Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Front Endocrinol (Lausanne) ; 14: 1143261, 2023.
Article in English | MEDLINE | ID: mdl-37056674

ABSTRACT

For many years, luteinizing hormone-releasing hormone or gonadotropin-releasing hormone (GnRH) analogs have been used to treat androgen or estrogen-dependent tumors. However, emerging evidence shows that the GnRH receptor (GnRH-R) is overexpressed in several cancer cells, including ovarian, endometrial, and prostate cancer cells, suggesting that GnRH analogs could exert direct antitumoral actions in tumoral tissues that express GnRH-R. Another recent approach based on this knowledge was the use of GnRH peptides for developing specific targeted therapies, improving the delivery and accumulation of drugs in tumoral cells, and decreasing most side effects of current treatments. In this review, we discuss the conventional uses of GnRH analogs, together with the recent advances in GnRH-based drug delivery for ovarian, breast, and prostatic cancer cells.


Subject(s)
Gonadotropin-Releasing Hormone , Prostatic Neoplasms , Male , Female , Humans , Prostatic Neoplasms/drug therapy , Prostate , Ovary , Drug Delivery Systems
2.
Int J Nanomedicine ; 18: 8169-8185, 2023.
Article in English | MEDLINE | ID: mdl-38169997

ABSTRACT

Introduction: The development of new materials and tools for radiology is key to the implementation of this diagnostic technique in clinics. In this work, we evaluated the differential accumulation of peptide-functionalized GNRs in a transgenic animal model (APPswe/PSENd1E9) of Alzheimer's disease (AD) by computed tomography (CT) and measured the pharmacokinetic parameters and bioaccumulation of the nanosystem. Methods: The GNRs were functionalized with two peptides, Ang2 and D1, which conferred on them the properties of crossing the blood-brain barrier and binding to amyloid aggregates, respectively, thus making them a diagnostic tool with great potential for AD. The nanosystem was administered intravenously in APPswe/PSEN1dE9 model mice of 4-, 8- and 18-months of age, and the accumulation of gold nanoparticles was observed by computed tomography (CT). The gold accumulation and biodistribution were determined by atomic absorption. Results: Our findings indicated that 18-month-old animals treated with our nanosystem (GNR-D1/Ang2) displayed noticeable differences in CT signals compared to those treated with a control nanosystem (GNR-Ang2). However, no such distinctions were observed in younger animals. This suggests that our nanosystem holds the potential to effectively detect AD pathology. Discussion: These results support the future development of gold nanoparticle-based technology as a more effective and accessible alternative for the diagnosis of AD and represent a significant advance in the development of gold nanoparticle applications in disease diagnosis.


Subject(s)
Alzheimer Disease , Metal Nanoparticles , Nanotubes , Mice , Animals , Gold/chemistry , Bioaccumulation , Tissue Distribution , Metal Nanoparticles/chemistry , Peptides/chemistry , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Amyloid/metabolism , Tomography, X-Ray Computed , Nanotubes/chemistry , Tomography , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Disease Models, Animal , Brain/metabolism
3.
Pharmaceutics ; 14(5)2022 Apr 28.
Article in English | MEDLINE | ID: mdl-35631544

ABSTRACT

One of the recent attractive therapeutic approaches for cancer treatment is restoring downregulated microRNAs. They play an essential muti-regulatory role in cellular processes such as proliferation, differentiation, survival, apoptosis, cell cycle, angiogenesis, and metastasis, among others. In this study, a gold nanoplatform (GNPF) carrying miR-145, a downregulated microRNA in many cancer types, including epithelial ovarian cancer, was designed and synthesized. For targeting purposes, the GNPF was functionalized with the FSH33 peptide, which provided selectivity for ovarian cancer, and loaded with the miR-145 to obtain the nanosystem GNPF-miR-145. The GNPF-mir-145 was selectively incorporated in A2780 and SKOV3 cells and significantly inhibited cell viability and migration and exhibited proliferative and anchor-independent growth capacities. Moreover, it diminished VEGF release and reduced the spheroid size of ovarian cancer through the damage of cell membranes, thus decreasing cell viability and possibly activating apoptosis. These results provide important advances in developing miR-based therapies using nanoparticles as selective vectors and provide approaches for in vivo evaluation.

4.
Nanomedicine ; 44: 102569, 2022 08.
Article in English | MEDLINE | ID: mdl-35595016

ABSTRACT

The ß-amyloid (Aß) peptide is one of the key etiological agents in Alzheimer's disease (AD). The in vivo detection of Aß species is challenging in all stages of the illness. Currently, the development of fluorescent probes allows the detection of Aß in animal models in the near-infrared region (NIR). However, considering future applications in biomedicine, it is relevant to develop strategies to improve detection of amyloid aggregates using NIR probes. An innovative approach to increase the fluorescence signal of these fluorophores is the use of plasmonic gold nanoparticles (surface-enhanced fluorescence effect). In this work, we improved the detection of Aß aggregates in C. elegans and mouse models of AD by co-administering functionalized gold nanorods (GNRs-PEG-D1) with the fluorescent probes CRANAD-2 or CRANAD-58, which bind selectively to different amyloid species (soluble and insoluble). This work shows that GNRs improve the detection of Aß using NIR probes in vivo.


Subject(s)
Alzheimer Disease , Metal Nanoparticles , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Animals , Caenorhabditis elegans , Fluorescent Dyes/chemistry , Gold , Metal Nanoparticles/chemistry , Mice
5.
Int J Mol Sci ; 23(5)2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35269699

ABSTRACT

Searching for adequate and effective compounds displaying antimicrobial activities, especially against Gram-positive bacteria, is an important research area due to the high hospitalization and mortality rates of these bacterial infections in both the human and veterinary fields. In this work, we explored (E)-4-amino-3-((3,5-di-tert-butyl-2-hydroxybenzylidene)amino) benzoic acid (SB-1, harboring an intramolecular hydrogen bond) and (E)-2-((4-nitrobenzilidene)amino)aniline (SB-2), two Schiff bases derivatives. Results demonstrated that SB-1 showed an antibacterial activity determined by the minimal inhibitory concentration (MIC) against Staphylococcus aureus, Enterococcus faecalis, and Bacillus cereus (Gram-positive bacteria involved in human and animal diseases such as skin infections, pneumonia, diarrheal syndrome, and urinary tract infections, among others), which was similar to that shown by the classical antibiotic chloramphenicol. By contrast, this compound showed no effect against Gram-negative bacteria (Klebsiella pneumoniae, Escherichia coli, and Salmonella enterica). Furthermore, we provide a comprehensive physicochemical and theoretical characterization of SB-1 (as well as several analyses for SB-2), including elemental analysis, ESMS, 1H and 13C NMR (assigned by 1D and 2D techniques), DEPT, UV-Vis, FTIR, and cyclic voltammetry. We also performed a computational study through the DFT theory level, including geometry optimization, TD-DFT, NBO, and global and local reactivity analyses.


Subject(s)
Gram-Positive Bacteria , Schiff Bases , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Escherichia coli , Gram-Negative Bacteria , Microbial Sensitivity Tests , Schiff Bases/chemistry , Schiff Bases/pharmacology
6.
Nanoscale ; 13(42): 17807-17821, 2021 Nov 04.
Article in English | MEDLINE | ID: mdl-34668502

ABSTRACT

Gold nanoparticles (GNPs) are an attractive nanomaterial for potential applications in therapy and diagnostics due to their capability to direct toward specific sites in the organism. However, when exposed to plasma, GNPs can interact with different biomolecules that form a dynamic nano-bio interface called a "protein corona" (PC). Remarkably, the PC could affect multiple biological processes, such as cell targeting and uptake, cytotoxicity, and nanoparticle (NP) clearance. The interaction of nanomaterials with plasmatic proteins has been widely studied under bulk conditions, however, under dynamic conditions, it has just recently been explored. Thus, to mimic a dynamic natural environment found in arteries and veins, microfluidic devices were used. In this work, gold nanorods (GNRs) were synthesized and conjugated with polyethylene glycol (PEG) to reduce their interaction with plasma proteins and increase their biocompatibility. Then, GNRs were functionalized with folic acid, a targeting ligand typically used to recognize tumor cells. The resulting nanosystem was exposed to fibrinogen (FB) to study the development and biological impact of PC formation through two strategies: bulk and laminar flow conditions. The obtained nanosystems were characterized by absorption spectrophotometry, DLS, laser Doppler microelectrophoresis, neutron activation analysis, circular dichroism spectroscopy and TEM. Finally, cell viability and cellular uptake assays were performed to study the influence of the PC on the cell viability and delivery of nanosystems.


Subject(s)
Metal Nanoparticles , Nanotubes , Neoplasms , Adsorption , Fibrinogen , Folic Acid , Gold , Metal Nanoparticles/toxicity , Microfluidics , Neoplasms/drug therapy , Polyethylene Glycols
7.
Pharmaceutics ; 13(8)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34452165

ABSTRACT

Gold nanoparticles (AuNPs) have been shown to be outstanding tools for drug delivery and biomedical applications, mainly owing to their colloidal stability, surface chemistry, and photothermal properties. The biocompatibility and stability of nanoparticles can be improved by capping the nanoparticles with endogenous proteins, such as albumin. Notably, protein coating of nanoparticles can interfere with and decrease their cell penetration. Therefore, in the present study, we functionalized albumin with the r8 peptide (All-D, octaarginine) and used it for coating NIR-plasmonic anisotropic gold nanoparticles. Gold nanoprisms (AuNPrs) and gold nanorods (AuNRs) were coated with bovine serum albumin (BSA) previously functionalized using a cell penetrating peptide (CPP) with the r8 sequence (BSA-r8). The effect of the coated and r8-functionalized AuNPs on HeLa cell viability was assessed by the MTS assay, showing a low effect on cell viability after BSA coating. Moreover, the internalization of the nanostructures into HeLa cells was assessed by confocal microscopy and transmission electron microscopy (TEM). As a result, both nanoconstructs showed an improved internalization level after being capped with BSA-r8, in contrast to the BSA-functionalized control, suggesting the predominant role of CPP functionalization in cell internalization. Thus, our results validate both novel nanoconstructs as potential candidates to be coated by endogenous proteins and functionalized with a CPP to optimize cell internalization. In a further approach, coating AuNPs with CPP-functionalized BSA can broaden the possibilities for biomedical applications by combining their optical properties, biocompatibility, and cell-penetration abilities.

8.
Front Microbiol ; 12: 672467, 2021.
Article in English | MEDLINE | ID: mdl-34025627

ABSTRACT

The appearance of multi-resistant strains has contributed to reintroducing polymyxin as the last-line therapy. Although polymyxin resistance is based on bacterial envelope changes, other resistance mechanisms are being reported. Outer membrane vesicles (OMVs) are nanosized proteoliposomes secreted from the outer membrane of Gram-negative bacteria. In some bacteria, OMVs have shown to provide resistance to diverse antimicrobial agents either by sequestering and/or expelling the harmful agent from the bacterial envelope. Nevertheless, the participation of OMVs in polymyxin resistance has not yet been explored in S. Typhi, and neither OMVs derived from hypervesiculating mutants. In this work, we explored whether OMVs produced by the hypervesiculating strains Salmonella Typhi ΔrfaE (LPS synthesis), ΔtolR (bacterial envelope) and ΔdegS (misfolded proteins and σ E activation) exhibit protective properties against polymyxin B. We found that the OMVs extracted from S. Typhi ΔtolR and ΔdegS protect S. Typhi WT from polymyxin B in a concentration-depending manner. By contrast, the protective effect exerted by OMVs from S. Typhi WT and S. Typhi ΔrfaE is much lower. This effect is achieved by the sequestration of polymyxin B, as assessed by the more positive Zeta potential of OMVs with polymyxin B and the diminished antibiotic's availability when coincubated with OMVs. We also found that S. Typhi ΔtolR exhibited an increased MIC of polymyxin B. Finally, we determined that S. Typhi ΔtolR and S. Typhi ΔdegS, at a lesser level, can functionally and transiently transfer the OMV-mediated polymyxin B resistance to susceptible bacteria in cocultures. This work shows that mutants in genes related to OMVs biogenesis can release vesicles with improved abilities to protect bacteria against membrane-active agents. Since mutations affecting OMV biogenesis can involve the bacterial envelope, mutants with increased resistance to membrane-acting agents that, in turn, produce protective OMVs with a high vesiculation rate (e.g., S. Typhi ΔtolR) can arise. Such mutants can functionally transfer the resistance to surrounding bacteria via OMVs, diminishing the effective concentration of the antimicrobial agent and potentially favoring the selection of spontaneous resistant strains in the environment. This phenomenon might be considered the source for the emergence of polymyxin resistance in an entire bacterial community.

9.
Biomater Sci ; 9(11): 4178-4190, 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-33982040

ABSTRACT

The development and use of nanosystems is an emerging strategy for the diagnosis and treatment of a broad number of diseases, such as Alzheimer's disease (AD). Here, we developed a neurotheranostic nanosystem based on gold nanorods (GNRs) that works as a therapeutic peptide delivery system and can be detected in vivo for microcomputed tomography (micro-CT), being a diagnostic tool. GNRs functionalized with the peptides Ang2 (a shuttle to the Central Nervous System) and D1 (that binds to the Aß peptide, also inhibiting its aggregation) allowed detecting differences in vivo between wild type and AD mice (APPswe/PSEN1dE9) 15 minutes after a single dose by micro-CT. Moreover, after a recurrent treatment for one month with GNRs-D1/Ang2, we observed a diminution of amyloid load and inflammatory markers in the brain. Thus, this new designed nanosystem exhibits promising properties for neurotheranostics of AD.


Subject(s)
Alzheimer Disease , Nanotubes , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Gold , Mice , Mice, Transgenic , X-Ray Microtomography
10.
Int J Nanomedicine ; 16: 2187-2201, 2021.
Article in English | MEDLINE | ID: mdl-33758506

ABSTRACT

The physicochemical and optical properties of silver nanoparticles (SNPs) and gold nanoparticles (GNPs) have allowed them to be employed for various biomedical applications, including delivery, therapy, imaging, and as theranostic agents. However, since they are foreign body systems, they are usually redistributed and accumulated in some vital organs, which can produce toxic effects; therefore, this a crucial issue that should be considered for potential clinical trials. This review aimed to summarize the reports from the past ten years that have used SNPs and GNPs for in vivo studies on the diagnosis and treatment of brain diseases and those related to the central nervous system, emphasizing their toxicity as a crucial topic address. The article focuses on the effect of the nanoparticle´s size and chemical composition as relevant parameters for in vivo toxicity. At the beginning of this review, the general toxicity and distribution studies are discussed separately for SNPs and GNPs. Subsequently, this manuscript analyzes the principal applications of both kinds of nanoparticles for glioma, neurodegenerative, and other brain diseases, and discusses the advances in clinical trials. Finally, we analyze research prospects towards clinical applications for both types of metallic nanoparticles.


Subject(s)
Central Nervous System Diseases/pathology , Gold/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Particle Size , Silver/chemistry , Toxicity Tests , Animals , Humans , Metal Nanoparticles/ultrastructure , Tissue Distribution/drug effects
11.
Int J Nanomedicine ; 15: 1837-1851, 2020.
Article in English | MEDLINE | ID: mdl-32256063

ABSTRACT

INTRODUCTION: Gold nanorods are highly reactive, have a large surface-to-volume ratio, and can be functionalized with biomolecules. Gold nanorods can absorb infrared electromagnetic radiation, which is subsequently dispersed as local heat. Gold nanoparticles can be used as powerful tools for the diagnosis and therapy of different diseases. To improve the biological barrier permeation of nanoparticles with low cytotoxicity, in this study, we conjugated gold nanorods with cell-penetrating peptides (oligoarginines) and with the amphipathic peptide CLPFFD. METHODS: We studied the interaction of the functionalized gold nanorods with biological membrane models (liposomes) by dynamic light scattering, transmission electron microscopy and the Langmuir balance. Furthermore, we evaluated the effects on cell viability and permeability with an MTS assay and TEM. RESULTS AND DISCUSSION: The interaction study by DLS, the Langmuir balance and cryo-TEM support that GNR-Arg7CLPFFD enhances the interactions between GNRs and biological membranes. In addition, cells treated with GNR-Arg7CLPFFD internalized 80% more nanoparticles than cells treated with GNR alone and did not induce cell damage. CONCLUSION: Our results indicate that incorporation of an amphipathic sequence into oligoarginines for the functionalization of gold nanorods enhances biological membrane nanoparticle interactions and nanoparticle cell permeability with respect to nanorods functionalized with oligoarginine. Overall, functionalized gold nanorods with amphipathic arginine rich peptides might be candidates for improving drug delivery by facilitating biological barrier permeation.


Subject(s)
Cell-Penetrating Peptides/chemistry , Liposomes/pharmacokinetics , Nanotubes/chemistry , Arginine/chemistry , Cell Line, Tumor , Cell Survival , Cell-Penetrating Peptides/pharmacokinetics , Drug Delivery Systems , Dynamic Light Scattering , Gold/chemistry , Humans , Liposomes/chemistry , Metal Nanoparticles/chemistry , Microscopy, Electron, Transmission , Peptides/chemistry
12.
J Nanobiotechnology ; 18(1): 20, 2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31973696

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs) have shown great potential for targeted therapy, as they have a natural ability to pass through biological barriers and, depending on their origin, can preferentially accumulate at defined sites, including tumors. Analyzing the potential of EVs to target specific cells remains challenging, considering the unspecific binding of lipophilic tracers to other proteins, the limitations of fluorescence for deep tissue imaging and the effect of external labeling strategies on their natural tropism. In this work, we determined the cell-type specific tropism of B16F10-EVs towards cancer cell and metastatic tumors by using fluorescence analysis and quantitative gold labeling measurements. Surface functionalization of plasmonic gold nanoparticles was used to promote indirect labeling of EVs without affecting size distribution, polydispersity, surface charge, protein markers, cell uptake or in vivo biodistribution. Double-labeled EVs with gold and fluorescent dyes were injected into animals developing metastatic lung nodules and analyzed by fluorescence/computer tomography imaging, quantitative neutron activation analysis and gold-enhanced optical microscopy. RESULTS: We determined that B16F10 cells preferentially take up their own EVs, when compared with colon adenocarcinoma, macrophage and kidney cell-derived EVs. In addition, we were able to detect the preferential accumulation of B16F10 EVs in small metastatic tumors located in lungs when compared with the rest of the organs, as well as their precise distribution between tumor vessels, alveolus and tumor nodules by histological analysis. Finally, we observed that tumor EVs can be used as effective vectors to increase gold nanoparticle delivery towards metastatic nodules. CONCLUSIONS: Our findings provide a valuable tool to study the distribution and interaction of EVs in mice and a novel strategy to improve the targeting of gold nanoparticles to cancer cells and metastatic nodules by using the natural properties of malignant EVs.


Subject(s)
Antineoplastic Agents/chemistry , Extracellular Vesicles/chemistry , Gold/chemistry , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Melanoma/chemistry , Metal Nanoparticles/chemistry , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/therapy , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Membrane Permeability , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/therapy , Fluorescent Dyes/chemistry , Humans , Lung/metabolism , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Optical Imaging , Surface Properties , Tissue Distribution
13.
Int J Nanomedicine ; 14: 6387-6406, 2019.
Article in English | MEDLINE | ID: mdl-31496693

ABSTRACT

Nanotechnology is an emerging field which has created great opportunities either through the creation of new materials or by improving the properties of existing ones. Nanoscale materials with a wide range of applications in areas ranging from engineering to biomedicine have been produced. Gold nanoparticles (AuNPs) have emerged as a therapeutic agent, and are useful for imaging, drug delivery, and photodynamic and photothermal therapy. AuNPs have the advantage of ease of functionalization with therapeutic agents through covalent and ionic binding. Combining AuNPs and other materials can result in nanoplatforms, which can be useful for biomedical applications. Biomaterials such as biomolecules, polymers and proteins can improve the therapeutic properties of nanoparticles, such as their biocompatibility, biodistribution, stability and half-life. Serum albumin is a versatile, non-toxic, stable, and biodegradable protein, in which structural domains and functional groups allow the binding and capping of inorganic nanoparticles. AuNPs coated with albumin have improved properties such as greater compatibility, bioavailability, longer circulation times, lower toxicity, and selective bioaccumulation. In the current article, we review the features of albumin, as well as its interaction with AuNPs, focusing on its biomedical applications.


Subject(s)
Albumins/chemistry , Drug Delivery Systems , Gold/chemistry , Metal Nanoparticles/chemistry , Humans , Theranostic Nanomedicine
14.
Molecules ; 23(5)2018 May 18.
Article in English | MEDLINE | ID: mdl-29783629

ABSTRACT

Here we report the incorporation of gold nanostructures (nanospheres or nanorods, functionalized with carboxylate-end PEG) and curcumin oil-in-water (O/W) nanoemulsions (CurNem) into alginate microgels using the dripping technique. While gold nanostructures are promising nanomaterials for photothermal therapy applications, CurNem possess important pharmacological activities as reported here. In this sense, we evaluated the effect of CurNem on cell viability of both cancerous and non-cancerous cell lines (AGS and HEK293T, respectively), demonstrating preferential toxicity in cancer cells and safety for the non-cancerous cells. After incorporating gold nanostructures and CurNem together into the microgels, microstructures with diameters of 220 and 540 µm were obtained. When stimulating microgels with a laser, the plasmon effect promoted a significant rise in the temperature of the medium; the temperature increase was higher for those containing gold nanorods (11⁻12 °C) than nanospheres (1⁻2 °C). Interestingly, the incorporation of both nanosystems in the microgels maintains the photothermal properties of the gold nanostructures unmodified and retains with high efficiency the curcumin nanocarriers. We conclude that these results will be of interest to design hydrogel formulations with therapeutic applications.


Subject(s)
Drug Carriers/chemistry , Gold/chemistry , Nanospheres/chemistry , Nanotubes/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Curcumin/administration & dosage , Curcumin/chemistry , Drug Liberation , Emulsions , Gels , HEK293 Cells , Humans , Lasers , Particle Size , Photochemotherapy/methods , Polyethylene Glycols/chemistry , Solubility , Surface Properties
15.
Colloids Surf B Biointerfaces ; 166: 323-329, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29625410

ABSTRACT

We studied the photothermal release of carboxyfluorescein (CF) linked to the gold surface of gold nanorods (GNRs) by two Diels-Alder adducts of different lengths (n = 4 and n = 9). The functionalized GNRs were irradiated with infrared light to produce photothermal release of CF by a retro-Diels-Alder reaction. The adducts were chemisorbed on the GNRs and the functionalized nanoparticles were characterized by UV-vis, DLS, zeta potential and Raman and surface-enhanced Raman spectroscopy (SERS). On the basis of the degree of nanoparticle functionalization and the SERS results, we inferred the orientation of CF on the surface of the gold nanoparticle. Moreover, we determined the photothermal release profiles of CF from the gold surface by laser irradiation. The release was faster for the longer linker (n = 9). SERS revealed that, for the shorter linker (n = 4), molecules are oriented perpendicularly with respect to the gold surface, thereby maintaining the CF far from the surface. In contrast, the longer linker was observed to be tilted, thus maintaining CF close to the gold surface and therefore potentially favoring the photothermal transfer of energy. These results are relevant for the future development of the spatial and temporal controlled release of drugs by means of gold nanoparticles.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Nanotubes/chemistry , Spectrum Analysis, Raman
16.
Nanomedicine (Lond) ; 12(20): 2503-2517, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28882086

ABSTRACT

AIM: To improve the in vivo delivery of gold nanorods (GNRs) to the central nervous system of rats, these gold nanoparticles were conjugated to Angiopep-2, a shuttle peptide that can cross the blood-brain barrier. MATERIALS & METHODS: GNRs were synthesized and modified using polyethylene glycol and Angiopep-2 (GNR-PEG-Angiopep-2). The physicochemical properties, in vitro cytotoxicity and ex vivo biodistribution of the conjugate were examined. RESULTS: GNR-PEG-Angiopep-2 was stable over the following days, and the different concentrations that were tested did not affect the viability of microvascular endothelial cells. The conjugation of Angiopep-2 to GNRs enhanced the endocytosis of these particles (in vitro) and the accumulation in brains (in vivo), when compared with GNRs modified only with PEG. CONCLUSION: This study provides evidence that Angiopep-2 improves the delivery of GNRs to the brain parenchyma. This property is highly relevant for future applications of GNRs as platforms for photothermal and theranostic purposes.


Subject(s)
Central Nervous System/drug effects , Gold/chemistry , Nanotubes/chemistry , Peptides/chemistry , Peptides/pharmacology , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Survival , Drug Carriers/chemistry , Drug Liberation , Endothelial Cells , Fluorescent Dyes/chemistry , Humans , Male , Microscopy, Electron, Transmission/methods , Optical Imaging/methods , Particle Size , Peptides/toxicity , Permeability , Polyethylene Glycols/chemistry , Rats , Rats, Sprague-Dawley , Surface Properties , Tissue Distribution
17.
Curr Pharm Des ; 23(13): 1916-1926, 2017.
Article in English | MEDLINE | ID: mdl-28056734

ABSTRACT

This review focuses on the application of metal nanoparticles in the diagnosis and treatment of Alzheimer's and Parkinson's diseases. Metal nanoparticles present interesting physicochemical properties that can be applied to increase biomarker detection sensitivities in vitro and in vivo. Furthermore, these nanoparticles could be used in different strategies for the treatment of central nervous system diseases, particularly in regards to drug delivery. Herein, specific potential applications of metal nanoparticles are separately discussed for the contexts of in vitro diagnoses and treatments. Briefly, research using surface plasmon resonance methodologies has mainly used these nanoparticles for the in vitro detection of Aß and, to a lesser extent, of α-synuclein. Regarding treatment approaches, in vitro studies have focused on using metal nanoparticles to manipulate the Aß aggregation, thus reducing toxicity. Furthermore, in vivo applications of metal nanoparticles are also discussed, with many of the existing studies focusing on a magnetic nanoparticle-detection of Aß through magnetic resonance imaging and, to a lesser degree, extension fluorescence techniques. Finally, conclusions and perspectives are provided regarding the real potential for using metal nanoparticles in the treatment and diagnosis of central nervous system diseases.


Subject(s)
Brain Diseases/diagnosis , Brain Diseases/drug therapy , Metal Nanoparticles/therapeutic use , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/drug therapy , Animals , Humans , Metal Nanoparticles/chemistry
18.
Int J Nanomedicine ; 10: 4919-36, 2015.
Article in English | MEDLINE | ID: mdl-26300639

ABSTRACT

An exciting and emerging field in nanomedicine involves the use of gold nanoparticles (AuNPs) in the preclinical development of new strategies for the treatment and diagnosis of brain-related diseases such as neurodegeneration and cerebral tumors. The treatment of many brain-related disorders with AuNPs, which possess useful physical properties, is limited by the blood-brain barrier (BBB). The BBB highly regulates the substances that can permeate into the brain. Peptides and proteins may represent promising tools to improve the delivery of AuNPs to the central nervous system (CNS). In this review, we summarize the potential applications of AuNPs to CNS disorders, discuss different strategies based on the use of peptides or proteins to improve the delivery of AuNPs to the brain, and examine the intranasal administration route, which bypasses the BBB. We also analyze the potential neurotoxicity of AuNPs and the perspectives and new challenges concerning the use of peptides and proteins to enhance the delivery of AuNPs to the brain. The majority of the work described in this review is in a preclinical stage of experimentation, or in select cases, in clinical trials in humans. We note that the use of AuNPs still requires substantial study before being translated into human applications. However, for further clinical research, the issues related to the potential use of AuNPs must be analyzed.


Subject(s)
Brain/metabolism , Drug Carriers , Gold , Metal Nanoparticles , Nanomedicine/methods , Peptides , Brain Diseases , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/therapeutic use , Drug Carriers/toxicity , Gold/chemistry , Gold/pharmacokinetics , Gold/therapeutic use , Gold/toxicity , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Metal Nanoparticles/toxicity , Peptides/chemistry , Peptides/pharmacokinetics , Peptides/therapeutic use , Peptides/toxicity , Proteins/chemistry , Proteins/pharmacokinetics , Proteins/therapeutic use , Proteins/toxicity
19.
Nanomedicine (Lond) ; 9(13): 2023-39, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25343351

ABSTRACT

In this article, we describe how nanoparticles work in photothermally triggered drug delivery, starting with a description of the plasmon resonance and the photothermal effect, and how this is used to release a drug. Then, we describe the four major functionalization strategies and each of their different applications. Finally, we discuss the biodistribution and toxicity of these systems and the necessary requirements for the use of gold nanoparticles for spatially and temporally controlling drug release through the photothermal effect.


Subject(s)
Drug Delivery Systems , Gold/therapeutic use , Metal Nanoparticles/therapeutic use , Neoplasms/drug therapy , Delayed-Action Preparations , Hot Temperature , Humans , Light , Nanotubes/chemistry , Neoplasms/pathology , Polyethylene Glycols/therapeutic use
20.
Nanomedicine (Lond) ; 5(6): 897-913, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20735225

ABSTRACT

BACKGROUND & AIMS: Gold nanoparticles (GNPs) have promising applications for drug delivery as well as for the diagnosis and treatment of several pathologies, such as those related to the CNS. However, GNPs are retained in a number of organs, such as the liver and spleen. Owing to their negative charge and/or processes of opsonization, GNPs are retained by the reticuloendothelial system, thereby decreasing their delivery to the brain. It is therefore crucial to modify the nanoparticle surface in order to increase its lipophilicity and reduce its negative charge, thus achieving enhanced delivery to the brain. RESULTS: In this article, we have shown that conjugation of 12 nm GNPs with the amphipathic peptide CLPFFD increases the in vivo penetration of these particles to the rat brain. The C(GNP)-LPFFD conjugates showed a smaller negative charge and a greater hydrophobic character than citrate-capped GNPs of the same size. We administered intraperitoneal injections of citrate GNPs and C(GNP)-LPFFD in rats, and determined the gold content in the tissues by neutron activation. Compared with citrate GNPs, the C(GNP)-LPFFD conjugate improved the delivery to the brain, increasing the concentration of gold by fourfold, while simultaneously reducing its retention by the spleen 1 and 2 h after injection. At 24 h, the conjugate was partially cleared from the brain, and mainly accumulated in the liver. The C(GNP)-LPFFD did not alter the integrity of the blood-brain barrier, and had no effect on cell viability.


Subject(s)
Brain/metabolism , Gold , Metal Nanoparticles , Peptides/chemistry , Animals , Rats
SELECTION OF CITATIONS
SEARCH DETAIL