61. Sayad, A.; Skafidas, E.; Kwan, P. Magneto-impedance biosensor sensitivity: Effect and enhancement. Sensors 2020, 20, 5213.
  62. Ramin, N.A.; Ramachandran, M.R.; Saleh, N.M.; Mat Ali, Z.M.; Asman, S. Magnetic Nanoparticles Molecularly Imprinted Polymers: A Review. Curr. Nanosci. 2022, 18, 1–29.
  63. Meseguer-Lloret, S.; Torres-Cartas, S.; Gómez-Benito, C.; Herrero-Martínez, J.M. Magnetic molecularly imprinted polymer for the simultaneous selective extraction of phenoxy acid herbicides from environmental water samples. Talanta 2022, 239, 123082.
  64. Fresco-Cala, B.; Batista, A.D.; Cárdenas, S. Molecularly imprinted polymer micro- And nano-particles: A review. Molecules 2020, 25, 4740.
  65. Dmitrienko, E.V.; Pyshnaya, I.A.; Martyanov, O.N.; Pyshnyi, D.V. Molecularly imprinted polymers for biomedical and biotechnological applications. Russ. Chem. Rev. 2016, 85, 513–536.
  66. Ariani, M.D.; Zuhrotun, A.; Manesiotis, P.; Hasanah, A.N. Magnetic Molecularly Imprinted Polymers: An Update on Their Use in the Separation of Active Compounds from Natural Products. Polymers 2022, 14, 1389.
  67. Dmitrienko, E.V.; Bulushev, R.D.; Haupt, K.; Kosolobov, S.S.; Latyshev, A.V.; Pyshnaya, I.A.; Pyshnyi, D.V. A simple approach to prepare molecularly imprinted polymers from nylon-6. J. Mol. Recognit. 2013, 26, 368–375.
  68. Dinc, M.; Esen, C.; Mizaikoff, B. Recent advances on core–shell magnetic molecularly imprinted polymers for biomacromolecules. Trends Anal. Chem. 2019, 114, 202–217.
  69. Dinc, M.; Basan, H.; Diemant, T.; Behm, R.J.; Lindén, M.; Mizaikoff, B. Inhibitor-assisted synthesis of silica-core microbeads with pepsin-imprinted nanoshells. J. Mater. Chem. B 2016, 4, 4462–4469.
  70. Gao, S.; Wang, W.; Wang, B. Molecularly imprinted polymers as recognition elements in optical sensors. In Molecularly Imprinted Materials; CRC: Boca Raton, FL, USA, 2008; pp. 701–726.
  71. Huynh, T.P.; Pieta, P.; D’Souza, F.; Kutner, W. Molecularly imprinted polymer for recognition of 5-fluorouracil by RNA-type nucleobase pairing. Anal. Chem. 2013, 85, 8304–8312.
  72. Babamiri, B.; Salimi, A.; Hallaj, R. A molecularly imprinted electrochemiluminescence sensor for ultrasensitive HIV-1 gene detection using EuS nanocrystals as luminophore. Biosens. Bioelectron. 2018, 117, 332–339.
  73. Slinchenko, O.; Rachkov, A.; Miyachi, H.; Ogiso, M.; Minoura, N. Imprinted polymer layer for recognizing double-stranded DNA. Biosens. Bioelectron. 2004, 20, 1091–1097.
  74. Huang, L.; Wang, X.; Xie, X.; Xie, W.; Li, X.; Gong, X.; Long, S.; Guo, H.; Liu, Z. Synthesis and DNA Adsorption of Poly(2-Vinyl-4,6-Diamino-1,3,5-Triazine) Coated Polystyrene Microspheres. J. Wuhan Univ. Technol. Mater. Sci. Ed. 2018, 33, 999–1006.
  75. Rutkowska, M.; Płotka-Wasylka, J.; Morrison, C.; Wieczorek, P.P.; Namieśnik, J.; Marć, M. Application of molecularly imprinted polymers in analytical chiral separations and analysis. Trends Anal. Chem. 2018, 102, 91–102.
  76. Ding, X.; Heiden, P.A. Recent developments in molecularly imprinted nanoparticles by surface imprinting techniques. Macromol. Mater. Eng. 2014, 299, 268–282.
  77. Whitcombe, M.J.; Kirsch, N.; Nicholls, I.A. Molecular imprinting science and technology: A survey of the literature for the years 2004–2011. J. Mol. Recognit. 2014, 27, 297–401.
  78. Garnier, M.; Sabbah, M.; Ménager, C.; Griffete, N. Hybrid molecularly imprinted polymers: The future of nanomedicine? Nanomaterials 2021, 11, 3091.
  79. Niu, M.; Pham-Huy, C.; He, H. Core-shell nanoparticles coated with molecularly imprinted polymers: A review. Microchim. Acta 2016, 183, 2677–2695.
  80. Huang, S.; Xu, J.; Zheng, J.; Zhu, F.; Xie, L.; Ouyang, G. Synthesis and application of magnetic molecularly imprinted polymers in sample preparation. Anal. Bioanal. Chem. 2018, 410, 3991–4014.
  81. Li, J.; Wang, Y.; Yu, X. Magnetic Molecularly Imprinted Polymers: Synthesis and Applications in the Selective Extraction of Antibiotics. Front. Chem. 2021, 9, 1–17.
  82. Li, J.; Zhou, Q.; Yuan, Y.; Wu, Y. Iron-based magnetic molecular imprinted polymers and their application in removal and determination of di-n-pentyl phthalate in aqueous media. R. Soc. Open Sci. 2017, 4, 170672.
  83. Goyal, G.; Bhakta, S.; Mishra, P. Surface Molecularly Imprinted Biomimetic Magnetic Nanoparticles for Enantioseparation. ACS Appl. Nano Mater. 2019, 2, 6747–6756.
  84. Dai, Q.; Wang, Y.; Xu, W.; Liu, Y.; Zhou, Y. Adsorption and specific recognition of DNA by using imprinted polymer layers grafted onto ionic liquid functionalized magnetic microspheres. Microchim. Acta 2017, 184, 4433–4441.
  85. Sanadgol, N.; Wackerlig, J. Developments of smart drug-delivery systems based on magnetic molecularly imprinted polymers for targeted cancer therapy: A short review. Pharmaceutics 2020, 12, 831.
  86. Nawaz, N.; Abu Bakar, N.K.; Mahmud, H.N.M.E.; Jamaludin, N.S. Molecularly imprinted polymers-based DNA biosensors. Anal. Biochem. 2021, 630, 114328.
  87. Zhang, Z.; Liu, J. Molecularly Imprinted Polymers with DNA Aptamer Fragments as Macromonomers. ACS Appl. Mater. Interfaces 2016, 8, 6371–6378.
  88. Brahmbhatt, H.; Poma, A.; Pendergraff, H.M.; Watts, J.K.; Turner, N.W. Improvement of DNA recognition through molecular imprinting: Hybrid oligomer imprinted polymeric nanoparticles (oligoMIP NPs). Biomater. Sci. 2016, 4, 281–287.
  89. Zhang, Z.; Liu, J. Molecular Imprinting with Functional DNA. Small 2019, 15, 1805246.
  90. Yazdanparast, S.; Benvidi, A.; Azimzadeh, M.; Tezerjani, M.D.; Ghaani, M.R. Experimental and theoretical study for miR-155 detection through resveratrol interaction with nucleic acids using magnetic core-shell nanoparticles. Microchim. Acta 2020, 187, 1–10.
  91. Wang, L.; Liu, Z.J.; Cao, H.X.; Liang, G.X. Ultrasensitive colorimetric miRNA detection based on magnetic 3D DNA walker and unmodified AuNPs. Sens. Actuators B Chem. 2021, 337, 3–9.
  92. Masud, M.K.; Umer, M.; Hossain, M.S.A.; Yamauchi, Y.; Nguyen, N.T.; Shiddiky, M.J.A. Nanoarchitecture Frameworks for Electrochemical miRNA Detection. Trends Biochem. Sci. 2019, 44, 433–452.
  93. Chen, Y.; Liu, Y.; Shi, Y.; Ping, J.; Wu, J.; Chen, H. Magnetic particles for integrated nucleic acid purification, amplification and detection without pipetting. TrAC Trends Anal. Chem. 2020, 127, 115912.
  94. Li, P.; Li, M.; Zhang, F.; Wu, M.; Jiang, X.; Ye, B.; Zhao, Z.; Yue, D.; Fan, Q.; Chen, H. High-efficient nucleic acid separation from animal tissue samples via surface modified magnetic nanoparticles. Sep. Purif. Technol. 2021, 262, 118348.
  95. Yang, Q.; Dong, Y.; Qiu, Y.; Yang, X.; Cao, H.; Wu, Y. Design of Functional Magnetic Nanocomposites for Bioseparation. Colloids Surf. B Biointerfaces 2020, 191, 111014.
  96. Mdlovu, N.V.; Lin, K.S.; Chen, Y.; Wu, C.M. Formulation of magnetic nanocomposites for intracellular delivery of micro-RNA for MYCN inhibition in neuroblastoma. Colloids Surf. A Physicochem. Eng. Asp. 2021, 615, 126264.
  97. Lawai, V.; Ngaini, Z. Chitosan magnetic nanocomposites for gene delivery. In Polysaccharide-Based Nanocomposites for Gene Delivery and Tissue Engineering; Woodhead: Sawston, UK, 2021; p. 335.
  98. Do, H.D.; Ménager, C.; Michel, A.; Seguin, J.; Korichi, T.; Dhotel, H.; Marie, C.; Doan, B.T.; Mignet, N. Development of theranostic cationic liposomes designed for image-guided delivery of nucleic acid. Pharmaceutics 2020, 12, 854.
  99. Tang, C.; He, Z.; Liu, H.; Xu, Y.; Huang, H.; Yang, G.; Xiao, Z.; Li, S.; Liu, H.; Deng, Y.; et al. Application of magnetic nanoparticles in nucleic acid detection. J. Nanobiotechnol. 2020, 18, 1–19.
  100. Szymczyk, A.; Drozd, M.; Kamińska, A.; Matczuk, M.; Trzaskowski, M.; Mazurkiewicz-Pawlicka, M.; Ziółkowski, R.; Malinowska, E. Comparative Evaluation of Different Surface Coatings of Fe3O4-Based Magnetic Nano Sorbent for Applications in the Nucleic Acids Extraction. Int. J. Mol. Sci. 2022, 23, 8860.
  101. Emaus, M.N.; Varona, M.; Eitzmann, D.R.; Hsieh, S.A.; Zeger, V.R.; Anderson, J.L. Nucleic acid extraction: Fundamentals of sample preparation methodologies, current advancements, and future endeavors. TrAC Trends Anal. Chem. 2020, 130, 115985.
  102. Zhang, M.; Li, L.; Li, B.; Tian, N.; Yang, M.; Zhang, H.; You, C.; Zhang, J. Adsorption of DNA by using polydopamine modified magnetic nanoparticles based on solid-phase extraction. Anal. Biochem. 2019, 579, 9–17.
  103. Bulgakova, A.; Chubarov, A.; Dmitrienko, E. Magnetic Nylon 6 Nanocomposites for the Microextraction of Nucleic Acids from Biological Samples. Magnetochemistry 2022, 8, 85.
  104. Zheng, H.; Lin, H.; Chen, X.; Sui, J.; Ullah Khan, M.; Ramesh Pavase, T.; Han, X.; Cao, L. Tailor-made magnetic nanocomposite with pH and thermo-dual responsive copolymer brush for bacterial separation. Food Chem. 2021, 358, 129907.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 315
Revisions: 2 times (View History)
Update Date: 29 Jan 2023
1000/1000