Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Bull Exp Biol Med ; 176(3): 399-402, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38342809

RESUMO

A morphological analysis of the liver of Wistar rats was performed 2 months after a single intravenous injection of porous silicon particles of different sizes (60-80, 250-300, and 500-600 nm; 2 mg/ml, 1 ml). Histological, immunohistochemical, and electron microscopic methods showed the development of CD68+ granulomas in all experimental groups. Injection of 60-80-nm porous silicon particles led to the formation of single large granulomas (>2000 µm2), while 500-600-nm nanoparticles caused the formation of numerous smaller granulomas. The mechanism of involution of granulomas by apoptosis of Kupffer cells and the absence of subsequent connective tissue remodeling of the organ tissue is shown.


Assuntos
Fígado , Silício , Ratos , Animais , Ratos Wistar , Fígado/patologia , Granuloma/induzido quimicamente , Granuloma/patologia , Células de Kupffer
2.
Mol Pharm ; 20(1): 545-560, 2023 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-36484477

RESUMO

Clinical treatment of glioblastoma (GBM) remains a major challenge because of the blood-brain barrier, chemotherapeutic resistance, and aggressive tumor metastasis. The development of advanced nanoplatforms that can efficiently deliver drugs and gene therapies across the BBB to the brain tumors is urgently needed. The protein "downregulated in renal cell carcinoma" (DRR) is one of the key drivers of GBM invasion. Here, we engineered porous silicon nanoparticles (pSiNPs) with antisense oligonucleotide (AON) for DRR gene knockdown as a targeted gene and drug delivery platform for GBM treatment. These AON-modified pSiNPs (AON@pSiNPs) were selectively internalized by GBM and human cerebral microvascular endothelial cells (hCMEC/D3) cells expressing Class A scavenger receptors (SR-A). AON was released from AON@pSiNPs, knocked down DRR and inhibited GBM cell migration. Additionally, a penetration study in a microfluidic-based BBB model and a biodistribution study in a glioma mice model showed that AON@pSiNPs could specifically cross the BBB and enter the brain. We further demonstrated that AON@pSiNPs could carry a large payload of the chemotherapy drug temozolomide (TMZ, 1.3 mg of TMZ per mg of NPs) and induce a significant cytotoxicity in GBM cells. On the basis of these results, the nanocarrier and its multifunctional strategy provide a strong potential for clinical treatment of GBM and research for targeted drug and gene delivery.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Camundongos , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Silício , Porosidade , Células Endoteliais , Distribuição Tecidual , Linhagem Celular Tumoral , Temozolomida/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Resistencia a Medicamentos Antineoplásicos/genética
3.
J Nanobiotechnology ; 17(1): 47, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30935403

RESUMO

BACKGROUND: Glioma is a common brain tumor with a high mortality rate. A small population of cells expressing stem-like cell markers in glioma contributes to drug resistance and tumor recurrence. METHODS: Porous silicon nanoparticles (PSi NPs) as photothermal therapy (PTT) agents loaded with TMZ (TMZ/PSi NPs), was combined with hyperbaric oxygen (HBO) therapy in vitro and in vivo. To further investigate underlying mechanism, we detected the expression of stem-like cell markers and hypoxia related molecules in vitro and in vivo after treatment of TMZ/PSi NPs in combination with PTT and HBO. RESULTS: NCH-421K and C6 cells were more sensitive to the combination treatment. Moreover, the expression of stem-like cell markers and hypoxia related molecules were decreased after combination treatment. The in vivo results were in line with in vitro. The combination treatment presents significant antitumor effects in mice bearing C6 tumor compared with the treatment of TMZ, PTT or TMZ/PSi NPs only. CONCLUSION: These results suggested the TMZ/PSi NPs combined with HBO and PTT could be a potential therapeutic strategy for glioma.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/terapia , Glioma/terapia , Nanopartículas/química , Células-Tronco Neoplásicas/patologia , Silício/química , Temozolomida/farmacologia , Animais , Antineoplásicos Alquilantes/química , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Glioma/patologia , Humanos , Oxigenoterapia Hiperbárica , Hipertermia Induzida , Camundongos Nus , Transplante de Neoplasias , Células-Tronco Neoplásicas/efeitos dos fármacos , Tamanho da Partícula , Porosidade , Ratos , Temozolomida/química
4.
Nano Lett ; 18(2): 1448-1453, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29382198

RESUMO

Porous silicon nanoparticles (PSiNPs) and gold nanorods (AuNRs) can be used as biocompatible nanocarriers for delivery of therapeutics but undesired leakage makes them inefficient. By encapsulating the PSiNPs and AuNRs in a hydrogel shell, we create a biocompatible functional nanocarrier that enables sustained release of therapeutics. Here, we report the fabrication of AuNRs-conjugated PSi nanoparticles (AuNRsPSiNPs) through two-step chemical reaction for high-capacity loading of hydrophobic and hydrophilic therapeutics with photothermal property. Furthermore, using water-in-oil microemulsion templates, we encapsulate the AuNRsPSiNPs within a calcium alginate hydrogel nanoshell, creating a versatile biocompatible nanocarrier to codeliver therapeutics for biomedical applications. We find that the functionalized nanohydrogel effectively controls the release rate of the therapeutics while maintaining a high loading efficiency and tunable loading ratios. Notably, combinations of therapeutics coloaded in the functionalized nanohydrogels significantly enhance inhibition of multidrug resistance through synergism and promote faster cancer cell death when combined with photothermal therapy. Moreover, the AuNRs can mediate the conversion of near-infrared laser radiation into heat, increasing the release of therapeutics as well as thermally inducing cell damage to promote faster cancer cell death. Our AuNRsPSiNPs functionalized calcium alginate nanohydrogel holds great promise for photothermal combination therapy and other advanced biomedical applications.

5.
Chemphyschem ; 19(17): 2143-2147, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29779258

RESUMO

Porous silicon nanoparticles have recently garnered attention as potentially-promising biomedical platforms for drug delivery and medical diagnostics. Here, we demonstrate porous silicon nanoparticles as contrast agents for 29 Si magnetic resonance imaging. Size-controlled porous silicon nanoparticles were synthesized by magnesiothermic reduction of silica nanoparticles and were surface activated for further functionalization. Particles were hyperpolarized via dynamic nuclear polarization to enhance their 29 Si MR signals; the particles demonstrated long 29 Si spin-lattice relaxation (T1 ) times (∼25 mins), which suggests potential applicability for medical imaging. Furthermore, 29 Si hyperpolarization levels were sufficient to allow 29 Si MRI in phantoms. These results underscore the potential of porous silicon nanoparticles that, when combined with hyperpolarized magnetic resonance imaging, can be a powerful theragnostic deep tissue imaging platform to interrogate various biomolecular processes in vivo.


Assuntos
Imageamento por Ressonância Magnética , Nanopartículas/química , Silício/química , Meios de Contraste/química , Isótopos/química , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Tamanho da Partícula , Porosidade , Propilaminas/química , Silanos/química
6.
Bioorg Med Chem Lett ; 27(3): 403-405, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28057421

RESUMO

Ethionamide (ETH) is an important second-line antituberculosis drug used for the treatment of patients infected with multidrug-resistant Mycobacterium tuberculosis. Recently, we reported that the loading of ETH into thermally carbonized-porous silicon (TCPSi) nanoparticles enhanced the solubility and permeability of ETH at different pH-values and also increased its metabolization process. Based on these results, we synthesized carboxylic acid functionalized thermally hydrocarbonized porous silicon nanoparticles (UnTHCPSi NPs) conjugated with ETH and its antimicrobial effect was evaluated against Mycobacterium tuberculosis strain H37Rv. The activity of the conjugate was increased when compared to free-ETH, which suggests that the nature of the synergy between the NPs and ETH is likely due to the weakening of the bacterial cell wall that improves conjugate-penetration. These ETH-conjugated NPs have great potential in reducing dosing frequency of ETH in the treatment of multidrug-resistant tuberculosis (MDR-TB).


Assuntos
Antituberculosos/química , Etionamida/química , Nanopartículas/química , Silício/química , Antituberculosos/farmacologia , Antituberculosos/uso terapêutico , Etionamida/farmacologia , Etionamida/uso terapêutico , Humanos , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/efeitos dos fármacos , Tamanho da Partícula , Porosidade , Solubilidade , Tuberculose Resistente a Múltiplos Medicamentos/tratamento farmacológico
7.
Adv Healthc Mater ; : e2400171, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38657207

RESUMO

Strategies incorporating mesenchymal stromal cells (MSC), hydrogels and osteoinductive signals offer promise for bone repair. Osteoinductive signals such as growth factors face challenges in clinical translation due to their high cost, low stability and immunogenicity leading to interest in microRNAs as a simple, inexpensive and powerful alternative. The selection of appropriate miRNA candidates and their efficient delivery must be optimised to make this a reality. This study evaluated pro-osteogenic miRNAs and used porous silicon nanoparticles modified with polyamidoamine dendrimers (PAMAM-pSiNP) to deliver these to MSC encapsulated within gelatin-PEG hydrogels. miR-29b-3p, miR-101-3p and miR-125b-5p are strongly pro-osteogenic and are shown to target FASN and ELOVL4 in the fatty acid biosynthesis pathway to modulate MSC osteogenesis. Hydrogel delivery of miRNA:PAMAM-pSiNP complexes enhanced transfection compared to 2D. The osteogenic potential of hBMSC in hydrogels with miR125b:PAMAM-pSiNP complexes is evaluated. Importantly, a dual-effect on osteogenesis occurred, with miRNAs increasing expression of alkaline phosphatase (ALP) and Runt-related transcription factor 2 (RUNX2) whilst the pSiNPs enhanced mineralisation, likely via degradation into silicic acid. Overall, this work presents insights into the role of miRNAs and fatty acid signalling in osteogenesis, providing future targets to improve bone formation and a promising system to enhance bone tissue engineering.

8.
Int J Biol Macromol ; 259(Pt 2): 129231, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38185310

RESUMO

Bioactive scaffolds fabricated from a combination of organic and inorganic biomaterials are a promising approach for addressing defects in bone tissue engineering. In the present study, a self-crosslinked nanocomposite hydrogel, composed of gelatin/aldehyde-modified xanthan (Gel-AXG) is successfully developed by varying concentrations of porous silicon nanoparticles (PSiNPs). The effect of PSiNPs incorporation on physical, mechanical, and biological performance of the nanocomposite hydrogel is evaluated. Morphological analysis reveals formation of highly porous 3D microstructures with interconnected pores in all nanocomposite hydrogels. Increased content of PSiNPs results in a lower swelling ratio, reduced porosity and pore size, which in turn impeded media penetration and slowed down the degradation process. In addition, remarkable enhancements in dynamic mechanical properties are observed in Gel-AXG-8%Si (compressive strength: 0.6223 MPa at 90 % strain and compressive modulus: 0.054 MPa), along with improved biomineralization ability via hydroxyapatite formation after immersion in simulated body fluid (SBF). This optimized nanocomposite hydrogel provides a sustained release of Si ions at safe dose levels. Furthermore, in-vitro cytocompatibility studies using MG-63 cells exhibited remarkable performance in terms of cell attachment, proliferation, and ALP activity for Gel-AXG-8%Si. These findings suggest that the prepared nanocomposite hydrogel holds promising potential as a scaffold for bone tissue engineering.


Assuntos
Nanopartículas , Polissacarídeos Bacterianos , Engenharia Tecidual , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Hidrogéis/farmacologia , Hidrogéis/química , Gelatina/química , Silício , Nanogéis , Porosidade
9.
ACS Nano ; 17(17): 16412-16431, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37582231

RESUMO

The complexity of CRISPR machinery is a challenge to its application for nonviral in vivo therapeutic gene editing. Here, we demonstrate that proteins, regardless of size or charge, efficiently load into porous silicon nanoparticles (PSiNPs). Optimizing the loading strategy yields formulations that are ultrahigh loading─>40% cargo by volume─and highly active. Further tuning of a polymeric coating on the loaded PSiNPs yields nanocomposites that achieve colloidal stability under cryopreservation, endosome escape, and gene editing efficiencies twice that of the commercial standard Lipofectamine CRISPRMAX. In a mouse model of arthritis, PSiNPs edit cells in both the cartilage and synovium of knee joints, and achieve 60% reduction in expression of the therapeutically relevant MMP13 gene. Administered intramuscularly, they are active over a broad dose range, with the highest tested dose yielding nearly 100% muscle fiber editing at the injection site. The nanocomposite PSiNPs are also amenable to systemic delivery. Administered intravenously in a model that mimics muscular dystrophy, they edit sites of inflamed muscle. Collectively, the results demonstrate that the PSiNP nanocomposites are a versatile system that can achieve high loading of diverse cargoes and can be applied for gene editing in both local and systemic delivery applications.


Assuntos
Sistemas CRISPR-Cas , Nanopartículas , Camundongos , Animais , Sistemas CRISPR-Cas/genética , Silício , Porosidade , Polímeros
10.
Bioact Mater ; 24: 497-506, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36685808

RESUMO

There has been a lot of basic and clinical research on Alzheimer's disease (AD) over the last 100 years, but its mechanisms and treatments have not been fully clarified. Despite some controversies, the amyloid-beta hypothesis is one of the most widely accepted causes of AD. In this study, we disclose a new amyloid-beta plaque disaggregating agent and an AD brain-targeted delivery system using porous silicon nanoparticles (pSiNPs) as a therapeutic nano-platform to overcome AD. We hypothesized that the negatively charged sulfonic acid functional group could disaggregate plaques and construct a chemical library. As a result of the in vitro assay of amyloid plaques and library screening, we confirmed that 6-amino-2-naphthalenesulfonic acid (ANA) showed the highest efficacy for plaque disaggregation as a hit compound. To confirm the targeted delivery of ANA to the AD brain, a nano-platform was created using porous silicon nanoparticles (pSiNPs) with ANA loaded into the pore of pSiNPs and biotin-polyethylene glycol (PEG) surface functionalization. The resulting nano-formulation, named Biotin-CaCl2-ANA-pSiNPs (BCAP), delivered a large amount of ANA to the AD brain and ameliorated memory impairment of the AD mouse model through the disaggregation of amyloid plaques in the brain. This study presents a new bioactive small molecule for amyloid plaque disaggregation and its promising therapeutic nano-platform for AD brain-targeted delivery.

11.
Pharmaceutics ; 15(5)2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-37242631

RESUMO

Despite the clinical benefits that chemotherapeutics has had on the treatment of breast cancer, drug resistance remains one of the main obstacles to curative cancer therapy. Nanomedicines allow therapeutics to be more targeted and effective, resulting in enhanced treatment success, reduced side effects, and the possibility of minimising drug resistance by the co-delivery of therapeutic agents. Porous silicon nanoparticles (pSiNPs) have been established as efficient vectors for drug delivery. Their high surface area makes them an ideal carrier for the administration of multiple therapeutics, providing the means to apply multiple attacks to the tumour. Moreover, immobilising targeting ligands on the pSiNP surface helps direct them selectively to cancer cells, thereby reducing harm to normal tissues. Here, we engineered breast cancer-targeted pSiNPs co-loaded with an anticancer drug and gold nanoclusters (AuNCs). AuNCs have the capacity to induce hyperthermia when exposed to a radiofrequency field. Using monolayer and 3D cell cultures, we demonstrate that the cell-killing efficacy of combined hyperthermia and chemotherapy via targeted pSiNPs is 1.5-fold higher than applying monotherapy and 3.5-fold higher compared to using a nontargeted system with combined therapeutics. The results not only demonstrate targeted pSiNPs as a successful nanocarrier for combination therapy but also confirm it as a versatile platform with the potential to be used for personalised medicine.

12.
Colloids Surf B Biointerfaces ; 225: 113273, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36965332

RESUMO

Porous silicon nanoparticles (pSiNPs) have gained attention from drug delivery systems (DDS) due to their biocompatibility, high drug-loading efficiency, and facile surface modification. To date, many surface chemistries of pSiNPs have been developed to maximize the merits and overcome the drawbacks of pSiNPs. In this work, we newly disclosed a formulation, iron-silicate-coated pSiNPs (Fe-pSiNPs-NCS), using the surface modification method with iron-silicate and 3-isothiocyanatopropyltriethoxysilane (TEPITC). Fe-pSiNPs-NCS demonstrated effective reactive-oxygen species (ROS) self-generation ability via a Fenton-like reaction of iron-silicate and in situ hydrogen peroxide (H2O2) generation of TEPITC on the surface of pSiNPs, resulting in excellent anticancer effect in U87MG cancer cells. Moreover, we confirmed that Fe-pSiNPs-NCS could be used as a drug delivery carrier as it was proven that anticancer drugs (doxorubicin, SN-38) were loaded into Fe-pSiNPs-NCS with high-loading efficiency. These findings could offer efficient strategies for developing nanotherapeutics in biomedical fields.


Assuntos
Nanopartículas , Silício , Silício/farmacologia , Espécies Reativas de Oxigênio , Ferro , Porosidade , Peróxido de Hidrogênio , Silicatos , Portadores de Fármacos , Dióxido de Silício
13.
Heliyon ; 9(12): e23105, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38149182

RESUMO

The present work focuses on the structural, morphological, electrical characteristics, and antibacterial activity of mesoporous silicon (PS) against S. aureus and E. coli. We depict the structural and antimicrobial activity of PS as a result of different etching times (10.0, 20.0, 30.0, 40.0, 50.0, and 60.0 min) with a current density of 100 mA/cm2. The structural and morphological characteristics of synthesized PS have been examined with Fourier-transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), scanning electron microscopy (SEM), transmission electron microscopy (TEM), and atomic force microscopy (AFM). FTIR spectra have been used to confirmed the Si-O, Si-O-Si bond and the adsorption on the surface of PS nanoparticles. The formation of pores on the c-Si wafer results in an analysis of a photoluminescence (PL) band at 712 nm, which changes with etching time in a process similar to current density. The correlation exist among etching times and the ideality factor (η) and barrier height (фb). Gram-positive (S. aureus) and Gram-negative (E. coli) bacteria showed enhanced antimicrobial activity against the PS nanoparticles. The synthesized of PS has been shown with good electrical and antimicrobial activities.

14.
ACS Nano ; 2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36598186

RESUMO

The development of nanosystems with intrinsic immunomodulatory effects on macrophage polarization is important for the macrophage-targeted immunotherapy. Here, mitochondria-targeted bovine serum albumins (BSAs) via the conjugation of fluorescent, lipophilic, and cationic rhodamine 110 molecules can efficiently enhance the gene expression of the proinflammatory phenotype of macrophages and correspondingly inhibit the gene expression of their anti-inflammatory phenotype. On this basis, porous silicon nanocarriers can further boost the immunomodulation of these mitochondria-targeted BSAs in vitro or in vivo, accompanied by the secretion of proinflammatory mediators including tumor necrosis factor α, nitric oxide, and reactive oxygen species (ROS). Meanwhile, BSA coatings can also improve the biocompatibility of porous silicon nanoparticulate cores on macrophages. Finally, the mechanism investigations demonstrate that porous silicon nanocarriers can efficiently deliver mitochondria-targeted BSA into macrophages to generate mitochondrial ROS via the interference with mitochondrial respiratory chains, which can further trigger the downstream signaling transduction pathways for the proinflammatory transition. Considering the good biosafety and versatile loading capability, this developed porous silicon@BSA nanosystem with a strong proinflmmatory regulatory effect has important potential on the combinatorial chemoimmunotherapy against cancer or viral/bacterial-related infectious diseases.

15.
Adv Healthc Mater ; 12(27): e2301052, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37499629

RESUMO

The concept of using two-photon excitation in the NIR for the spatiotemporal control of biological processes holds great promise. However, its use for the delivery of nucleic acids has been very scarcely described and the reported procedures are not optimal as they often involve potentially toxic materials and irradiation conditions. This work prepares a simple system made of biocompatible porous silicon nanoparticles (pSiNP) for the safe siRNA photocontrolled delivery and gene silencing in cells upon two-photon excitation. PSiNP are linked to an azobenzene moiety, which possesses a lysine group (pSiNP@ICPES-azo@Lys) to efficiently complex siRNA. Non-linear excitation of the two-photon absorber system (pSiNP) followed by intermolecular energy transfer (FRET) to trans azobenzene moiety, result in the photoisomerization of the azobenzene from trans to cis and in the destabilization of the azobenzene-siRNA complex, thus inducing the delivery of the cargo siRNA to the cytoplasm of cells. Efficient silencing in MCF-7 expressing stable firefly luciferase with siRNAluc against luciferase is observed. Furthermore, siRNA against inhibitory apoptotic protein (IAP) leads to over 70% of MCF-7 cancer cell death. The developed technique using two-photon light allows a unique high spatiotemporally controlled and safe siRNA delivery in cells in few seconds of irradiation.


Assuntos
Nanopartículas , Neoplasias , Humanos , RNA Interferente Pequeno/genética , Silício , Porosidade , Transfecção , Linhagem Celular Tumoral
16.
Pharmaceutics ; 15(9)2023 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-37765240

RESUMO

Overcoming the blood-brain barrier (BBB) remains a significant challenge with regard to drug delivery to the brain. By incorporating targeting ligands, and by carefully adjusting particle sizes, nanocarriers can be customized to improve drug delivery. Among these targeting ligands, transferrin stands out due to the high expression level of its receptor (i.e., transferrin receptor) on the BBB. Porous silicon nanoparticles (pSiNPs) are a promising drug nanocarrier to the brain due to their biodegradability, biocompatibility, and exceptional drug-loading capacity. However, an in-depth understanding of the optimal nanoparticle size and transferrin surface density, in order to maximize BBB penetration, is still lacking. To address this gap, a diverse library of pSiNPs was synthesized using bifunctional poly(ethylene glycol) linkers with methoxy or/and carboxyl terminal groups. These variations allowed us to explore different transferrin surface densities in addition to particle sizes. The effects of these parameters on the cellular association, uptake, and transcytosis in immortalized human brain microvascular endothelial cells (hCMEC/D3) were investigated using multiple in vitro systems of increasing degrees of complexity. These systems included the following: a 2D cell culture, a static Transwell model, and a dynamic BBB-on-a-chip model. Our results revealed the significant impact of both the ligand surface density and size of pSiNPs on their ability to penetrate the BBB, wherein intermediate-level transferrin densities and smaller pSiNPs exhibited the highest BBB transportation efficiency in vitro. Moreover, notable discrepancies emerged between the tested in vitro assays, further emphasizing the necessity of using more physiologically relevant assays, such as a microfluidic BBB-on-a-chip model, for nanocarrier testing and evaluation.

17.
Bioact Mater ; 7: 39-46, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34179568

RESUMO

New viral infections, due to their rapid spread, lack of effective antiviral drugs and vaccines, kill millions of people every year. The global pandemic SARS-CoV-2 in 2019-2021 has shown that new strains of viruses can widespread very quickly, causing disease and death, with significant socio-economic consequences. Therefore, the search for new methods of combating different pathogenic viruses is an urgent task, and strategies based on nanoparticles are of significant interest. This work demonstrates the antiviral adsorption (virucidal) efficacy of nanoparticles of porous silicon (PSi NPs) against various enveloped and non-enveloped pathogenic human viruses, such as Influenza A virus, Poliovirus, Human immunodeficiency virus, West Nile virus, and Hepatitis virus. PSi NPs sized 60 nm with the average pore diameter of 2 nm and specific surface area of 200 m2/g were obtained by ball-milling of electrochemically-etched microporous silicon films. After interaction with PSi NPs, a strong suppression of the infectious activity of the virus-contaminated fluid was observed, which was manifested in a decrease in the infectious titer of all studied types of viruses by approximately 104 times, and corresponded to an inactivation of 99.99% viruses in vitro. This sorption capacity of PSi NPs is possible due to their microporous structure and huge specific surface area, which ensures efficient capture of virions, as confirmed by ELISA analysis, dynamic light scattering measurements and transmission electron microscopy images. The results obtained indicate the great potential of using PSi NPs as universal viral sorbents and disinfectants for the detection and treatment of viral diseases.

18.
ACS Appl Mater Interfaces ; 14(49): 54539-54549, 2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36469497

RESUMO

An approach to differentially modify the internal surface of porous silicon nanoparticles (pSiNPs) with hydrophobic dodecene and the external surface with antifouling poly-N-(2-hydroxypropyl) acrylamide (polyHPAm) as well as a cell-targeting peptide was developed. Specifically, to generate these core-shell pSiNPs, the interior surface of a porous silicon (pSi) film was hydrosilylated with 1-dodecene, followed by ultrasonication to create pSiNPs. The new external surfaces were modified by silanization with a polymerization initiator, and surface-initiated atom transfer radical polymerization was performed to introduce polyHPAm brushes. Afterward, a fraction of the polymer side chain hydroxyl groups was activated to conjugate cRGDfK─a peptide with a high affinity and selectivity for the ανß3 integrin receptor that is overexpressed in prostate and melanoma cancers. Finally, camptothecin, a hydrophobic anti-cancer drug, was successfully loaded into the pores. This drug delivery system showed excellent colloidal stability in a cell culture medium, and the in vitro drug release kinetics could be fine-tuned by the combination of internal and external surface modifications. In vitro studies by confocal microscopy and flow cytometry revealed improved cellular association attributed to cRGDfK. Furthermore, the cell viability results showed that the drug-loaded and peptide-functionalized nanoparticles had enhanced cytotoxicity toward a C4-2B prostate carcinoma cell line in both 2D cell culture and a 3D spheroid model.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Silício/química , Porosidade , Nanopartículas/química , Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Linhagem Celular Tumoral
19.
Adv Healthc Mater ; 11(12): e2200076, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35306736

RESUMO

Porous silicon nanoparticles (pSiNPs) are widely utilized as drug carriers due to their excellent biocompatibility, large surface area, and versatile surface chemistry. However, the dispersion in pore size and biodegradability of pSiNPs arguably have hindered the application of pSiNPs for controlled drug release. Here, a step-changing solution to this problem is described involving the design, synthesis, and application of three different linker-drug conjugates comprising anticancer drug doxorubicin (DOX) and different stimulus-cleavable linkers (SCLs) including the photocleavable linker (ortho-nitrobenzyl), pH-cleavable linker (hydrazone), and enzyme-cleavable linker (ß-glucuronide). These SCL-DOX conjugates are covalently attached to the surface of pSiNP via copper (I)-catalyzed alkyne-azide cycloaddition (CuAAC, i.e., click reaction) to afford pSiNP-SCL-DOXs. The mass loading of the covalent conjugation approach for pSiNP-SCL-DOX reaches over 250 µg of DOX per mg of pSiNPs, which is notably twice the mass loading achieved by noncovalent loading. Moreover, the covalent conjugation between SCL-DOX and pSiNPs endows the pSiNPs with excellent stability and highly controlled release behavior. When tested in both in vitro and in vivo tumor models, the pSiNP-SCL-DOXs induces excellent tumor growth inhibition.


Assuntos
Nanopartículas , Neoplasias , Doxorrubicina/farmacologia , Portadores de Fármacos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Porosidade , Silício
20.
Front Pharmacol ; 13: 962763, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36016563

RESUMO

Nanomaterials are a central pillar in modern medicine. They are thought to optimize drug delivery, enhance therapeutic efficacy, and reduce side-effects. To foster this technology, analytical methods are needed to validate not only the localization and distribution of these nanomaterials, but also their compatibility with cells, drugs, and drug release. In the present work, we assessed nanoparticles based on porous silicon (pSiNPs) loaded with the clinically used tyrosine kinase inhibitor sunitinib for their effectiveness of drug delivery, release, and toxicity in colon cancer cells (HCT 116 cells) and cardiac myoblast cells (H9c2) using Raman micro-spectroscopy, high-resolution fluorescence microscopy, along with biological methods for toxicological effects. We produced pSiNPs with a size of about 100 nm by grinding mesoporous silicon layers. pSiNPs allowed an effective loading of sunitinib due to their high porosity. Photoluminescence properties of the nanoparticles within the visible spectrum allowed the visualization of their uptake in cardiac cells. Raman micro-spectroscopy allowed not only the detection of the uptake and distribution of pSiNPs within the cells via a characteristic silicon Raman band at about 518-520 cm-1, but also the localization of the drug based on its characteristic molecular fingerprints. Cytotoxicity studies by Western blot analyses of apoptotic marker proteins such as caspase-3, and the detection of apoptosis by subG1-positive cell fractions in HCT 116 and MTT analyses in H9c2 cells, suggest a sustained release of sunitinib from pSiNPs and delayed cytotoxicity of sunitinib in HCT 116 cells. The analyses in cardiac cells revealed that pSiNPs are well tolerated and that they may even protect from toxic effects in these cells to some extent. Analyses of the integrity of mitochondrial networks as an early indicator for apoptotic cellular effects seem to validate these observations. Our study suggests pSiNPs-based nanocontainers for efficient and safe drug delivery and Raman micro-spectroscopy as a reliable method for their detection and monitoring. Thus, the herein presented nanocontainers and analytical methods have the potential to allow an efficient advancement of nanoparticles for targeted and sustained intracellular drug release that is of need, e.g., in chronic diseases and for the prevention of cardiac toxicity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA