Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 502
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(36): e2409955121, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39190351

ABSTRACT

Facing complex and variable emerging antibiotic pollutants, the traditional development of functional materials is a "trial-and-error" process based on physicochemical principles, where laborious steps and long timescales make it difficult to accelerate technical breakthroughs. Notably, natural biomolecular coronas derived from highly tolerant organisms under significant contamination scenarios can be used in conjunction with nanotechnology to tackling emerging contaminants of concern. Here, super worms (Tubifex tubifex) with high pollutant tolerance were integrated with nano-zero valent iron (nZVI) to effectively reduce the content of 17 antibiotics in wastewater within 7 d. Inspired by the synergistic remediation, nZVI-augmented worms were constructed as biological nanocomposites. Neither nZVI (0.3 to 3 g/L) nor worms (104 to 105 per liter) alone efficiently degraded florfenicol (FF, as a representative antibiotic), while their composite removed 87% of FF (3 µmol/L). Under antibiotic exposure, biomolecules secreted by worms formed a corona on and modified the nZVI particle surface, enabling the nano-bio interface greater functionality, including responsiveness, enrichment, and reduction. Mechanistically, FF exposure activated glucose-alanine cycle pathways that synthesize organic acids and amines as major metabolites, which were assembled into vesicles and secreted, thereby interacting with nZVI in a biologically response design strategy. Lactic acid and urea formed hydrogen bonds with FF, enriched analyte presence at the heterogeneous interface. Succinic and lactic acids corroded the nZVI passivation layer and promoted electron transfer through surface conjugation. This unique strategy highlights biomolecular coronas as a complex resource to augment nano-enabled technologies and will provide shortcuts for rational manipulation of nanomaterial surfaces with coordinated multifunctionalities.


Subject(s)
Anti-Bacterial Agents , Iron , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Animals , Iron/chemistry , Iron/metabolism , Protein Corona/chemistry , Protein Corona/metabolism , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/metabolism , Oligochaeta/metabolism , Biodegradation, Environmental , Environmental Restoration and Remediation/methods , Nanocomposites/chemistry
2.
Proc Natl Acad Sci U S A ; 119(11): e2106053119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35275789

ABSTRACT

SignificanceDeep profiling of the plasma proteome at scale has been a challenge for traditional approaches. We achieve superior performance across the dimensions of precision, depth, and throughput using a panel of surface-functionalized superparamagnetic nanoparticles in comparison to conventional workflows for deep proteomics interrogation. Our automated workflow leverages competitive nanoparticle-protein binding equilibria that quantitatively compress the large dynamic range of proteomes to an accessible scale. Using machine learning, we dissect the contribution of individual physicochemical properties of nanoparticles to the composition of protein coronas. Our results suggest that nanoparticle functionalization can be tailored to protein sets. This work demonstrates the feasibility of deep, precise, unbiased plasma proteomics at a scale compatible with large-scale genomics enabling multiomic studies.


Subject(s)
Blood Proteins , Deep Learning , Nanoparticles , Proteomics , Blood Proteins/chemistry , Nanoparticles/chemistry , Protein Corona/chemistry , Proteome , Proteomics/methods
3.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35173043

ABSTRACT

Safe and efficacious systemic delivery of messenger RNA (mRNA) to specific organs and cells in vivo remains the major challenge in the development of mRNA-based therapeutics. Targeting of systemically administered lipid nanoparticles (LNPs) coformulated with mRNA has largely been confined to the liver and spleen. Using a library screening approach, we identified that N-series LNPs (containing an amide bond in the tail) are capable of selectively delivering mRNA to the mouse lung, in contrast to our previous discovery that O-series LNPs (containing an ester bond in the tail) that tend to deliver mRNA to the liver. We analyzed the protein corona on the liver- and lung-targeted LNPs using liquid chromatography-mass spectrometry and identified a group of unique plasma proteins specifically absorbed onto the surface that may contribute to the targetability of these LNPs. Different pulmonary cell types can also be targeted by simply tuning the headgroup structure of N-series LNPs. Importantly, we demonstrate here the success of LNP-based RNA therapy in a preclinical model of lymphangioleiomyomatosis (LAM), a destructive lung disease caused by loss-of-function mutations in the Tsc2 gene. Our lung-targeting LNP exhibited highly efficient delivery of the mouse tuberous sclerosis complex 2 (Tsc2) mRNA for the restoration of TSC2 tumor suppressor in tumor and achieved remarkable therapeutic effect in reducing tumor burden. This research establishes mRNA LNPs as a promising therapeutic intervention for the treatment of LAM.


Subject(s)
Drug Delivery Systems/methods , Lymphangioleiomyomatosis/drug therapy , RNA, Messenger/administration & dosage , Animals , Female , Gene Transfer Techniques , Genetic Engineering/methods , Liposomes/chemistry , Liposomes/pharmacology , Lung/cytology , Lung/pathology , Lung Diseases/drug therapy , Lung Diseases/metabolism , Lymphangioleiomyomatosis/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Protein Corona/chemistry , Protein Corona/metabolism , RNA, Messenger/genetics , RNA, Messenger/pharmacology , RNA, Small Interfering/metabolism
4.
Nano Lett ; 24(32): 9874-9881, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39096192

ABSTRACT

We recently revealed significant variability in protein corona characterization across various proteomics facilities, indicating that data sets are not comparable between independent studies. This heterogeneity mainly arises from differences in sample preparation protocols, mass spectrometry workflows, and raw data processing. To address this issue, we developed standardized protocols and unified sample preparation workflows, distributing uniform protein corona digests to several top-performing proteomics centers from our previous study. We also examined the influence of using similar mass spectrometry instruments on data homogeneity and standardized database search parameters and data processing workflows. Our findings reveal a remarkable stepwise improvement in protein corona data uniformity, increasing overlaps in protein identification from 11% to 40% across facilities using similar instruments and through a uniform database search. We identify the key parameters behind data heterogeneity and provide recommendations for designing experiments. Our findings should significantly advance the robustness of protein corona analysis for diagnostic and therapeutics applications.


Subject(s)
Nanomedicine , Protein Corona , Proteomics , Protein Corona/chemistry , Protein Corona/analysis , Humans , Proteomics/methods , Reproducibility of Results , Mass Spectrometry/methods , Workflow
5.
Nano Lett ; 24(30): 9202-9211, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39037031

ABSTRACT

The formation of a protein corona gives nanomedicines a distinct biological identity, profoundly influencing their fate in the body. Nonspecific nanoparticle-protein interactions are typically highly heterogeneous, which can lead to unique biological behaviors and in vivo fates for individual nanoparticles that remain underexplored. To address this, we have established an in situ approach that allows quantitative examination of nanoparticle-protein adsorption at the individual nanoparticle level. This method integrates dual fluorescence quantification techniques, wherein the nanoparticles are first individually analyzed via nanoflow cytometry to detect fluorescent signals from adsorbed proteins. The obtained fluorescence intensity is then translated into protein quantities through calibration with microplate reader quantification. Consequently, this approach enables analysis of interparticle heterogeneity of nano-protein interactions, as well as in situ monitoring of protein adsorption kinetics and nanoparticle aggregation status in blood serum, preconditioning for a comprehensive understanding of nano-bio interactions, and predicting in vivo fate of nanomedicines.


Subject(s)
Blood Proteins , Nanoparticles , Adsorption , Nanoparticles/chemistry , Blood Proteins/chemistry , Blood Proteins/analysis , Humans , Protein Corona/chemistry , Fluorescence , Kinetics
6.
J Proteome Res ; 23(10): 4694-4703, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39312774

ABSTRACT

The dynamic range challenge for the detection of proteins and their proteoforms in human plasma has been well documented. Here, we use the nanoparticle protein corona approach to enrich low-abundance proteins selectively and reproducibly from human plasma and use top-down proteomics to quantify differential enrichment for the 2841 detected proteoforms from 114 proteins. Furthermore, nanoparticle enrichment allowed top-down detection of proteoforms between ∼1 µg/mL and ∼10 pg/mL in absolute abundance, providing up to a 105-fold increase in proteome depth over neat plasma in which only proteoforms from abundant proteins (>1 µg/mL) were detected. The ability to monitor medium and some low-abundant proteoforms through reproducible enrichment significantly extends the applicability of proteoform research by adding depth beyond albumin, immunoglobins, and apolipoproteins to uncover many involved in immunity and cell signaling. As proteoforms carry unique information content relative to peptides, this report opens the door to deeper proteoform sequencing in clinical proteomics of disease or aging cohorts.


Subject(s)
Blood Proteins , Nanoparticles , Proteomics , Humans , Proteomics/methods , Blood Proteins/analysis , Blood Proteins/chemistry , Nanoparticles/chemistry , Proteome/analysis , Protein Corona/chemistry
7.
J Proteome Res ; 23(8): 3649-3658, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39007500

ABSTRACT

Noninvasive detection of protein biomarkers in plasma is crucial for clinical purposes. Liquid chromatography-mass spectrometry (LC-MS) is the gold standard technique for plasma proteome analysis, but despite recent advances, it remains limited by throughput, cost, and coverage. Here, we introduce a new hybrid method that integrates direct infusion shotgun proteome analysis (DISPA) with nanoparticle (NP) protein corona enrichment for high-throughput and efficient plasma proteomic profiling. We realized over 280 protein identifications in 1.4 min collection time, which enables a potential throughput of approximately 1000 samples daily. The identified proteins are involved in valuable pathways, and 44 of the proteins are FDA-approved biomarkers. The robustness and quantitative accuracy of this method were evaluated across multiple NPs and concentrations with a mean coefficient of variation of 17%. Moreover, different protein corona profiles were observed among various NPs based on their distinct surface modifications, and all NP protein profiles exhibited deeper coverage and better quantification than neat plasma. Our streamlined workflow merges coverage and throughput with precise quantification, leveraging both DISPA and NP protein corona enrichment. This underscores the significant potential of DISPA when paired with NP sample preparation techniques for plasma proteome studies.


Subject(s)
Blood Proteins , Nanoparticles , Protein Corona , Proteome , Proteomics , Humans , Blood Proteins/analysis , Blood Proteins/chemistry , Nanoparticles/chemistry , Protein Corona/chemistry , Protein Corona/analysis , Proteome/analysis , Proteomics/methods , Chromatography, Liquid/methods , Mass Spectrometry/methods , Biomarkers/blood
8.
J Am Chem Soc ; 146(29): 19874-19885, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39007743

ABSTRACT

Detection of serum protein biomarkers is extremely challenging owing to the superior complexity of serum. Here, we report a method of proteome fishing from the serum. It uses a magnetic nanoparticle-protein corona and a multiplexed aptamer panel, which we incubated with the nanoparticle-protein corona for biomarker recognition. To transfer protein biomarker detection to aptamer detection, we established a CRISPR/Cas12a-based orthogonal multiplex aptamer sensing (COMPASS) platform by profiling the aptamers of protein corona with clinical nonsmall cell lung cancer (NSCLC) serum samples. Furthermore, we determined the four out of nine (FOON) panel (including HE4, NSE, AFP, and VEGF165) to be the most cost-effective and accurate panel for COMPASS in NSCLC diagnosis. The diagnostic accuracy of NSCLC by the FOON panel with internal and external cohorts was 95.56% (ROC-AUC = 99.40%) and 89.58% (ROC-AUC = 95.41%), respectively. Our developed COMPASS technology circumvents the otherwise challenging multiplexed serum protein amplification problem and avoids aptamer degradation in serum. Therefore, this novel COMPASS could lead to the development of a facile, cost-effective, intelligent, and high-throughput diagnostic platform for large-cohort cancer screening.


Subject(s)
Aptamers, Nucleotide , CRISPR-Cas Systems , Carcinoma, Non-Small-Cell Lung , Aptamers, Nucleotide/chemistry , Humans , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/blood , Proteome/analysis , Lung Neoplasms/blood , Lung Neoplasms/diagnosis , Biomarkers, Tumor/blood , Magnetite Nanoparticles/chemistry , Protein Corona/chemistry
9.
J Am Chem Soc ; 146(22): 15096-15107, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38773940

ABSTRACT

For cationic nanoparticles, the spontaneous nanoparticle-protein corona formation and aggregation in biofluids can trigger unexpected biological reactions. Herein, we present a biomimetic strategy for camouflaging the cationic peptide/siRNA nanocomplex (P/Si) with single or dual proteins, which exploits the unique properties of endogenous proteins and stabilizes the cationic P/Si complex for safe and targeted delivery. An in-depth study of the P/Si protein corona (P/Si-PC) formation and protein binding was conducted. The results provided insights into the biochemical and toxicological properties of cationic nanocomplexes and the rationales for engineering biomimetic protein camouflages. Based on this, the human serum albumin (HSA) and apolipoprotein AI (Apo-AI) ranked within the top 20 abundant protein species of P/Si-PC were selected to construct biomimetic HSA-dressed P/Si (P/Si@HSA) and dual protein (HSA and Apo-AI)-dressed P/Si (P/Si@HSA_Apo), given that the dual-protein camouflage plays complementary roles in efficient delivery. A branched cationic peptide (b-HKR) was tailored for siRNA delivery, and their nanocomplexes, including the cationic P/Si and biomimetic protein-dressed P/Si, were produced by a precise microfluidic technology. The biomimetic anionic protein camouflage greatly enhanced P/Si biostability and biocompatibility, which offers a reliable strategy for overcoming the limitation of applying cationic nanoparticles in biofluids and systemic delivery.


Subject(s)
Biomimetic Materials , Nanoparticles , Peptides , RNA, Small Interfering , Serum Albumin, Human , Humans , RNA, Small Interfering/chemistry , Peptides/chemistry , Biomimetic Materials/chemistry , Nanoparticles/chemistry , Serum Albumin, Human/chemistry , Protein Engineering , Apolipoprotein A-I/chemistry , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Protein Corona/chemistry , Biomimetics/methods
10.
Anal Chem ; 96(36): 14363-14371, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39192740

ABSTRACT

Highly abundant proteins present in biological fluids and tissues significantly interfere with low-abundance protein identification by mass spectrometry (MS), limiting proteomic depth and hindering protein biomarker discovery. Herein, to enhance the coverage of tissue proteomics, we developed a nanoparticle-protein corona (NP-PC)-based method for the aging mouse proteome atlas. Based on this method, we investigated the complexity of life process of 5 major organs, including the heart, liver, spleen, lungs, and kidneys, from 4 groups of mice at different ages. Compared with the conventional strategy, NP-PC-based proteomics significantly increased the number of identified protein groups in the heart (from 3007 to 3927; increase of 30.6%), liver (from 2982 to 4610; increase of 54.6%), spleen (from 5047 to 7351; increase of 45.7%), lungs (from 4984 to 6903; increase of 38.5%), and kidneys (from 3550 to 5739; increase of 61.7%), and we identified a total of 10 104 protein groups. The overall data indicated that 3-week-old mice showed more differences compared with the other three age groups. The proteins of amino acid-related metabolism were increased in aged mice compared with those in the 3-week-old mice. Protein-related infections were increased in the spleen of the aged mice. Interestingly, the spliceosome-related pathway significantly changed from youth to elders in the liver, spleen, and lungs, indicating the vital role of the spliceosome during the aging process. Our established aging mouse organ proteome atlas provides comprehensive insights into understanding the aging process, and it may help in prevention and treatment of age-related diseases.


Subject(s)
Aging , Nanoparticles , Protein Corona , Proteome , Proteomics , Animals , Mice , Aging/metabolism , Proteome/analysis , Proteome/metabolism , Nanoparticles/chemistry , Protein Corona/chemistry , Protein Corona/metabolism , Mice, Inbred C57BL , Kidney/metabolism , Kidney/chemistry , Male , Liver/metabolism , Liver/chemistry
11.
Small ; 20(26): e2305684, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38247186

ABSTRACT

Understanding the conformation of proteins in the nanoparticle corona has important implications in how organisms respond to nanoparticle-based drugs. These proteins coat the nanoparticle surface, and their properties will influence the nanoparticle's interaction with cell targets and the immune system. While some coronas are thought to be disordered, two key unanswered questions are the degree of disorder and solvent accessibility. Here, a model is developed for protein corona disorder in polystyrene nanoparticles of varying size. For two different proteins, it is found that binding affinity decreases as nanoparticle size increases. The stoichiometry of binding, along with changes in the hydrodynamic size, supports a highly solvated, disordered protein corona anchored at a small number of attachment sites. The scaling of the stoichiometry versus nanoparticle size is consistent with disordered polymer dimensions. Moreover, it is found that proteins are destabilized less in the presence of larger nanoparticles, and hydrophobic exposure decreases at lower curvatures. The observations hold for proteins on flat polystyrene surfaces, which have the lowest hydrophobic exposure. The model provides an explanation for previous observations of increased amyloid fibrillation rates in the presence of larger nanoparticles, and it may rationalize how cell receptors can recognize protein disorder in therapeutic nanoparticles.


Subject(s)
Nanoparticles , Polystyrenes , Protein Binding , Protein Corona , Polystyrenes/chemistry , Nanoparticles/chemistry , Protein Corona/chemistry , Solvents/chemistry , Hydrophobic and Hydrophilic Interactions , Particle Size
12.
Small ; 20(15): e2306474, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38085683

ABSTRACT

Gold nanoparticles (GNPs) are largely employed in diagnostics/biosensors and are among the most investigated nanomaterials in biology/medicine. However, few GNP-based nanoformulations have received FDA approval to date, and promising in vitro studies have failed to translate to in vivo efficacy. One key factor is that biological fluids contain high concentrations of proteins, lipids, sugars, and metabolites, which can adsorb/interact with the GNP's surface, forming a layer called biomolecular corona (BMC). The BMC can mask prepared functionalities and target moieties, creating new surface chemistry and determining GNPs' biological fate. Here, the current knowledge is summarized on GNP-BMCs, analyzing the factors driving these interactions and the biological consequences. A partial fingerprint of GNP-BMC analyzing common patterns of composition in the literature is extrapolated. However, a red flag is also risen concerning the current lack of data availability and regulated form of knowledge on BMC. Nanomedicine is still in its infancy, and relying on recently developed analytical and informatic tools offers an unprecedented opportunity to make a leap forward. However, a restart through robust shared protocols and data sharing is necessary to obtain "stronger roots". This will create a path to exploiting BMC for human benefit, promoting the clinical translation of biomedical nanotools.


Subject(s)
Metal Nanoparticles , Nanoparticles , Protein Corona , Humans , Metal Nanoparticles/chemistry , Gold/chemistry , Protein Corona/chemistry , Nanoparticles/chemistry , Proteins , Nanomedicine
13.
Small ; 20(10): e2306168, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37880910

ABSTRACT

Coronary artery disease (CAD) is the most common type of heart disease and represents the leading cause of death in both men and women worldwide. Early detection of CAD is crucial for decreasing mortality, prolonging survival, and improving patient quality of life. Herein, a non-invasive is described, nanoparticle-based diagnostic technology which takes advantages of proteomic changes in the nano-bio interface for CAD detection. Nanoparticles (NPs) exposed to biological fluids adsorb on their surface a layer of proteins, the "protein corona" (PC). Pathological changes that alter the plasma proteome can directly result in changes in the PC. By forming disease-specific PCs on six NPs with varying physicochemical properties, a PC-based sensor array is developed for detection of CAD using specific PC pattern recognition. While the PC of a single NP may not provide the required specificity, it is reasoned that multivariate PCs across NPs with different surface chemistries, can provide the desirable information to selectively discriminate the condition under investigation. The results suggest that such an approach can detect CAD with an accuracy of 92.84%, a sensitivity of 87.5%, and a specificity of 82.5%. These new findings demonstrate the potential of PC-based sensor array detection systems for clinical use.


Subject(s)
Coronary Artery Disease , Nanoparticles , Protein Corona , Female , Humans , Protein Corona/chemistry , Coronary Artery Disease/diagnosis , Proteomics , Quality of Life , Nanoparticles/chemistry , Proteome
14.
Small ; 20(34): e2311115, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38556634

ABSTRACT

Engineering of catalytically active inorganic nanomaterials holds promising prospects for biomedicine. Catalytically active metal oxides show applications in enhancing wound healing but have also been employed to induce cell death in photodynamic or radiation therapy. Upon introduction into a biological system, nanomaterials are exposed to complex fluids, causing interaction and adsorption of ions and proteins. While protein corona formation on nanomaterials is acknowledged, its modulation of nanomaterial catalytic efficacy is less understood. In this study, proteomic analyses and nano-analytic methodologies quantify and characterize adsorbed proteins, correlating this protein layer with metal oxide catalytic activity in vitro and in vivo. The protein corona comprises up to 280 different proteins, constituting up to 38% by weight. Enhanced complement factors and other opsonins on nanocatalyst surfaces lead to their uptake into macrophages when applied topically, localizing >99% of the nanomaterials in tissue-resident macrophages. Initially, the formation of the protein corona significantly reduces the nanocatalysts' activity, but this activity can be partially recovered in endosomal conditions due to the proteolytic degradation of the corona. Overall, the research reveals the complex relationship between physisorbed proteins and the catalytic characteristics of specific metal oxide nanoparticles, providing design parameters for optimizing nanocatalysts in complex biological environments.


Subject(s)
Oxides , Protein Corona , Oxides/chemistry , Animals , Catalysis , Protein Corona/chemistry , Protein Corona/metabolism , Protein Aggregates , Macrophages/metabolism , Mice , Metals/chemistry , Humans , RAW 264.7 Cells
15.
Small ; 20(38): e2309616, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38564782

ABSTRACT

Radiolabeling and nuclear imaging techniques are used to investigate the biodistribution patterns of the soft and hard protein corona around poly (lactic-co-glycolic acid) nanoparticles (PLGA NPs) after administration to healthy mice. Soft and hard protein coronas of 131I-labeled BSA or 131I-labeled serum are formed on PLGA NPs functionalized with either polyehtylenimine (PEI) or bovine serum albumin (BSA). The exchangeability of hard and soft corona is assessed in vitro by gamma counting exposing PLGA NPs with corona to non-labeled BSA, serum, or simulated body fluid. PEI PLGA NPs form larger and more stable coronas than BSA PLGA NPs. Soft coronas are more exchangeable than hard ones. The in vivo fate of PEI PLGA NPs coated with preformed 18F-labeled BSA hard and soft coronas is assessed by positron emission tomography (PET) following intravenous administration. While the soft corona shows a biodistribution similar to free 18F BSA with high activity in blood and kidney, the hard corona follows patterns characteristic of nanoparticles, accumulating in the lungs, liver, and spleen. These results show that in vivo fates of soft and hard corona are different, and that soft corona is more easily exchanged with proteins from the body, while hard corona is largely retained on the nanoparticle surface.


Subject(s)
Nanoparticles , Positron-Emission Tomography , Protein Corona , Serum Albumin, Bovine , Animals , Protein Corona/chemistry , Positron-Emission Tomography/methods , Nanoparticles/chemistry , Mice , Tissue Distribution , Serum Albumin, Bovine/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Cattle , Models, Animal
16.
Chembiochem ; 25(13): e202400188, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38743506

ABSTRACT

Plastics, omnipresent in the environment, have become a global concern due to their durability and limited biodegradability, especially in the form of microparticles and nanoparticles. Polystyrene (PS), a key plastic type, is susceptible to fragmentation and surface alterations induced by environmental factors or industrial processes. With widespread human exposure through pollution and diverse industrial applications, understanding the physiological impact of PS, particularly in nanoparticle form (PS-NPs), is crucial. This study focuses on the interaction of PS-NPs with model blood proteins, emphasising the formation of a protein corona, and explores the subsequent contact with platelet membrane mimetics using experimental and theoretical approaches. The investigation involves αIIbß3-expressing cells and biomimetic membranes, enabling real-time and label-free nanoscale precision. By employing quartz-crystal microbalance with dissipation monitoring studies, the concentration-dependent cytotoxic effects of differently functionalised ~210 nm PS-NPs on HEK293 cells overexpressing αIIbß3 are evaluated in detail. The study unveils insights into the molecular details of PS-NP interaction with supported lipid bilayers, demonstrating that a protein corona formed in the presence of exemplary blood proteins offers protection against membrane damage, mitigating PS-NP cytotoxicity.


Subject(s)
Nanoparticles , Polystyrenes , Protein Corona , Humans , Polystyrenes/chemistry , Protein Corona/chemistry , Protein Corona/metabolism , Nanoparticles/chemistry , HEK293 Cells , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry
17.
Nat Chem Biol ; 18(9): 1023-1031, 2022 09.
Article in English | MEDLINE | ID: mdl-35953550

ABSTRACT

Nanotechnology provides platforms to deliver medical agents to specific cells. However, the nanoparticle's surface becomes covered with serum proteins in the blood after administration despite engineering efforts to protect it with targeting or blocking molecules. Here, we developed a strategy to identify the main interactions between nanoparticle-adsorbed proteins and a cell by integrating mass spectrometry with pooled genome screens and Search Tool for the Retrieval of Interacting Genes analysis. We found that the low-density lipoprotein (LDL) receptor was responsible for approximately 75% of serum-coated gold nanoparticle uptake in U-87 MG cells. Apolipoprotein B and complement C8 proteins on the nanoparticle mediated uptake through the LDL receptor. In vivo, nanoparticle accumulation correlated with LDL receptor expression in the organs of mice. A detailed understanding of how adsorbed serum proteins bind to cell receptors will lay the groundwork for controlling the delivery of nanoparticles at the molecular level to diseased tissues for therapeutic and diagnostic applications.


Subject(s)
Metal Nanoparticles , Protein Corona , Animals , Blood Proteins , Gold , Mice , Protein Corona/chemistry , Protein Corona/metabolism , Receptors, Cell Surface , Receptors, LDL/genetics
18.
Mol Pharm ; 21(5): 2272-2283, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38607681

ABSTRACT

Over the years, there has been significant interest in PEGylated lipid-based nanocarriers within the drug delivery field. The inevitable interplay between the nanocarriers and plasma protein plays a pivotal role in their in vivo biological fate. Understanding the factors influencing lipid-based nanocarrier and protein corona interactions is of paramount importance in the design and clinical translation of these nanocarriers. Herein, discoid-shaped lipid nanodiscs (sNDs) composed of different phospholipids with varied lipid tails and head groups were fabricated. We investigated the impact of phospholipid components on the interaction between sNDs and serum proteins, particle stability, and biodistribution. The results showed that all of these lipid nanodiscs remained stable over a 15 day storage period, while their stability in the blood serum demonstrated significant differences. The sND composed of POPG exhibited the least stability due to its potent complement activation capability, resulting in rapid blood clearance. Furthermore, a negative correlation between the complement activation capability and serum stability was identified. Pharmacokinetic and biodistribution experiments indicated that phospholipid composition did not influence the capability of sNDs to evade the accelerated blood clearance phenomenon. Complement deposition on the sND was inversely associated with the area under the curve. Additionally, all lipid nanodiscs exhibited dominant adsorption of apolipoprotein. Remarkably, the POPC-based lipid nanodisc displayed a significantly higher deposition of apolipoprotein E, contributing to an obvious brain distribution, which provides a promising tool for brain-targeted drug delivery.


Subject(s)
Nanoparticles , Phospholipids , Protein Corona , Protein Corona/chemistry , Animals , Phospholipids/chemistry , Tissue Distribution , Mice , Nanoparticles/chemistry , Drug Carriers/chemistry , Nanostructures/chemistry , Male , Complement Activation/drug effects , Lipids/chemistry , Drug Delivery Systems/methods , Blood Proteins/metabolism , Blood Proteins/chemistry
19.
Langmuir ; 40(8): 4531-4543, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38357868

ABSTRACT

Conventional gold nanoparticles (Au NPs) have many limitations, such as aggregation and subsequent precipitation in the medium of high ionic strength and protein molecules. Furthermore, when exposed to biological fluids, nanoparticles form a protein corona, which controls different biological processes such as the circulation lifetime, drug release profile, biodistribution, and in vivo cellular distribution. These limitations reduce the functionality of Au NPs in targeted delivery, bioimaging, gene delivery, drug delivery, and other biomedical applications. To circumvent these problems, there are numerous attempts to design corona-free and stable nanoparticles. Here, we report for the first time that lipid corona (coating of lipid) formation on phenylalanine-functionalized Au NPs (AuPhe NPs) imparts excellent stability against the high ionic strength of bivalent metal ions, amino acids, and proteins of different charges as compared to bare nanoparticles. Moreover, this work is focused on the ability of lipid corona formation on AuPhe NPs to prevent protein adsorption in the presence of cell culture medium (CCM), oppositely charged protein (e.g., histone 3), and human serum albumin (HSA). The results demonstrate that the lipid corona successfully protects the AuPhe NPs from protein adsorption, leading to the development of corona-free character. This unique achievement has profound implications for enhancing the biomedical utility and safety of these nanoparticles.


Subject(s)
Metal Nanoparticles , Nanoparticles , Protein Corona , Humans , Gold/chemistry , Metal Nanoparticles/chemistry , Phenylalanine , Tissue Distribution , Nanoparticles/chemistry , Proteins , Protein Corona/chemistry , Lipids
20.
Langmuir ; 40(23): 11843-11857, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38787578

ABSTRACT

The interaction of nanoparticles (NPs) with biological environments triggers the formation of a protein corona (PC), which significantly influences their behavior in vivo. This review explores the evolving understanding of PC formation, focusing on the opportunity for decreasing or suppressing protein-NP interactions by macromolecular engineering of NP shells. The functionalization of NPs with a dense, hydrated polymer brush shell is a powerful strategy for imparting stealth properties in order to elude recognition by the immune system. While poly(ethylene glycol) (PEG) has been extensively used for this purpose, concerns regarding its stability and immunogenicity have prompted the exploration of alternative polymers. The stealth properties of brush shells can be enhanced by tailoring functionalities and structural parameters, including the molar mass, grafting density, and polymer topology. Determining correlations between these parameters and biopassivity has enabled us to obtain polymer-grafted NPs with high colloidal stability and prolonged circulation time in biological media.


Subject(s)
Nanoparticles , Nanoparticles/chemistry , Polymers/chemistry , Protein Corona/chemistry , Polyethylene Glycols/chemistry , Proteins/chemistry , Humans , Animals
SELECTION OF CITATIONS
SEARCH DETAIL