Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Science ; 372(6537)2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33795432

RESUMEN

Multivalent display of receptor-engaging antibodies or ligands can enhance their activity. Instead of achieving multivalency by attachment to preexisting scaffolds, here we unite form and function by the computational design of nanocages in which one structural component is an antibody or Fc-ligand fusion and the second is a designed antibody-binding homo-oligomer that drives nanocage assembly. Structures of eight nanocages determined by electron microscopy spanning dihedral, tetrahedral, octahedral, and icosahedral architectures with 2, 6, 12, and 30 antibodies per nanocage, respectively, closely match the corresponding computational models. Antibody nanocages targeting cell surface receptors enhance signaling compared with free antibodies or Fc-fusions in death receptor 5 (DR5)-mediated apoptosis, angiopoietin-1 receptor (Tie2)-mediated angiogenesis, CD40 activation, and T cell proliferation. Nanocage assembly also increases severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudovirus neutralization by α-SARS-CoV-2 monoclonal antibodies and Fc-angiotensin-converting enzyme 2 (ACE2) fusion proteins.


Asunto(s)
Anticuerpos/química , Anticuerpos/inmunología , Nanoestructuras , Ingeniería de Proteínas , Transducción de Señal , Angiopoyetinas/química , Angiopoyetinas/inmunología , Angiopoyetinas/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Antígenos CD40/química , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Línea Celular Tumoral , Proliferación Celular , Simulación por Computador , Genes Sintéticos , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Activación de Linfocitos , Modelos Moleculares , Unión Proteica , Receptor TIE-2/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Linfocitos T/fisiología
2.
bioRxiv ; 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33299994

RESUMEN

Antibodies are widely used in biology and medicine, and there has been considerable interest in multivalent antibody formats to increase binding avidity and enhance signaling pathway agonism. However, there are currently no general approaches for forming precisely oriented antibody assemblies with controlled valency. We describe the computational design of two-component nanocages that overcome this limitation by uniting form and function. One structural component is any antibody or Fc fusion and the second is a designed Fc-binding homo-oligomer that drives nanocage assembly. Structures of 8 antibody nanocages determined by electron microscopy spanning dihedral, tetrahedral, octahedral, and icosahedral architectures with 2, 6, 12, and 30 antibodies per nanocage match the corresponding computational models. Antibody nanocages targeting cell-surface receptors enhance signaling compared to free antibodies or Fc-fusions in DR5-mediated apoptosis, Tie2-mediated angiogenesis, CD40 activation, and T cell proliferation; nanocage assembly also increases SARS-CoV-2 pseudovirus neutralization by α-SARS-CoV-2 monoclonal antibodies and Fc-ACE2 fusion proteins. We anticipate that the ability to assemble arbitrary antibodies without need for covalent modification into highly ordered assemblies with different geometries and valencies will have broad impact in biology and medicine.

3.
J Am Chem Soc ; 142(42): 17938-17943, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33022172

RESUMEN

A nanoparticle can hold multiple types of therapeutic and imaging agents for disease treatment and diagnosis. However, controlling the storage of molecules in nanoparticles is challenging, because nonspecific intermolecular interactions are used for encapsulation. Here, we used specific DNA interactions to store molecules in nanoparticles. We made nanoparticles containing DNA anchors to capture DNA-conjugated small molecules. By changing the sequences and stoichiometry of DNA anchors, we can control the amount and ratio of molecules with different chemical properties in the nanoparticles. We modified the cytotoxicity of our nanoparticles to cancer cells by changing the ratio of encapsulated drugs (mertansine and doxorubicin). Specifically controlling the storage of multiple types of molecules allows us to optimize the properties of combination drug and imaging nanoparticles.


Asunto(s)
ADN/química , Nanopartículas/química , Proteínas/química , Bibliotecas de Moléculas Pequeñas/química , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Células HeLa , Humanos , Maitansina/farmacología , Imagen Óptica , Tamaño de la Partícula , Propiedades de Superficie
4.
J Am Chem Soc ; 142(19): 8827-8836, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32293877

RESUMEN

Blood proteins adsorb onto the surface of nanoparticles after intravenous injection to form a protein corona. The underlying organization and binding function of these adsorbed proteins remain unclear. This can impact how the corona mediates cell and tissue interactions. Here, we investigated the function and structural organization of the protein corona using an immunoassay approach. We discovered that only 27% of the adsorbed proteins examined are functional for binding to their target protein. This is because the corona architecture is not a monolayer, but an assembly of proteins that are bound to each other. We further demonstrated that we can control the binding functionality of a protein by changing the organization of proteins in the assembly. We show that manipulation of the corona protein composition and assembly can influence their interactions with macrophage cells in culture. This study provides detailed functional and structural insights into the protein corona on nanomaterials and offers a new strategy to manipulate it for controlled interactions with the biological system.


Asunto(s)
Corona de Proteínas/química , Adsorción , Células Cultivadas , Humanos , Macrófagos/química , Nanoestructuras/química , Tamaño de la Partícula , Unión Proteica , Conformación Proteica , Propiedades de Superficie
5.
Nano Lett ; 19(10): 7226-7235, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31508968

RESUMEN

Lymph node follicles capture and retain antigens to induce germinal centers and long-lived humoral immunity. However, control over antigen retention has been limited. Here we discovered that antigen conjugated to nanoparticle carriers of different sizes impacts the intralymph node transport and specific cell interaction. We found that follicular dendritic cell (FDC) networks determine the intralymph node follicle fate of these nanoparticles by clearing smaller ones (5-15 nm) within 48 h and retaining larger ones (50-100 nm) for over 5 weeks. The 50-100 nm-sized nanoparticles had 175-fold more delivery of antigen at the FDC dendrites, 5-fold enhanced humoral immune responses of germinal center B cell formation, and 5-fold more antigen-specific antibody production over 5-15 nm nanoparticles. Our results show that we can tune humoral immunity by simply manipulating the carrier size design to produce effectiveness of vaccines.


Asunto(s)
Antígenos/inmunología , Inmunidad Humoral , Ganglios Linfáticos/inmunología , Nanoconjugados/química , Ovalbúmina/inmunología , Animales , Antígenos/administración & dosificación , Linfocitos B/inmunología , Células Dendríticas/inmunología , Centro Germinal/inmunología , Oro/química , Proteínas Inmovilizadas/inmunología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Tamaño de la Partícula , Vacunas/administración & dosificación , Vacunas/inmunología
6.
ACS Nano ; 13(7): 8023-8034, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31268684

RESUMEN

The surface of nanoparticles changes immediately after intravenous injection because blood proteins adsorb on the surface. How this interface changes during circulation and its impact on nanoparticle distribution within the body is not understood. Here, we developed a workflow to show that the evolution of proteins on nanoparticle surfaces predicts the biological fate of nanoparticles in vivo. This workflow involves extracting nanoparticles at multiple time points from circulation, isolating the proteins off the surface and performing proteomic mass spectrometry. The mass spectrometry protein library served as inputs, while blood clearance and organ accumulation were used as outputs to train a supervised deep neural network that predicts nanoparticle biological fate. In a double-blinded study, we tested the network by predicting nanoparticle spleen and liver accumulation with upward of 94% accuracy. Our neural network discovered that the mechanism of liver and spleen uptake is due to patterns of a multitude of nanoparticle surface adsorbed proteins. There are too many combinations to change these proteins manually using chemical or biological inhibitors to alter clearance. Therefore, we developed a technique that uses the host to act as a bioreactor to prepare nanoparticles with predictable clearance patterns that reduce liver and spleen uptake by 50% and 70%, respectively. These techniques provide opportunities to both predict nanoparticle behavior and also to engineer surface chemistries that are specifically designed by the body.


Asunto(s)
Proteínas Sanguíneas/química , Oro/química , Nanopartículas del Metal/química , Aprendizaje Automático Supervisado , Adsorción , Animales , Espectrometría de Masas , Imagen Óptica , Tamaño de la Partícula , Biblioteca de Péptidos , Proteómica , Ratas , Ratas Sprague-Dawley , Propiedades de Superficie
7.
Nano Lett ; 19(1): 116-123, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30525697

RESUMEN

Nanoparticles are engineered from materials such as metals, polymers, and different carbon allotropes that do not exist within the body. Exposure to these exogenous compounds raises concerns surrounding toxicity, inflammation, and immune activation. These responses could potentially be mitigated by synthesizing nanoparticles directly from molecules derived from the host. However, efforts to assemble patient-derived macromolecules into structures with the same degree of size and shape tunability as their exogenous counterparts remains a significant challenge. Here we solve this problem by creating a new class of size- and shape-tunable personalized protein nanoparticles (PNP) made entirely from patient-derived proteins. PNPs are built into different sizes and shapes with the same degree of tunability as gold nanoparticles. They are biodegradable and do not activate innate or adaptive immunity following single and repeated administrations in vivo. PNPs can be further modified with specific protein cargos that remain catalytically active even after intracellular delivery in vivo. Finally, we demonstrate that PNPs created from different human patients have unique molecular fingerprints encoded directly into the structure of the nanoparticle. This new class of personalized nanomaterial has the potential to revolutionize how we treat patients and can become an integral component in the diagnostic and therapeutic toolbox.


Asunto(s)
Nanopartículas del Metal/química , Nanoestructuras/química , Medicina de Precisión , Proteínas/química , Carbono/química , Oro/química , Humanos , Tamaño de la Partícula , Polímeros/química , Corona de Proteínas/química , Proteínas/síntesis química , Proteínas/genética
8.
Chem Commun (Camb) ; 51(14): 2756-67, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26829150

RESUMEN

Nanoparticles are suitable platforms for cancer targeting and diagnostic applications. Typically, less than 10% of all systemically administered nanoparticles accumulate in the tumour. Here we explore the interactions of blood components with nanoparticles and describe how these interactions influence solid tumour targeting. In the blood, serum proteins adsorb onto nanoparticles to form a protein corona in a manner dependent on nanoparticle physicochemical properties. These serum proteins can block nanoparticle tumour targeting ligands from binding to tumour cell receptors. Additionally, serum proteins can also encourage nanoparticle uptake by macrophages, which decreases nanoparticle availability in the blood and limits tumour accumulation. The formation of this protein corona will also increase the nanoparticle hydrodynamic size or induce aggregation, which makes nanoparticles too large to enter into the tumour through pores of the leaky vessels, and prevents their deep penetration into tumours for cell targeting. Recent studies have focused on developing new chemical strategies to reduce or eliminate serum protein adsorption, and rescue the targeting potential of nanoparticles to tumour cells. An in-depth and complete understanding of nanoparticle-blood interactions is key to designing nanoparticles with optimal physicochemical properties with high tumour accumulation. The purpose of this review article is to describe how the protein corona alters the targeting of nanoparticles to solid tumours and explains current solutions to solve this problem.


Asunto(s)
Proteínas Sanguíneas/química , Nanopartículas/química , Neoplasias/química , Adsorción , Relación Estructura-Actividad Cuantitativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...