Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 27(19)2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36235009

RESUMEN

The development of biomimetic drug delivery systems for biomedical applications has attracted significant research attention. As the use of cell membrane as a surface coating has shown to be a promising platform for several disease treatments. Cell-membrane-coated nanoparticles exhibit enhanced immunocompatibility and prolonged circulation time. Herein, human red blood cell (RBC) membrane-cloaked nanoparticles with enhanced targeting functionality were designed as a targeted nanotheranostic against cancer. Naturally, derived human RBC membrane modified with targeting ligands coated onto polymeric nanoparticle cores containing both chemotherapy and imaging agent. Using epithelial cell adhesion molecule (EpCAM)-positive MCF-7 breast cancer cells as a disease model, the nature-inspired targeted theranostic human red blood cell membrane-coated polymeric nanoparticles (TT-RBC-NPs) platform was capable of not only specifically binding to targeted cancer cells, effectively delivering doxorubicin (DOX), but also visualizing the targeted cancer cells. The TT-RBC-NPs achieved an extended-release profile, with the majority of the drug release occurring within 5 days. The TT-RBC-NPs enabled enhanced cytotoxic efficacy against EpCAM positive MCF-7 breast cancer over the non-targeted NPs. Additionally, fluorescence images of the targeted cancer cells incubated with the TT-RBC-NPs visually indicated the increased cellular uptake of TT-RBC-NPs inside the breast cancer cells. Taken together, this TT-RBC-NP platform sets the foundation for the next-generation stealth theranostic platforms for systemic cargo delivery for treatment and diagnostic of cancer.


Asunto(s)
Neoplasias de la Mama , Nanopartículas , Biomimética , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina , Sistemas de Liberación de Medicamentos/métodos , Molécula de Adhesión Celular Epitelial/análisis , Membrana Eritrocítica , Femenino , Humanos , Nanopartículas/química , Medicina de Precisión , Nanomedicina Teranóstica/métodos
2.
Nature ; 526(7571): 118-21, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26374997

RESUMEN

Development of functional nanoparticles can be encumbered by unanticipated material properties and biological events, which can affect nanoparticle effectiveness in complex, physiologically relevant systems. Despite the advances in bottom-up nanoengineering and surface chemistry, reductionist functionalization approaches remain inadequate in replicating the complex interfaces present in nature and cannot avoid exposure of foreign materials. Here we report on the preparation of polymeric nanoparticles enclosed in the plasma membrane of human platelets, which are a unique population of cellular fragments that adhere to a variety of disease-relevant substrates. The resulting nanoparticles possess a right-side-out unilamellar membrane coating functionalized with immunomodulatory and adhesion antigens associated with platelets. Compared to uncoated particles, the platelet membrane-cloaked nanoparticles have reduced cellular uptake by macrophage-like cells and lack particle-induced complement activation in autologous human plasma. The cloaked nanoparticles also display platelet-mimicking properties such as selective adhesion to damaged human and rodent vasculatures as well as enhanced binding to platelet-adhering pathogens. In an experimental rat model of coronary restenosis and a mouse model of systemic bacterial infection, docetaxel and vancomycin, respectively, show enhanced therapeutic efficacy when delivered by the platelet-mimetic nanoparticles. The multifaceted biointerfacing enabled by the platelet membrane cloaking method provides a new approach in developing functional nanoparticles for disease-targeted delivery.


Asunto(s)
Antibacterianos/administración & dosificación , Plaquetas/citología , Membrana Celular/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/administración & dosificación , Nanopartículas/química , Adhesividad Plaquetaria , Animales , Antibacterianos/farmacocinética , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Colágeno/química , Colágeno/inmunología , Activación de Complemento/inmunología , Reestenosis Coronaria/sangre , Reestenosis Coronaria/tratamiento farmacológico , Reestenosis Coronaria/metabolismo , Modelos Animales de Enfermedad , Docetaxel , Humanos , Macrófagos/inmunología , Masculino , Ratones , Polímeros/química , Ratas , Ratas Sprague-Dawley , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/citología , Staphylococcus aureus/metabolismo , Taxoides/administración & dosificación , Taxoides/farmacocinética , Liposomas Unilamelares/química , Vancomicina/administración & dosificación , Vancomicina/farmacocinética
3.
Proc Natl Acad Sci U S A ; 114(43): 11488-11493, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073076

RESUMEN

Sepsis, resulting from uncontrolled inflammatory responses to bacterial infections, continues to cause high morbidity and mortality worldwide. Currently, effective sepsis treatments are lacking in the clinic, and care remains primarily supportive. Here we report the development of macrophage biomimetic nanoparticles for the management of sepsis. The nanoparticles, made by wrapping polymeric cores with cell membrane derived from macrophages, possess an antigenic exterior the same as the source cells. By acting as macrophage decoys, these nanoparticles bind and neutralize endotoxins that would otherwise trigger immune activation. In addition, these macrophage-like nanoparticles sequester proinflammatory cytokines and inhibit their ability to potentiate the sepsis cascade. In a mouse Escherichia coli bacteremia model, treatment with macrophage mimicking nanoparticles, termed MΦ-NPs, reduced proinflammatory cytokine levels, inhibited bacterial dissemination, and ultimately conferred a significant survival advantage to infected mice. Employing MΦ-NPs as a biomimetic detoxification strategy shows promise for improving patient outcomes, potentially shifting the current paradigm of sepsis management.


Asunto(s)
Membrana Celular/química , Citocinas/química , Endotoxinas/química , Infecciones por Escherichia coli/terapia , Nanopartículas/química , Sepsis/terapia , Animales , Bacteriemia/terapia , Línea Celular , Lipopolisacáridos/farmacología , Lipoproteínas/química , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Receptor Toll-Like 4
4.
Proc Natl Acad Sci U S A ; 111(49): 17600-5, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25422427

RESUMEN

Helicobacter pylori infection is marked by a vast prevalence and strong association with various gastric diseases, including gastritis, peptic ulcers, and gastric cancer. Because of the rapid emergence of H. pylori strains resistant to existing antibiotics, current treatment regimens show a rapid decline of their eradication rates. Clearly, novel antibacterial strategies against H. pylori are urgently needed. Here, we investigated the in vivo therapeutic potential of liposomal linolenic acid (LipoLLA) for the treatment of H. pylori infection. The LipoLLA formulation with a size of ∼ 100 nm was prone to fusion with bacterial membrane, thereby directly releasing a high dose of linolenic acids into the bacterial membrane. LipoLLA penetrated the mucus layer of mouse stomach, and a significant portion of the administered LipoLLA was retained in the stomach lining up to 24 h after the oral administration. In vivo tests further confirmed that LipoLLA was able to kill H. pylori and reduce bacterial load in the mouse stomach. LipoLLA treatment was also shown to reduce the levels of proinflammatory cytokines including interleukin 1ß, interleukin 6, and tumor necrosis factor alpha, which were otherwise elevated because of the H. pylori infection. Finally, a toxicity test demonstrated excellent biocompatibility of LipoLLA to normal mouse stomach. Collectively, results from this study indicate that LipoLLA is a promising, effective, and safe therapeutic agent for the treatment of H. pylori infection.


Asunto(s)
Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/efectos de los fármacos , Inflamación/tratamiento farmacológico , Liposomas/química , Ácido alfa-Linolénico/administración & dosificación , Animales , Antibacterianos , Carcinoma/tratamiento farmacológico , Carcinoma/microbiología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Análisis Costo-Beneficio , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Farmacorresistencia Bacteriana , Ácidos Grasos no Esterificados/química , Mucosa Gástrica/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Nanomedicina , Células Madre , Estómago/microbiología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/microbiología
5.
Angew Chem Int Ed Engl ; 56(8): 2156-2161, 2017 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-28105785

RESUMEN

The highly acidic gastric environment creates a physiological barrier for using therapeutic drugs in the stomach. While proton pump inhibitors have been widely used for blocking acid-producing enzymes, this approach can cause various adverse effects. Reported herein is a new microdevice, consisting of magnesium-based micromotors which can autonomously and temporally neutralize gastric acid through efficient chemical propulsion in the gastric fluid by rapidly depleting the localized protons. Coating these micromotors with a cargo-containing pH-responsive polymer layer leads to autonomous release of the encapsulated payload upon gastric-acid neutralization by the motors. Testing in a mouse model demonstrate that these motors can safely and rapidly neutralize gastric acid and simultaneously release payload without causing noticeable acute toxicity or affecting the stomach function, and the normal stomach pH is restored within 24 h post motor administration.


Asunto(s)
Preparaciones de Acción Retardada/química , Ácido Gástrico/química , Magnesio/química , Polímeros/química , Animales , Liberación de Fármacos , Colorantes Fluorescentes/administración & dosificación , Oro/química , Concentración de Iones de Hidrógeno , Ratones , Ácidos Polimetacrílicos/química , Rodaminas/administración & dosificación
6.
Adv Funct Mater ; 26(10): 1628-1635, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-27325913

RESUMEN

With the rising threat of antibiotic-resistant bacteria, vaccination is becoming an increasingly important strategy to prevent and manage bacterial infections. Made from deactivated bacterial toxins, toxoid vaccines are widely used in the clinic as they help to combat the virulence mechanisms employed by different pathogens. Herein, the efficacy of a biomimetic nanoparticle-based anti-virulence vaccine is examined in a mouse model of methicillin-resistant Staphylococcus aureus (MRSA) skin infection. Vaccination with nanoparticle-detained staphylococcal α-hemolysin (Hla) effectively triggers the formation of germinal centers and induces high anti-Hla titers. Compared to mice vaccinated with control samples, those vaccinated with the nanoparticle toxoid show superior protective immunity against MRSA skin infection. The vaccination not only inhibits lesion formation at the site of bacterial challenge, but also reduces the invasiveness of MRSA, preventing dissemination into other organs. Overall, this biomimetic nanoparticle-based toxin detainment strategy is a promising method for the design of potent anti-virulence vaccines for managing bacterial infections.

7.
Nano Lett ; 15(2): 1403-9, 2015 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-25615236

RESUMEN

Synthetic nanoparticles coated with cellular membranes have been increasingly explored to harness natural cell functions toward the development of novel therapeutic strategies. Herein, we report on a unique bacterial membrane-coated nanoparticle system as a new and exciting antibacterial vaccine. Using Escherichia coli as a model pathogen, we collect bacterial outer membrane vesicles (OMVs) and successfully coat them onto small gold nanoparticles (AuNPs) with a diameter of 30 nm. The resulting bacterial membrane-coated AuNPs (BM-AuNPs) show markedly enhanced stability in biological buffer solutions. When injected subcutaneously, the BM-AuNPs induce rapid activation and maturation of dendritic cells in the lymph nodes of the vaccinated mice. In addition, vaccination with BM-AuNPs generates antibody responses that are durable and of higher avidity than those elicited by OMVs only. The BM-AuNPs also induce an elevated production of interferon gamma (INFγ) and interleukin-17 (IL-17), but not interleukin-4 (IL-4), indicating its capability of generating strong Th1 and Th17 biased cell responses against the source bacteria. These observed results demonstrate that using natural bacterial membranes to coat synthetic nanoparticles holds great promise for designing effective antibacterial vaccines.


Asunto(s)
Membrana Celular , Células Dendríticas/inmunología , Escherichia coli/patogenicidad , Nanopartículas , Animales , Citometría de Flujo , Ratones , Microscopía Electrónica de Rastreo
8.
Infect Immun ; 82(1): 356-63, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24166959

RESUMEN

Approximately 50% of the world's population is infected with Helicobacter pylori, leading to chronic inflammation, which increases the risk for gastric adenocarcinoma. MyD88 is a key adaptor molecule in inflammatory pathways involved in interleukin 1 (IL-1)/IL-18/Toll-like receptor signaling and has been shown to have divergent effects in carcinogenesis. The role of MyD88 in Helicobacter-induced gastric malignancy is unknown. Using a mouse model of Helicobacter-induced gastric cancer, we assessed the role of MyD88 in cancer development by evaluating gastric histopathology, apoptosis, proliferation, and cytokine expression. Infection of MyD88-deficient (Myd88(-/-)) mice with Helicobacter resulted in early and rapid advancement to gastric dysplasia as early as 25 weeks postinfection. The progression of Helicobacter-induced disease to precancerous and cancerous lesions in the absence of MyD88 signaling was accompanied by increased gastric epithelial apoptosis and proliferation. In addition, inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), gamma interferon (IFN-γ), IL-6, and IL-1ß were highly expressed in association with the development of gastric dysplasia. These data suggest that MyD88 signaling retards development and progression to cancer during Helicobacter infection. This is the first study to show evidence of MyD88 protection in an infection-driven inflammation-associated cancer model.


Asunto(s)
Infecciones por Helicobacter/complicaciones , Helicobacter pylori , Síndromes de Inmunodeficiencia/metabolismo , Factor 88 de Diferenciación Mieloide/fisiología , Neoplasias Gástricas/microbiología , Animales , Apoptosis/fisiología , Proliferación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales/citología , Mucosa Gástrica/citología , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/genética , Ratones , Ratones Endogámicos C57BL , Lesiones Precancerosas/microbiología , Lesiones Precancerosas/patología , Enfermedades de Inmunodeficiencia Primaria , Neoplasias Gástricas/patología , Neoplasias Gástricas/fisiopatología
9.
Langmuir ; 29(39): 12228-33, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23987129

RESUMEN

We report a novel pH-responsive gold nanoparticle-stabilized liposome system for gastric antimicrobial delivery. By adsorbing small chitosan-modified gold nanoparticles (diameter ~10 nm) onto the outer surface of negatively charged phospholipid liposomes (diameter ~75 nm), we show that at gastric pH the liposomes have excellent stability with limited fusion ability and negligible cargo releases. However, when the stabilized liposomes are present in an environment with neutral pH, the gold stabilizers detach from the liposomes, resulting in free liposomes that can actively fuse with bacterial membranes. Using Helicobacter pylori as a model bacterium and doxycycline as a model antibiotic, we demonstrate such pH-responsive fusion activity and drug release profile of the nanoparticle-stabilized liposomes. Particularly, at neutral pH the gold nanoparticles detach, and thus the doxycycline-loaded liposomes rapidly fuse with bacteria and cause superior bactericidal efficacy as compared to the free doxycycline counterpart. Our results suggest that the reported liposome system holds a substantial potential for gastric drug delivery; it remains inactive (stable) in the stomach lumen but actively interacts with bacteria once it reaches the mucus layer of the stomach where the bacteria may reside.


Asunto(s)
Antibacterianos/farmacología , Doxiciclina/farmacología , Sistemas de Liberación de Medicamentos , Helicobacter pylori/efectos de los fármacos , Liposomas/química , Estómago/efectos de los fármacos , Antibacterianos/química , Membrana Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxiciclina/química , Oro/química , Helicobacter pylori/citología , Humanos , Concentración de Iones de Hidrógeno , Liposomas/síntesis química , Nanopartículas del Metal/química , Pruebas de Sensibilidad Microbiana , Tamaño de la Partícula , Estómago/química , Estómago/microbiología , Relación Estructura-Actividad , Propiedades de Superficie
10.
Biosens Bioelectron ; 230: 115247, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37023552

RESUMEN

The release of cytokines by chimeric antigen receptor (CAR) T-cells and tumor resident immune cells defines a significant part of CAR T-cell functional activity and patient immune responses during CAR T-cell therapy. However, few studies have so far precisely characterized the cytokine secretion dynamics in the tumor niche during CAR T-cell therapy, which requires multiplexed, and timely biosensing platforms and integration with biomimetic tumor microenvironment. Herein, we implemented a digital nanoplasmonic microarray immunosensor with a microfluidic biomimetic Leukemia-on-a-Chip model to monitor cytokine secretion dynamics during CD19 CAR T-cell therapy against precursor B-cell acute lymphocytic leukemia (B-ALL). The integrated nanoplasmonic biosensors achieved precise multiplexed cytokine measurements with low operating sample volume, short assay time, heightened sensitivity, and negligible sensor crosstalk. Using the digital nanoplasmonic biosensing approach, we measured the concentrations of six cytokines (TNF-α, IFN-γ, MCP-1, GM-CSF, IL-1ß, and IL-6) during first 5 days of CAR T-cell treatment in the microfluidic Leukemia-on-a-Chip model. Our results revealed a heterogeneous secretion profile of various cytokines during CAR T-cell therapy and confirmed a correlation between the cytokine secretion profile and the CAR T-cell cytotoxic activity. The capability to monitor immune cell cytokine secretion dynamics in a biomimetic tumor microenvironment could further help in study of cytokine release syndrome during CAR T-cell therapy and in development of more efficient and safer immunotherapies.


Asunto(s)
Técnicas Biosensibles , Leucemia , Humanos , Inmunoterapia Adoptiva/métodos , Citocinas , Microambiente Tumoral , Inmunoensayo
11.
Mol Pharm ; 9(9): 2677-85, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22827534

RESUMEN

Helicobacter pylori (H. pylori) infection with its vast prevalence is responsible for various gastric diseases including gastritis, peptic ulcers, and gastric malignancy. While effective, current treatment regimens are challenged by a fast-declining eradication rate due to the increasing emergence of H. pylori strains resistant to existing antibiotics. Therefore, there is an urgent need to develop novel antibacterial strategies against H. pylori. In this study, we developed a liposomal nanoformulation of linolenic acid (LipoLLA) and evaluated its bactericidal activity against resistant strains of H. pylori. Using a laboratory strain of H. pylori, we found that LipoLLA was effective in killing both spiral and coccoid forms of the bacteria via disrupting bacterial membranes. Using a metronidazole-resistant strain of H. pylori and seven clinically isolated strains, we further demonstrated that LipoLLA eradicated all strains of the bacteria regardless of their antibiotic resistance status. Furthermore, under our experimental conditions, the bacteria did not develop drug resistance when cultured with LipoLLA at various sub-bactericidal concentrations, whereas they rapidly acquired resistance to both metronidazole and free linolenic acid (LLA). Our findings suggest that LipoLLA is a promising antibacterial nanotherapeutic to treat antibiotic-resistant H. pylori infection.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Helicobacter pylori/efectos de los fármacos , Ácidos Linolénicos/química , Ácidos Linolénicos/farmacología , Liposomas/química , Liposomas/farmacología , Farmacorresistencia Microbiana , Infecciones por Helicobacter/tratamiento farmacológico , Metronidazol/farmacología
12.
Acta Pharm Sin B ; 10(11): 2054-2074, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33304779

RESUMEN

Cancer immunotherapy has veered the paradigm of cancer treatment. Despite recent advances in immunotherapy for improved antitumor efficacy, the complicated tumor microenvironment (TME) is highly immunosuppressive, yielding both astounding and unsatisfactory clinical successes. In this regard, clinical outcomes of currently available immunotherapy are confined to the varied immune systems owing in large part to the lack of understanding of the complexity and diversity of the immune context of the TME. Various advanced designs of nanomedicines could still not fully surmount the delivery barriers of the TME. The immunosuppressive TME may even dampen the efficacy of antitumor immunity. Recently, some nanotechnology-related strategies have been inaugurated to modulate the immunosuppressive cells within the tumor immune microenvironment (TIME) for robust immunotherapeutic responses. In this review, we will highlight the current understanding of the immunosuppressive TIME and identify disparate subclasses of TIME that possess an impact on immunotherapy, especially those unique classes associated with the immunosuppressive effect. The immunoregulatory cell types inside the immunosuppressive TIME will be delineated along with the existing and potential approaches for immunosuppressive cell modulation. After introducing the various strategies, we will ultimately outline both the novel therapeutic targets and the potential issues that affect the efficacy of TIME-based nanomedicines.

13.
Adv Ther (Weinh) ; 1(2)2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30320205

RESUMEN

Inspired by the natural pathogen-host interactions and adhesion, this study reports on the development of a novel targeted nanotherapeutics for the treatment of Helicobacter pylori (H. pylori) infection. Specifically, plasma membranes of gastric epithelial cells (e.g. AGS cells) are collected and coated onto antibiotic-loaded polymeric cores, the resulting biomimetic nanoparticles (denoted AGS-NPs) bear the same surface antigens as the source AGS cells and thus have inherent adhesion to H. pylori bacteria. When incubated with H. pylori bacteria in vitro, the AGS-NPs preferentially accumulate on the bacterial surfaces. Using clarithromycin (CLR) as a model antibiotic and a mouse model of H. pylori infection, the CLR-loaded AGS-NPs demonstrate superior therapeutic efficacy as compared the free drug counterpart as well as non-targeted nanoparticle control group. Overall, this work illustrates the promise and strength of using natural host cell membranes to functionalize drug nanocarriers for targeted drug delivery to pathogens that colonize on the host cells. As host-pathogen adhesion represents a common biological event for various types of pathogenic bacteria, the bioinspired nanotherapeutic strategy reported here represents a versatile delivery platform that may be applied to treat numerous infectious diseases.

14.
Nat Commun ; 8(1): 272, 2017 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-28814725

RESUMEN

Advances in bioinspired design principles and nanomaterials have led to tremendous progress in autonomously moving synthetic nano/micromotors with diverse functionalities in different environments. However, a significant gap remains in moving nano/micromotors from test tubes to living organisms for treating diseases with high efficacy. Here we present the first, to our knowledge, in vivo therapeutic micromotors application for active drug delivery to treat gastric bacterial infection in a mouse model using clarithromycin as a model antibiotic and Helicobacter pylori infection as a model disease. The propulsion of drug-loaded magnesium micromotors in gastric media enables effective antibiotic delivery, leading to significant bacteria burden reduction in the mouse stomach compared with passive drug carriers, with no apparent toxicity. Moreover, while the drug-loaded micromotors reach similar therapeutic efficacy as the positive control of free drug plus proton pump inhibitor, the micromotors can function without proton pump inhibitors because of their built-in proton depletion function associated with their locomotion.Nano- and micromotors have been demonstrated in vitro for a range of applications. Here the authors demonstrate the in-vivo therapeutic use of micromotors to treat H. pylori infection.


Asunto(s)
Antibacterianos/administración & dosificación , Claritromicina/administración & dosificación , Dilatación Gástrica/tratamiento farmacológico , Infecciones por Helicobacter/tratamiento farmacológico , Inhibidores de la Bomba de Protones/uso terapéutico , Animales , Sistemas de Liberación de Medicamentos , Quimioterapia Combinada , Helicobacter pylori , Magnesio , Ratones , Nanotecnología
15.
Nat Commun ; 8(1): 1299, 2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29089506

RESUMEN

Marygorret Obonyo, who provided the H. pylori SS1 strain for this work and participated in the design of H. pylori infection studies, was inadvertently omitted from the author list. This has now been corrected in both the PDF and HTML versions of the Article.

16.
Nat Photonics ; 11: 352-355, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29576804

RESUMEN

Ultrasensitive nanomechanical instruments, including the atomic force microscope (AFM)1-4 and optical and magnetic tweezers5-8, have helped shed new light on the complex mechanical environments of biological processes. However, it is difficult to scale down the size of these instruments due to their feedback mechanisms9, which, if overcome, would enable high-density nanomechanical probing inside materials. A variety of molecular force probes including mechanophores10, quantum dots11, fluorescent pairs12,13 and molecular rotors14-16 have been designed to measure intracellular stresses; however, fluorescence-based techniques can have short operating times due to photo-instability and it is still challenging to quantify the forces with high spatial and mechanical resolution. Here, we develop a compact nanofibre optic force transducer (NOFT) that utilizes strong near-field plasmon-dielectric interactions to measure local forces with a sensitivity of <200 fN. The NOFT system is tested by monitoring bacterial motion and heart-cell beating as well as detecting infrasound power in solution.

17.
ACS Appl Mater Interfaces ; 8(28): 18367-74, 2016 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-27352845

RESUMEN

Effective antibacterial treatment at the infection site associated with high shear forces remains challenging, owing largely to the lack of durably adhesive and safe delivery platforms that can enable localized antibiotic accumulation against bacterial colonization. Inspired by delivery systems mimicking marine mussels for adhesion, herein, we developed a bioadhesive nanoparticle-hydrogel hybrid (NP-gel) to enhance localized antimicrobial drug delivery. Antibiotics were loaded into polymeric nanoparticles and then embedded into a 3D hydrogel network that confers adhesion to biological surfaces. The combination of two distinct delivery platforms, namely, nanoparticles and hydrogel, allows the hydrogel network properties to be independently tailored for adhesion while maintaining controlled and prolonged antibiotic release profile from the nanoparticles. The bioadhesive NP-gel developed here showed superior adhesion and antibiotic retention under high shear stress on a bacterial film, a mammalian cell monolayer, and mouse skin tissue. Under a flow environment, the NP-gel inhibited the formation of an Escherichia coli bacterial film. When applied on mouse skin tissue for 7 consecutive days, the NP-gel did not generate any observable skin reaction or toxicity, implying its potential as a safe and effective local delivery platform against microbial infections.


Asunto(s)
Antibacterianos/administración & dosificación , Antibacterianos/química , Sistemas de Liberación de Medicamentos/métodos , Hidrogeles/química , Nanopartículas/química , Acrilamidas/química , Adhesivos/administración & dosificación , Adhesivos/química , Animales , Biopelículas/efectos de los fármacos , Ciprofloxacina/administración & dosificación , Ciprofloxacina/química , Dopamina/química , Sistemas de Liberación de Medicamentos/instrumentación , Escherichia coli/efectos de los fármacos , Células HEK293 , Humanos , Hidrogeles/administración & dosificación , Ácido Láctico/administración & dosificación , Ácido Láctico/química , Ratones , Ratones Endogámicos ICR , Nanopartículas/administración & dosificación , Ácido Poliglicólico/administración & dosificación , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Piel/efectos de los fármacos
18.
Theranostics ; 6(7): 1004-11, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27217833

RESUMEN

The therapeutic potential of nanoparticle-based drug carriers depends largely on their ability to evade the host immune system while delivering their cargo safely to the site of action. Of particular interest are simple strategies for the functionalization of nanoparticle surfaces that are both inherently safe and can also bestow immunoevasive properties, allowing for extended blood circulation times. Here, we evaluated a recently reported cell membrane-coated nanoparticle platform as a drug delivery vehicle for the treatment of a murine model of lymphoma. These biomimetic nanoparticles, consisting of a biodegradable polymeric material cloaked with natural red blood cell membrane, were shown to efficiently deliver a model chemotherapeutic, doxorubicin, to solid tumor sites for significantly increased tumor growth inhibition compared with conventional free drug treatment. Importantly, the nanoparticles also showed excellent immunocompatibility as well as an advantageous safety profile compared with the free drug, making them attractive for potential translation. This study demonstrates the promise of using a biomembrane-coating approach as the basis for the design of functional, safe, and immunocompatible nanocarriers for cancer drug delivery.


Asunto(s)
Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Portadores de Fármacos/metabolismo , Membrana Eritrocítica/química , Linfoma/tratamiento farmacológico , Nanoestructuras , Animales , Antineoplásicos/farmacología , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Xenoinjertos , Linfoma/patología , Ratones Endogámicos C57BL , Resultado del Tratamiento
19.
ACS Nano ; 10(10): 9536-9542, 2016 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-27648483

RESUMEN

The gastrointestinal (GI) tract, which hosts hundreds of bacteria species, becomes the most exciting organ for the emerging microbiome research. Some of these GI microbes are hostile and cause a variety of diseases. These bacteria colonize in different segments of the GI tract dependent on the local physicochemical and biological factors. Therefore, selectively locating therapeutic or imaging agents to specific GI segments is of significant importance for studying gut microbiome and treating various GI-related diseases. Herein, we demonstrate an enteric micromotor system capable of precise positioning and controllable retention in desired segments of the GI tract. These motors, consisting of magnesium-based tubular micromotors coated with an enteric polymer layer, act as a robust nanobiotechnology tool for site-specific GI delivery. The micromotors can deliver payload to a particular location via dissolution of their enteric coating to activate their propulsion at the target site toward localized tissue penetration and retention.

20.
Vaccines (Basel) ; 3(4): 814-28, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26457720

RESUMEN

Nanoparticles have demonstrated unique advantages in enhancing immunotherapy potency and have drawn increasing interest in developing safe and effective vaccine formulations. Recent technological advancement has led to the discovery and development of cell membrane-coated nanoparticles, which combine the rich functionalities of cellular membranes and the engineering flexibility of synthetic nanomaterials. This new class of biomimetic nanoparticles has inspired novel vaccine design strategies with strong potential for modulating antibacterial immunity. This article will review recent progress on using cell membrane-coated nanoparticles for antibacterial vaccination. Specifically, two major development strategies will be discussed, namely (i) vaccination against virulence factors through bacterial toxin sequestration; and (ii) vaccination against pathogens through mimicking bacterial antigen presentation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA