Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 469
Filtrar
1.
Biomed Mater ; 19(4)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38838693

RESUMO

To improve the translational and clinical applications of gold nanoparticles (GNPs) in medicine there is a need for better understanding of physicochemical properties of the nanoparticles in relation to the systemic parameters andin-vivoperformance. This review presents the influence of physicochemical properties (surface charges and size) and route of administration on the biodistribution of GNPs. The role of protein corona (PC) (a unique biological identifier) as a barrier to biodistribution of GNPs, and the advances in engineered GNPs towards improving biodistribution are presented. Proteins can easily adsorb on charged (anionic and cationic) functionalized GNPs in circulation and shape the dynamics of their biodistribution. Non-ionic coatings such as PEG experience accelerated blood clearance (ABC) due to immunogenic response. While zwitterionic coatings provide stealth effects to formation of PC on the GNPs. GNPs with sizes less than 50 nm were found to circulate to several organs while the route of administration of the GNPs determines the serum protein that adsorbs on the nanoparticles.


Assuntos
Ouro , Nanopartículas Metálicas , Tamanho da Partícula , Propriedades de Superfície , Animais , Humanos , Ouro/química , Ouro/farmacocinética , Nanopartículas Metálicas/química , Coroa de Proteína/química , Distribuição Tecidual
2.
Nat Commun ; 15(1): 5070, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38871729

RESUMO

In acute ischemic stroke, even when successful recanalization is obtained, downstream microcirculation may still be obstructed by microvascular thrombosis, which is associated with compromised brain reperfusion and cognitive decline. Identifying these microthrombi through non-invasive methods remains challenging. We developed the PHySIOMIC (Polydopamine Hybridized Self-assembled Iron Oxide Mussel Inspired Clusters), a MRI-based contrast agent that unmasks these microthrombi. In a mouse model of thromboembolic ischemic stroke, our findings demonstrate that the PHySIOMIC generate a distinct hypointense signal on T2*-weighted MRI in the presence of microthrombi, that correlates with the lesion areas observed 24 hours post-stroke. Our microfluidic studies reveal the role of fibrinogen in the protein corona for the thrombosis targeting properties. Finally, we observe the biodegradation and biocompatibility of these particles. This work demonstrates that the PHySIOMIC particles offer an innovative and valuable tool for non-invasive in vivo diagnosis and monitoring of microthrombi, using MRI during ischemic stroke.


Assuntos
Meios de Contraste , Modelos Animais de Doenças , Compostos Férricos , Indóis , Imageamento por Ressonância Magnética , Polímeros , Trombose , Animais , Polímeros/química , Imageamento por Ressonância Magnética/métodos , Indóis/química , Camundongos , Meios de Contraste/química , Compostos Férricos/química , Trombose/diagnóstico por imagem , Masculino , Acidente Vascular Cerebral/diagnóstico por imagem , Humanos , Fibrinogênio/metabolismo , AVC Isquêmico/diagnóstico por imagem , Camundongos Endogâmicos C57BL , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/patologia
3.
J Am Chem Soc ; 146(22): 15096-15107, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38773940

RESUMO

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.


Assuntos
Materiais Biomiméticos , Nanopartículas , Peptídeos , RNA Interferente Pequeno , Albumina Sérica Humana , Humanos , RNA Interferente Pequeno/química , Peptídeos/química , Materiais Biomiméticos/química , Nanopartículas/química , Albumina Sérica Humana/química , Engenharia de Proteínas , Apolipoproteína A-I/química , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Coroa de Proteína/química , Biomimética/métodos
4.
Biomater Sci ; 12(13): 3411-3422, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38809118

RESUMO

Bacteria have evolved survival mechanisms that enable them to live within host cells, triggering persistent intracellular infections that present significant clinical challenges due to the inability for conventional antibiotics to permeate cell membranes. In recent years, antibiotic nanocarriers or 'nanoantibiotics' have presented a promising strategy for overcoming intracellular infections by facilitating cellular uptake of antibiotics, thus improving targeting to the bacteria. However, prior to reaching host cells, nanocarriers experience interactions with proteins that form a corona and alter their physiological response. The influence of this protein corona on the cellular uptake, drug release and efficacy of nanoantibiotics for intracellular infections is poorly understood and commonly overlooked in preclinical studies. In this study, protein corona influence on cellular uptake was investigated for two nanoparticles; liposomes and cubosomes in macrophage and epithelial cells that are commonly infected with pathogens. Studies were conducted in presence of fetal bovine serum (FBS) to form a biologically relevant protein corona in an in vitro setting. Protein corona impact on cellular uptake was shown to be nanoparticle-dependent, where reduced internalization was observed for liposomes, the opposite was observed for cubosomes. Subsequently, vancomycin-loaded cubosomes were explored for their drug delivery performance against intracellular small colony variants of Staphylococcus aureus. We demonstrated improved bacterial killing in macrophages, with greater reduction in bacterial viability upon internalization of cubosomes mediated by the protein corona. However, no differences in efficacy were observed in epithelial cells. Thus, this study provides insights and evidence to the role of protein corona in modulating the performance of nanoparticles in a dynamic manner; these findings will facilitate improved understanding and translation of future investigations from in vitro to in vivo.


Assuntos
Antibacterianos , Lipossomos , Nanopartículas , Coroa de Proteína , Staphylococcus aureus , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/administração & dosagem , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Animais , Humanos , Lipossomos/química , Nanopartículas/química , Vancomicina/farmacologia , Vancomicina/química , Vancomicina/administração & dosagem , Camundongos , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Células RAW 264.7 , Testes de Sensibilidade Microbiana , Lipídeos/química , Portadores de Fármacos/química
5.
J Mater Chem B ; 12(23): 5573-5588, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38757190

RESUMO

Lipid nanoparticles (LNPs) are commonly employed for drug delivery owing to their considerable drug-loading capacity, low toxicity, and excellent biocompatibility. Nevertheless, the formation of protein corona (PC) on their surfaces significantly influences the drug's in vivo fate (such as absorption, distribution, metabolism, and elimination) upon administration. PC denotes the phenomenon wherein one or multiple strata of proteins adhere to the external interface of nanoparticles (NPs) or microparticles within the biological milieu, encompassing ex vivo fluids (e.g., serum-containing culture media) and in vivo fluids (such as blood and tissue fluids). Hence, it is essential to claim the PC formation behaviors and mechanisms on the surface of LNPs. This overview provided a comprehensive examination of crucial aspects related to such issues, encompassing time evolution, controllability, and their subsequent impacts on LNPs. Classical studies of PC generation on the surface of LNPs were additionally integrated, and its decisive role in shaping the in vivo fate of LNPs was explored. The mechanisms underlying PC formation, including the adsorption theory and alteration theory, were introduced to delve into the formation process. Subsequently, the existing experimental outcomes were synthesized to offer insights into the research and application facets of PC, and it was concluded that the manipulation of PC held substantial promise in the realm of targeted delivery.


Assuntos
Lipídeos , Nanopartículas , Coroa de Proteína , Coroa de Proteína/química , Nanopartículas/química , Humanos , Lipídeos/química , Animais , Propriedades de Superfície , Lipossomos
6.
Langmuir ; 40(23): 11843-11857, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38787578

RESUMO

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.


Assuntos
Nanopartículas , Nanopartículas/química , Polímeros/química , Coroa de Proteína/química , Polietilenoglicóis/química , Proteínas/química , Humanos , Animais
7.
Nanoscale Horiz ; 9(7): 1070-1071, 2024 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-38695198

RESUMO

This article highlights the recent work of Castagnola, Armirotti, et al. (Nanoscale Horiz., 2024, https://doi.org/10.1039/D3NH00510K) on demonstrating that the widespread use of 10% fetal bovine serum in an in vitro assay cannot recapitulate the complexity of in vivo systemic administration.


Assuntos
Nanoestruturas , Coroa de Proteína , Nanoestruturas/química , Humanos , Coroa de Proteína/química , Animais , Bovinos
8.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1448-1468, 2024 May 25.
Artigo em Chinês | MEDLINE | ID: mdl-38783808

RESUMO

Nanoparticles, as a novel material, have a wide range of applications in the food and biomedical fields. Nanoparticles spontaneously adsorb proteins in the biological environment, and tens or even hundreds of proteins can form protein corona on the surface of nanoparticles. The formation of protein corona on the surface of nanoparticles is one of the key factors affecting the stability, biocompatibility, targeting, and drug release properties of nanoparticles. The formation mechanism of protein corona is affected by a variety of factors, including the surface chemical properties, sizes, and shapes of nanoparticles and the types, concentrations, and pH of proteins. Studies have shown that the protein structure is associated with protein distribution on the nanoparticle surface, while the protein conformation affects the binding mode and stability of the protein on the nanoparticle surface. Since the mechanism of the formation of protein corona on the surface of nanoparticles is complex, the roles of multiple factors need to be considered comprehensively. Understanding the mechanisms and influencing factors of the formation of protein corona will help us to understand the process of protein corona formation and control the formation of protein corona for specific needs. In this paper, we summarize the recent studies on the mechanisms and influencing factors of the formation of protein corona on the surface of nanoparticles, with a view to providing a theoretical basis for in-depth research on protein corona.


Assuntos
Nanopartículas , Coroa de Proteína , Propriedades de Superfície , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Nanopartículas/química , Adsorção , Conformação Proteica , Humanos
9.
ACS Appl Mater Interfaces ; 16(20): 25977-25993, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38741563

RESUMO

Environmental pollution with plastic polymers has become a global problem, leaving no continent and habitat unaffected. Plastic waste is broken down into smaller parts by environmental factors, which generate micro- and nanoplastic particles (MNPPs), ultimately ending up in the human food chain. Before entering the human body, MNPPs make their first contact with saliva in the human mouth. However, it is unknown what proteins attach to plastic particles and whether such protein corona formation is affected by the particle's biophysical properties. To this end, we employed polystyrene MNPPs of two different sizes and three different charges and incubated them individually with saliva donated by healthy human volunteers. Particle zeta potential and size analyses were performed using dynamic light scattering complemented by nanoliquid chromatography high-resolution mass spectrometry (nLC/HRMS) to qualitatively and quantitatively reveal the protein soft and hard corona for each particle type. Notably, protein profiles and relative quantities were dictated by plastic particle size and charge, which in turn affected their hydrodynamic size, polydispersity, and zeta potential. Strikingly, we provide evidence of the latter to be dynamic processes depending on exposure times. Smaller particles seemed to be more reactive with the surrounding proteins, and cultures of the particles with five different cell lines (HeLa, HEK293, A549, HepG2, and HaCaT) indicated protein corona effects on cellular metabolic activity and genotoxicity. In summary, our data suggest nanoplastic size and surface chemistry dictate the decoration by human saliva proteins, with important implications for MNPP uptake in humans.


Assuntos
Tamanho da Partícula , Poliestirenos , Saliva , Proteínas e Peptídeos Salivares , Propriedades de Superfície , Humanos , Saliva/química , Saliva/metabolismo , Proteínas e Peptídeos Salivares/química , Proteínas e Peptídeos Salivares/metabolismo , Poliestirenos/química , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Nanopartículas/química , Microplásticos/química
10.
Nat Commun ; 15(1): 4267, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769317

RESUMO

The membrane-fusion-based internalization without lysosomal entrapment is advantageous for intracellular delivery over endocytosis. However, protein corona formed on the membrane-fusogenic liposome surface converts its membrane-fusion performance to lysosome-dependent endocytosis, causing poorer delivery efficiency in biological conditions. Herein, we develop an antifouling membrane-fusogenic liposome for effective intracellular delivery in vivo. Leveraging specific lipid composition at an optimized ratio, such antifouling membrane-fusogenic liposome facilitates fusion capacity even in protein-rich conditions, attributed to the copious zwitterionic phosphorylcholine groups for protein-adsorption resistance. Consequently, the antifouling membrane-fusogenic liposome demonstrates robust membrane-fusion-mediated delivery in the medium with up to 38% fetal bovine serum, outclassing two traditional membrane-fusogenic liposomes effective at 4% and 6% concentrations. When injected into mice, antifouling membrane-fusogenic liposomes can keep their membrane-fusion-transportation behaviors, thereby achieving efficient luciferase transfection and enhancing gene-editing-mediated viral inhibition. This study provides a promising tool for effective intracellular delivery under complex physiological environments, enlightening future nanomedicine design.


Assuntos
Lipossomos , Fusão de Membrana , Lipossomos/metabolismo , Animais , Camundongos , Humanos , Endocitose , Transfecção , Edição de Genes/métodos , Coroa de Proteína/metabolismo , Coroa de Proteína/química , Incrustação Biológica/prevenção & controle , Feminino , Lipídeos/química
11.
Talanta ; 275: 126172, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38692050

RESUMO

Nanoparticles (NPs) could be uptake orally and exposed to digestive tract through various sources such as particulate pollutant, nanomedicine and food additive. Inflammatory bowel disease (IBD), as a global disease, induced disruption of the intestinal mucosal barrier and thus altered in vivo distribution of NPs as a possible consequence. However, related information was relatively scarce. Herein, in vivo distribution of typical silica (SiO2) and titania (TiO2) NPs was investigated in healthy and IBD models at cell and animal levels via a surface-enhanced Raman scattering (SERS) tag labeling technique. The labeled NPs were composed of gold SERS tag core and SiO2 (or TiO2) shell, demonstrating sensitive and characteristic SERS signals ideal to trace the NPs in vivo. Cell SERS mapping revealed that protein corona from IBD intestinal fluid decreased uptake of NPs by lipopolysaccharide-induced RAW264.7 cells compared with normal intestinal fluid protein corona. SERS signal detection combined with inductively coupled plasma mass spectrometry (ICP-MS) analysis of mouse tissues (heart, liver, spleen, lung and kidney) indicated that both NPs tended to accumulate in lung specifically after oral administration for IBD mouse (6 out of 20 mice for SiO2 and 4 out of 16 mice for TiO2 were detected in lung). Comparatively, no NP signals were detected in all tissues from healthy mice. These findings suggested that there might be a greater risk associated with the oral uptake of NPs in IBD patients due to altered in vivo distribution of NPs.


Assuntos
Doenças Inflamatórias Intestinais , Dióxido de Silício , Análise Espectral Raman , Titânio , Animais , Análise Espectral Raman/métodos , Camundongos , Titânio/química , Dióxido de Silício/química , Células RAW 264.7 , Doenças Inflamatórias Intestinais/metabolismo , Administração Oral , Nanopartículas/química , Distribuição Tecidual , Nanopartículas Metálicas/química , Ouro/química , Masculino , Coroa de Proteína/química , Coroa de Proteína/análise , Coroa de Proteína/metabolismo
12.
Nanoscale Horiz ; 9(5): 799-816, 2024 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-38563642

RESUMO

The biological fate of nanomaterials (NMs) is driven by specific interactions through which biomolecules, naturally adhering onto their surface, engage with cell membrane receptors and intracellular organelles. The molecular composition of this layer, called the biomolecular corona (BMC), depends on both the physical-chemical features of the NMs and the biological media in which the NMs are dispersed and cells grow. In this work, we demonstrate that the widespread use of 10% fetal bovine serum in an in vitro assay cannot recapitulate the complexity of in vivo systemic administration, with NMs being transported by the blood. For this purpose, we undertook a comparative journey involving proteomics, lipidomics, high throughput multiparametric in vitro screening, and single molecular feature analysis to investigate the molecular details behind this in vivo/in vitro bias. Our work indirectly highlights the need to introduce novel, more physiological-like media closer in composition to human plasma to produce realistic in vitro screening data for NMs. We also aim to set the basis to reduce this in vitro-in vivo mismatch, which currently limits the formulation of NMs for clinical settings.


Assuntos
Nanoestruturas , Coroa de Proteína , Humanos , Nanoestruturas/química , Coroa de Proteína/química , Animais , Proteômica/métodos , Lipidômica/métodos , Bovinos
13.
Drug Deliv Transl Res ; 14(8): 2188-2202, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38578378

RESUMO

Nanoparticles (NPs) engineered as drug delivery systems continue to make breakthroughs as they offer numerous advantages over free therapeutics. However, the poor understanding of the interplay between the NPs and biomolecules, especially blood proteins, obstructs NP translation to clinics. Nano-bio interactions determine the NPs' in vivo fate, efficacy and immunotoxicity, potentially altering protein function. To fulfill the growing need to investigate nano-bio interactions, this study provides a systematic understanding of two key aspects: (i) protein corona (PC) formation and (ii) NP-induced modifications on protein's structure and stability. A methodology was developed by combining orthogonal techniques to analyze both quantitative and qualitative aspects of nano-bio interactions, using human serum albumin (HSA) as a model protein. Protein quantification via liquid chromatography-mass spectrometry, and capillary zone electrophoresis (CZE) clarified adsorbed protein quantity and stability. CZE further unveiled qualitative insights into HSA forms (native, glycated HSA and cysteinylated), while synchrotron radiation circular dichroism enabled analyzing HSA's secondary structure and thermal stability. Comparative investigations of NP cores (organic vs. hybrid), and shells (with or without polyethylene glycol (PEG)) revealed pivotal factors influencing nano-bio interactions. Polymeric NPs based on poly(lactic-co-glycolic acid) (PLGA) and hybrid NPs based on metal-organic frameworks (nanoMOFs) presented distinct HSA adsorption profiles. PLGA NPs had protein-repelling properties while inducing structural modifications on HSA. In contrast, HSA exhibited a high affinity for nanoMOFs forming a PC altering thereby the protein structure. A shielding effect was gained through PEGylation for both types of NPs, avoiding the PC formation as well as the alteration of unbound HSA structure.


Assuntos
Nanopartículas , Albumina Sérica Humana , Humanos , Nanopartículas/química , Albumina Sérica Humana/química , Coroa de Proteína/química , Estabilidade Proteica , Polímeros/química , Eletroforese Capilar , Dicroísmo Circular
14.
Water Res ; 256: 121574, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38593606

RESUMO

The ecological risk of combined pollution from microplastics (MPs) and associated contaminants usually depends on their interactions and environmental behavior, which was also disturbed by varying surface modifications of MPs. In this study, the significance of surface functionalization and protein-corona on the cotransport of nanoplastics (NPs; 100 nm) and the related additive bisphenol AF (BPAF) was examined in simulated unsaturated hyporheic zone (quartz sand; 250-425 µm). The electronegative bovine serum albumin (BSA) and electropositive trypsin were chosen as representative proteins, while pristine (PNPs), amino-modified (ANPs), and carboxyl-modified NPs (CNPs) were representative NPs with different charges. The presence of BPAF inhibited the mobility of PNPs/CNPs, but enhanced the release of ANPs in hyporheic zone, which was mainly related to their hydrophobicity changes and electrostatic interactions. Meanwhile, the NPs with high mobility and strong affinity to BPAF became effective carriers, promoting the cotransport of BPAF by 16.4 %-26.4 %. The formation of protein-coronas altered the mobility of NPs alone and their cotransport with BPAF, exhibiting a coupling effect with functional groups. BSA-corona promoted the transport of PNPs/CNPs, but this promoting effect was weakened by the presence of BPAF via increasing particle aggregation and hydrophobicity. Inversely, trypsin-corona aggravated the deposition of PNPs/CNPs, but competition deposition sites and increased energy barrier caused by coexisting BPAF reversed this effect, facilitating the cotransport of trypsin-PNPs/CNPs in hyporheic zone. However, BPAF and protein-coronas synergistically promoted the mobility of ANPs, owing to competition deposition sites and decreased electrostatic attraction. Although all of the NPs with two protein-coronas reduced dissolved BPAF in the effluents via providing deposition sites, the cotransport of total BPAF was improved by the NPs with high mobility (BSA-PNPs/CNPs) or high affinity to BPAF (BSA/trypsin-ANPs). However, the trypsin-PNPs/CNPs inhibited the transport of BPAF due to their weak mobility and adsorption with BPAF. The results provide new insights into the role of varying surface modifications on NPs in the vertical cotransport of NPs and associated contaminants in unsaturated hyporheic zone.


Assuntos
Plásticos , Plásticos/química , Coroa de Proteína/química , Microplásticos/química , Poluentes Químicos da Água/química , Fenóis/química , Soroalbumina Bovina/química , Compostos Benzidrílicos/química , Nanopartículas/química
15.
Biomed Pharmacother ; 175: 116627, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38653112

RESUMO

Nanoparticles (NPs) serve as versatile delivery systems for anticancer, antibacterial, and antioxidant agents. The manipulation of protein-NP interactions within biological systems is crucial to the application of NPs in drug delivery and cancer nanotherapeutics. The protein corona (PC) that forms on the surface of NPs is the interface between biomacromolecules and NPs and significantly influences their pharmacokinetics and pharmacodynamics. Upon encountering proteins, NPs undergo surface alterations that facilitate their clearance from circulation by the mononuclear phagocytic system (MPS). PC behavior depends largely on the biological microenvironment and the physicochemical properties of the NPs. This review describes various strategies employed to engineer PC compositions on NP surfaces. The effects of NP characteristics such as size, shape, surface modification and protein precoating on PC performance were explored. In addition, this study addresses these challenges and guides the future directions of this evolving field.


Assuntos
Nanopartículas , Coroa de Proteína , Coroa de Proteína/metabolismo , Coroa de Proteína/química , Humanos , Animais , Sistemas de Liberação de Medicamentos/métodos , Engenharia de Proteínas/métodos , Propriedades de Superfície
16.
Nanoscale ; 16(19): 9348-9360, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38651870

RESUMO

Understanding nanoparticle-cell interaction is essential for advancing research in nanomedicine and nanotoxicology. Apart from the transcytotic pathway mediated by cellular recognition and energetics, nanoparticles (including nanomedicines) may harness the paracellular route for their transport by inducing endothelial leakiness at cadherin junctions. This phenomenon, termed as NanoEL, is correlated with the physicochemical properties of the nanoparticles in close association with cellular signalling, membrane mechanics, as well as cytoskeletal remodelling. However, nanoparticles in biological systems are transformed by the ubiquitous protein corona and yet the potential effect of the protein corona on NanoEL remains unclear. Using confocal fluorescence microscopy, biolayer interferometry, transwell, toxicity, and molecular inhibition assays, complemented by molecular docking, here we reveal the minimal to significant effects of the anionic human serum albumin and fibrinogen, the charge neutral immunoglobulin G as well as the cationic lysozyme on negating gold nanoparticle-induced endothelial leakiness in vitro and in vivo. This study suggests that nanoparticle-cadherin interaction and hence the extent of NanoEL may be partially controlled by pre-exposing the nanoparticles to plasma proteins of specific charge and topology to facilitate their biomedical applications.


Assuntos
Caderinas , Fibrinogênio , Ouro , Nanopartículas Metálicas , Coroa de Proteína , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Humanos , Caderinas/metabolismo , Caderinas/química , Ouro/química , Nanopartículas Metálicas/química , Fibrinogênio/química , Fibrinogênio/metabolismo , Animais , Células Endoteliais da Veia Umbilical Humana , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Muramidase/química , Muramidase/metabolismo , Simulação de Acoplamento Molecular , Camundongos
17.
Mol Pharm ; 21(5): 2272-2283, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38607681

RESUMO

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.


Assuntos
Nanopartículas , Fosfolipídeos , Coroa de Proteína , Coroa de Proteína/química , Animais , Fosfolipídeos/química , Distribuição Tecidual , Camundongos , Nanopartículas/química , Portadores de Fármacos/química , Nanoestruturas/química , Masculino , Ativação do Complemento/efeitos dos fármacos , Lipídeos/química , Sistemas de Liberação de Medicamentos/métodos , Proteínas Sanguíneas/metabolismo , Proteínas Sanguíneas/química
18.
Int J Biol Macromol ; 267(Pt 2): 131546, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38614172

RESUMO

Chitosan-based nanoparticles inevitably adsorb numerous proteins in the bloodstream, forming a protein corona that significantly influences their functionality. This study employed a pre-coated protein corona using cyclic Arg-Gly-Asp peptide (cRGD)-modified bovine serum albumin (BcR) to confer tumor-targeting capabilities on siVEGF-loaded chitosan-based nanoparticles (CsR/siVEGF NPs) and actively manipulated the serum protein corona composition to enhance their anti-tumor angiogenesis. Consequently, BcR effectively binds to the nanoparticles' surface, generating nanocarriers of appropriate size and stability that enhance the inhibition of endothelial cell proliferation, migration, invasion, and tube formation, as well as suppress tumor proliferation and angiogenesis in tumor-bearing nude mice. Proteomic analysis indicated a significant enrichment of serotransferrin, albumin, and proteasome subunit alpha type-1 in the protein corona of BcR-precoated NPs formed in the serum of tumor-bearing nude mice. Additionally, there was a decrease in proteins associated with complement activation, immunoglobulins, blood coagulation, and acute-phase responses. This modification resulted in an enhanced impact on anti-tumor angiogenesis, along with a reduction in opsonization and inflammatory responses. Therefore, pre-coating of nanoparticles with a functionalized albumin corona to manipulate the composition of serum protein corona emerges as an innovative approach to improve the delivery effectiveness of chitosan-based carriers for siVEGF, targeting the inhibition of tumor angiogenesis.


Assuntos
Quitosana , Nanopartículas , Neovascularização Patológica , Coroa de Proteína , Soroalbumina Bovina , Quitosana/química , Animais , Nanopartículas/química , Camundongos , Humanos , Coroa de Proteína/química , Soroalbumina Bovina/química , Neovascularização Patológica/tratamento farmacológico , Camundongos Nus , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos/química , Bovinos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Movimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/química , Angiogênese
19.
Curr Opin Biotechnol ; 87: 103101, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38461749

RESUMO

Upon administration, nanomedicines adsorb a corona of endogenous biomolecules on their surface, which can affect nanomedicine interactions with cells, targeting, and efficacy. While strategies to reduce protein binding are available, the high selectivity of the adsorbed corona is enabling novel applications, such as for biomarker discovery and rare protein identification. Additionally, the adsorbed molecules can promote interactions with specific cell receptors, thus conferring the nanomedicine new endogenous targeting capabilities. This has been reported for Onpattro, a lipid nanoparticle targeting the hepatocytes via apolipoproteins in its corona. Recently, selective organ-targeting (SORT) nanoparticles have been proposed, which exploit corona-mediated interactions to deliver nanoparticles outside the liver. Strategies for corona seeding and corona engineering are emerging to increase the selectivity of similar endogenous targeting mechanisms.


Assuntos
Nanomedicina , Coroa de Proteína , Nanomedicina/métodos , Humanos , Coroa de Proteína/química , Coroa de Proteína/metabolismo , Nanopartículas/química , Animais
20.
Nat Nanotechnol ; 19(6): 846-855, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38366223

RESUMO

Extracellular vesicles (EVs) derived from mesenchymal stem cells are promising nanotherapeutics in liver diseases due to their regenerative and immunomodulatory properties. Nevertheless, a concern has been raised regarding the rapid clearance of exogenous EVs by phagocytic cells. Here we explore the impact of protein corona on EVs derived from two culturing conditions in which specific proteins acquired from media were simultaneously adsorbed on the EV surface. Additionally, by incubating EVs with serum, simulating protein corona formation upon systemic delivery, further resolved protein corona-EV complex patterns were investigated. Our findings reveal the potential influences of corona composition on EVs under in vitro conditions and their in vivo kinetics. Our data suggest that bound albumin creates an EV signature that can retarget EVs from hepatic macrophages. This results in markedly improved cellular uptake by hepatocytes, liver sinusoidal endothelial cells and hepatic stellate cells. This phenomenon can be applied as a camouflage strategy by precoating EVs with albumin to fabricate the albumin-enriched protein corona-EV complex, enhancing non-phagocytic uptake in the liver. This work addresses a critical challenge facing intravenously administered EVs for liver therapy by tailoring the protein corona-EV complex for liver cell targeting and immune evasion.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Coroa de Proteína , Vesículas Extracelulares/metabolismo , Coroa de Proteína/metabolismo , Coroa de Proteína/química , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Animais , Humanos , Camundongos , Hepatócitos/metabolismo , Hepatócitos/citologia , Fígado/metabolismo , Macrófagos/metabolismo , Macrófagos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...