Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 266
Filter
1.
Nano Lett ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38855905

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-37990378

ABSTRACT

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.


Subject(s)
Metal-Organic Frameworks , Nanoparticles , Animals , Mice , Humans , Neurotoxins , Cell Membrane , Carrier Proteins , Nanoparticles/chemistry , Neurons
3.
Nano Lett ; 23(17): 7941-7949, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37602707

ABSTRACT

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.


Subject(s)
Melanoma , Vaccination , Animals , Mice , Cell Membrane , Membranes , Antigen-Presenting Cells , Adjuvants, Immunologic/therapeutic use
4.
Small ; 19(52): e2305551, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37635117

ABSTRACT

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.


Subject(s)
Drug Delivery Systems , Nanoparticles , Mice , Animals , Drug Delivery Systems/methods , Macrophages/metabolism , Cell Membrane/metabolism , Cytoplasm
5.
Small ; : e2308327, 2023 Dec 03.
Article in English | MEDLINE | ID: mdl-38044300

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-36138145

ABSTRACT

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.


Subject(s)
Nanoparticles , Pneumonia, Bacterial , Mice , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/microbiology , Pseudomonas aeruginosa , Lung
7.
Mol Cell ; 59(6): 931-40, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26365380

ABSTRACT

Glaucoma, a blinding neurodegenerative disease, whose risk factors include elevated intraocular pressure (IOP), age, and genetics, is characterized by accelerated and progressive retinal ganglion cell (RGC) death. Despite decades of research, the mechanism of RGC death in glaucoma is still unknown. Here, we demonstrate that the genetic effect of the SIX6 risk variant (rs33912345, His141Asn) is enhanced by another major POAG risk gene, p16INK4a (cyclin-dependent kinase inhibitor 2A, isoform INK4a). We further show that the upregulation of homozygous SIX6 risk alleles (CC) leads to an increase in p16INK4a expression, with subsequent cellular senescence, as evidenced in a mouse model of elevated IOP and in human POAG eyes. Our data indicate that SIX6 and/or IOP promotes POAG by directly increasing p16INK4a expression, leading to RGC senescence in adult human retinas. Our study provides important insights linking genetic susceptibility to the underlying mechanism of RGC death and provides a unified theory of glaucoma pathogenesis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Glaucoma, Open-Angle/metabolism , Homeodomain Proteins/physiology , Retinal Ganglion Cells/physiology , Trans-Activators/physiology , Amino Acid Sequence , Animals , Case-Control Studies , Cell Death , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/pathology , Humans , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutation, Missense , Up-Regulation
8.
Nano Lett ; 22(17): 7057-7065, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35998891

ABSTRACT

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.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Sepsis , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Animals , Biomimetics , Disease Models, Animal , Mice , Neutrophils
9.
Nano Lett ; 22(23): 9672-9678, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36448694

ABSTRACT

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.


Subject(s)
Anthrax Vaccines , Anthrax , Bacterial Toxins , Animals , Humans , Mice , Anthrax/prevention & control , Antigens, Bacterial , Bacillus anthracis , Nanotechnology
10.
J Infect Dis ; 226(2): 319-323, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35262728

ABSTRACT

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.


Subject(s)
Giardia lamblia , Giardiasis , Nanoparticles , Parasites , Adjuvants, Immunologic , Animals , Giardiasis/parasitology , Giardiasis/prevention & control , Humans , Immunity, Mucosal , Mice
11.
Angew Chem Int Ed Engl ; 62(21): e202301566, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36853913

ABSTRACT

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.


Subject(s)
Bacterial Toxins , Methicillin-Resistant Staphylococcus aureus , Humans , Animals , Mice , Erythrocytes , Erythrocyte Membrane , Lipid Bilayers
12.
J Am Chem Soc ; 144(38): 17700-17708, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36112651

ABSTRACT

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.


Subject(s)
Biosensing Techniques , Antigens , Cell Membrane , Electrochemical Techniques , Electrodes , Humans , Immunoassay , Tumor Necrosis Factor-alpha
13.
Bioconjug Chem ; 33(4): 586-593, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35285617

ABSTRACT

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.


Subject(s)
Biomimetic Materials , Lung Diseases , Nanoparticles , Biomimetic Materials/chemistry , Biomimetic Materials/therapeutic use , Biomimetics , Drug Delivery Systems , Humans , Nanomedicine , Nanoparticles/chemistry , Nanotechnology
14.
Nature ; 531(7594): 323-8, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26958831

ABSTRACT

The repair and regeneration of tissues using endogenous stem cells represents an ultimate goal in regenerative medicine. To our knowledge, human lens regeneration has not yet been demonstrated. Currently, the only treatment for cataracts, the leading cause of blindness worldwide, is to extract the cataractous lens and implant an artificial intraocular lens. However, this procedure poses notable risks of complications. Here we isolate lens epithelial stem/progenitor cells (LECs) in mammals and show that Pax6 and Bmi1 are required for LEC renewal. We design a surgical method of cataract removal that preserves endogenous LECs and achieves functional lens regeneration in rabbits and macaques, as well as in human infants with cataracts. Our method differs conceptually from current practice, as it preserves endogenous LECs and their natural environment maximally, and regenerates lenses with visual function. Our approach demonstrates a novel treatment strategy for cataracts and provides a new paradigm for tissue regeneration using endogenous stem cells.


Subject(s)
Cataract/therapy , Lens, Crystalline/cytology , Lens, Crystalline/physiology , Recovery of Function , Regeneration/physiology , Stem Cells/cytology , Vision, Ocular/physiology , Animals , Cataract/congenital , Cataract/pathology , Cataract/physiopathology , Cataract Extraction , Epithelial Cells/cytology , Epithelial Cells/metabolism , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Homeostasis , Humans , Macaca , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism
15.
Nano Lett ; 21(6): 2603-2609, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33687220

ABSTRACT

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.


Subject(s)
Mitochondrial Membranes , Nanoparticles , Animals , Apoptosis , Cell Membrane , Mice , Mitochondria , Mitochondrial Membranes/metabolism
16.
Angew Chem Int Ed Engl ; 61(24): e202203115, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35395111

ABSTRACT

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.


Subject(s)
Metal-Organic Frameworks , Nanoparticles , Zeolites , Cell Membrane/metabolism , Humans , Metal-Organic Frameworks/metabolism
17.
Angew Chem Int Ed Engl ; 61(2): e202113671, 2022 01 10.
Article in English | MEDLINE | ID: mdl-34694684

ABSTRACT

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.


Subject(s)
Cytosol
18.
Biochemistry ; 60(13): 941-955, 2021 04 06.
Article in English | MEDLINE | ID: mdl-32452667

ABSTRACT

There has been significant interest in developing cell membrane-coated nanoparticles due to their unique abilities of biomimicry and biointerfacing. As the technology progresses, it becomes clear that the application of these nanoparticles can be drastically broadened if additional functions beyond those derived from the natural cell membranes can be integrated. Herein, we summarize the most recent advances in the functionalization of cell membrane-coated nanoparticles. In particular, we focus on emerging methods, including (1) lipid insertion, (2) membrane hybridization, (3) metabolic engineering, and (4) genetic modification. These approaches contribute diverse functions in a nondisruptive fashion while preserving the natural function of the cell membranes. They also improve on the multifunctional and multitasking ability of cell membrane-coated nanoparticles, making them more adaptive to the complexity of biological systems. We hope that these approaches will serve as inspiration for more strategies and innovations to advance cell membrane coating technology.


Subject(s)
Cell Membrane/chemistry , Nanoparticles/chemistry , Nanotechnology/methods
19.
J Am Chem Soc ; 143(42): 17615-17621, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34647745

ABSTRACT

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.


Subject(s)
COVID-19 Drug Treatment , Heparin/administration & dosage , Nanostructures/therapeutic use , Polysaccharides/administration & dosage , SARS-CoV-2/metabolism , COVID-19/virology , Heparin/metabolism , Humans , Polysaccharides/metabolism
20.
J Am Chem Soc ; 143(31): 12194-12201, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34291944

ABSTRACT

The coronavirus SARS-CoV-2 can survive in wastewater for several days with a potential risk of waterborne human transmission, hence posing challenges in containing the virus and reducing its spread. Herein, we report on an active biohybrid microrobot system that offers highly efficient capture and removal of target virus from various aquatic media. The algae-based microrobot is fabricated by using click chemistry to functionalize microalgae with angiotensin-converting enzyme 2 (ACE2) receptor against the SARS-CoV-2 spike protein. The resulting ACE2-algae-robot displays fast (>100 µm/s) and long-lasting (>24 h) self-propulsion in diverse aquatic media including drinking water and river water, obviating the need for external fuels. Such movement of the ACE2-algae-robot offers effective "on-the-fly" removal of SARS-CoV-2 spike proteins and SARS-CoV-2 pseudovirus. Specifically, the active biohybrid microrobot results in 95% removal of viral spike protein and 89% removal of pseudovirus, significantly exceeding the control groups such as static ACE2-algae and bare algae. These results suggest considerable promise of biologically functionalized algae toward the removal of viruses and other environmental threats from wastewater.


Subject(s)
Angiotensin-Converting Enzyme 2/chemistry , Biotechnology/methods , Microalgae/chemistry , SARS-CoV-2/isolation & purification , Wastewater/virology , Water Purification/methods , Angiotensin-Converting Enzyme 2/metabolism , Biotechnology/instrumentation , Cell Line , Click Chemistry , Humans , Receptors, Virus/chemistry , Receptors, Virus/metabolism , SARS-CoV-2/metabolism , Water Purification/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL