Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Methods Mol Biol ; 2668: 301-311, 2023.
Artículo en Inglés | MEDLINE | ID: covidwho-2316082

RESUMEN

Extracellular vesicles (EVs) enable cell-to-cell communication and, by delivering antigens, can stimulate the immune response strongly. Approved in use SARS-CoV-2 vaccine, candidates immunize with the viral spike protein delivered via viral vectors, translated by injected mRNAs, or as a pure protein. Here, we outline a novel methodological approach for generating SARS-CoV-2 vaccine using exosome that delivers antigens from the SARS-CoV-2 structural proteins. Engineered EVs can be loaded with viral antigens, thus acting as antigens presenting EVs, eliciting strong and targeted CD8(+) T cell and B cell, offering a unique approach to vaccine development. Engineered EVs thus portray a safe, adaptable, and effective approach for a virus-free vaccine development.


Asunto(s)
COVID-19 , Exosomas , Vesículas Extracelulares , Humanos , Vacunas contra la COVID-19/metabolismo , Exosomas/metabolismo , SARS-CoV-2/genética , COVID-19/prevención & control , COVID-19/metabolismo , Vesículas Extracelulares/metabolismo , Antígenos/metabolismo , Proteínas Virales/metabolismo
2.
Front Immunol ; 13: 1107366, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-2244988

RESUMEN

Common flow cytometry-based methods used for functional assessment of antigen-specific T cells rely on de novo expression of intracellular cytokines or cell surface activation induced markers. They come with some limitations such as complex experimental setting, loss of cell viability and often high unspecific background which impairs assay sensitivity. We have previously shown that staining of activated ß2-integrins either with multimers of their ligand ICAM-1 or with a monoclonal antibody can serve as a functional marker detectable on T cells after minutes (CD8+) or few hours (CD4+) of activation. Here, we present a simple method for detection of activated ß2-integrins in combination with established cell surface activation induced markers. We observed that activated ß2-integrins were still detectable after 14 hours of stimulation, allowing their detection together with CD137 and CD154. Combinatorial gating of cells expressing activated ß2-integrins and CD137 or CD154 reduced background in unstimulated samples, increasing the signal-to-noise ratio and allowing improved assessment of low-frequency T cell responses. Extracellular staining of these markers highly correlated with production of intracellular cytokines IL-2, TNF or IFNγ in CD4+ and CD8+ T cells. As an exemplary application, SARS-CoV-2 spike-specific T cell responses were assessed in individuals after COVID-19 vaccination. This method should be useful for epitope discovery projects and for the simultaneous monitoring of low-frequency antigen-specific CD4+ and CD8+ T cell responses in various physiological situations.


Asunto(s)
Linfocitos T CD8-positivos , COVID-19 , Humanos , Linfocitos T CD4-Positivos , Integrinas/metabolismo , Vacunas contra la COVID-19/metabolismo , COVID-19/metabolismo , SARS-CoV-2 , Antígenos/metabolismo , Ligando de CD40 , Citocinas/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(34): e2207841119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: covidwho-1991768

RESUMEN

The targeted delivery of messenger RNA (mRNA) to desired organs remains a great challenge for in vivo applications of mRNA technology. For mRNA vaccines, the targeted delivery to the lymph node (LN) is predicted to reduce side effects and increase the immune response. In this study, we explored an endogenously LN-targeting lipid nanoparticle (LNP) without the modification of any active targeting ligands for developing an mRNA cancer vaccine. The LNP named 113-O12B showed increased and specific expression in the LN compared with LNP formulated with ALC-0315, a synthetic lipid used in the COVID-19 vaccine Comirnaty. The targeted delivery of mRNA to the LN increased the CD8+ T cell response to the encoded full-length ovalbumin (OVA) model antigen. As a result, the protective and therapeutic effect of the OVA-encoding mRNA vaccine on the OVA-antigen-bearing B16F10 melanoma model was also improved. Moreover, 113-O12B encapsulated with TRP-2 peptide (TRP2180-188)-encoding mRNA also exhibited excellent tumor inhibition, with the complete response of 40% in the regular B16F10 tumor model when combined with anti-programmed death-1 (PD-1) therapy, revealing broad application of 113-O12B from protein to peptide antigens. All the treated mice showed long-term immune memory, hindering the occurrence of tumor metastatic nodules in the lung in the rechallenging experiments that followed. The enhanced antitumor efficacy of the LN-targeting LNP system shows great potential as a universal platform for the next generation of mRNA vaccines.


Asunto(s)
Vacunas contra el Cáncer , Nanopartículas , Neoplasias , Vacunas de ARNm , Amino Alcoholes , Animales , Antígenos/metabolismo , Linfocitos T CD8-positivos , Vacunas contra el Cáncer/uso terapéutico , Decanoatos , Memoria Inmunológica , Liposomas , Ganglios Linfáticos , Ratones , Metástasis de la Neoplasia/prevención & control , Neoplasias/terapia , Ovalbúmina , Vacunas de ARNm/uso terapéutico
4.
J Vis Exp ; (185)2022 07 25.
Artículo en Inglés | MEDLINE | ID: covidwho-1988090

RESUMEN

Biomimetic nanoparticles obtained from bacteria or viruses have attracted substantial interest in vaccine research and development. Outer membrane vesicles (OMVs) are mainly secreted by gram-negative bacteria during average growth, with a nano-sized diameter and self-adjuvant activity, which may be ideal for vaccine delivery. OMVs have functioned as a multifaceted delivery system for proteins, nucleic acids, and small molecules. To take full advantage of the biological characteristics of OMVs, bioengineered Escherichia coli-derived OMVs were utilized as a carrier and SARS-CoV-2 receptor-binding domain (RBD) as an antigen to construct a "Plug-and-Display" vaccine platform. The SpyCatcher (SC) and SpyTag (ST) domains in Streptococcus pyogenes were applied to conjugate OMVs and RBD. The Cytolysin A (ClyA) gene was translated with the SC gene as a fusion protein after plasmid transfection, leaving a reactive site on the surface of the OMVs. After mixing RBD-ST in a conventional buffer system overnight, covalent binding was formed between the OMVs and RBD. Thus, a multivalent-displaying OMV vaccine was achieved. By replacing with diverse antigens, the OMVs vaccine platform can efficiently display a variety of heterogeneous antigens, thereby potentially rapidly preventing infectious disease epidemics. This protocol describes a precise method for constructing the OMV vaccine platform, including production, purification, bioconjugation, and characterization.


Asunto(s)
COVID-19 , Nanopartículas , Vacunas , Antígenos/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , SARS-CoV-2
5.
Front Immunol ; 12: 732298, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1506693

RESUMEN

Immune modulating therapies and vaccines are in high demand, not least to the recent global spread of SARS-CoV2. To achieve efficient activation of the immune system, professional antigen presenting cells have proven to be key coordinators of such responses. Especially targeted approaches, actively directing antigens to specialized dendritic cells, promise to be more effective and accompanied by reduced payload due to less off-target effects. Although antibody and glycan-based targeting of receptors on dendritic cells have been employed, these are often expensive and time-consuming to manufacture or lack sufficient specificity. Thus, we applied a small-molecule ligand that specifically binds Langerin, a hallmark receptor on Langerhans cells, conjugated to a model protein antigen. Via microneedle injection, this construct was intradermally administered into intact human skin explants, selectively loading Langerhans cells in the epidermis. The ligand-mediated cellular uptake outpaces protein degradation resulting in intact antigen delivery. Due to the pivotal role of Langerhans cells in induction of immune responses, this approach of antigen-targeting of tissue-resident immune cells offers a novel way to deliver highly effective vaccines with minimally invasive administration.


Asunto(s)
Antígenos CD/metabolismo , Antígenos/administración & dosificación , Proteínas Fluorescentes Verdes/administración & dosificación , Células de Langerhans/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Animales , Antígenos/inmunología , Antígenos/metabolismo , Células COS , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Inyecciones Intradérmicas , Células de Langerhans/inmunología , Ligandos , Miniaturización , Nanomedicina , Agujas , Unión Proteica , Transporte de Proteínas , Proteolisis , Células THP-1 , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/metabolismo
7.
Sci Rep ; 10(1): 18149, 2020 10 23.
Artículo en Inglés | MEDLINE | ID: covidwho-1387454

RESUMEN

Antigens displayed on self-assembling nanoparticles can stimulate strong immune responses and have been playing an increasingly prominent role in structure-based vaccines. However, the development of such immunogens is often complicated by inefficiencies in their production. To alleviate this issue, we developed a plug-and-play platform using the spontaneous isopeptide-bond formation of the SpyTag:SpyCatcher system to display trimeric antigens on self-assembling nanoparticles, including the 60-subunit Aquifex aeolicus lumazine synthase (LuS) and the 24-subunit Helicobacter pylori ferritin. LuS and ferritin coupled to SpyTag expressed well in a mammalian expression system when an N-linked glycan was added to the nanoparticle surface. The respiratory syncytial virus fusion (F) glycoprotein trimer-stabilized in the prefusion conformation and fused with SpyCatcher-could be efficiently conjugated to LuS-SpyTag or ferritin-SpyTag, enabling multivalent display of F trimers with prefusion antigenicity. Similarly, F-glycoprotein trimers from human parainfluenza virus-type 3 and spike-glycoprotein trimers from SARS-CoV-2 could be displayed on LuS nanoparticles with decent yield and antigenicity. Notably, murine vaccination with 0.08 µg of SARS-CoV-2 spike-LuS nanoparticle elicited similar neutralizing responses as 2.0 µg of spike, which was ~ 25-fold higher on a weight-per-weight basis. The versatile platform described here thus allows for multivalent plug-and-play presentation on self-assembling nanoparticles of trimeric viral antigens, with SARS-CoV-2 spike-LuS nanoparticles inducing particularly potent neutralizing responses.


Asunto(s)
Antígenos/inmunología , Betacoronavirus/metabolismo , Nanopartículas/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Antígenos/genética , Antígenos/metabolismo , Aquifex , Bacterias/enzimología , Proteínas Bacterianas/genética , Betacoronavirus/aislamiento & purificación , COVID-19 , Infecciones por Coronavirus , Ferritinas/genética , Helicobacter pylori/metabolismo , Humanos , Ratones , Complejos Multienzimáticos/genética , Pruebas de Neutralización , Pandemias , Neumonía Viral , Multimerización de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Propiedades de Superficie
8.
Biophys Chem ; 265: 106441, 2020 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1343142

RESUMEN

The possibility of immobilizing a protein with antigenic properties on a solid support offers significant possibilities in the development of immunosensors and vaccine formulations. For both applications, the orientation of the antigen should ensure ready accessibility of the antibodies to the epitope. However, an experimental assessment of the orientational preferences necessarily proceeds through the preparation/isolation of the antigen, the immobilization on different surfaces and one or more biophysical characterization steps. To predict a priori whether favorable orientations can be achieved or not would allow one to select the most promising experimental routes, partly mitigating the time cost towards the final product. In this manuscript, we apply a simple computational model, based on united-residue modelling, to the prediction of the orientation of the receptor binding domain of the SARS-CoV-2 spike protein on surfaces commonly used in lateral-flow devices. These calculations can account for the experimental observation that direct immobilization on gold gives sufficient exposure of the epitope to obtain a response in immunochemical assays.


Asunto(s)
Betacoronavirus/metabolismo , Epítopos/química , Modelos Moleculares , Glicoproteína de la Espiga del Coronavirus/metabolismo , Antígenos/química , Antígenos/inmunología , Antígenos/metabolismo , Epítopos/inmunología , Simulación del Acoplamiento Molecular , Dominios Proteicos , SARS-CoV-2 , Dióxido de Silicio/química , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Propiedades de Superficie
9.
J Immunol Methods ; 496: 113099, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1292808

RESUMEN

Bispecific antibodies (BsAbs) are engineered to simultaneously bind two different antigens, and offer promising clinical outcomes for various diseases. The dual binding properties of BsAbs may enable superior efficacies and/or potencies compared to standard monoclonal antibodies (mAbs) or combination mAb therapies. Characterizing BsAb binding properties is critical during biotherapeutic development, where data is leveraged to predict efficacy and potency, assess critical quality attributes and improve antibody design. Traditional single-target, single-readout approaches (e.g., ELISA) have limited usefulness for interpreting complex bispecific binding, and double the benchwork. To address these deficiencies, we developed and implemented a new dual-target/readout binding assay that accurately dissects the affinities of both BsAb binding domains directly and simultaneously. This new assay uses AlphaPlex® technology, which eliminates traditional ELISA wash steps and can be miniaturized for automated workflows. The optimized BsAb AlphaPlex assay demonstrates 99-107% accuracy within a 50-150% linear range, and detected >50% binding degradation from photo- and thermal stress conditions. To the best of our knowledge, this is the first instance of a dual-target/readout BsAb AlphaPlex assay with GMP-suitable linear range, accuracy, specificity, and stability-indicating properties. As a highly customizable and efficient assay, BsAb AlphaPlex may be applicable to numerous bispecific formats and/or co-formulations against a variety of antigens beyond the clinical therapeutic space.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Especificidad de Anticuerpos , Antígenos/inmunología , Antígeno CTLA-4/inmunología , Inmunoensayo , Receptor de Muerte Celular Programada 1/inmunología , Anticuerpos Biespecíficos/metabolismo , Complejo Antígeno-Anticuerpo , Antígenos/metabolismo , Sitios de Unión de Anticuerpos , Tampones (Química) , Antígeno CTLA-4/metabolismo , Ensayo de Inmunoadsorción Enzimática , Epítopos , Humanos , Concentración de Iones de Hidrógeno , Cinética , Valor Predictivo de las Pruebas , Receptor de Muerte Celular Programada 1/metabolismo , Unión Proteica , Reproducibilidad de los Resultados
10.
Immunol Rev ; 296(1): 120-131, 2020 07.
Artículo en Inglés | MEDLINE | ID: covidwho-819984

RESUMEN

The majority of all vaccines work by inducing protective antibody responses. The mechanisms by which the B cells responsible for producing protective antibodies are elicited to respond are not well understood. Interclonal B cell competition to complex antigens, particularly in germinal centers, has emerged as an important hurdle in designing effective vaccines. This review will focus on recent advances in understanding the roles of B cell precursor frequency, B cell receptor affinity for antigen, antigen avidity, and other factors that can substantially alter the outcomes of B cell responses to complex antigens. Understanding the interdependence of these fundamental factors that affect B cell responses can inform current vaccine design efforts for pathogens with complex proteins as candidate immunogens such as HIV, influenza, and coronaviruses.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Competencia Celular/inmunología , Epítopos Inmunodominantes/inmunología , Inmunomodulación , Animales , Antígenos/metabolismo , Linfocitos B/citología , Competencia Celular/genética , Diferenciación Celular/inmunología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Humanos , Receptores de Antígenos de Linfocitos B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA