Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Nanotechnol ; 19(4): 554-564, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38225356

RESUMO

Bladder cancer treatment via intravesical drug administration achieves reasonable survival rates but suffers from low therapeutic efficacy. To address the latter, self-propelled nanoparticles or nanobots have been proposed, taking advantage of their enhanced diffusion and mixing capabilities in urine when compared with conventional drugs or passive nanoparticles. However, the translational capabilities of nanobots in treating bladder cancer are underexplored. Here, we tested radiolabelled mesoporous silica-based urease-powered nanobots in an orthotopic mouse model of bladder cancer. In vivo and ex vivo results demonstrated enhanced nanobot accumulation at the tumour site, with an eightfold increase revealed by positron emission tomography in vivo. Label-free optical contrast based on polarization-dependent scattered light-sheet microscopy of cleared bladders confirmed tumour penetration by nanobots ex vivo. Treating tumour-bearing mice with intravesically administered radio-iodinated nanobots for radionuclide therapy resulted in a tumour size reduction of about 90%, positioning nanobots as efficient delivery nanosystems for bladder cancer therapy.


Assuntos
Urease , Neoplasias da Bexiga Urinária , Camundongos , Animais , Neoplasias da Bexiga Urinária/diagnóstico por imagem , Neoplasias da Bexiga Urinária/tratamento farmacológico , Administração Intravesical , Radioisótopos/uso terapêutico
2.
Small ; 20(11): e2309387, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38200672

RESUMO

Over the past decades, the development of nanoparticles (NPs) to increase the efficiency of clinical treatments has been subject of intense research. Yet, most NPs have been reported to possess low efficacy as their actuation is hindered by biological barriers. For instance, synovial fluid (SF) present in the joints is mainly composed of hyaluronic acid (HA). These viscous media pose a challenge for many applications in nanomedicine, as passive NPs tend to become trapped in complex networks, which reduces their ability to reach the target location. This problem can be addressed by using active NPs (nanomotors, NMs) that are self-propelled by enzymatic reactions, although the development of enzyme-powered NMs, capable of navigating these viscous environments, remains a considerable challenge. Here, the synergistic effects of two NMs troops, namely hyaluronidase NMs (HyaNMs, Troop 1) and urease NMs (UrNMs, Troop 2) are demonstrated. Troop 1 interacts with the SF by reducing its viscosity, thus allowing Troop 2 to swim more easily through the SF. Through their collective motion, Troop 2 increases the diffusion of macromolecules. These results pave the way for more widespread use of enzyme-powered NMs, e.g., for treating joint injuries and improving therapeutic effectiveness compared with traditional methods.


Assuntos
Nanopartículas , Viscosidade , Substâncias Macromoleculares
3.
Nanoscale Adv ; 6(1): 126-135, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38125604

RESUMO

Magnetic nanoparticles have been extensively explored as theranostic agents both in academic and clinical settings. Their self-assembly into nanohybrids using block copolymers can lead to new nanostructures with high functionalities and performances. Herein, we demonstrate a high-throughput and scalable method to elaborate magnetic micelles by the assembly of iron oxide magnetite nanoflowers, an efficient nanoheater, and the block copolymer Poly(styrene)-block-poly(acrylic acid) via a microfluidic-assisted nanoprecipitation method. We show that the size and shape of the magnetomicelles can be easily tuned by modulating the residence time in the microfluidic channel. In addition to their biocompatibility, we demonstrate the potential of these magnetic nanohybrids as multimodal theranostic platforms capable of generating heat by photothermia and functioning as negative contrast agents in magnetic resonance imaging and as imaging tracers in magnetic particle imaging. Notably, they outperform currently commercially available particles in terms of imaging functionalities.

4.
ACS Nano ; 17(8): 7180-7193, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37058432

RESUMO

Targeted drug delivery depends on the ability of nanocarriers to reach the target site, which requires the penetration of different biological barriers. Penetration is usually low and slow because of passive diffusion and steric hindrance. Nanomotors (NMs) have been suggested as the next generation of nanocarriers in drug delivery due to their autonomous motion and associated mixing hydrodynamics, especially when acting collectively as a swarm. Here, we explore the concept of enzyme-powered NMs designed as such that they can exert disruptive mechanical forces upon laser irradiation. The urease-powered motion and swarm behavior improve translational movement compared to passive diffusion of state-of-the-art nanocarriers, while optically triggered vapor nanobubbles can destroy biological barriers and reduce steric hindrance. We show that these motors, named Swarm 1, collectively displace through a microchannel blocked with type 1 collagen protein fibers (barrier model), accumulate onto the fibers, and disrupt them completely upon laser irradiation. We evaluate the disruption of the microenvironment induced by these NMs (Swarm 1) by quantifying the efficiency by which a second type of fluorescent NMs (Swarm 2) can move through the cleared microchannel and be taken up by HeLa cells at the other side of the channel. Experiments showed that the delivery efficiency of Swarm 2 NMs in a clean path was increased 12-fold in the presence of urea as fuel compared to when no fuel was added. When the path was blocked with the collagen fibers, delivery efficiency dropped considerably and only depicted a 10-fold enhancement after pretreatment of the collagen-filled channel with Swarm 1 NMs and laser irradiation. The synergistic effect of active motion (chemically propelled) and mechanical disruption (light-triggered nanobubbles) of a biological barrier represents a clear advantage for the improvement of therapies which currently fail due to inadequate passage of drug delivery carriers through biological barriers.


Assuntos
Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Humanos , Células HeLa
5.
Sci Robot ; 6(52)2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-34043548

RESUMO

Enzyme-powered nanomotors are an exciting technology for biomedical applications due to their ability to navigate within biological environments using endogenous fuels. However, limited studies into their collective behavior and demonstrations of tracking enzyme nanomotors in vivo have hindered progress toward their clinical translation. Here, we report the swarming behavior of urease-powered nanomotors and its tracking using positron emission tomography (PET), both in vitro and in vivo. For that, mesoporous silica nanoparticles containing urease enzymes and gold nanoparticles were used as nanomotors. To image them, nanomotors were radiolabeled with either 124I on gold nanoparticles or 18F-labeled prosthetic group to urease. In vitro experiments showed enhanced fluid mixing and collective migration of nanomotors, demonstrating higher capability to swim across complex paths inside microfabricated phantoms, compared with inactive nanomotors. In vivo intravenous administration in mice confirmed their biocompatibility at the administered dose and the suitability of PET to quantitatively track nanomotors in vivo. Furthermore, nanomotors were administered directly into the bladder of mice by intravesical injection. When injected with the fuel, urea, a homogeneous distribution was observed even after the entrance of fresh urine. By contrast, control experiments using nonmotile nanomotors (i.e., without fuel or without urease) resulted in sustained phase separation, indicating that the nanomotors' self-propulsion promotes convection and mixing in living reservoirs. Active collective dynamics, together with the medical imaging tracking, constitute a key milestone and a step forward in the field of biomedical nanorobotics, paving the way toward their use in theranostic applications.


Assuntos
Nanopartículas Metálicas , Robótica/instrumentação , Bexiga Urinária/diagnóstico por imagem , Administração Intravesical , Animais , Desenho de Equipamento , Feminino , Ouro , Camundongos , Camundongos Endogâmicos C57BL , Movimento (Física) , Imagens de Fantasmas , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Medicina de Precisão , Pesquisa Translacional Biomédica , Urease
6.
ACS Appl Mater Interfaces ; 13(13): 14964-14973, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33769023

RESUMO

The low efficacy of current conventional treatments for bacterial infections increases mortality rates worldwide. To alleviate this global health problem, we propose drug-free enzyme-based nanomotors for the treatment of bacterial urinary-tract infections. We develop nanomotors consisting of mesoporous silica nanoparticles (MSNPs) that were functionalized with either urease (U-MSNPs), lysozyme (L-MSNPs), or urease and lysozyme (M-MSNPs), and use them against nonpathogenic planktonic Escherichia coli. U-MSNPs exhibited the highest bactericidal activity due to biocatalysis of urea into NaHCO3 and NH3, which also propels U-MSNPs. In addition, U-MSNPs in concentrations above 200 µg/mL were capable of successfully reducing 60% of the biofilm biomass of a uropathogenic E. coli strain. This study thus provides a proof-of-concept, demonstrating that enzyme-based nanomotors are capable of fighting infectious diseases. This approach could potentially be extended to other kinds of diseases by selecting appropriate biomolecules.


Assuntos
Antibacterianos/farmacologia , Escherichia coli/efeitos dos fármacos , Muramidase/farmacologia , Nanopartículas/química , Dióxido de Silício/química , Urease/farmacologia , Antibacterianos/administração & dosagem , Biocatálise , Biofilmes/efeitos dos fármacos , Canavalia/enzimologia , Portadores de Fármacos/química , Escherichia coli/fisiologia , Infecções por Escherichia coli/tratamento farmacológico , Humanos , Muramidase/administração & dosagem , Urease/administração & dosagem , Infecções Urinárias/tratamento farmacológico
7.
ACS Nano ; 13(10): 12171-12183, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31580642

RESUMO

The introduction of stimuli-responsive cargo release capabilities on self-propelled micro- and nanomotors holds enormous potential in a number of applications in the biomedical field. Herein, we report the preparation of mesoporous silica nanoparticles gated with pH-responsive supramolecular nanovalves and equipped with urease enzymes which act as chemical engines to power the nanomotors. The nanoparticles are loaded with different cargo molecules ([Ru(bpy)3]Cl2 (bpy = 2,2'-bipyridine) or doxorubicin), grafted with benzimidazole groups on the outer surface, and capped by the formation of inclusion complexes between benzimidazole and cyclodextrin-modified urease. The nanomotor exhibits enhanced Brownian motion in the presence of urea. Moreover, no cargo is released at neutral pH, even in the presence of the biofuel urea, due to the blockage of the pores by the bulky benzimidazole:cyclodextrin-urease caps. Cargo delivery is only triggered on-command at acidic pH due to the protonation of benzimidazole groups, the dethreading of the supramolecular nanovalves, and the subsequent uncapping of the nanoparticles. Studies with HeLa cells indicate that the presence of biofuel urea enhances nanoparticle internalization and both [Ru(bpy)3]Cl2 or doxorubicin intracellular release due to the acidity of lysosomal compartments. Gated enzyme-powered nanomotors shown here display some of the requirements for ideal drug delivery carriers such as the capacity to self-propel and the ability to "sense" the environment and deliver the payload on demand in response to predefined stimuli.


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , Dióxido de Silício/química , Benzimidazóis/química , Ciclodextrinas/química , Sistemas de Liberação de Medicamentos/métodos , Células HeLa , Humanos , Lisossomos/metabolismo , Nanofios/química
8.
Angew Chem Int Ed Engl ; 58(24): 7992-7996, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-30990243

RESUMO

We report lipase-based nanomotors that are capable of enhanced Brownian motion over long periods of time in triglyceride solution and of degrading triglyceride droplets that mimic "blood lipids". We achieved about 40 min of enhanced diffusion of lipase-modified mesoporous silica nanoparticles (MSNPs) through a biocatalytic reaction between lipase and its corresponding water-soluble oil substrate (triacetin) as fuel, which resulted in an enhanced diffusion coefficient (ca. 50 % increase) at low triacetin concentration (<10 mm). Lipase not only serves as the power engine but also as a highly efficient cleaner for the triglyceride droplets (e.g., tributyrin) in PBS solution, which could yield potential biomedical applications, for example, for dealing with diseases related to the accumulation of triglycerides, or for environmental remediation, for example, for the degradation of oil spills.


Assuntos
Enzimas Imobilizadas/química , Lipase/química , Nanopartículas/química , Dióxido de Silício/química , Triglicerídeos/química , Candida/enzimologia , Enzimas Imobilizadas/metabolismo , Lipase/metabolismo , Nanotecnologia/métodos , Triglicerídeos/metabolismo
9.
ACS Nano ; 13(1): 429-439, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30588798

RESUMO

Cancer is one of the main causes of death around the world, lacking efficient clinical treatments that generally present severe side effects. In recent years, various nanosystems have been explored to specifically target tumor tissues, enhancing the efficacy of cancer treatment and minimizing the side effects. In particular, bladder cancer is the ninth most common cancer worldwide and presents a high survival rate but serious recurrence levels, demanding an improvement in the existent therapies. Here, we present urease-powered nanomotors based on mesoporous silica nanoparticles that contain both polyethylene glycol and anti-FGFR3 antibody on their outer surface to target bladder cancer cells in the form of 3D spheroids. The autonomous motion is promoted by urea, which acts as fuel and is inherently present at high concentrations in the bladder. Antibody-modified nanomotors were able to swim in both simulated and real urine, showing a substrate-dependent enhanced diffusion. The internalization efficiency of the antibody-modified nanomotors into the spheroids in the presence of urea was significantly higher compared with antibody-modified passive particles or bare nanomotors. Furthermore, targeted nanomotors resulted in a higher suppression of spheroid proliferation compared with bare nanomotors, which could arise from the local ammonia production and the therapeutic effect of anti-FGFR3. These results hold significant potential for the development of improved targeted cancer therapy and diagnostics using biocompatible nanomotors.


Assuntos
Nanopartículas/metabolismo , Esferoides Celulares/metabolismo , Urease/metabolismo , Neoplasias da Bexiga Urinária/patologia , Anticorpos/imunologia , Células Cultivadas , Humanos , Movimento (Física) , Nanopartículas/química , Polietilenoglicóis/química , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/imunologia , Dióxido de Silício/química , Células Tumorais Cultivadas , Ureia/metabolismo , Urease/química
10.
J Am Chem Soc ; 138(42): 13782-13785, 2016 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-27718566

RESUMO

The motion of self-propelled tubular micro- and nanojets has so far been achieved by bubble propulsion, e.g., O2 bubbles formed by catalytic decomposition of H2O2, which renders future biomedical applications inviable. An alternative self-propulsion mechanism for tubular engines on the nanometer scale is still missing. Here, we report the fabrication and characterization of bubble-free propelled tubular nanojets (as small as 220 nm diameter), powered by an enzyme-triggered biocatalytic reaction using urea as fuel. We studied the translational and rotational dynamics of the nanojets as functions of the length and location of the enzymes. Introducing tracer nanoparticles into the system, we demonstrated the presence of an internal flow that extends into the external fluid via the cavity opening, leading to the self-propulsion. One-dimensional nanosize, longitudinal self-propulsion, and biocompatibility make the tubular nanojets promising for future biomedical applications.

11.
ACS Nano ; 10(10): 9111-9122, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27666121

RESUMO

Enzymes play a crucial role in many biological processes which require harnessing and converting free chemical energy into kinetic forces in order to accomplish tasks. Enzymes are considered to be molecular machines, not only because of their capability of energy conversion in biological systems but also because enzymatic catalysis can result in enhanced diffusion of enzymes at a molecular level. Enlightened by nature's design of biological machinery, researchers have investigated various types of synthetic micro/nanomachines by using enzymatic reactions to achieve self-propulsion of micro/nanoarchitectures. Yet, the mechanism of motion is still under debate in current literature. Versatile proof-of-concept applications of these enzyme-powered micro/nanodevices have been recently demonstrated. In this review, we focus on discussing enzymes not only as stochastic swimmers but also as nanoengines to power self-propelled synthetic motors. We present an overview on different enzyme-powered micro/nanomachines, the current debate on their motion mechanism, methods to provide motion and speed control, and an outlook of the future potentials of this multidisciplinary field.


Assuntos
Catálise , Nanoestruturas , Difusão , Cinética , Movimento (Física)
12.
Biomacromolecules ; 16(11): 3562-73, 2015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26443892

RESUMO

In this work, we introduce dipeptides containing tryptophan N-capped with the nonsteroidal anti-inflammatory drug naproxen and C-terminal dehydroamino acids, dehydrophenylalanine (ΔPhe), dehydroaminobutyric acid (ΔAbu), and dehydroalanine (ΔAla) as efficacious protease resistant hydrogelators. Optimized conditions for gel formation are reported. Transmission electron microscopy experiments revealed that the hydrogels consist of networks of micro/nanosized fibers formed by peptide self-assembly. Fluorescence and circular dichroism spectroscopy indicate that the self-assembly process is driven by stacking interactions of the aromatic groups. The naphthalene groups of the naproxen moieties are highly organized in the fibers through chiral stacking. Rheological experiments demonstrated that the most hydrophobic peptide (containing C-terminal ΔPhe) formed more elastic gels at lower critical gelation concentrations. This gel revealed irreversible breakup, while the C-terminal ΔAbu and ΔAla gels, although less elastic, exhibited structural recovery and partial healing of the elastic properties. A potential antitumor thieno[3,2-b]pyridine derivative was incorporated (noncovalently) into the gel formed by the hydrogelator containing C-terminal ΔPhe residue. Fluorescence and Förster resonance energy transfer measurements indicate that the drug is located in a hydrophobic environment, near/associated with the peptide fibers, establishing this type of hydrogel as a good drug-nanocarrier candidate.


Assuntos
Portadores de Fármacos/química , Hidrogéis/química , Naproxeno/química , Triptofano/química , Alanina/análogos & derivados , Alanina/química , Linhagem Celular Tumoral , Dicroísmo Circular , Humanos , Interações Hidrofóbicas e Hidrofílicas , Células MCF-7 , Microscopia Eletrônica de Transmissão , Modelos Teóricos , Naftalenos/química , Fenilalanina/análogos & derivados , Fenilalanina/química , Reologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...