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 + 3300 word(s) 3300 2022-01-20 07:58:01 |
2 The format is correct Meta information modification 3300 2022-01-25 03:32:51 |

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.
Paul, A. Therapeutic Applications of Nano-Phytopharmaceuticals. Encyclopedia. Available online: https://encyclopedia.pub/entry/18733 (accessed on 21 July 2024).
Paul A. Therapeutic Applications of Nano-Phytopharmaceuticals. Encyclopedia. Available at: https://encyclopedia.pub/entry/18733. Accessed July 21, 2024.
Paul, Alok. "Therapeutic Applications of Nano-Phytopharmaceuticals" Encyclopedia, https://encyclopedia.pub/entry/18733 (accessed July 21, 2024).
Paul, A. (2022, January 24). Therapeutic Applications of Nano-Phytopharmaceuticals. In Encyclopedia. https://encyclopedia.pub/entry/18733
Paul, Alok. "Therapeutic Applications of Nano-Phytopharmaceuticals." Encyclopedia. Web. 24 January, 2022.
Therapeutic Applications of Nano-Phytopharmaceuticals
Edit

Phytopharmaceuticals have been widely used globally since ancient times and acknowledged by healthcare professionals and patients for their superior therapeutic value and fewer side effects compared to modern medicines. Dose reduction, improved bioavailability, receptor-selective binding, and targeted delivery of phytopharmaceuticals can be likely achieved by molding them into specific nano-formulations. 

nano-formulations phytopharmaceuticals herbal medications nanomaterials

1. Introduction

Nanotechnology is a vital tool for medical sciences. The introduction of nanomedicine, using nanotechnology combined with drugs or diagnostic molecules, has improved the ability to target specific cells or tissues that require treatment and repair. These nanomaterials have been proven to be produced at a nanoscale level and are safe to introduce into the body. It is possible to modify a variety of nanocarriers’ characteristics that include their constituents, size, shape, bioavailability, surface properties, and target specificity to achieve or enhance desirable pharmacological targets [1][2] Several strategies have been implemented to increase the drug-target specificity. Recently, several studies have reported improved efficacy of therapy when combined with nanomaterials [3]. Pure herbal medicines are often considered less effective compared to pure constituents that are mainly demonstrated to have reduced intestinal absorption when administered orally [4]. This is the reason behind the pharmacological activity/loss associated with pure constituents and such problems can be overcome using new drug delivery systems, such as nanotechnology.

Nanoparticulate delivery of drugs can generally improve drugs’ solubility, bioavailability, stability, pharmacological activity, increase target specificity, promote transport across membrane, prolong circulation times, and reduce systemic and organ toxicity [5][6]. Various treatments are being investigated with the use of nanoparticle drug delivery systems for diseases, such as infectious diseases, autoimmune diseases, cardiovascular diseases, neurodegenerative diseases, ocular diseases, fungal infections, iron deficiency, and pulmonary diseases [7][8]. However, the greatest advances were seen in the treatment of cancer with several nano strategies being used clinically after approval by the FDA in the United States of America [9]. Recently, more attention has shifted towards novel drug delivery systems using nanoparticles for herbal and plant-based drugs [10].

Plant-based medicine has some limitations which hinder its use and production in the mainstream disease treatment and therapy. There are several chemical constituents in a plant’s extract that lead to its medicinal properties. The active constituents of plant extracts like tannins, flavonoids, alkaloids, phenylpropanoids, and terpenoids are water-soluble but show poor absorption from their inability to cross lipid membranes and have large molecular sizes, which then results in low bioavailability and efficacy [5][11][12]. There are also concerns of safety due to the incompatibility of some plant extracts with other components in a drug formulation which can lead to undesirable effects [11]. High systemic clearance of these compounds also leads to low therapeutic levels in the blood resulting in no therapeutic effect [13]. Furthermore, poor reproducibility of in vitro effects in vivo prevents many plant-based medicines from clearing clinical trial phases [14]. Nanomedicine aims to overcome these limitations and to improve the delivery of plant-based medicines to treat various diseases. Nanoparticle drug delivery systems can potentially improve the stability, solubility, and bioavailability of encapsulated plant extracts, promote its movement across lipid membranes, and prolong its circulation, all while delivering the active constituent to a specific target site [5][6][11] (Figure 1). Liposomes, dendrimers, polymeric NCs, polymeric micelles, metallic NPs (magnetic, gold), SLNs, nanocapsules, nanospheres, and nanogels are some of the examples of nano-based drug delivery systems that are presently under investigation [10].

Figure 1. Representation of delivery of phytopharmaceuticals using nanotechnology. The figure was made with www.biorender.com (access date: 17 December 2021) and adapted from DOI: 10.3390/nano12020238.

2. Therapeutic Applications of Nano-Phytopharmaceuticals

2.1. Nano-Phytopharmaceuticals in Neurological (CNS) Disorders

Despite enduring efforts to deliver medicinal drugs to the brain tissue, these treatment regimens are compromised by low bioavailability due to the blood-brain barrier (BBB), a natural protective layer consisting of capillary endothelial cells, pericytes, and tight junctions [15]. This near-impermeable barrier impedes the entry of most macromolecules and allows only the minutest particles (<400 Da) to cross into the nerve tissue. Indeed, less than 5% of conventional therapeutic molecules in various stages of pharmaceutical development may penetrate this physiological barrier [16]. Therefore, CNS-targeted natural product formulations in nanocarriers hold infinite promise. Evidence suggests that nutraceuticals and phytochemicals exert therapeutic effects in neurological diseases, owing to their antioxidant, anti-inflammatory, and neuroprotective mechanisms [17][18]. Coupled with nanoscale delivery systems which improve solubility, enhance retention rates [19], and with the ability to permeate through the BBB, there is hope yet for effective treatment of neurological disorders. The following topic discusses two herbal compounds, curcumin and ginseng, and their significant potential for use in neurotherapeutics through nano-encapsulation.

A derivative of the South Asian turmeric plant rhizome, Curcuma longa (turmeric), the polyphenol curcumin has amassed quite a reputation in nutraceutical development for various diseases, including cancer and inflammatory conditions [20]. The healing properties of this yellow spice date back to ancient civilizations, featuring strongly in Ayurvedic therapy. Nevertheless, the relatively poor bioavailability due to dismal water solubility and rapid intestinal clearance impedes the therapeutic effects of conventional drug delivery, paving the way for the advent of nano-formulations.
The role of nanoparticulate systems to treat glioblastoma was recently highlighted using different strategies, such as improving their diffusion through the BBB [21]. In a glioblastoma study, Schmitt et al. demonstrated that curcumin encased in liposomal carriers (LipoCur) proved to be superior in reducing the proliferation and reactivity of human microglia and astrocytes (human fetal astrocyte cell line, SVGA) compared to the free compound. Immunostaining of murine organotypic brain segments challenged with lipopolysaccharide (LPS) for eight days, revealed that LipoCur was equally effective in reducing glial scarring [20].
Aside from CNS cancers, chronic degenerative conditions such as Alzheimer’s disease (AD) fall under intense scrutiny from researchers. Characterized by neurofibrillary tangles of tau proteins and “senile”amyloid-β (Aβ) plaques in the brain, the condition is notoriously challenging to diagnose and treat due to the late onset of symptoms, usually decades after the onset of pathological changes [22]. Neuro-inflammation is recognized as one of the primary mechanisms underlying neuronal degeneration, and thus is a target for natural product (NP) research. Malvajerd and colleagues encapsulated curcumin in solid lipid nanoparticles (SLNs) (entrapment efficiency 82 ± 0.49%) and nanostructured lipid carriers (NLCs) (94 ± 0.74%), and found that NLCs resulted in the highest bioaccumulation of curcumin in rat brains compared to free compound and SLNs [23].
Poly (d,l-lactic acid-co-glycolic acid, PLGA) and polymer NPs are known to be biocompatible and thus show great potential in disarming neuroinflammation in another 5xFAD mice Alzheimer’s disease model [24].
Kim et al. established a formulation of red ginseng water extract with gold nanoparticles (WERGGN) and established its antioxidant activity through DPPH, ORAC, and ABTS assays. WERGGN treatment on neuron-like PC-12 cells revealed cytoprotective effects, mainly due to the decreased intracellular oxidative stress. In addition, levels of neurotransmitter degradation enzymes such as acetylcholinesterase and butyrylcholinesterase were also inhibited, suggesting that WERGGN promoted synaptic impulse transmission [25]. Nonetheless, the dosage of ginseng extract, when co-administered with a synthetic drug (e.g. selegiline), determines the pharmacokinetics in the body: lower doses result in poorer bioavailability due to CYP1A2 induction, while higher concentrations cause inhibition of CYP3A4 and thus enhanced systemic exposure [26].
It is essential to note that nanomaterials have potential limitations and the adverse effects on CNS were described previously [27][28]. For example, one study showed that female mice that were intranasally treated with titanium dioxide nanoparticles (TiO2 NPs) (0.5 mg daily for 1 month) produced apoptosis, affected brain development, and oxidative stress (caused increased lipid peroxidation, protein oxidation, catalase expressions, and release of glutamic acid and nitric oxide) [28]. These authors also concluded that an intranasal spray of TiO2 NPs could be translocated into the CNS and cause potential lesions of the brain [28]. Similarly, Fe2O3 magnetic NPs (0.15–15 mM) also showed toxicity and c viability in a neuronal cell line in vitro [29]. Whereas another study showed that toxicity of zinc and silicon NPs is relatively low, but the neuronal toxicity of NPs depends on the materials used [27][30][31]. Therefore, the toxicity of NPs is present, and depends on the formulation, particle size and concentration or treatment protocols of specific NPs.

2.2. Nano-Phytopharmaceuticals in Cardiovascular Disorders

Since CVD require most often long-term medication, treatment regimens become complex and create a burden for the patient especially when multiple medicines are prescribed and must be taken for life [32][33] Although these therapeutic drugs have been successful in halting the progression of the disease, thereby improving the quality of life of patients, most of these cures only the symptoms and may not repair or regenerate the damaged tissues. In addition, CVD medication has different side effects, such as antiplatelet drugs may cause diarrhea, rash, or itching [32]. Others can cause abdominal pain, headache, chest pain, muscle aches, and dizziness. In the case of anticoagulants, their side effects can lead to bleeding and necrotic or gangrenous skin. Given the excessive side effects of current therapies, alternative therapeutic approaches like medicinal plants and natural products are preferred. Against this premise, a better treatment for CVD that would not burden the patients is necessary. Hence, it is important to explore new technologies and drugs to lessen the use of conventional treatments. The lower toxicity, chemical diversity, cost-effectiveness, and therapeutic potentials of natural products make them the popular choice of medicine compared to other products [34]. With the combination of nanoformulation methods to deliver phytomedicines, it becomes more effective with improved solubility, bioavailability, circulation time, surface area-to-volume ratio, nil systemic adverse side effects, and drug delivery efficiency of these medications. The introduction of nanomedicine using a combined nanotechnology with drugs or diagnostic molecules has improved the ability to target specific cells or tissues that require treatment and repair. These nanomaterials are produced on a nanoscale level and are safe to introduce into the body. Hence, applications for nanotechnology in medicine include imaging, diagnosis, or the delivery of drugs that will help medical professionals treat various diseases including cardiovascular diseases [35][36]. The functionality of the most recent nano-formulated medicinal plants and/or natural products against various cardiovascular conditions such as hypertension, atherosclerosis, thrombosis, and myocardial infarction is expected to be maximized.

Some plant extracts/compounds used for treatment of CVD that are nano-formulated for efficient delivery to their target are listed in Table 1. Among the extracts used for CVD, curcumin, quercetin, and resveratrol were the most applied natural products, respectively. However, curcumin, despite its curative potential, has poor aqueous solubility and consequently, minimal systemic bioavailability along with rapid degradation [37]. These characteristics restrict the utilization of curcumin a medical perspective. Liposomes have found uses in drug delivery of poorly water-soluble drugs. Such drug-loaded liposomes can be fabricated by a wide variety of nanotechnology methods such as ethanol injection, thin-film hydration, sonication, high-pressure extrusion, reverse-phase evaporation, calcium-induced fusion, and supercritical fluid methods, among others [37].

Table 1. Nano-phytopharmaceuticals for therapeutic applications in CVDs.
Nanoformulation Phyto-Pharmaceutical Effects References
Liposomes Curcumin Anti-hypercholesterolemic, anti-atherosclerotic and protective against cardiac ischemia and reperfusion. [38]
PLGA nanoparticle Quercetin Anti-hypercholesterolemia, better cell rescue by lowering oxidized thiols and sustaining superior ATP production, improved therapeutics for ROS-based cardiac diseases. [39]
Solid lipid Nanoparticle Resveratrol Protective action of vascular walls towards oxidation, inflammation, platelet oxidation and thrombus formation [40][41]
1,2-diacyl-Sn-glycero-3-phosphocholine [EPC] and 1,2-dipalmitoyl-Sn-glycero-3-phosphocholine (DPPC) liposomes Magnolol Enhanced inhibitory effect on migration and hyperplasia of vascular smooth-muscle cells; Anti-platelet, anti-thrombotic, and anti-hypertensive via inhibiting MAPK family activation, Akt/ERK1/2/GSK3 β-catenin pathway, and angiotensin-converting enzyme (ACE)/angiotensin II (Ang II)/Ang II type 1 receptor (AT-1R) cascade and upregulating PPAR-β/γ and NO/guanosine 3′,5′-cyclic phosphate/PKG. [34][42][43]
Nano-micelles Tilianin Protective effects of cardiomyocytes by inhibiting inflammation and oxidative stress during myocardial ischemia-reperfusion injury [44]
PEGylated nanostructured lipid carriers Baicalin Improved myocardial ischemia; beneficial roles against the initiation and progression of CVDs such as atherosclerosis, hypertension, myocardial infarction, reperfusion and heart failure [45][46]
Liposomes Berberine Effect of protecting heart failure, hypertension, hyperlipidemia, insulin resistance, arrhythmias, and platelet aggregation. [47][48]
Quercetin acts as an antioxidant on cells but is hindered by its high metabolism rate. In order to control this, quercetin is encapsulated in poly [lactic-co-glycolic] acid (PLGA) nanoparticles [39]. This guarantees secure and controlled release of the quercetin, and permits cell enlistment, attachment, expansion, and articulation of heart proteins in local myocardium.
Resveratrol’s capacity for protective action makes it one of the commonly used compounds against CVD. However, its poor pharmacokinetic properties, such as low aqueous solubility, low photostability and extensive first-pass metabolism, result in poor bioavailability and hinder its clinical potential [49][50]. To resolve this issue, the use of lipid nanoparticles offer the possibility to develop new therapeutics. Further, solid lipid nanoparticles hold great promise for reaching the goal of controlled and site-specific drug delivery [34].
Other compounds used for CVDs are Magnolols, Berberine, Tilianin and Baicalin. These compounds require nanoformulation to maximize their use. Magnolols as a phenolic polyhydroxy compound have poor aqueous solubility and low oral bioavailability which limit their clinical use. Therefore, various formulations such as liposomes [51], solid dispersions [52], emulsions [53], and nanoparticles [54] have been developed to ameliorate the water solubility and bioavailability of it.

2.3. Nano-Phytopharmaceuticals in Pulmonary Disorders

To curb the increasing threat of respiratory diseases, there is an immediate need to explore some alternatives to the use of antibiotics, and an ecofriendly, cost effective, efficacious and durable strategy to combat these global respiratory problems. Thus, the field of nanotechnology provides a promising technology against respiratory diseases. In the past, various studies have shown very effective and interesting results. In addition to their antiviral properties, the application of nanoparticles (NPs) provides a potential strategy to manage infections caused by multidrug-resistant organisms (MDROs) [55][56][57]. NPs exhibiting antibacterial activities can target multiple biomolecules and have the potential to reduce or eliminate the evolution of MDROs [58]. Plant extracts are complex mixtures that provide a rich arsenal of molecules, such as flavanones, flavones, flavonols and chalcones, fatty acids, amino acids, terpenoids, aldehydes, and alcohols [59], with high redox potential [60]. Furthermore, biogenic synthesis produces potential, stable, and better-defined materials [61][62].
Silver nanoparticles (AgNPs) have shown broad applications in the medical system, such as anti-inflammatory, anti-angiogenesis, antiplatelet, antifungal, anticancer, and antibacterial activities [63][64][65][66]. Nowadays, AgNPs have been reported as biomedical therapeutic agents, such as wound dressings, and long-term burn care products and antibacterial lotions [67]; They also exhibit antiviral activities against influenza A virus, hepatitis B virus, human parainfluenza virus, herpes simplex virus, and human immunodeficiency virus [68][69]. They prevent the anchoring and binding of the virus to the host and cell receptor, respectively, and hence they deactivate the virus by denaturing the surface.
AgNPs inhibit the binding of the virus by interacting with the glycoprotein (gp120) of the sulfur-bearing groups distributed in the lipid membrane of the virus [70]. In another study, the virus inactivation was observed when the nano-silver was combined with the nucleic acid of the virus and modified the structure of the capsid and affected the replication [71]. Along with such surface modification, AgNPs also show synergistic antiviral activity. In one study, it was reported that curcumin, when used as both a reducing and stabilizing agent prevented viral replication, and blocked the budding of viruses [72]. Similarly, the AgNP surface, when modified through chemical methods with drugs like zanamivir, amantadine and oseltamivir, was able to interact with virus particles directly, which destroyed the virus and blocked its entry [73].
Gold nanoparticles (AuNPs) provide superior properties [74], and various methods for the preparation of gold nanoparticles have been well reported [63]. Compared to silver nanoparticles, AuNPs display better results in in vivo studies. AuNPs can interact with the haemagglutinin (HA) glycoprotein and can oxidize the disulfide bond, resulting in the inactivation of the virus [75]. Surface properties are useful in targeting the virus. The sulphonates are organic sulphates that were used to interact with the capsid proteins of the virus cell and prevent the HA activity [76].
In nanoscience, the carbon dots (CDs) also play an important role due to their unique properties. CDs prevent the viral infection as they have hydroxyl and carboxyl groups on their surfaces that interact with viral membranes [77]. To improve the activities of CDs, antiviral agents like plant extracts are grafted on the surface that involves a two-step reaction. The functionalized antiviral agent shows the inhibition of the virus into the cell that has a broad-spectrum action for the enveloped and the non-enveloped viruses [78].
Another important nanotechnology-based materials are based on graphene, (GO), which also have biomedical applications [79]. To prevent viral infection in the host cell, GO has been used with organic and metallic nanoparticles that show antiviral activities [80].

2.4. Nano-Phytopharmaceuticals in Gastro-Intestinal Disorders

Berberis vulgaris and Curcuma longa extracts encapsulated in cationic polymer EPO (Figure 5), demonstrated significant anti-parasitic activity against Entamoeba histolytica. Furthermore, B. vulgaris encapsulated within EPO showed IC50 of 26 ppm in comparison with the free extract having IC50 of 34 µg/mL. The IC50 of Curcuma longa loaded within EPO was found to be 19 ppm in comparison with the free extract having IC50 of 38 µg/mL [81]. Another group of researchers used cerium oxide nanoparticles encapsulated with Nelumbo nucifera flower extract against the human colon cancer cell line (HCT 116), exhibited IC50 of 4.16 mg/mL [81]. Garcinia mangostana has been widely used for medical applications. G. mangostana extract loaded within ethylcellulose and methylcellulose nanoparticles proved a highly protective agent for stomach ulcers with a MIC value of 6.25 ug/mL against Helicobacter pylori, which is almost the same as metronidazole. A study done by Saravanakumar and team found that silver nanoparticles encapsulated with Toxicodendron vernicifluum extract were effective against enteropathogenic bacteria, E. coli and H. Pylori, with MIC 8.12 µg/mL and 18.14 µg/mL. Acorus calamus Lim extract loaded within silver nanoparticles has shown significant inhibition in the formation of H. pylori at a concentration of 350 µg/mL. Metallic nanoparticles demonstrated promising results in inhibition of enteropathogenic bacteria in various studies. Gold nanoparticles loaded with extracts of Tribulus terrestris showed effective activity against H. pylori in a size-dependent manner, with MIC of 16.75 µg/mL at 55 nm and MIC of 18 µg/mL at 7 nm size of gold nanoparticles, respectively [81] (Figure 2; Table 2).
Figure 2. Encapsulation of bioactive compounds from various herbal extracts with silver, gold, cellulose, and polymeric nanoparticles to improve delivery in the gastrointestinal tract, and to prevent or cure various diseases. The figure was made with www.biorender.com (access date: 12 January 2022) and adapted from DOI: 10.3390/nano12020238.
Table 2. Nano-phytopharmaceuticals for therapeutic applications in GI disorders.
Nanoformulation Phyto-Pharmaceutical Enteropathogen/GI Cell Lines; IC50/MIC Reference
Polymeric (EPO) Berberis vulgaris Entamoeba histolytica; 26 ppm [81]
Polymeric (EPO) Curcuma longa Entamoeba histolytica; 19 ppm [81]
Polymeric (Cerium oxide) Nelumbo nucifera Human colon cancer (HCT 116); 4.16 µg/mL [81]
Cellulose (Ethyl) Garcinia mangostana Helicobacter pylori; 62.5 µg/mL [81]
Metallic (Silver) Toxicodendron vernicifluum Helicobacter pylori; 18.14 µg/mL [81]
Metallic (Silver) Toxicodendron vernicifluum E.coli; 8.12 µg/mL [81]
Metallic (Gold) Tribulus terrestris Helicobacter pylori; 16.75 µg/mL [81]
Apart from benefits, NPs showed adverse events, such as selenium and selenite nanoparticles in mice (2 or 4 mg/kg body weight daily) treated over a period of 15 days which showed more toxicity (growth suppression, increased liver toxicity, and reduced superoxide dismutase activity) in selenite NPs [82].
Nanotechnology is a vital tool for its applications in medical science, given that it is possible to obtain a variety of nanocarrier characteristics that include their constituents, size, shape, bioavailability, surface properties, and target specificity to achieve or enhance desirable pharmacological targets [81]. A number of strategies have been implemented to increase the drug-target specificity. Recently, several studies have reported the improved efficacy of herbal extracts for gastrointestinal disorders when associated with nanomaterials mainly due to oral absorption, greater stability, and simulated intestinal fluid [3][4]. Pure herbal medicines are often considered less effective in comparison due to their size and reduced intestinal absorption when administered orally. However, plant extracts loaded on nanoparticles are more stable in high protein environments and help to increase target specificity [4]. Solubility is always considered a major concern associated with plant extracts, which can be successfully upgraded using nanosystems [82]. These are the reasons behind a pharmacological loss associated with plant extracts and such problems can be overcome using novel drug delivery systems such as nanotechnology. The advantages of using these novel delivery systems include better absorption by smoothing diffusion through the epithelium, modification of pharmacokinetics, enhancement of intracellular penetration, and distribution with reduced toxicity, overcoming resistance and lowering cost. Nanoparticles are stable in harsh conditions like sterilization temperatures [82].

References

  1. Mobasser, S.; Firoozi, A.A. Review of nanotechnology applications in science and engineering. J. Civil. Eng. Urban. 2017, 64, 84–93.
  2. Chen, G.; Roy, I.; Yang, C.; Prasad, P.N. Nanochemistry and nanomedicine for nanoparticle-based diagnostics and therapy. Chem. Rev. 2016, 116, 2826–2885.
  3. Morelli, L.; Gimondi, S.; Sevieri, M.; Salvioni, L.; Guizzetti, M.; Colzani, B.; Palugan, L.; Foppoli, A.; Talamini, L.; Morosi, L.; et al. Monitoring the fate of orally administered PLGA nanoformulation for local delivery of therapeutic drugs. Pharmaceutics 2019, 11, 658.
  4. Zhao, Q.; Luan, X.; Zheng, M.; Tian, X.-H.; Zhao, J.; Zhang, W.-D.; Ma, B.-L. Synergistic mechanisms of constituents in herbal extracts during intestinal absorption: Focus on natural occurring nanoparticles. Pharmaceutics 2020, 12, 128.
  5. Kumari, P.; Luqman, S.; Meena, A. Application of the combinatorial approaches of medicinal and aromatic plants with nanotechnology and its impacts on healthcare. DARU J. Pharm. Sci. 2019, 27, 475–489.
  6. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124.
  7. Mishra, V.; Kesharwani, P.; Amin, M.C.M.; Iyer, A. (Eds.) Nanotechnology-Based Approaches for Targeting and Delivery of Drugs and Genes, 1st ed.; 2021; Available online: https://www.elsevier.com/books/nanotechnology-based-approaches-for-targeting-and-delivery-of-drugs-and-genes/mishra/978-0-12-809717-5 (accessed on 20 August 2021).
  8. Yetisgin, A.A.; Cetinel, S.; Zuvin, M.; Kosar, A.; Kutlu, O. Therapeutic nanoparticles and their targeted delivery applications. Molecules 2020, 25, 2193.
  9. Pelaz, B.; Alexiou, C.; Alvarez-Puebla, R.A.; Alves, F.; Andrews, A.M.; Ashraf, S.; Balogh, L.P.; Ballerini, L.; Bestetti, A.; Brendel, C.; et al. Diverse applications of nanomedicine. ACS Nano 2017, 11, 2313–2381.
  10. Prairna; Paramanya, A.; Sharma, S.; Bagdat, R.B.; Ali, A. Recent practices of medicinal and aromatic plants in nanotechnology. In Nanomaterials for Agriculture and Forestry Applications; Elsevier: Amsterdam, The Netherlands, 2020; pp. 435–467.
  11. Bonifácio, B.V.; Silva, P.B.; Ramos, M.A.; Negri, K.M.; Bauab, T.M.; Chorilli, M. Nanotechnology-based drug delivery systems and herbal medicines: A review. Int. J. Nanomed. 2013, 9, 1–15.
  12. Yuan, H.; Ma, Q.; Ye, L.; Piao, G. The traditional medicine and modern medicine from natural products. Molecules 2016, 21, 559.
  13. Namdari, M.; Eatemadi, A.; Soleimaninejad, M.; Hammed, A.T. A brief review on the application of nanoparticle enclosed herbal medicine for the treatment of infective endocarditis. Biomed. Pharmacother. 2017, 87, 321–331.
  14. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; del Pilar Rodriguez-Torres, M.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71.
  15. Guerra, M.; Blázquez, J.L.; Rodríguez, E.M. Blood–brain barrier and foetal-onset hydrocephalus, with a view on potential novel treatments beyond managing CSF flow. Fluids Barriers CNS 2017, 14, 1–15.
  16. Dong, X. Current strategies for brain drug delivery. Theranostics 2018, 8, 1481–1493.
  17. Makkar, R.; Behl, T.; Bungau, S.; Zengin, G.; Mehta, V.; Kumar, A.; Uddin, S.; Ashraf, G.M.; Abdel-Daim, M.M.; Arora, S.; et al. Nutraceuticals in neurological disorders. Int. J. Mol. Sci. 2020, 21, 4424.
  18. Uddin, S.; Hossain, F.; Al Mamun, A.; Shah, M.A.; Hasana, S.; Bulbul, I.J.; Sarwar, S.; Mansouri, R.A.; Ashraf, G.M.; Rauf, A.; et al. Exploring the multimodal role of phytochemicals in the modulation of cellular signaling pathways to combat age-related neurodegeneration. Sci. Total Environ. 2020, 725, 138313.
  19. Zhu, F.-D.; Hu, Y.-J.; Yu, L.; Zhou, X.-G.; Wu, J.-M.; Tang, Y.; Qin, D.-L.; Fan, Q.-Z.; Wu, A.-G. Nanoparticles: A hope for the treatment of inflammation in CNS. Front. Pharmacol. 2021, 12, 683935.
  20. Schmitt, C.; Lechanteur, A.; Cossais, F.; Bellefroid, C.; Arnold, P.; Lucius, R.; Held-Feindt, J.; Piel, G.; Hattermann, K. Liposomal encapsulated curcumin effectively attenuates neuroinflammatory and reactive astrogliosis reactions in glia cvells and organotypic brain slices. Int. J. Nanomed. 2020, ume 15, 3649–3667.
  21. Alphandéry, E. Nano-therapies for glioblastoma treatment. Cancers 2020, 12, 242.
  22. Ege, D. Action mechanisms of curcumin in Alzheimer’s disease and its brain targeted delivery. Materials 2021, 14, 3332.
  23. Sadegh Malvajerd, S.; Azadi, A.; Izadi, Z.; Kurd, M.; Dara, T.; Dibaei, M.; Zadeh, M.S.; Javar, H.A.; Hamidi, M. Brain delivery of curcumin using solid lipid nanoparticles and nanostructured lipid carriers: Preparation, optimization, and pharmacokinetic evaluation. ACS Chem. Neurosci. 2019, 10, 728–739.
  24. Jeon, S.G.; Cha, M.-Y.; Kim, J.-I.; Hwang, T.W.; Kim, K.A.; Kim, T.H.; Song, K.C.; Kim, J.-J.; Moon, M. Vitamin D-binding protein-loaded PLGA nanoparticles suppress Alzheimer’s disease-related pathology in 5XFAD mice. Nanomed. Nanotechnol. Biol. Med. 2019, 17, 297–307.
  25. Kim, J.W.; Cho, C.H.; Hwang, Y.-G.; Park, W.J.; Kang, H.; Kim, D.-O. Protective effects of red ginseng treated with gold nanoparticles against H2O2 -induced oxidative stress in neuronal PC-12 cells. Korean J. Food Sci. Technol. 2017, 49, 222–227.
  26. Yang, L.; Li, C.-L.; Tsai, T.-H. Preclinical herb-drug pharmacokinetic interaction of Panax ginseng extract and selegiline in freely moving rats. ACS Omega 2020, 5, 4682–4688.
  27. Pattan, G.; Kaul, G. Health hazards associated with nanomaterials. Toxicol. Ind. Heal. 2012, 30, 499–519.
  28. Wang, J.; Chen, C.; Liu, Y.; Jiao, F.; Li, W.; Lao, F.; Li, Y.-F.; Li, B.; Ge, C.; Zhou, G. Potential neurological lesion after nasal instillation of TiO2 nanoparticles in the anatase and rutile crystal phases. Toxicol. Lett. 2008, 183, 72–80.
  29. Pisanic, T.R.; Blackwell, J.D.; Shubayev, V.I.; Fiñones, R.R.; Jin, S. Nanotoxicity of iron oxide nanoparticle internalization in growing neurons. Biomaterials 2007, 28, 2572–2581.
  30. Zhao, J.; Xu, L.; Zhang, T.; Ren, G.; Yang, Z. Influences of nanoparticle zinc oxide on acutely isolated rat hippocampal CA3 pyramidal neurons. NeuroToxicology 2009, 30, 220–230.
  31. Cha, K.E.; Myung, H. Cytotoxic effects of nanoparticles assessed in vitro and in vivo. J. Microbiol. Biotechnol. 2007, 17, 1573–1578.
  32. Van Der Laan, D.M.; Elders, P.J.M.; Boons, C.C.L.M.; Nijpels, G.; Krska, J.; Hugtenburg, J.G. The impact of cardiovascular medication use on patients’ daily lives: A cross-sectional study. Int. J. Clin. Pharm. 2018, 40, 412–420.
  33. Chronic kidney disease in the United States. 2021. Available online: https://www.cdc.gov/kidneydisease/publications-resources/ckd-national-facts.html (accessed on 2 June 2021).
  34. Hesari, M.; Mohammadi, P.; Khademi, F.; Shackebaei, D.; Momtaz, S.; Moasefi, N.; Farzaei, M.H.; Abdollahi, M. Current Advances in the use of nanophytomedicine therapies for human cardiovascular diseases. Int. J. Nanomed. 2021, 16, 3293–3315.
  35. Owen, A.; Dufès, C.; Moscatelli, D.; Mayes, E.; Lovell, J.F.; Katti, K.V.; Sokolov, K.; Mazza, M.; Fontaine, O.; Rannard, S.; et al. The application of nanotechnology in medicine: Treatment and diagnostics. Nanomedicine 2014, 9, 1291–1294.
  36. Wang, D.K.; Rahimi, M.; Filgueira, C.S. Nanotechnology applications for cardiovascular disease treatment: Current and future perspectives. Nanomed. Nanotechnol. Biol. Med. 2021, 34, 102387.
  37. Gera, M.; Sharma, N.; Ghosh, M.; Huynh, D.L.; Lee, S.J.; Min, T.; Kwon, T.; Jeong, D.K. Nanoformulations of curcumin: An emerging paradigm for improved remedial application. Oncotarget 2017, 8, 66680–66698.
  38. Salehi, B.; Prado-Audelo, D.; María, L.; Cortés, H.; Leyva-Gómez, G.; Stojanović-Radić, Z.; Singh, Y.D.; Patra, J.K.; Das, G.; Martins, N.; et al. Therapeutic applications of curcumin nanomedicine formulations in cardiovascular diseases. J. Clin. Med. 2020, 9, 746.
  39. Lozano, O.; Lázaro-Alfaro, A.; Silva-Platas, C.; Oropeza-Almazán, Y.; Torres-Quintanilla, A.; Bernal-Ramirez, J.; Figueiredo, H.; García-Rivas, G. Nanoencapsulated quercetin improves cardioprotection during hypoxia-reoxygenation injury through preservation of mitochondrial function. Oxidative Med. Cell. Longev. 2019, 2019, 1–14.
  40. Bonnefont-Rousselot, D. Resveratrol and Cardiovascular Diseases. Nutrients 2016, 8, 250.
  41. Duan, Y.; Dhar, A.; Patel, C.; Khimani, M.; Neogi, S.; Sharma, P.; Kumar, N.S.; Vekariya, R.L. A brief review on solid lipid nanoparticles: Part and parcel of contemporary drug delivery systems. RSC Adv. 2020, 10, 26777–26791.
  42. Lin, Y.; Li, Y.; Zeng, Y.; Tian, B.; Qu, X.; Yuan, Q.; Song, Y. Pharmacology, toxicity, bioavailability, and formulation of magnolol: An update. Front. Pharmacol. 2021, 12, 632767.
  43. Chen, C.Y.-C.; Wu, C.-H. Magnolol encapsulated by liposome in inhibiting smooth muscle cell proliferation. J. Chin. Chem. Soc. 2008, 55, 517–521.
  44. Zhang, J.; Wang, H.; Yan, X.; Zhang, L. Comparison of short-term toxicity between Nano-Se and selenite in mice. Life Sci. 2005, 76, 1099–1109.
  45. Zhang, S.; Wang, J.; Pan, J. Baicalin-loaded PEGylated lipid nanoparticles: Characterization, pharmacokinetics, and protective effects on acute myocardial ischemia in rats. Drug Deliv. 2016, 23, 3696–3703.
  46. Xin, L.; Gao, J.; Lin, H.; Qu, Y.; Shang, C.; Wang, Y.; Lu, Y.; Cui, X. Regulatory mechanisms of baicalin in cardiovascular diseases: A review. Front. Pharmacol. 2020, 11, 583200.
  47. Xia, L.-M.; Luo, M.-H. Study progress of berberine for treating cardiovascular disease. Chronic Dis. Transl. Med. 2015, 1, 231–235.
  48. Duong, T.; Isomäki, A.; Paaver, U.; Laidmäe, I.; Tõnisoo, A.; Yen, T.; Kogermann, K.; Raal, A.; Heinämäki, J.; Pham, T.-M.-H. Nanoformulation and evaluation of oral berberine-loaded liposomes. Molecules 2021, 26, 2591.
  49. Summerlin, N.; Soo, E.; Thakur, S.; Qu, Z.; Jambhrunkar, S.; Popat, A. Resveratrol nanoformulations: Challenges and opportunities. Int. J. Pharm. 2015, 479, 282–290.
  50. Shahraki, A.; Bahadorikhalili, S.; Hashemzaei, M.; Hajinezhad, M.; Afsharimoghaddam, A.; Sarani, F.; Tajrobekar, O. Resveratrol nano-capsule as an efficient tool for blood pressure regulation: A study on metabolic syndrome induced mice. Biosci Biotechnol. Res Commun. 2017, 10, 623–630.
  51. Shen, P.; Zhang, Z.; He, Y.; Gu, C.; Zhu, K.; Li, S.; Li, Y.; Lu, X.; Liu, J.; Zhang, N.; et al. Magnolol treatment attenuates dextran sulphate sodium-induced murine experimental colitis by regulating inflammation and mucosal damage. Life Sci. 2018, 196, 69–76.
  52. Stefanache, A.; Ignat, M.; Peptu, C.A.; Diaconu, A.; Stoleriu, I.; Ochiuz, L. Development of a prolonged-release drug delivery system with magnolol loaded in amino-functionalized mesoporous silica. Appl. Sci. 2017, 7, 237.
  53. Sheng, Y.-L.; Xu, J.-H.; Shi, C.-H.; Li, W.; Xu, H.-Y.; Li, N.; Zhao, Y.-Q.; Zhang, X.-R. UPLC-MS/MS-ESI assay for simultaneous determination of magnolol and honokiol in rat plasma: Application to pharmacokinetic study after administration emulsion of the isomer. J. Ethnopharmacol. 2014, 155, 1568–1574.
  54. Wang, Y.-J.; Chien, Y.-C.; Wu, C.-H.; Liu, D.-M. Magnolol-loaded core–shell hydrogel nanoparticles: Drug release, intracellular uptake, and controlled cytotoxicity for the inhibition of migration of vascular Ssooth muscle cells. Mol. Pharm. 2011, 8, 2339–2349.
  55. Singh, R.; Smitha, M.S.; Singh, S.P. The Role of Nanotechnology in combating multi-drug resistant bacteria. J. Nanosci. Nanotechnol. 2014, 14, 4745–4756.
  56. Natan, M.; Banin, E. From Nano to Micro: Using nanotechnology to combat microorganisms and their multidrug resistance. FEMS Microbiol. Rev. 2017, 41, 302–322.
  57. Hayat, S.; Muzammil, S.; Fakhar-E-Alam, M.; Aslam, B.; Siddique, M.H.; Nisar, M.A.; Saqalein, M.; Atif, M.; Sarwar, A.; Khurshid, A.; et al. Nanoantibiotics future nanotechnologies to combat antibiotic resistance. Front. Biosci. 2018, 10, 352–374.
  58. Slavin, Y.N.; Asnis, J.; Häfeli, U.O.; Bach, H. Metal nanoparticles: Understanding the mechanisms behind antibacterial activity. J. Nanobiotechnol. 2017, 15, 65.
  59. Borase, H.P.; Salunke, B.K.; Salunkhe, R.B.; Patil, C.D.; Hallsworth, J.E.; Kim, B.S.; Patil, S.V. Plant Extract: A Promising biomatrix for ecofriendly, controlled synthesis of silver nanoparticles. Appl. Biochem. Biotechnol. 2014, 173, 1–29.
  60. Mohammadinejad, R.; Shavandi, A.; Raie, D.S.; Sangeetha, J.; Soleimani, M.; Hajibehzad, S.S.; Thangadurai, D.; Hospet, R.; Popoola, J.O.; Arzani, A.; et al. Plant molecular farming: Production of metallic nanoparticles and therapeutic proteins using green factories. Green Chem. 2019, 21, 1845–1865.
  61. Iravani, S. Green synthesis of metal nanoparticles using plants. Green Chem. 2011, 13, 2638–2650.
  62. Mittal, A.K.; Chisti, Y.; Banerjee, U.C. Synthesis of metallic nanoparticles using plant extracts. Biotechnol. Adv. 2013, 31, 346–356.
  63. Lee, S.Y.; Krishnamurthy, S.; Cho, C.-W.; Yun, Y.-S. Biosynthesis of gold nanoparticles Using Ocimum sanctum extracts by solvents with different polarity. ACS Sustain. Chem. Eng. 2016, 4, 2651–2659.
  64. Lü, S.; Wu, Y.; Liu, H. Silver nanoparticles synthesized using Eucommia ulmoides bark and their antibacterial efficacy. Mater. Lett. 2017, 196, 217–220.
  65. Aiad, I.; Marzouk, M.; Shaker, S.A.; Ebrahim, N.E.; Abd-Elaal, A.; Tawfik, S.M. Antipyrine cationic surfactants capping silver nanoparticles as potent antimicrobial agents against pathogenic bacteria and fungi. J. Mol. Liq. 2017, 243, 572–583.
  66. Kuppusamy, P.; Ichwan, S.J.A.; Parine, N.R.; Yusoff, M.; Maniam, G.P.; Govindan, N. Intracellular biosynthesis of Au and Ag nanoparticles using ethanolic extract of Brassica oleracea L. and studies on their physicochemical and biological properties. J. Environ. Sci. 2015, 29, 151–157.
  67. Rosa, R.M.; Silva, J.C.; Sanches, I.S.; Henriques, C. Simultaneous photo-induced cross-linking and silver nanoparticle formation in a PVP electrospun wound dressing. Mater. Lett. 2017, 207, 145–148.
  68. Huy, T.Q.; Thanh, N.T.H.; Thuy, N.T.; Van Chung, P.; Hung, P.N.; Le, A.-T. Cytotoxicity and antiviral activity of electrochemical-synthesized silver nanoparticles against poliovirus. J. Virol. Methods 2017, 241, 52–57.
  69. Jannat, K.; Paul, A.K.; Bondhon, T.A.; Hasan, A.; Nawaz, M.; Jahan, R.; Mahboob, T.; Nissapatorn, V.; Wilairatana, P.; Pereira, M.D.L.; et al. Nanotechnology applications of flavonoids for viral diseases. Pharmaceutics 2021, 13, 1895.
  70. Sofy, A.R.; Hmed, A.A.; El Haliem, N.F.A.; Zein, M.A.-E.; Elshaarawy, R.F. Polyphosphonium-oligochitosans decorated with nanosilver as new prospective inhibitors for common human enteric viruses. Carbohydr. Polym. 2019, 226, 115261.
  71. Li, Y.; Lin, Z.; Xu, T.; Wang, C.; Zhao, M.; Xiao, M.; Wang, H.; Deng, N.; Zhu, B. Delivery of VP1 siRNA to inhibit the EV71 virus using functionalized silver nanoparticles through ROS-mediated signaling pathways. RSC Adv. 2017, 7, 1453–1463.
  72. Yang, X.X.; Li, C.M.; Huang, C.Z. Curcumin modified silver nanoparticles for highly efficient inhibition of respiratory syncytial virus infection. Nanoscale 2016, 8, 3040–3048.
  73. Li, Y.; Lin, Z.; Zhao, M.; Xu, T.; Wang, C.; Hua, L.; Wang, H.; Xia, H.; Zhu, B. Silver Nanoparticle based codelivery of ooeltamivir to inhibit the activity of the H1N1 influenza virus through ROS-mediated signaling pathways. ACS Appl. Mater. Interfaces 2016, 8, 24385–24393.
  74. Bartczak, D.; Muskens, O.L.; Sanchez-Elsner, T.; Kanaras, A.G.; Millar, T.M. Manipulation of in vitro angiogenesis using peptide-coated gold nanoparticles. ACS Nano 2013, 7, 5628–5636.
  75. El-Gaffary, M.; Bashandy, M.M.; Ahmed, A.R.; El-Borady, O.M. Self-assembled gold nanoparticles for in-vitro inhibition of bovine viral diarrhea virus as surrogate model for HCV. Mater. Res. Express 2019, 6, 075075.
  76. Sametband, M.; Shukla, S.; Meningher, T.; Hirsh, S.; Mendelson, E.; Sarid, R.; Gedanken, A.; Mandelboim, M. Effective multi-strain inhibition of influenza virus by anionic gold nanoparticles. Med. Chem. Comm. 2011, 2, 421–423.
  77. Fahmi, M.Z.; Sukmayani, W.; Khairunisa, S.Q.; Witaningrum, A.M.; Indriati, D.W.; Matondang, M.Q.Y.; Chang, J.-Y.; Kotaki, T.; Kameoka, M. Design of boronic acid-attributed carbon dots on inhibits HIV-1 entry. RSC Adv. 2016, 6, 92996–93002.
  78. Tong, T.; Hu, H.; Zhou, J.; Deng, S.; Zhang, X.; Tang, W.; Fang, L.; Xiao, S.; Liang, J. Glycyrrhizic-acid-based carbon dots with high antiviral activity by multisite inhibition mechanisms. Small 2020, 16, e1906206.
  79. Du, X.; Xiao, R.; Fu, H.; Yuan, Z.; Zhang, W.; Yin, L.; He, C.; Li, C.; Zhou, J.; Liu, G.; et al. Hypericin-loaded graphene oxide protects ducks against a novel duck reovirus. Mater. Sci. Eng. C 2019, 105, 110052.
  80. Donskyi, I.S.; Azab, W.; Cuellar-Camacho, J.L.; Guday, G.; Lippitz, A.; Unger, W.E.S.; Osterrieder, K.; Adeli, M.; Haag, R. Functionalized nanographene sheets with high antiviral activity through synergistic electrostatic and hydrophobic interactions. Nanoscale 2019, 11, 15804–15809.
  81. Heya, M.; David Torres-Hernández, Á.; Heya, M.S.; Torres Hernández, A.D.; Cordero Díaz, A.; García Coronado, P.L. Biomedical apllication of nanoformulated plant Diagnóstico de las dermatofitosis View project Biomedical apllication of nanoformulated plant View project Biomedical apllication of nanoformulated plant. Artic. Int. J. Pharm. Sci. Res. 2022, 13, 1000.
  82. Madureira, A.R.; Pereira, A.; Pintado, M. Current state on the development of nanoparticles for use against bacterial gastrointestinal pathogens. Focus on chitosan nanoparticles loaded with phenolic compounds. Carbohydr. Polym. 2015, 130, 429–439.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 791
Revisions: 2 times (View History)
Update Date: 25 Jan 2022
1000/1000
Video Production Service