Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nano Lett ; 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38855905

RESUMO

Neurotoxins are known for their extreme lethality. However, due to their enormous diversity, effective and broad-spectrum countermeasures are lacking. This study presents a dual-modal cellular nanoparticle (CNP) formulation engineered for continuous neurotoxin neutralization. The formulation involves encapsulating the metabolic enzyme N-sulfotransferase (SxtN) into metal-organic framework (MOF) nanoparticle cores and coating them with a natural neuronal membrane, termed "Neuron-MOF/SxtN-NPs". The resulting nanoparticles combine membrane-enabled broad-spectrum neurotoxin neutralization with enzyme payload-enabled continuous neurotoxin neutralization. The studies confirm the protection of the enzyme payload by the MOF core and validate the continuous neutralization of saxitoxin (STX). In vivo studies conducted using a mouse model of STX intoxication reveal markedly improved survival rates compared with control groups. Furthermore, acute toxicity assessments show no adverse effects associated with the administration of Neuron-MOF/SxtN-NPs in healthy mice. Overall, Neuron-MOF/SxtN-NPs represent a unique biomimetic nanomedicine platform poised to effectively neutralize neurotoxins, marking an important advancement in the field of countermeasure nanomedicine.

2.
Small ; 20(14): e2309635, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37990378

RESUMO

Neurotoxins present a substantial threat to human health and security as they disrupt and damage the nervous system. Their potent and structurally diverse nature poses challenges in developing effective countermeasures. In this study, a unique nanoparticle design that combines dual-biomimicry mechanisms to enhance the detoxification efficacy of neurotoxins is introduced. Using saxitoxin (STX), one of the deadliest neurotoxins, and its natural binding protein saxiphilin (Sxph) as a model system, human neuronal membrane-coated and Sxph-loaded metal-organic framework (MOF) nanosponges (denoted "Neuron-MOF/Sxph-NS") are successfully developed. The resulting Neuron-MOF/Sxph-NS exhibit a biomimetic design that not only emulates host neurons for function-based detoxification through the neuronal membrane coating, but also mimics toxin-resistant organisms by encapsulating the Sxph protein within the nanoparticle core. The comprehensive in vitro assays, including cell osmotic swelling, calcium flux, and cytotoxicity assays, demonstrate the improved detoxification efficacy of Neuron-MOF/Sxph-NS. Furthermore, in mouse models of STX intoxication, the application of Neuron-MOF/Sxph-NS shows significant survival benefits in both therapeutic and prophylactic regimens, without any apparent acute toxicity. Overall, the development of Neuron-MOF/Sxph-NS represents an important advancement in neurotoxin detoxification, offering promising potential for treating injuries and diseases caused by neurotoxins and addressing the current limitations in neurotoxin countermeasures.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Animais , Camundongos , Humanos , Neurotoxinas , Membrana Celular , Proteínas de Transporte , Nanopartículas/química , Neurônios
3.
Nano Lett ; 23(17): 7941-7949, 2023 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-37602707

RESUMO

Cell membrane-based nanovaccines have demonstrated attractive features due to their inherently multiantigenic nature and ability to be formulated with adjuvants. Here, we report on cellular nanodiscs fabricated from cancer cell membranes and incorporated with a lipid-based adjuvant for antitumor vaccination. The cellular nanodiscs, with their small size and discoidal shape, are readily taken up by antigen-presenting cells and drain efficiently to the lymph nodes. Due to its highly immunostimulatory properties, the nanodisc vaccine effectively stimulates the immune system and promotes tumor-specific immunity. Using a murine colorectal cancer model, strong control of tumor growth is achieved in both prophylactic and therapeutic settings, particularly in combination with checkpoint blockades. Considerable therapeutic efficacy is also observed in treating a weakly immunogenic metastatic melanoma model. This work presents a new paradigm for the design of multiantigenic nanovaccines that can effectively activate antitumor immune responses and may be applicable to a wide range of cancers.


Assuntos
Melanoma , Vacinação , Animais , Camundongos , Membrana Celular , Membranas , Células Apresentadoras de Antígenos , Adjuvantes Imunológicos/uso terapêutico
4.
Small ; 19(52): e2305551, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37635117

RESUMO

Nanoparticles coated with natural cell membranes have emerged as a promising class of biomimetic nanomedicine with significant clinical potential. Among them, macrophage membrane-coated nanoparticles hold particular appeal due to their versatility in drug delivery and biological neutralization applications. This study employs a genetic engineering approach to enhance their in vivo residence times, aiming to further improve their performance. Specifically, macrophages are engineered to express proline-alanine-serine (PAS) peptide chains, which provide additional protection against opsonization and phagocytosis. The resulting modified nanoparticles demonstrate prolonged residence times when administered intravenously or introduced intratracheally, surpassing those coated with the wild-type membrane. The longer residence times also contribute to enhanced nanoparticle efficacy in inhibiting inflammatory cytokines in mouse models of lipopolysaccharide-induced lung injury and sublethal endotoxemia, respectively. This study underscores the effectiveness of genetic modification in extending the in vivo residence times of macrophage membrane-coated nanoparticles. This approach can be readily extended to modify other cell membrane-coated nanoparticles toward more favorable biomedical applications.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas , Camundongos , Animais , Sistemas de Liberação de Medicamentos/métodos , Macrófagos/metabolismo , Membrana Celular/metabolismo , Citoplasma
5.
Small ; : e2308327, 2023 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-38044300

RESUMO

The multifaceted functions of platelets in various physiological processes have long inspired the development of therapeutic nanoparticles that mimic specific platelet features for disease treatment. Here, the development and characterization of platelet membrane-derived nanodiscs (PLT-NDs) as platelet decoys for biological neutralization is reported. In one application, PLT-NDs effectively bind with anti-platelet autoantibodies, thus blocking them from interacting with platelets. In a mouse model of thrombocytopenia, PLT-NDs successfully neutralize pathological anti-platelet antibodies, preventing platelet depletion and maintaining hemostasis. In another application, PLT-NDs effectively neutralize the cytotoxicity of bacterial virulence factors secreted by methicillin-resistant Staphylococcus aureus (MRSA). In a mouse model of MRSA infection, treatment with PLT-NDs leads to significant survival benefits for the infected mice. Additionally, PLT-NDs show good biocompatibility and biosafety, as demonstrated in acute toxicity studies conducted in mice. These findings underscore the potential of PLT-NDs as a promising platelet mimicry for neutralizing various biological agents that target platelets. Overall, this work expands the repertoire of platelet-mimicking nanomedicine by creating a unique disc-like nanostructure made of natural platelet membranes.

6.
Nat Mater ; 21(11): 1324-1332, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36138145

RESUMO

Bioinspired microrobots capable of actively moving in biological fluids have attracted considerable attention for biomedical applications because of their unique dynamic features that are otherwise difficult to achieve by their static counterparts. Here we use click chemistry to attach antibiotic-loaded neutrophil membrane-coated polymeric nanoparticles to natural microalgae, thus creating hybrid microrobots for the active delivery of antibiotics in the lungs in vivo. The microrobots show fast speed (>110 µm s-1) in simulated lung fluid and uniform distribution into deep lung tissues, low clearance by alveolar macrophages and superb tissue retention time (>2 days) after intratracheal administration to test animals. In a mouse model of acute Pseudomonas aeruginosa pneumonia, the microrobots effectively reduce bacterial burden and substantially lessen animal mortality, with negligible toxicity. Overall, these findings highlight the attractive functions of algae-nanoparticle hybrid microrobots for the active in vivo delivery of therapeutics to the lungs in intensive care unit settings.


Assuntos
Nanopartículas , Pneumonia Bacteriana , Camundongos , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/microbiologia , Pseudomonas aeruginosa , Pulmão
7.
Nano Lett ; 22(17): 7057-7065, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35998891

RESUMO

Acinetobacter baumannii is a leading cause of antibiotic-resistant nosocomial infections with high mortality rates, yet there is currently no clinically approved vaccine formulation. During the onset of A. baumannii infection, neutrophils are the primary responders and play a major role in resisting the pathogen. Here, we design a biomimetic nanotoxoid for antivirulence vaccination by using neutrophil membrane-coated nanoparticles to safely capture secreted A. baumannii factors. Vaccination with the nanotoxoid formulation rapidly mobilizes innate immune cells and promotes pathogen-specific adaptive immunity. In murine models of pneumonia, septicemia, and superficial wound infection, immunization with the nanovaccine offers significant protection, improving survival and reducing signs of acute inflammation. Lower bacterial burdens are observed in vaccinated animals regardless of the infection route. Altogether, neutrophil nanotoxoids represent an effective platform for eliciting multivalent immunity to protect against multidrug-resistant A. baumannii in a wide range of disease conditions.


Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii , Sepse , Infecções por Acinetobacter/tratamento farmacológico , Infecções por Acinetobacter/microbiologia , Animais , Biomimética , Modelos Animais de Doenças , Camundongos , Neutrófilos
8.
Nano Lett ; 22(23): 9672-9678, 2022 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-36448694

RESUMO

Anthrax infections caused by Bacillus anthracis are an ongoing bioterrorism and livestock threat worldwide. Current approaches for management, including extended passive antibody transfusion, antibiotics, and prophylactic vaccination, are often cumbersome and associated with low patient compliance. Here, we report on the development of an adjuvanted nanotoxoid vaccine based on macrophage membrane-coated nanoparticles bound with anthrax toxins. This design leverages the natural binding interaction of protective antigen, a key anthrax toxin, with macrophages. In a murine model, a single low-dose vaccination with the nanotoxoids generates long-lasting immunity that protects against subsequent challenge with anthrax toxins. Overall, this work provides a new approach to address the ongoing threat of anthrax outbreaks and bioterrorism by taking advantage of an emerging biomimetic nanotechnology.


Assuntos
Vacinas contra Antraz , Antraz , Toxinas Bacterianas , Animais , Humanos , Camundongos , Antraz/prevenção & controle , Antígenos de Bactérias , Bacillus anthracis , Nanotecnologia
9.
J Infect Dis ; 226(2): 319-323, 2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-35262728

RESUMO

The protozoan pathogen Giardia lamblia is an important worldwide cause of diarrheal disease and malabsorption. Infection is managed with antimicrobials, although drug resistance and treatment failures are a clinical challenge. Prior infection provides significant protection, yet a human vaccine has not been realized. Individual antigens can elicit partial protection in experimental models, but protection is weaker than after prior infection. Here, we developed a multivalent nanovaccine by coating membranes derived from the parasite onto uniform and stable polymeric nanoparticles loaded with a mucosal adjuvant. Intranasal immunization with the nanovaccine induced adaptive immunity and effectively protected mice from G. lamblia infection.


Assuntos
Giardia lamblia , Giardíase , Nanopartículas , Parasitos , Adjuvantes Imunológicos , Animais , Giardíase/parasitologia , Giardíase/prevenção & controle , Humanos , Imunidade nas Mucosas , Camundongos
10.
Angew Chem Int Ed Engl ; 62(21): e202301566, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36853913

RESUMO

Nanodiscs are a compelling nanomedicine platform due to their ultrasmall size and distinct disc shape. Current nanodisc formulations are made primarily with synthetic lipid bilayers and proteins. Here, we report a cellular nanodisc made with human red blood cell (RBC) membrane (denoted "RBC-ND") and show its effective neutralization against bacterial toxins. In vitro, RBC-ND neutralizes the hemolytic activity and cytotoxicity caused by purified α-toxin or complex whole secreted proteins (wSP) from methicillin-resistant Staphylococcus aureus bacteria. In vivo, RBC-ND confers significant survival benefits for mice intoxicated with α-toxin or wSP in both therapeutic and prevention regimens. Moreover, RBC-ND shows good biocompatibility and biosafety in vivo. Overall, RBC-ND distinguishes itself by inheriting the biological functions of the source cell membrane for bioactivity. The design strategy of RBC-ND can be generalized to other types of cell membranes for broad applications.


Assuntos
Toxinas Bacterianas , Staphylococcus aureus Resistente à Meticilina , Humanos , Animais , Camundongos , Eritrócitos , Membrana Eritrocítica , Bicamadas Lipídicas
11.
J Am Chem Soc ; 144(38): 17700-17708, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36112651

RESUMO

Conventional sandwich immunosensors rely on antibody recognition layers to selectively capture and detect target antigen analytes. However, the fabrication of these traditional affinity sensors is typically associated with lengthy and multistep surface modifications of electrodes and faces the challenge of nonspecific adsorption from complex sample matrices. Here, we report on a unique design of bioelectronic affinity sensors by using natural cell membranes as recognition layers for protein detection and prevention of biofouling. Specifically, we employ the human macrophage (MΦ) membrane together with the human red blood cell (RBC) membrane to coat electrochemical transducers through a one-step process. The natural protein receptors on the MΦ membrane are used to capture target antigens, while the RBC membrane effectively prevents nonspecific surface binding. In an attempt to detect tumor necrosis factor alpha (TNF-α) cytokine using the bioelectronic affinity sensor, it demonstrates a remarkable limit of detection of 150 pM. This new sensor design integrates natural cell membranes and electronic transduction, which offers synergistic functionalities toward a broad range of biosensing applications.


Assuntos
Técnicas Biossensoriais , Antígenos , Membrana Celular , Técnicas Eletroquímicas , Eletrodos , Humanos , Imunoensaio , Fator de Necrose Tumoral alfa
12.
Bioconjug Chem ; 33(4): 586-593, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35285617

RESUMO

Active targeting strategies aimed at improving drug homing while reducing systemic toxicity are widely being pursued in the growing field of nanomedicine. While they can be effective, these approaches often require the identification of cell-specific targets and in-depth knowledge of receptor binding interactions. More recently, there has been significant interest in biomimetic nanoformulations capable of replicating the properties of naturally occurring systems. In particular, the advent of cell membrane coating nanotechnology has enabled researchers to leverage the inherent tropisms displayed by living cells, bypassing many of the challenges associated with traditional bottom-up nanoengineering. In this work, we report on a biomimetic organotropic nanodelivery system for localizing therapeutic payloads to the lungs. Metastatic breast cancer exosomes, which are lung tropic due to their unique surface marker expression profile, are used to coat nanoparticle cores loaded with the anti-inflammatory drug dexamethasone. In vivo, these nanoparticles demonstrate enhanced accumulation in lung tissue and significantly reduce proinflammatory cytokine burden in a lung inflammation model. Overall, this work highlights the potential of using biomimetic organ-level delivery strategies for the management of certain disease conditions.


Assuntos
Materiais Biomiméticos , Pneumopatias , Nanopartículas , Materiais Biomiméticos/química , Materiais Biomiméticos/uso terapêutico , Biomimética , Sistemas de Liberação de Medicamentos , Humanos , Nanomedicina , Nanopartículas/química , Nanotecnologia
13.
Nano Lett ; 21(6): 2603-2609, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33687220

RESUMO

Natural cell membranes derived from various cell sources have been successfully utilized to coat nanomaterials for functionalization. However, intracellular membranes from the organelles of eukaryotes remain unexplored. Herein, we choose mitochondrion as a representative cell organelle and coat outer mitochondrial membrane (OMM) from mouse livers onto nanoparticles and field-effect transistors (FETs) through a membrane vesicle-substrate fusion process. Polymeric nanoparticles coated with OMM (OMM-NPs) can bind with ABT-263, a B-cell lymphoma protein 2 (Bcl-2) inhibitor that targets the OMM. As a result, OMM-NPs effectively protect the cells from ABT-263 induced cell death and apoptosis in vitro and attenuated ABT-263-induced thrombocytopenia in vivo. Meanwhile, FET sensors coated with OMM (OMM-FETs) can detect and distinguish anti-Bcl-2 antibody and small molecule agonists. Overall, these results show that OMM can be coated onto the surfaces of both nanoparticles and functional devices, suggesting that intracellular membranes can be used as coating materials for novel biointerfacing.


Assuntos
Membranas Mitocondriais , Nanopartículas , Animais , Apoptose , Membrana Celular , Camundongos , Mitocôndrias , Membranas Mitocondriais/metabolismo
14.
Angew Chem Int Ed Engl ; 61(24): e202203115, 2022 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-35395111

RESUMO

Metal-organic-framework nanoparticles (MOF NPs) have been increasingly used to encapsulate therapeutic enzymes for delivery. To better interface these MOF NPs with biological systems, researchers have coated them with natural cell membranes, enabling biomimicking properties suitable for innovative biomedical applications. Herein, we report that the enzymatic activity of cell-membrane-coated MOF NPs can be significantly enhanced by reducing membrane cholesterol content. We demonstrate such cholesterol-enzymatic activity correlation using zeolitic imidazolate framework-8 MOF NPs to encapsulate catalase, horseradish peroxidase, and organophosphate hydrolase, respectively. MOF NPs coated with membranes of human red blood cells or macrophages show similar outcomes, illustrating the broad applicability of this finding. The mechanistic investigation further reveals that reducing cholesterol levels effectively enhances membrane permeability likely responsible for the increased enzymatic activity. These results also imply a facile approach to tailoring the enzymatic activity of cell-membrane-coated MOF NPs by simply tuning the membrane cholesterol level.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Zeolitas , Membrana Celular/metabolismo , Humanos , Estruturas Metalorgânicas/metabolismo
15.
Angew Chem Int Ed Engl ; 61(2): e202113671, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-34694684

RESUMO

Effective endosomal escape after cellular uptake represents a major challenge in the field of nanodelivery, as the majority of drug payloads must localize to subcellular compartments other than the endosomes in order to exert activity. In nature, viruses can readily deliver their genetic material to the cytosol of host cells by triggering membrane fusion after endocytosis. For the influenza A virus, the hemagglutinin (HA) protein found on its surface fuses the viral envelope with the surrounding membrane at endosomal pH values. Biomimetic nanoparticles capable of endosomal escape were fabricated using a membrane coating derived from cells engineered to express HA on their surface. When evaluated in vitro, these virus-mimicking nanoparticles were able to deliver an mRNA payload to the cytosolic compartment of target cells, resulting in the successful expression of the encoded protein. When the mRNA-loaded nanoparticles were administered in vivo, protein expression levels were significantly increased in both local and systemic delivery scenarios. We therefore conclude that utilizing genetic engineering approaches to express viral fusion proteins on the surface of cell membrane-coated nanoparticles is a viable strategy for modulating the intracellular localization of encapsulated cargoes.


Assuntos
Citosol
16.
J Am Chem Soc ; 143(42): 17615-17621, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34647745

RESUMO

Cellular binding and entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are mediated by its spike glycoprotein (S protein), which binds with not only the human angiotensin-converting enzyme 2 (ACE2) receptor but also glycosaminoglycans such as heparin. Cell membrane-coated nanoparticles ("cellular nanosponges") mimic the host cells to attract and neutralize SARS-CoV-2 through natural cellular receptors, leading to a broad-spectrum antiviral strategy. Herein, we show that increasing surface heparin density on the cellular nanosponges can promote their inhibition against SARS-CoV-2. Specifically, cellular nanosponges are made with azido-expressing host cell membranes followed by conjugating heparin to the nanosponge surfaces. Cellular nanosponges with a higher heparin density have a larger binding capacity with viral S proteins and a significantly higher inhibition efficacy against SARS-CoV-2 infectivity. Overall, surface glycan engineering of host-mimicking cellular nanosponges is a facile method to enhance SARS-CoV-2 inhibition. This approach can be readily generalized to promote the inhibition of other glycan-dependent viruses.


Assuntos
Tratamento Farmacológico da COVID-19 , Heparina/administração & dosagem , Nanoestruturas/uso terapêutico , Polissacarídeos/administração & dosagem , SARS-CoV-2/metabolismo , COVID-19/virologia , Heparina/metabolismo , Humanos , Polissacarídeos/metabolismo
17.
Nature ; 526(7571): 118-21, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26374997

RESUMO

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.


Assuntos
Antibacterianos/administração & dosagem , Plaquetas/citologia , Membrana Celular/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/administração & dosagem , Nanopartículas/química , Adesividade Plaquetária , Animais , Antibacterianos/farmacocinética , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Colágeno/química , Colágeno/imunologia , Ativação do Complemento/imunologia , Reestenose Coronária/sangue , Reestenose Coronária/tratamento farmacológico , Reestenose Coronária/metabolismo , Modelos Animais de Doenças , Docetaxel , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Polímeros/química , Ratos , Ratos Sprague-Dawley , Infecções Estafilocócicas/sangue , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/citologia , Staphylococcus aureus/metabolismo , Taxoides/administração & dosagem , Taxoides/farmacocinética , Lipossomas Unilamelares/química , Vancomicina/administração & dosagem , Vancomicina/farmacocinética
18.
Nano Lett ; 20(5): 4051-4058, 2020 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-32352801

RESUMO

Therapeutic enzymes used for genetic disorders or metabolic diseases oftentimes suffer from suboptimal pharmacokinetics and stability. Nanodelivery systems have shown considerable promise for improving the performance of enzyme therapies. Here, we develop a cell membrane-camouflaged metal-organic framework (MOF) system with enhanced biocompatibility and functionality. The MOF core can efficiently encapsulate enzymes while maintaining their bioactivity. After the introduction of natural cell membrane coatings, the resulting nanoformulations can be safely administered in vivo. The surface receptors on the membrane can also provide additional functionalities that synergize with the encapsulated enzyme to target disease pathology from multiple dimensions. Employing uricase as a model enzyme, we demonstrate the utility of this approach in multiple animal disease models. The results support the use of cell membrane-coated MOFs for enzyme delivery, and this strategy could be leveraged to improve the usefulness of enzyme-based therapies for managing a wide range of important human health conditions.


Assuntos
Membrana Celular , Enzimas/administração & dosagem , Estruturas Metalorgânicas , Nanopartículas , Animais , Sistemas de Liberação de Medicamentos , Humanos
19.
Nano Lett ; 20(7): 5570-5574, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32551679

RESUMO

We report cellular nanosponges as an effective medical countermeasure to the SARS-CoV-2 virus. Two types of cellular nanosponges are made of the plasma membranes derived from human lung epithelial type II cells or human macrophages. These nanosponges display the same protein receptors, both identified and unidentified, required by SARS-CoV-2 for cellular entry. It is shown that, following incubation with the nanosponges, SARS-CoV-2 is neutralized and unable to infect cells. Crucially, the nanosponge platform is agnostic to viral mutations and potentially viral species, as well. As long as the target of the virus remains the identified host cell, the nanosponges will be able to neutralize the virus.


Assuntos
Betacoronavirus , Infecções por Coronavirus/prevenção & controle , Nanoestruturas , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Betacoronavirus/patogenicidade , Betacoronavirus/fisiologia , COVID-19 , Membrana Celular/virologia , Infecções por Coronavirus/virologia , Células Epiteliais/virologia , Interações entre Hospedeiro e Microrganismos , Humanos , Pulmão/citologia , Pulmão/virologia , Macrófagos/virologia , Nanoestruturas/ultraestrutura , Nanotecnologia , Pneumonia Viral/virologia , Receptores Virais/fisiologia , SARS-CoV-2 , Internalização do Vírus
20.
Proc Natl Acad Sci U S A ; 114(43): 11488-11493, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-29073076

RESUMO

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.


Assuntos
Membrana Celular/química , Citocinas/química , Endotoxinas/química , Infecções por Escherichia coli/terapia , Nanopartículas/química , Sepse/terapia , Animais , Bacteriemia/terapia , Linhagem Celular , Lipopolissacarídeos/farmacologia , Lipoproteínas/química , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Receptor 4 Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA