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 -- 2393 2023-05-02 20:54:05 |
2 format change Meta information modification 2393 2023-05-04 04:39:09 |

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.
Islam, T.; Rahaman, M.M.; Mia, M.N.; Ara, I.; Islam, M.T.; Alam Riaz, T.; Araújo, A.C.J.; De Lima Silva, J.M.F.; De Lacerda, B.C.G.V.; De Andrade, E.M.; et al. Therapeutic Perspectives of Metal Nanoformulations. Encyclopedia. Available online: https://encyclopedia.pub/entry/43685 (accessed on 18 August 2024).
Islam T, Rahaman MM, Mia MN, Ara I, Islam MT, Alam Riaz T, et al. Therapeutic Perspectives of Metal Nanoformulations. Encyclopedia. Available at: https://encyclopedia.pub/entry/43685. Accessed August 18, 2024.
Islam, Tawhida, Md. Mizanur Rahaman, Md. Nayem Mia, Iffat Ara, Md. Tariqul Islam, Thoufiqul Alam Riaz, Ana C. J. Araújo, João Marcos Ferreira De Lima Silva, Bruna Caroline Gonçalves Vasconcelos De Lacerda, Edlane Martins De Andrade, et al. "Therapeutic Perspectives of Metal Nanoformulations" Encyclopedia, https://encyclopedia.pub/entry/43685 (accessed August 18, 2024).
Islam, T., Rahaman, M.M., Mia, M.N., Ara, I., Islam, M.T., Alam Riaz, T., Araújo, A.C.J., De Lima Silva, J.M.F., De Lacerda, B.C.G.V., De Andrade, E.M., Khan, M.A., Coutinho, H.D.M., Husain, Z., & Islam, M.T. (2023, May 02). Therapeutic Perspectives of Metal Nanoformulations. In Encyclopedia. https://encyclopedia.pub/entry/43685
Islam, Tawhida, et al. "Therapeutic Perspectives of Metal Nanoformulations." Encyclopedia. Web. 02 May, 2023.
Therapeutic Perspectives of Metal Nanoformulations
Edit

Acceptance of nanoparticles (NPs) in therapeutic applications has increased because of their outstanding physicochemical features. By overcoming the drawbacks of conventional therapy, the utilization of metal NPs, metal-oxide, or metal supported nanomaterials have shown to have significant therapeutic applications in medicine. This is proved by a lot of clinical and laboratory investigations that show improved treatment outcomes, site-specific drug delivery, and fewer side effects compared to traditional medicine. The metal NPs interaction with living cells (animal and plant) showed many ways to develop therapeutic models with the NPs.

metal nanoparticles nanotherapy therapeutic uses

1. Introduction

The implementation of nanoparticles (NPs) for the treatment and diagnosis of disease is a revolutionary concept that has been developed over the past few decades. The nanotechnological approach can be divided into two branches: one is nanodevices and the other is nanomaterials. The nanodevice can be defined as such tiny devices at the nanoscale range, which includes microarrays and some devices such as respirocytes [1][2][3].
Particles smaller than 100 nanometers (nm) in any one of the dimensions are considered nanomaterials. Biomedical science found successful result by using nanoparticles as therapeutic agents in the treatment of various diseases. As it is selective on the target organ and receptors, it overcomes several limitations of conventional therapy, such as nonspecificity, unwanted side effects, less efficiency, and low bioavailability [4].
Therefore, current research projects are considerably more focused on developing and designing new drug delivery systems, and the most promising area is ensured by NPs for their uniqueness in biological and physicochemical characteristics, as they can deliver molecules to specific locations in the body [5].
The therapeutic molecules which are insoluble in water can be complexed with NPs, resulting in greater bioavailability and significantly fewer physiological barriers; for example, NP carriers assist medication in passing the blood–brain barrier (BBB) [6][7][8]. Nevertheless, because it is targeted, it will require lower doses than conventional therapy, and the therapeutic index will be higher as it will minimize the toxicity in the biological system. The utilization of NPs in various fields of the health sector is possible because of their ability to provide a visual image of the targeted delivery location by using some agents; moreover, their pathway can be tracked.
Several studies have recently focused on the method of producing metal NPs using green synthesis, which has shown positive results against pathogens, cancer cells, helminths, fungi, etc. using the metal NPs Zn, Ag, Au, Pt, Mn, Ni, and Ti [9]. Currently, among other NPs, Ag-NPs are one of the top listed compounds being researched [10]. In 1857, Michael Faraday was the first person to study Au-NPs in a colloidal system and report Au-NP’s optical features [11].

2. Therapeutic Applications of Metal NPs

2.1. Therapeutic Interventions of Gold Nanoparticles (Au-NPs)

When Robert Koch discovered that gold cyanide had a bacteriostatic effect on Mycobacterium TB, the medical use of gold for the treatment of tuberculosis was established for the first time. This led to the introduction of gold as a medicine in the 1920s [12].
Au-NPs have a tendency to aggregate at tumor sites [13]. Tumor cells can be killed by Au-NPs in a variety of ways, including as drug delivery systems for mechanical damage, anticancer medicines, and photothermal ablation [14].
In particular, Au-NPs are used in drug delivery, imaging, photo-thermal therapy, sensing, catalysis, and antimicrobials [15]. The list of applications of Au-NPs is much longer because of their unique properties. The biocompatibility of gold nanoparticles has been well documented; however, the typical reduction procedures used to create them can leave behind harmful chemical species [16]. Consequently, Au-NPs manufactured in an environmentally friendly manner hold far more promise in a variety of settings. Although Au-NPs are not as widely used as Ag-NPs as antibacterial agents, they nonetheless have considerable impact against a wide range of diseases due to their inherent biocidal qualities [15][17].
Au-NPs of 60 nm showed a positive result in retinoblastoma treatment [18], Au nanopopcorn 28 nm in size is used to diagnose prostate and breast cancer [19], and Au nanostars (Au-NS) 30 and 60 nm in size can be used to identify brain tumors, and this same NP showed a satisfying result against bladder cancer [20].
Silica-coated Au nanorods showed effective antitumor activity, both in vivo and in vitro, against breast cancer by targeting CD44+ receptors [21]. Colloidal Au-NPs are of interest as nontoxic carriers for drug delivery [22][23][24]. In a study, it was found that the internalization of the 50 nm spherical gold nanoparticles (AuNPs) was the best of all the nanoparticles investigated [25]. TrxR (thioredoxin reductase) function can be inhibited by gold compounds, which causes tumor cells to accumulate reactive oxygen species (ROS) and experience oxidative stress, which ultimately kills the tumor cells [26][27] and the proposed anticancer mechanism of Au-NPs is illustrated in Figure 1.
Figure 1. Proposed anticancer mechanism of gold nanoparticles. Here, Au-NPs pass through the cancer cell membrane by endocytosis, and endosomal release causes ROS (reactive oxygen species) production. These ROS cause mitochondrial dysfunction and result in caspase 3, 9, and 8 activations, which results in DNA damage and finally cell death [26][27][28][29].
  • Nanotherapeutic Application of Gold

2.2. Therapeutic Interventions of Silver Nanoparticles (Ag-NPs)

Silver has excellent physicochemical features, such as catalytic, optical, electric, and, of course, antibacterial capabilities, and these qualities make silver nanoparticles the most marketable nanoparticles. In the presence of Ag-NPs, the synergistic impact of antibiotics such as cefotaxime, azithromycin, cefuroxime, chloramphenicol, and fosfomycin against E. coli was greatly boosted as compared to antibiotics alone [30].
Other metal NPs may exhibit equivalent efficacy against particular germs, but overall, silver is said to be the most effective material against a variety of pathogens. Ag-NPs inhibit the extracellular activity of severe acute respiratory distress syndrome coronavirus 2 (SARS-CoV-2) [31].
Ag-NPs are the preferred metal when antibacterial characteristics are required. The antibacterial, antiviral, antioxidant, and anticancer characteristics of silver are well recognized, and it has the potential to be developed into a unique therapeutic agent. Ag also has antiparasitic, antiviral, and anticancer qualities [32][33], and the mechanisms of action of these effects are illustrated in Figure 2. Ag-NPs, after entering cells by endocytosis, produce ROS that damage the endoplasmic reticulum and mitochondria. The cellular pathways NF-kB, PI3K/AKT/mTOR, Wnt/beta-catenin, MAPK/ERK, and ERK activation result in DNA fragmentation, cell cycle arrest, and cell apoptosis [34][35][36][37][38]
Figure 2. Proposed anticancer mechanisms of silver nanoparticles (NF-kB: nuclear factor kappa-light-chain-enhancer of activated B cells; PI3K: phosphoinositide 3-kinases AKT: protein kinase B; mTOR: mammalian target of rapamycin; Wnt: wingless and Int-1; MAPK: mitogen-activated protein kinase; ERK: extra-cellular receptor kinase).
  • Nanotherapeutic Application of Silver

2.3. Therapeutic Interventions of Copper Nanoparticles (Cu-NPs)

Researchers and health care professionals have been drawn to cupric oxide (CuO) NPs for their physical, chemical, high temperature, and photocatalytic capabilities, but most notably for their antibacterial properties [39]. Copper nanoparticles’ synergistic activity with amoxicillin, ampicillin, ciprofloxacin, and gentamicin against both Gram-positive and Gram-negative bacteria was investigated, and ampicillin showed comparatively improved activity compared to alone [40]. Cu-NPs inactivate glycosidase to provide an antidiabetic effect, and the study found that Cu-NPs showed an anticancer effect by activating BAX and p53 and by decreasing Bcl-2 expression, which result in apoptosis in cancer [41]. Cu-NPs increase ROS production in bacterial cells and cause bacterial DNA and protein destruction; on the other hand, accumulation of Cu-NPs in the bacterial cell wall causes cell wall disruption [42][43][44][45][46][47][48].
The mechanisms underlying these effects are depicted in Figure 3.
Figure 3. Proposed mechanism of nanotherapeutic applications of copper. Here Cu-NPs showed an anticancer effect by increasing BAX and p53 expression and Bcl-2 downregulating, an antidiabetic effect by glycosidase inactivation, an antimicrobial effect by ROS production cell wall disruption, and a larvicidal effect against Aedes aegypti (Dengue virus carrier).
  • Nanotherapeutic Application of Copper

2.4. Therapeutic Interventions of Zinc Nanoparticles (Zn-NPs)

Zinc is a material that is frequently used in biomedical applications due to its unique features, such as electric conductivity, optical capabilities, and piezoelectric qualities [49]. Beyth et al. defined the method of killing bacteria using zinc oxide (ZnO) NPs as having two pathways of action [50]. The first involves cell wall penetration, and the second includes the formation of ROS. Zn-NPs follow the Bcl-2/BAX/BAK pathway to cell apoptosis by caspase-3 and -9 and ROS-induced DNA fragmentation leading to cell cycle arrest and apoptosis, and also follow the mitochondrial disruption for an anticancer effect [51][52][53], as shown in Figure 4.
Figure 4. Proposed anticancer mechanism of Zn-NPs (ZnO-NPs create stress in endoplasmic reticulum, and produce ROS, which results DNA fragmentation and cell cycle arrest; on the other hand, produced ROS disrupts mitochondrial membrane and activates caspase 3, 7, and 9, which results in apoptosis).
ZnO-NPs have antibacterial, antifungal, anticancer, antidiabetic, and antitubercular activity, and breast cancer inhibition is an optimistic property that this research observed in a number of studies. Even 100 nm Zn-NPs supplemented at 30 ppm improved growth and serum glucose levels in layer chicks [54].
  • Nanotherapeutic Application of Zinc

2.5. Therapeutic Interventions of Nickel Nanoparticles (Ni-NPs)

Ni-NPs have anticancer action [55][56]. A complex structure of Qu–PEG–NiGs (48–72 nm), green synthesized by Ocimum sanctum leaf extract, showed mitochondrial-mediated apoptosis against the MCF-7 cell line [57], antimicrobial activity, antioxidant action, and activity against human ovarian cancer, liver and spleen injury [55][58][59][60], lung inflammation [61], human lung cancer [62], lymphatic filariasis [63], and larvicidal parasitic activity [64]. Bacterial protein leakage induced by ROS activation [65] and disruption of the cell membrane [66] is one way of causing bacterial cell death. The antimicrobial mechanism is shown in Figure 5. It has numerous other therapeutic properties in a single formulation or a complex formulation.
Figure 5. Antimicrobial mechanism of action of Ni-NPs. Ni-NPs cause ROS production that cause oxidative damage of the cell wall and destroy the membrane. ROS cause protein leakage and interrupt electron transport; these processes result in the antimicrobial effect of Ni-NPs.
  • Nanotherapeutic Application of Nickel

2.6. Therapeutic Interventions of Iron Nanoparticles (Fe-NPs)

Among the Fe-NPs, prominently used NPs include magnetite (Fe3O4), hematite, or iron (III) oxide (Fe2O3), and the less abundant iron (II) oxide (FeO) [67]. Magnetite (Fe3O4) NPs are used in biomedical applications due to their magnetic characteristics, biocompatibility, and, in particular, their superparamagnetic capabilities [68].
Magnetic NPs, also known as superparamagnetic iron oxide, are used in drug delivery [69][70] and hyperthermia therapy [71][72][73]. Magnetite NPs can produce receptive oxygen species (ROS), which kill microbes, making them a promising contender for an antimicrobial agent. Lung cancer cells terminated by ferroptosis as a result of Zerovalent Fe-NPs (ZVI-NPs) induce mitochondrial malfunction, intracellular oxidative stress, and lipid peroxidation; here, AMPK/mTOR activated by ZVI-NPs cause upregulation of GSK3/β-TrCP, which results in NRF2 degradation and ultimately results ferroptosis, which causes cancer cell damage [74][75][76][77][78], as shown in Figure 6.
Figure 6. Possible anticancer mechanisms of iron (Fe) nanoparticles (zerovalent Fe-NPs cause ROS production, AMPK/mTOR activation, NRF2 degradation by GSK3/β-TrCP, and mitochondrial disfunction, which results in ferroptosis.
Superparamagnetic iron oxide nanoparticles (SPIONs) provide action against the human breast cancer cell MCF7 [70].
In the treatment of different types of cancer, ferroptosis, a new Fe- and ROS-dependent form of controlled cell death, has received a lot of attention. The potential of ferroptosis in combination with NPs for cancer therapy is becoming more and more clear as a result of the development of nanomaterials [79]. After cells consume Fe-based NPs, an excess of iron ions released from the lysosome in an acidic environment activates the fenton reaction, which causes ROS formation and cell ferroptosis [80].
Importantly, when antibiotic drugs are coupled with the iron nanoparticles of neem extract, the dose of traditional antibiotics can be decreased by nearly half without affecting efficiency. As a result, the use of natural antibiotics aids in the reduction of regular antibiotic doses [81]. There was also a trial of producing bimetallic NPs (Ag-Fe) that established the synergistic antibacterial (bactericidal) impact of the two metals forming the bimetallic nanoparticles when compared to the effects of the monometallic nanoparticles against yeast and both Gram-positive and Gram-negative multidrug-resistant bacteria [82].
  • Nanotherapeutic Application of Iron

3. Metal Nanoparticles Elimination from Body

The elimination of NPs depends on their particle size, intrinsic biodegradability, core density, surface charge, and surface chemistry [83]. The liver is the major clearance organ in the oral administration of NPs. Intravenously administered NPs are cleared from the bloodstream by two main mechanisms: (i) renal elimination and (ii) hepatobiliary elimination. Choi et al. [84] reported that smaller-sized (<5.5 nm diameter) quantum dots undergo efficient urinary excretion due to the pore size limit of glomerular filtration in the kidneys. According to estimates of Si-NPs in rats, 7–8% of NPs were eliminated in urine and 75–80% were expelled in feces [85]. Nonbiodegradable and larger-sized (>5.5 nm) NPs are supposed to be eliminated through the hepatobiliary route. The hepatobiliary elimination involved the following pathways: (1) the liver sinusoid; (2) the space of Disse, a tiny perisinusoidal space containing blood plasma, nutrients, oxygen, and body waste that has become crucial in the treatment of liver disease, which is located between endothelial cells and hepatocytes; (3) hepatocytes; (4) bile ducts; (5) intestines; and finally (6) out of the body, as shown in Figure 7. In hepatobiliary elimination, the liver nonparenchymal cells (e.g., Kupffer cells and liver sinusoidal endothelial cells) influence and determine the elimination fate. The removal of Kupffer cells increased the fecal elimination of NPs by more than 10-fold [86].
Figure 7. Proposed metal nanoparticles hepatobiliary clearance pathway (when metal NPs pass through the liver sinusoid, they enter the space of Disse via Kupffer cells, and then enter the bile duct, followed by fecal elimination.
NPs can enter the body through multiple routes, including the skin, respiratory tract, dermal exposure, mucosal, oral, intravenous, subcutaneous, intramuscular, etc., and can induce acute or chronic toxicities [87]. The anionic NPs are less toxic than the cationic NPs, which cause hemolysis and clotting [88]. Singh et al. [89] reported that ceramic NPs, commonly used for drug delivery, exhibit oxidative stress and cytotoxic activity in the lungs, liver, heart, and brain, as well as having teratogenic or carcinogenic effects. NPs have been shown, both in vivo and in vitro, to increase cellular reactive oxygen species, induce multiple minor and severe toxicities, and even disrupt host homeostasis [87]. Although NPs are useful for numerous medical applications, there are still some concerns for ecosystems and living organisms due to their uncontrollable use and discharge to the natural environment; thus, it should be considered to make the use of NPs more convenient and environmentally friendly. Preclinical studies have revealed the importance of renal-clearable luminous metal NPs in cancer therapy, which offers tremendous promise for potential clinical translation [90]. The retention of NPs in the body, especially in the vital organs, usually depends on the density of the particles. In a study of gold and silver NPs by Tang et al., it was demonstrated that the lower-density metal NPs have a higher distribution and shorter retention time than the higher-density metal NPs [91].

References

  1. Chandrasekhar, S.; Iyer, L.K.; Panchal, J.P.; Topp, E.M.; Cannon, J.B.; Ranade, V.V. Microarrays and microneedle arrays for delivery of peptides, proteins, vaccines and other applications. Expert Opin. Drug Deliv. 2013, 10, 1155–1170.
  2. Rabl, P.; Kolkowitz, S.J.; Koppens, F.H.L.; Harris, J.G.E.; Zoller, P.; Lukin, M.D. A quantum spin transducer based on nanoelectromechanical resonator arrays. Nat. Phys. 2010, 6, 602–608.
  3. Shabnashmi, P.S.; Naga Kani, S.; Vithya, V.; Vijaya Lakshmi, B.; Jasmine, R. Therapeutic applications of nanorobots-respirocytes and microbivores. J. Chem. Pharm. Res. 2016, 8, 605–609.
  4. Kadam, R.S.; Bourne, D.W.; Kompella, U.B. Nano-advantage in enhanced drug delivery with biodegradable nanoparticles: Contribution of reduced clearance. Drug Metab. Dispos. 2012, 40, 1380–1388.
  5. Jahan, S.T.; Sadat, S.; Walliser, M.; Haddadi, A. Targeted therapeutic nanoparticles: An immense promise to fight against cancer. J. Drug Deliv. 2017, 2017, 1–24.
  6. Zhou, Y.; Peng, Z.; Seven, E.S.; Leblanc, R.M. Crossing the blood-brain barrier with nanoparticles. J. Control. Release 2018, 270, 290–303.
  7. Teleanu, D.M.; Chircov, C.; Grumezescu, A.M.; Volceanov, A.; Teleanu, R.I. Impact of nanoparticles on brain health: An up to date overview. J. Clin. Med. 2018, 7, 490.
  8. Rizvi, S.A.; Saleh, A.M. Applications of nanoparticle systems in drug delivery technology. Saudi Pharm. J. 2018, 26, 64–70.
  9. Thakkar, K.N.; Mhatre, S.S.; Parikh, R.Y. Biological synthesis of metallic nanoparticles. Nanomed. Nanotechnol. Biol. Med. 2010, 6, 257–262.
  10. Firdhouse, M.J.; Lalitha, P. Biosynthesis of silver nanoparticles and its applications. J. Nanotechnol. 2015, 2015, 18.
  11. Faraday, M.X. The Bakerian Lecture.—Experimental relations of gold (and other metals) to light. Philos. Trans. R. Soc. Lond. 1857, 147, 145–181.
  12. Daniel, M.C.; Astruc, D. Gold nanoparticles: Assembly, supramolecular chemistry, quantum-size-related properties, and applications toward biology, catalysis, and nanotechnology. Chem. Rev. 2004, 104, 293–346.
  13. Sun, W.; Karmakar, B.; Ibrahium, H.A.; Awwad, N.S.; El-Kott, A.F. Design and synthesis of nano Cu/chitosan-starch bio-composite for the treatment of human thyroid carcinoma. Arab. J. Chem. 2022, 15, 103465.
  14. Kodiha, M.; Wang, Y.M.; Hutter, E.; Maysinger, D.; Stochaj, U. Off to the organelles-killing cancer cells with targeted gold nanoparticles. Theranostics 2015, 5, 357.
  15. Basavegowda, N.; Idhayadhulla, A.; Lee, Y.R. Preparation of Au and Ag nanoparticles using Artemisia annua and their in vitro antibacterial and tyrosinase inhibitory activities. Mater. Sci. Eng. 2014, 43, 58–64.
  16. Bar, H.; Bhui, D.K.; Sahoo, G.P.; Sarkar, P.; Pyne, S.; Chattopadhyay, D.; Misra, A. Synthesis of gold nanoparticles of variable morphologies using aqueous leaf extracts of Cocculus hirsutus. J. Exp. Nanosci. 2012, 7, 109–119.
  17. Khan, A.U.; Yuan, Q.; Wei, Y.; Khan, G.M.; Khan, Z.U.H.; Khan, S.; Ali, F.; Tahir, K.; Ahmad, A.; Khan, F.U. Photocatalytic and antibacterial response of biosynthesized gold nanoparticles. J. Photochem. Photobiol. B Biol. 2016, 162, 273–277.
  18. Moradi, S.; Mokhtari-Dizaji, M.; Ghassemi, F.; Sheibani, S.; Amoli, F.A. The effect of ultrasound hyperthermia with gold nanoparticles on retinoblastoma Y79 cells. Gold Bull. 2020, 53, 111–120.
  19. Lu, W.; Singh, A.K.; Khan, S.A.; Senapati, D.; Yu, H.; Ray, P.C. Gold nano-popcorn-based targeted diagnosis, nanotherapy treatment, and in situ monitoring of photothermal therapy response of prostate cancer cells using surface-enhanced Raman spectroscopy. J. Am. Chem. Soc. 2010, 132, 18103–18114.
  20. Odion, R.; Liu, Y.; Vo-Dinh, T. Plasmonic gold nanostar-mediated photothermal immunotherapy. IEEE J. Sel. Top. Quantum Electron. 2021, 27, 4800109.
  21. Cheng, D.; Ji, Y.; Wang, B.; Wang, Y.; Tang, Y.; Fu, Y.; Xu, Y.; Qian, X.; Zhu, W. Dual-responsive nanohybrid based on degradable silica-coated gold nanorods for triple-combination therapy for breast cancer. Acta Biomat. 2021, 128, 435–446.
  22. Peng, C.; Xu, J.; Yu, M.; Ning, X.; Huang, Y.; Du, B.; Hernandez, E.; Kapur, P.; Hsieh, J.T.; Zheng, J. Tuning the in vivo transport of anticancer drugs using renal-clearable gold nanoparticles. Angew. Chem. 2019, 131, 8567–8571.
  23. Kalimuthu, K.; Lubin, B.C.; Bazylevich, A.; Gellerman, G.; Shpilberg, O.; Luboshits, G.; Firer, M.A. Gold nanoparticles stabilize peptide-drug-conjugates for sustained targeted drug delivery to cancer cells. J. Nanobiotechnol. 2018, 16, 34.
  24. Farooq, M.U.; Novosad, V.; Rozhkova, E.A.; Wali, H.; Ali, A.; Fateh, A.A.; Neogi, P.B.; Neogi, A.; Wang, Z. Gold nanoparticles-enabled efficient dual delivery of anticancer therapeutics to HeLa cells. Sci. Rep. 2018, 8, 2907.
  25. Chithrani, B.D.; Ghazani, A.A.; Chan, W.C. Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. Nano Lett. 2006, 6, 662–668.
  26. Sze, J.H.; Raninga, P.V.; Nakamura, K.; Casey, M.; Khanna, K.K.; Berners-Price, S.J.; Di Trapani, G.; Tonissen, K.F. Anticancer activity of a Gold (I) phosphine thioredoxin reductase inhibitor in multiple myeloma. Redox Biol. 2020, 28, 101310.
  27. Patil, M.P.; Kim, G.D. Eco-friendly approach for nanoparticles synthesis and mechanism behind antibacterial activity of silver and anticancer activity of gold nanoparticles. Appl. Microbiol. Biotechnol. 2017, 101, 79–92.
  28. Baharara, J.; Ramezani, T.; Divsalar, A.; Mousavi, M.; Seyedarabi, A. Induction of apoptosis by green synthesized gold nanoparticles through activation of caspase-3 and 9 in human cervical cancer cells. Avicenna J. Med. Biotechnol. 2016, 8, 75.
  29. Arshad, M.; Ozaslan, M.; Ali, H.K.; Safdar, M.; Junejo, Y.; Babar, M.E. Molecular Investigation of Gold Nanoparticles Toxicity in Mice Model and p53 Activation. J. Biol. Sci. 2019, 19, 391–395.
  30. Abo-Shama, U.H.; El-Gendy, H.; Mousa, W.S.; Hamouda, R.A.; Yousuf, W.E.; Hetta, H.F.; Abdeen, E.E. Synergistic and antagonistic effects of metal nanoparticles in combination with antibiotics against some reference strains of pathogenic microorganisms. Infect. Drug Resist. 2020, 2020, 351–362.
  31. Jeremiah, S.S.; Miyakawa, K.; Morita, T.; Yamaoka, Y.; Ryo, A. Potent antiviral effect of silver nanoparticles on SARS-CoV-2. Biochem. Biophys. Res. Commun. 2020, 533, 195–200.
  32. Lara, H.H.; Ayala-Nuñez, N.V.; Ixtepan-Turrent, L.; Rodriguez-Padilla, C. Mode of antiviral action of silver nanoparticles against HIV-1. J. Nanobiotechnol. 2010, 8, 1–10.
  33. Rajan, A.; Vilas, V.; Philip, D. Studies on catalytic, antioxidant, antibacterial and anticancer activities of biogenic gold nanoparticles. J. Mol. Liq. 2015, 212, 331–339.
  34. Stępkowski, T.M.; Brzóska, K.; Kruszewski, M. Silver nanoparticles induced changes in the expression of NF-κB related genes are cell type specific and related to the basal activity of NF-κB. Toxicol. In Vitro 2014, 28, 473–478.
  35. Chang, X.; Wang, X.; Li, J.; Shang, M.; Niu, S.; Zhang, W.; Li, Y.; Sun, Z.; Gan, J.; Li, W.; et al. Silver nanoparticles induced cytotoxicity in HT22 cells through autophagy and apoptosis via PI3K/AKT/mTOR signaling pathway. Ecotoxicol. Environ. Saf. 2021, 8, 111696.
  36. Reddy, V.N.; Nyamathulla, S.; Pahirulzaman, K.A.K.; Mokhtar, S.I.; Giribabu, N.; Pasupuleti, V.R. Gallocatechin-silver nanoparticles embedded in cotton gauze patches accelerated wound healing in diabetic rats by promoting proliferation and inhibiting apoptosis through the Wnt/β-catenin signaling pathway. PLoS ONE 2022, 17, e0268505.
  37. Spitzer, N.; Patterson, K.C.K.; Kipps, D.W. Akt and MAPK/ERK signaling regulate neurite extension in adult neural progenitor cells but do not directly mediate disruption of cytoskeletal structure and neurite dynamics by low-level silver nanoparticles. Toxicol. In Vitro 2021, 74, 105151.
  38. Parnsamut, C.; Brimson, S. Effects of silver nanoparticles and gold nanoparticles on IL-2, IL-6, and TNF-α production via MAPK pathway in leukemic cell lines. Genet. Mol. Res. 2015, 14, 3650–3668.
  39. Ponmurugan, P.; Manjukarunambika, K.; Elango, V.; Gnanamangai, B.M. Antifungal activity of biosynthesised copper nanoparticles evaluated against red root-rot disease in tea plants. J. Exp. Nanosci. 2016, 11, 1019–1031.
  40. Kiranmai, M.; Kadimcharla, K.; Keesara, N.R.; Fatima, S.N.; Bommena, P.; Batchu, U.R. Green synthesis of stable copper nanoparticles and synergistic activity with antibiotics. Indian J. Pharm. Sci. 2017, 79, 695–700.
  41. Ghasemi, P.; Shafiee, G.; Ziamajidi, N.; Abbasalipourkabir, R. Copper Nanoparticles Induce Apoptosis and Oxidative Stress in SW480 Human Colon Cancer Cell Line. Biol. Trace Elem. Res. 2022, 2022, 1–9.
  42. Ghosh, S.; More, P.; Nitnavare, R.; Jagtap, S.; Chippalkatti, R.; Derle, A.; Kitture, R.; Asok, A.; Kale, S.; Singh, S.; et al. Antidiabetic and antioxidant properties of copper nanoparticles synthesized by medicinal plant Dioscorea bulbifera. J. Nanomed. Nanotechnol. 2015, S6, 1.
  43. Jung, S.; Yang, J.Y.; Byeon, E.Y.; Kim, D.G.; Lee, D.G.; Ryoo, S.; Lee, S.; Shin, C.W.; Jang, H.W.; Kim, H.J.; et al. Copper-coated polypropylene filter face mask with SARS-COV-2 antiviral ability. Polymers 2021, 13, 1367.
  44. Azizi, M.; Ghourchian, H.; Yazdian, F.; Dashtestani, F.; AlizadehZeinabad, H. Cytotoxic effect of albumin coated copper nanoparticle on human breast cancer cells of MDA-MB 231. PLoS ONE 2017, 12, e0188639.
  45. Lalitha, K.; Kalaimurgan, D.; Nithya, K.; Venkatesan, S.; Shivakumar, M.S. Antibacterial, antifungal and mosquitocidal efficacy of copper nanoparticles synthesized from entomopathogenic nematode: Insect–host relationship of bacteria in secondary metabolites of Morganella morganii sp. (PMA1). Arabian J. Sci. Eng. 2020, 45, 4489–4501.
  46. Sharon, E.A.; Velayutham, K.; Ramanibai, R. Biosynthesis of copper nanoparticles using Artocarpus heterophyllus against dengue vector Aedes aegypti. Int. J. Life Sci. Sci. Res. 2018, 2455, 1716.
  47. Hassanien, R.; Husein, D.Z.; Al-Hakkani, M.F. Biosynthesis of copper nanoparticles using aqueous Tilia extract: Antimicrobial and anticancer activities. Heliyon 2018, 4, e01077.
  48. Bramhanwade, K.; Shende, S.; Bonde, S.; Gade, A.; Rai, M. Fungicidal activity of Cu nanoparticles against Fusarium causing crop diseases. Environ. Chem. Lett. 2016, 14, 229–235.
  49. Ramesh, M.; Anbuvannan, M.; Viruthagiri, G. Green synthesis of ZnO nanoparticles using Solanum nigrum leaf extract and their antibacterial activity. Acta Part A Mol. Biomol. Spectrosc. 2015, 136, 864–870.
  50. Beyth, N.; Houri-Haddad, Y.; Domb, A.; Khan, W.; Hazan, R. Alternative antimicrobial approach: Nano-antimicrobial materials. Evid.-Based Complement. Altern. Med. 2015, 2015, 16.
  51. Wang, S.W.; Lee, C.H.; Lin, M.S.; Chi, C.W.; Chen, Y.J.; Wang, G.S.; Liao, K.W.; Chiu, L.P.; Wu, S.H.; Huang, D.M.; et al. ZnO nanoparticles induced caspase-dependent apoptosis in gingival squamous cell carcinoma through mitochondrial dysfunction and p70S6K signaling pathway. Int. J. Mol. Sci. 2020, 21, 1612.
  52. Gao, F.; Ma, N.; Zhou, H.; Wang, Q.; Zhang, H.; Wang, P.; Hou, H.; Wen, H.; Li, L. Zinc oxide nanoparticles-induced epigenetic change and G2/M arrest are associated with apoptosis in human epidermal keratinocytes. Int. J. Nanomed. 2016, 11, 3859.
  53. Patrón-Romero, L.; Luque-Morales, P.A.; Loera-Castañeda, V.; Lares-Asseff, I.; Leal-Ávila, M.Á.; Alvelais-Palacios, J.A.; Plasencia-López, I.; Almanza-Reyes, H. Mitochondrial Dysfunction Induced by Zinc Oxide Nanoparticles. Crystals 2022, 12, 1089.
  54. Mishra, A.; Swain, R.K.; Mishra, S.K.; Panda, N.; Sethy, K. Growth performance and serum biochemical parameters as affected by nano zinc supplementation in layer chicks. Indian J. Anim. Nutr. 2014, 31, 384–388.
  55. Adwin Jose, P.; Sankarganesh, M.; Dhaveethu Raja, J.; Senthilkumar, G.S.; Nandini Asha, R.; Raja, S.J.; Sheela, C.D. Bio-inspired nickel nanoparticles of pyrimidine-Schiff base: In vitro anticancer, BSA and DNA interactions, molecular docking and antioxidant studies. J. Biomol. Struct. Dyn. 2022, 40, 10715–10729.
  56. Jaji, N.D.; Lee, H.L.; Hussin, M.H.; Akil, H.M.; Zakaria, M.R.; Othman, M.B.H. Advanced nickel nanoparticles technology: From synthesis to applications. Nanotechnol. Rev. 2020, 9, 1456–1480.
  57. Rameshthangam, P.; Chitra, J.P. Synergistic anticancer effect of green synthesized nickel nanoparticles and quercetin extracted from Ocimum sanctum leaf extract. J. Mater. Sci. Technol. 2018, 34, 508–522.
  58. Gomaji Chaudhary, R.; Tanna, J.A.; Gandhare, N.V.; Rai, A.R.; Juneja, H.D. Synthesis of nickel nanoparticles: Microscopic investigation, an efficient catalyst and effective antibacterial activity. Adv. Mater. Lett. 2015, 6, 990–998.
  59. Ahghari, M.R.; Soltaninejad, V.; Maleki, A. Synthesis of nickel nanoparticles by a green and convenient method as a magnetic mirror with antibacterial activities. Sci. Rep. 2020, 10, 12627.
  60. Huang, Y.; Zhu, C.; Xie, R.; Ni, M. Green synthesis of nickel nanoparticles using Fumaria officinalis as a novel chemotherapeutic drug for the treatment of ovarian cancer. J. Exp. Nanosci. 2021, 16, 368–381.
  61. Magaye, R.R.; Yue, X.; Zou, B.; Shi, H.; Yu, H.; Liu, K.; Lin, X.; Xu, J.; Yang, C.; Wu, A.; et al. Acute toxicity of nickel nanoparticles in rats after intravenous injection. Int. J. Nanomed. 2014, 9, 1393.
  62. Shwetha, U.R.; CR, R.K.; Kiran, M.S.; Betageri, V.S.; Latha, M.S.; Veerapur, R.; Lamraoui, G.; Al-Kheraif, A.A.; Elgorban, A.M.; Syed, A.; et al. Biogenic synthesis of NiO nanoparticles using areca catechu leaf extract and their antidiabetic and cytotoxic effects. Molecules 2021, 26, 2448.
  63. Angajala, G.; Ramya, R.; Subashini, R. In-vitro anti-inflammatory and mosquito larvicidal efficacy of nickel nanoparticles phytofabricated from aqueous leaf extracts of Aegle marmelos Correa. Acta Trop. 2014, 135, 19–26.
  64. Rajakumar, G.; Rahuman, A.A.; Velayutham, K.; Ramyadevi, J.; Jeyasubramanian, K.; Marikani, A.; Elango, G.; Kamaraj, C.; Santhoshkumar, T.; Marimuthu, S.; et al. Novel and simple approach using synthesized nickel nanoparticles to control blood-sucking parasites. Vet. Parasitol. 2013, 191, 332–339.
  65. Jeyaraj Pandian, C.; Palanivel, R.; Dhanasekaran, S. Screening antimicrobial activity of nickel nanoparticles synthesized using Ocimum sanctum leaf extract. J. Nanopart. 2016, 2016, 4694367.
  66. Zarenezhad, E.; Abdulabbas, H.T.; Marzi, M.; Ghazy, E.; Ekrahi, M.; Pezeshki, B.; Ghasemian, A.; Moawad, A.A. Nickel Nanoparticles: Applications and Antimicrobial Role against Methicillin-Resistant Staphylococcus aureus Infections. Antibiotics 2022, 11, 1208.
  67. Tombuloglu, H.; Albenayyan, N.; Slimani, Y.; Akhtar, S.; Tombuloglu, G.; Almessiere, M.; Baykal, A.; Ercan, I.; Sabit, H.; Manikandan, A. Fate and impact of maghemite (γ-Fe2O3) and magnetite (Fe3O4) nanoparticles in barley (Hordeum vulgare L.). Environ. Sci. Pollut. Res. 2022, 29, 4710–4721.
  68. Marand, Z.R.; Farimani, M.H.R.; Shahtahmasebi, N. Study of magnetic and structural and optical properties of Zn doped Fe3O4 nanoparticles synthesized by co-precipitation method for biomedical application. Akush. Ginekol. 2014, 15, 238–247.
  69. Ding, W.; Guo, L. Immobilized transferrin Fe3O4@SiO2 nanoparticle with high doxorubicin loading for dual-targeted tumor drug delivery. Int. J. Nanomed. 2013, 8, 4631–4639.
  70. Zhang, H.; Li, T.; Luo, W.; Peng, G.X.; Xiong, J. Green synthesis of Ag nanoparticles from Leucus aspera and its application in anticancer activity against alveolar cancer. J. Exp. Nanosci. 2021, 17, 47–60.
  71. Arriortua, O.K.; Garaio, E.; de la Parte, B.H.; Insausti, M.; Lezama, L.; Plazaola, F.; García, J.A.; Aizpurua, J.M.; Sagartzazu, M.; Irazola, M.; et al. Antitumor magnetic hyperthermia induced by RGD-functionalized Fe3O4 nanoparticles, in an experimental model of colorectal liver metastases. Beilstein J. Nanotech. 2016, 7, 1532–1542.
  72. Kalber, T.L.; Ordidge, K.L.; Southern, P.; Loebinger, M.R.; Kyrtatos, P.G.; Pankhurst, Q.A.; Lythgoe, M.F.; Janes, S.M. Hyperthermia treatment of tumors by mesenchymal stem cell-delivered superparamagnetic iron oxide nanoparticles. Int. J. Nanomed. 2016, 11, 1973.
  73. Hedayatnasab, Z.; Dabbagh, A.; Abnisa, F.; Daud, W.M.A.W. Polycaprolactone-coated superparamagnetic iron oxide nanoparticles for in vitro magnetic hyperthermia therapy of cancer. Eur. Poly. J. 2020, 133, 109789.
  74. Hsieh, C.H.; Hsieh, H.C.; Shih, F.H.; Wang, P.W.; Yang, L.X.; Shieh, D.B.; Wang, Y.C. An innovative NRF2 nano-modulator induces lung cancer ferroptosis and elicits an immunostimulatory tumor microenvironment. Theranostics 2021, 11, 7072.
  75. Ismail, R.A.; Sulaiman, G.M.; Abdulrahman, S.A.; Marzoog, T.R. Antibacterial activity of magnetic iron oxide nanoparticles synthesized by laser ablation in liquid. Mater. Sci. Eng. 2015, 53, 286–297.
  76. Mahdy, S.A.; Raheed, Q.J.; Kalaichelvan, P.T. Antimicrobial activity of zero-valent iron nanoparticles. Int. J. Mod. Eng. Res. 2012, 2, 578–581.
  77. Kumar, R.; Nayak, M.; Sahoo, G.C.; Pandey, K.; Sarkar, M.C.; Ansari, Y.; Das, V.N.R.; Topno, R.K.; Madhukar, M.; Das, P. Iron oxide nanoparticles based antiviral activity of H1N1 influenza A virus. J. Infect. Chemother. 2019, 25, 325–329.
  78. Parveen, S.; Wani, A.H.; Shah, M.A.; Devi, H.S.; Bhat, M.Y.; Koka, J.A. Preparation, characterization and antifungal activity of iron oxide nanoparticles. Microb. Pathog. 2018, 115, 287–292.
  79. Wang, Y.; Liu, T.; Li, X.; Sheng, H.; Ma, X.; Hao, L. Ferroptosis-inducing nanomedicine for cancer therapy. Front. Pharm. 2021, 12, 3638.
  80. Wen, J.; Chen, H.; Ren, Z.; Zhang, P.; Chen, J.; Jiang, S. Ultrasmall iron oxide nanoparticles induced ferroptosis via Beclin1/ATG5-dependent autophagy pathway. Nano Converg. 2021, 8, 10.
  81. Bhinge, S.; Bhutkar, M.; Randive, D.; Wadkar, G.; Todkar, S. Synergistic effects of synthesized iron nanoparticles of neem extract with conventional antibiotic against gram positive negative microorganism. Int. J. Infect. Dis. 2020, 101, 48.
  82. Padilla-Cruz, A.L.; Garza-Cervantes, J.A.; Vasto-Anzaldo, X.G.; García-Rivas, G.; León-Buitimea, A.; Morones-Ramírez, J.R. Synthesis and design of Ag–Fe bimetallic nanoparticles as antimicrobial synergistic combination therapies against clinically relevant pathogens. Sci. Rep. 2021, 11, 5351.
  83. Du, B.; Yu, M.; Zheng, J. Transport and interactions of nanoparticles in the kidneys. Nat. Rev. Mater. 2018, 3, 358–374.
  84. Soo Choi, H.; Liu, W.; Misra, P.; Tanaka, E.; Zimmer, J.P.; Itty Ipe, B.; Bawendi, M.G.; Frangioni, J.V. Renal clearance of quantum dots. Nat. Biotechnol. 2007, 25, 1165–1170.
  85. Lee, J.A.; Kim, M.K.; Paek, H.J.; Kim, Y.R.; Kim, M.K.; Lee, J.K.; Jeong, J.; Choi, S.J. Tissue distribution and excretion kinetics of orally administered silica nanoparticles in rats. Int. J. Nanomed. 2014, 9, 251.
  86. Poon, W.; Zhang, Y.N.; Ouyang, B.; Kingston, B.R.; Wu, J.L.; Wilhelm, S.; Chan, W.C. Elimination pathways of nanoparticles. ACS Nano 2019, 13, 5785–5798.
  87. Chenthamara, D.; Subramaniam, S.; Ramakrishnan, S.G.; Krishnaswamy, S.; Essa, M.M.; Lin, F.H.; Qoronfleh, M.W. Therapeutic efficacy of nanoparticles and routes of administration. Biomater. Res. 2019, 23, 20.
  88. De Jong, W.H.; Hagens, W.I.; Krystek, P.; Burger, M.C.; Sips, A.J.; Geertsma, R.E. Particle size-dependent organ distribution of gold nanoparticles after intravenous administration. Biomaterials 2008, 29, 1912–1919.
  89. Singh, D.; Singh, S.; Sahu, J.; Srivastava, S.; Singh, M.R. Ceramic nanoparticles: Recompense, cellular uptake and toxicity concerns. Artif. Cells Nanomed. Biotechnol. 2016, 44, 401–409.
  90. Liu, T.; Chao, Y.; Gao, M.; Liang, C.; Chen, Q.; Song, G.; Cheng, L.; Liu, Z. Ultra-small MoS2 nanodots with rapid body clearance for photothermal cancer therapy. Nano Res. 2016, 9, 3003–3017.
  91. Tang, S.; Peng, C.; Xu, J.; Du, B.; Wang, Q.; Vinluan, R.D., 3rd; Yu, M.; Kim, M.J.; Zheng, J. Tailoring renal clearance and tumor targeting of ultrasmall metal nanoparticles with particle density. Angew. Chem. Int. Ed. 2016, 55, 16039–16043.
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: 421
Revisions: 2 times (View History)
Update Date: 06 May 2023
1000/1000
Video Production Service