Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 234
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 55(4): 718-733.e8, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35349789

RESUMEN

Resident memory B (BRM) cells develop and persist in the lungs of influenza-infected mice and humans; however, their contribution to recall responses has not been defined. Here, we used two-photon microscopy to visualize BRM cells within the lungs of influenza -virus immune and reinfected mice. Prior to re-exposure, BRM cells were sparsely scattered throughout the tissue, displaying limited motility. Within 24 h of rechallenge, these cells increased their migratory capacity, localized to infected sites, and subsequently differentiated into plasma cells. Alveolar macrophages mediated this process, in part by inducing expression of chemokines CXCL9 and CXCL10 from infiltrating inflammatory cells. This led to the recruitment of chemokine receptor CXCR3-expressing BRM cells to infected regions and increased local antibody concentrations. Our study uncovers spatiotemporal mechanisms that regulate lung BRM cell reactivation and demonstrates their capacity to rapidly deliver antibodies in a highly localized manner to sites of viral replication.


Asunto(s)
Gripe Humana , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Animales , Anticuerpos , Humanos , Memoria Inmunológica , Células B de Memoria , Ratones
2.
Allergy ; 79(8): 2207-2221, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38445568

RESUMEN

BACKGROUND: Anti-IgE immunotherapy with monoclonal antibodies represents a breakthrough in treatment of severe allergic diseases. However, drawbacks such as short half-life and high price are not negligible. Our objective is to develop an anti-IgE vaccine based on virus-like particles (VLPs) which can induce long-lasting neutralizing IgG anti-IgE antibodies reducing allergic responses without causing intrinsic mast cell activation due to IgE cross-linking. METHODS: The vaccines were made by chemically coupling three synthetic mouse IgE-Fc fragments to plant-derived immunologically optimized CuMVTT VLPs. The immunogenicity of the vaccines was tested by immunizing naive or allergic mice either with the coupled vaccines or the VLP control followed by systemic or local allergen challenge. RESULTS: Mice immunized with the vaccines exhibited high titers of anti-IgE antibodies in the sera and high levels of anti-IgE secreting plasma cells in lymphoid organs. Moreover, free IgE in serum were reduced by the induced anti-IgE antibodies; therefore, less IgE was bound to FcεRI on the surface of basophils. In line with these reduced IgE levels on effector cells after vaccination, immunized mice were protected from challenge with allergens. Importantly, despite presence of anti-IgE antibodies, no signs of acute or chronic allergic response were seen in immunized allergic mice. CONCLUSION: The generated vaccines can effectively induce anti-IgE antibodies that did not cause allergic responses in sensitized mice but were able to decrease the level of free and cell bound IgE and protected sensitized animals from allergic responses upon allergen challenge.


Asunto(s)
Inmunoglobulina E , Vacunas de Partículas Similares a Virus , Animales , Ratones , Inmunoglobulina E/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Alérgenos/inmunología , Anticuerpos Antiidiotipos/inmunología , Hipersensibilidad/inmunología , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C
3.
Allergy ; 79(1): 184-199, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37815010

RESUMEN

BACKGROUND: Virus-like particle (VLP) Peanut is a novel immunotherapeutic vaccine candidate for the treatment of peanut allergy. The active pharmaceutical ingredient represents cucumber mosaic VLPs (CuMVTT -VLPs) that are genetically fused with one of the major peanut allergens, Ara h 2 (CuMVTT -Ara h 2). We previously demonstrated the immunogenicity and the protective capacity of VLP Peanut-based immunization in a murine model for peanut allergy. Moreover, a Phase I clinical trial has been initiated using VLP Peanut material manufactured following a GMP-compliant manufacturing process. Key product characterization studies were undertaken here to understand the role and contribution of critical quality attributes that translate as predictive markers of immunogenicity and protective efficacy for clinical vaccine development. METHOD: The role of prokaryotic RNA encapsulated within VLP Peanut on vaccine immunogenicity was assessed by producing a VLP Peanut batch with a reduced RNA content (VLP Peanut low RNA). Immunogenicity and peanut allergen challenge studies were conducted with VLP Peanut low RNA, as well as with VLP Peanut in WT and TLR 7 KO mice. Furthermore, mass spectrometry and SDS-PAGE based methods were used to determine Ara h 2 antigen density on the surface of VLP Peanut particles. This methodology was subsequently applied to investigate the relationship between Ara h 2 antigen density and immunogenicity of VLP Peanut. RESULTS: A TLR 7 dependent formation of Ara h 2 specific high-avidity IgG antibodies, as well as a TLR 7 dependent change in the dominant IgG subclass, was observed following VLP Peanut vaccination, while total allergen-specific IgG remained relatively unaffected. Consistently, a missing TLR 7 signal caused only a weak decrease in allergen tolerability after vaccination. In contrast, a reduced RNA content for VLP Peanut resulted in diminished total Ara h 2 specific IgG responses, followed by a significant impairment in peanut allergen tolerability. The discrepant effect on allergen tolerance caused by an absent TLR 7 signal versus a reduced RNA content is explained by the observation that VLP Peanut-derived RNA not only stimulates TLR 7 but also TLR 3. Additionally, a strong correlation was observed between the number of Ara h 2 antigens displayed on the surface of VLP Peanut particles and the vaccine's immunogenicity and protective capacity. CONCLUSIONS: Our findings demonstrate that prokaryotic RNA encapsulated within VLP Peanut, including antigen density of Ara h 2 on viral particles, are key contributors to the immunogenicity and protective capacity of the vaccine. Thus, antigenicity and RNA content are two critical quality attributes that need to be determined at the stage of manufacturing, providing robust information regarding the immunogenicity and protective capacity of VLP Peanut in the mouse which has translational relevance to the human setting.


Asunto(s)
Hipersensibilidad al Cacahuete , Vacunas de Partículas Similares a Virus , Humanos , Animales , Ratones , Hipersensibilidad al Cacahuete/prevención & control , Receptor Toll-Like 7 , Alérgenos , Arachis , Inmunoglobulina G , ARN , Antígenos de Plantas
4.
Immunol Rev ; 296(1): 155-168, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32472710

RESUMEN

Vaccines need to be rationally designed in order be delivered to the immune system for maximizing induction of dynamic immune responses. Virus-like particles (VLPs) are ideal platforms for such 3D vaccines, as they allow the display of complex and native antigens in a highly repetitive form on their surface and can easily reach lymphoid organs in intact form for optimal activation of B and T cells. Adjusting size and zeta potential may allow investigators to further fine-tune delivery to lymphoid organs. An additional way to alter vaccine transfer to lymph nodes and spleen may be the formulation with micron-sized adjuvants that creates a local depot and results in a slow release of antigen and adjuvant. Ideally, the adjuvant in addition stimulates the innate immune system. The dynamics of the immune response may be further enhanced by inclusion of Toll-like receptor ligands, which many VLPs naturally package. Hence, considering the 3Ds in vaccine development may allow for enhancement of their attributes to tackle complex diseases, not usually amenable to conventional vaccine strategies.


Asunto(s)
Vacunación , Vacunas de Partículas Similares a Virus/inmunología , Vacunología , Animales , Antígenos/química , Antígenos/inmunología , Epítopos/química , Epítopos/inmunología , Ingeniería Genética , Historia del Siglo XVIII , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Ligandos , Vacunación/efectos adversos , Vacunación/métodos , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/historia , Vacunología/métodos
5.
Allergy ; 78(7): 1980-1996, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36883475

RESUMEN

BACKGROUND: Allergy to peanut is one of the leading causes of anaphylactic reactions among food allergic patients. Immunization against peanut allergy with a safe and protective vaccine holds a promise to induce durable protection against anaphylaxis caused by exposure to peanut. A novel vaccine candidate (VLP Peanut), based on virus-like particles (VLPs), is described here for the treatment of peanut allergy. METHODS AND RESULTS: VLP Peanut consists of two proteins: a capsid subunit derived from Cucumber mosaic virus engineered with a universal T-cell epitope (CuMVTT ) and a CuMVTT subunit fused with peanut allergen Ara h 2 (CuMVTT -Ara h 2), forming mosaic VLPs. Immunizations with VLP Peanut in both naïve and peanut-sensitized mice resulted in a significant anti-Ara h 2 IgG response. Local and systemic protection induced by VLP Peanut were established in mouse models for peanut allergy following prophylactic, therapeutic, and passive immunizations. Inhibition of FcγRIIb function resulted in a loss of protection, confirming the crucial role of the receptor in conferring cross protection against peanut allergens other than Ara h 2. CONCLUSION: VLP Peanut can be delivered to peanut-sensitized mice without triggering allergic reactions, while remaining highly immunogenic and offering protection against all peanut allergens. In addition, vaccination ablates allergic symptoms upon allergen challenge. Moreover, the prophylactic immunization setting conferred the protection against subsequent peanut-induced anaphylaxis, showing the potential for preventive vaccination. This highlights the effectiveness of VLP Peanut as a prospective break-through immunotherapy vaccine candidate toward peanut allergy. VLP Peanut has now entered clinical development with the study PROTECT.


Asunto(s)
Anafilaxia , Hipersensibilidad al Cacahuete , Ratones , Animales , Hipersensibilidad al Cacahuete/prevención & control , Estudios Prospectivos , Antígenos de Plantas , Alérgenos , Arachis
6.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36835301

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia and may contribute to 60-70% of cases. Worldwide, around 50 million people suffer from dementia and the prediction is that the number will more than triple by 2050, as the population ages. Extracellular protein aggregation and plaque deposition as well as accumulation of intracellular neurofibrillary tangles, all leading to neurodegeneration, are the hallmarks of brains with Alzheimer's disease. Therapeutic strategies including active and passive immunizations have been widely explored in the last two decades. Several compounds have shown promising results in many AD animal models. To date, only symptomatic treatments are available and because of the alarming epidemiological data, novel therapeutic strategies to prevent, mitigate, or delay the onset of AD are required. In this mini-review, we focus on our understanding of AD pathobiology and discuss current active and passive immunomodulating therapies targeting amyloid-ß protein.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Inmunoterapia , Animales , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Inmunoterapia/métodos , Ovillos Neurofibrilares/metabolismo , Placa Amiloide/metabolismo , Humanos , Modelos Animales de Enfermedad
7.
Allergy ; 77(1): 143-149, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34240429

RESUMEN

BACKGROUND: Several new variants of SARS-CoV-2 have emerged since fall 2020 which have multiple mutations in the receptor-binding domain (RBD) of the spike protein. It is unclear which mutations affect receptor affinity versus immune recognition. METHODS: We produced wild type RBD, RBD with single mutations (E484K, K417N, or N501Y) or with all three mutations combined and tested their binding to ACE2 by biolayer interferometry (BLI). The ability of convalescent sera to recognize RBDs and block their interaction with ACE2 was tested as well. RESULTS: We demonstrated that single mutation N501Y increased binding affinity to ACE2 but did not strongly affect its recognition by convalescent sera. In contrast, single mutation E484K had almost no impact on the binding kinetics, but essentially abolished recognition of RBD by convalescent sera. Interestingly, combining mutations E484K, K417N, and N501Y resulted in a RBD with both features: enhanced receptor binding and abolished immune recognition. CONCLUSIONS: Our data demonstrate that single mutations either affect receptor affinity or immune recognition while triple mutant RBDs combine both features.


Asunto(s)
Receptores Virales , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Enzima Convertidora de Angiotensina 2 , COVID-19 , Humanos , Inmunización Pasiva , Mutación , Unión Proteica , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Sueroterapia para COVID-19
8.
Allergy ; 77(1): 111-117, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34453338

RESUMEN

BACKGROUND: Emerged mutations can be attributed to increased transmissibility of the B.1.617 and B.1.36 Indian delta variants of SARS-CoV-2, most notably substitutions L452R/E484Q and N440K, respectively, which occur in the receptor-binding domain (RBD) of the Spike (S) fusion glycoprotein. OBJECTIVE: We aimed to assess the effects of mutations L452R/E484Q and N440K (as well as the previously studied mutation E484K present in variants B.1.351 and P.1) on the affinity of RBD for ACE2, SARS-CoV-2 main receptor. We also aimed to assess the ability of antibodies induced by natural infection or by immunization with BNT162b2 mRNA vaccine to recognize the mutated versions of the RBD, as well as blocking the interaction RBD-ACE2, an important surrogate readout for virus neutralization. METHODS: To this end, we produced recombinant wild-type RBD, as well as RBD containing each of the mutations L452R/E484Q, N440K, or E484K (the latest present in variants of concern B.1.351 and P.1), as well as the ectodomain of ACE2. Using Biolayer Interferometry (BLI), we measured the binding affinity of RBD for ACE2 and the ability of sera from COVID-19 convalescent donors or subjects immunized with BNT162b2 mRNA vaccine to block this interaction. Finally, we correlated these results with total anti-RBD IgG titers measured from the same sera by direct ELISA. RESULTS: The binding assays showed L452R/E484Q double-mutant RBD to interact with ACE2 with higher affinity (KD  = 4.6 nM) than wild-type (KD  = 21.3 nM) or single mutants N440K (KD  = 9.9 nM) and E484K (KD  = 19.7 nM) RBDs. Meanwhile, the anti-RBD IgG titration resulted in lower recognition of mutants E484K and L452R/E484Q by infection-induced antibodies, whereas only mutant E484K was recognized less by antibodies induced by vaccination. More interestingly, sera from convalescent as well as immunized subjects showed reduced ability to block the interaction between ACE2 and RBD mutants E484K and L452R/E484Q, as shown by the inhibition assays. CONCLUSION: Our data suggest that the newly emerged SARS-CoV-2 variant B.1.617, as well as the better-studied variants B.1.351 and P.1 (all containing a mutation at position E484) display increased transmissibility both due to their higher affinity for the cell receptor ACE2 and their ability to partially bypass immunity generated against the wild-type virus. For variant B.1.36 (with a point mutation at position N440), only increased affinity seems to play a role.


Asunto(s)
COVID-19 , SARS-CoV-2 , Vacuna BNT162 , Humanos , Evasión Inmune , Mutación , Unión Proteica , Glicoproteína de la Espiga del Coronavirus/metabolismo , Vacunas Sintéticas , Vacunas de ARNm
9.
Allergy ; 77(8): 2446-2458, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35403221

RESUMEN

BACKGROUND: The highly contagious SARS-CoV-2 is mainly transmitted by respiratory droplets and aerosols. Consequently, people are required to wear masks and maintain a social distance to avoid spreading of the virus. Despite the success of the commercially available vaccines, the virus is still uncontained globally. Given the tropism of SARS-CoV-2, a mucosal immune reaction would help to reduce viral shedding and transmission locally. Only seven out of hundreds of ongoing clinical trials are testing the intranasal delivery of a vaccine against COVID-19. METHODS: In the current study, we evaluated the immunogenicity of a traditional vaccine platform based on virus-like particles (VLPs) displaying RBD of SARS-CoV-2 for intranasal administration in a murine model. The candidate vaccine platform, CuMVTT -RBD, has been optimized to incorporate a universal T helper cell epitope derived from tetanus-toxin and is self-adjuvanted with TLR7/8 ligands. RESULTS: CuMVTT -RBD vaccine elicited a strong systemic RBD- and spike-IgG and IgA antibodies of high avidity. Local immune response was assessed, and our results demonstrate a strong mucosal antibody and plasma cell production in lung tissue. Furthermore, the induced systemic antibodies could efficiently recognize and neutralize different variants of concern (VOCs). CONCLUSION: Our data demonstrate that intranasal administration of CuMVTT -RBD induces a protective systemic and local specific antibody response against SARS-CoV-2 and its VOCs.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Vacunas de Partículas Similares a Virus , Administración Intranasal , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19/inmunología , Humanos , Ratones , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Vacunas de Partículas Similares a Virus/inmunología
10.
Allergy ; 77(1): 243-257, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34496033

RESUMEN

BACKGROUND: SARS-CoV-2 caused one of the most devastating pandemics in the recent history of mankind. Due to various countermeasures, including lock-downs, wearing masks, and increased hygiene, the virus has been controlled in some parts of the world. More recently, the availability of vaccines, based on RNA or adenoviruses, has greatly added to our ability to keep the virus at bay; again, however, in some parts of the world only. While available vaccines are effective, it would be desirable to also have more classical vaccines at hand for the future. Key feature of vaccines for long-term control of SARS-CoV-2 would be inexpensive production at large scale, ability to make multiple booster injections, and long-term stability at 4℃. METHODS: Here, we describe such a vaccine candidate, consisting of the SARS-CoV-2 receptor-binding motif (RBM) grafted genetically onto the surface of the immunologically optimized cucumber mosaic virus, called CuMVTT -RBM. RESULTS: Using bacterial fermentation and continuous flow centrifugation for purification, the yield of the production process is estimated to be >2.5 million doses per 1000-litre fermenter run. We demonstrate that the candidate vaccine is highly immunogenic in mice and rabbits and induces more high avidity antibodies compared to convalescent human sera. The induced antibodies are more cross-reactive to mutant RBDs of variants of concern (VoC). Furthermore, antibody responses are neutralizing and long-lived. In addition, the vaccine candidate was stable for at least 14 months at 4℃. CONCLUSION: Thus, the here presented VLP-based vaccine may be a good candidate for use as conventional vaccine in the long term.


Asunto(s)
COVID-19 , Vacunas de Partículas Similares a Virus , Animales , Anticuerpos Neutralizantes , Formación de Anticuerpos , Vacunas contra la COVID-19 , Control de Enfermedades Transmisibles , Humanos , Ratones , Conejos , SARS-CoV-2
11.
J Allergy Clin Immunol ; 147(4): 1430-1441, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33309740

RESUMEN

BACKGROUND: IgE causes anaphylaxis in type I hypersensitivity diseases by activating degranulation of effector cells such as mast cells and basophils. The mechanisms that control IgE activity and prevent anaphylaxis under normal conditions are still enigmatic. OBJECTIVE: We aimed to unravel how anti-IgE autoantibodies are induced and we aimed to understand their role in regulating serum IgE level and allergic anaphylaxis. METHODS: We immunized mice with different forms of IgE and tested anti-IgE autoantibody responses and their specificities. We then analyzed the effect of those antibodies on serum kinetics and their in vitro and in vivo impact on anaphylaxis. Finally, we investigated anti-IgE autoantibodies in human sera. RESULTS: Immunization of mice with IgE-immune complexes induced glycan-specific anti-IgE autoantibodies. The anti-IgE autoantibodies prevented effector cell sensitization, reduced total IgE serum levels, protected mice from passive and active IgE sensitization, and resulted in cross-protection against different allergens. Furthermore, glycan-specific anti-IgE autoantibodies were present in sera from subjects with allergy and subjects without allergy. CONCLUSION: In conclusion, this study provided the first evidence that in the murine model, the serum level and anaphylactic activity of IgE may be downregulated by glycan-specific IgG anti-IgE autoantibodies.


Asunto(s)
Anticuerpos Antiidiotipos/inmunología , Autoanticuerpos/inmunología , Hipersensibilidad/inmunología , Inmunoglobulina G/inmunología , Polisacáridos/inmunología , Alérgenos/administración & dosificación , Animales , Modelos Animales de Enfermedad , Glicoproteínas/administración & dosificación , Humanos , Inmunoglobulina E/administración & dosificación , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Int J Mol Sci ; 23(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35269805

RESUMEN

Intravenous (IV) iron nanoparticle preparations are widely used to treat iron deficiency. The mechanism of mononuclear phagocyte system-mediated clearance of IV iron nanoparticles is unknown. The early uptake and homeostasis of iron after injection of ferric carboxymaltose (FCM) in mice was studied. An increase in serum iron was observed at 2.5 h followed by a return to baseline by 24 h. An increase in circulating monocytes was observed, particularly Ly6Chi and Ly6Clow. FCM was also associated with a time-dependent decrease in liver Kupffer cells (KCs) and increase in liver monocytes. The increase in liver monocytes suggests an influx of iron-rich blood monocytes, while some KCs underwent apoptosis. Adoptive transfer experiments demonstrated that following liver infiltration, blood monocytes differentiated to KCs. KCs were also critical for IV iron uptake and biodegradation. Indeed, anti-Colony Stimulating Factor 1 Receptor (CSF1R)-mediated depletion of KCs resulted in elevated serum iron levels and impaired iron uptake by the liver. Gene expression profiling indicated that C-C chemokine receptor type 5 (CCR5) might be involved in monocyte recruitment to the liver, confirmed by pharmaceutical inhibition of CCR5. Liver KCs play a pivotal role in the clearance and storage of IV iron and KCs appear to be supported by the expanded blood monocyte population.


Asunto(s)
Macrófagos del Hígado , Nanopartículas , Animales , Carbohidratos , Hierro/metabolismo , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Ratones , Monocitos/metabolismo
13.
Eur J Immunol ; 50(4): 505-514, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31785153

RESUMEN

It is known that for achieving high affinity antibody responses, vaccines must be optimized for antigen dose/density, and the prime/boost interval should be at least 4 weeks. Similar knowledge is lacking for generating high avidity T-cell responses. The functional avidity (FA) of T cells, describing responsiveness to peptide, is associated with the quality of effector function and the protective capacity in vivo. Despite its importance, the FA is rarely determined in T-cell vaccination studies. We addressed the question whether different time intervals for short-term homologous vaccinations impact the FA of CD8 T-cell responses. Four-week instead of 2-week intervals between priming and boosting with potent subunit vaccines in C57BL/6 mice did not improve FA. Equally, similar FA was observed after vaccination with virus-like particles displaying low versus high antigen densities. Interestingly, FA was stable in vivo but not in vitro, depending on the antigen dose and the time interval since T-cell activation, as observed in murine monoclonal T cells. Our findings suggest dynamic in vivo modulation for equal FA. We conclude that low antigen density vaccines or a minimal 4-week prime/boost interval are not crucial for the T-cell's FA, in contrast to antibody responses.


Asunto(s)
Antígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Vacunas de Subunidad/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Formación de Anticuerpos , Presentación de Antígeno , Antígenos/inmunología , Células Cultivadas , Inmunización Secundaria , Ratones , Ratones Endogámicos C57BL , Péptidos/inmunología , Unión Proteica , Vacunación
14.
Curr Top Microbiol Immunol ; 428: 89-102, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30919087

RESUMEN

A large number of vaccines exist that control many of the most important infectious diseases. Despite these successes, there remain many pathogens without effective prophylactic vaccines. Notwithstanding strong difference in the biology of these infectious agents, there exist common problems in vaccine design. Many infectious agents have highly variable surface antigens and/or unusually high antibody levels are required for protection. Such high variability may be addressed by using conserved epitopes and these are, however, usually difficult to display with the right conformation in an immunogenic fashion. Exceptionally high antibody titers may be achieved using life vectors or virus-like display of the epitopes. Hence, an important goal in modern vaccinology is to induce high antibody responses against fragile antigens.


Asunto(s)
Anticuerpos/inmunología , Formación de Anticuerpos/inmunología , Epítopos Inmunodominantes/inmunología , Vacunas/inmunología , Humanos , Epítopos Inmunodominantes/química , Vacunas/química
15.
Allergy ; 76(8): 2565-2574, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33866583

RESUMEN

BACKGROUND: Allergy is a global disease with overall frequencies of >20%. Symptoms vary from irritating local itching to life-threatening systemic anaphylaxis. Even though allergies are allergen-specific, there is a wide range of cross-reactivities (eg apple and latex) that remain largely unexplained. Given the abilities of low-affinity IgG antibodies to inhibit mast cells activation, here we elucidate the minimal affinity of IgE antibodies to induce type I hypersensitivity. METHODS: Three mature (high-affinity) IgE antibodies recognizing three distinct epitopes on Fel d 1, the major cat allergen, were back-mutated to germline conformation, resulting in binding to Fel d 1 with low affinity. The ability of these IgE antibodies to activate mast cells in vitro and in vivo was tested. RESULTS: We demonstrate that affinities as low as 10-7  M are sufficient to activate mast cells in vitro and drive allergic reactions in vivo. Low-affinity IgE antibodies are able to do so, since they bind allergens bivalently on the surface of mast cells, leading to high-avidity interactions. CONCLUSIONS: These results suggest that the underlying mechanism of allergen cross-reactivity may be low-affinity but high-avidity binding between IgE antibodies and cross-reactive allergen.


Asunto(s)
Alérgenos , Hipersensibilidad , Animales , Gatos , Reacciones Cruzadas , Humanos , Inmunoglobulina E , Inmunoglobulina G
16.
Allergy ; 76(3): 853-865, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32997812

RESUMEN

BACKGROUND: Serological immunoassays that can identify protective immunity against SARS-CoV-2 are needed to adapt quarantine measures, assess vaccination responses, and evaluate donor plasma. To date, however, the utility of such immunoassays remains unclear. In a mixed-design evaluation study, we compared the diagnostic accuracy of serological immunoassays that are based on various SARS-CoV-2 proteins and assessed the neutralizing activity of antibodies in patient sera. METHODS: Consecutive patients admitted with confirmed SARS-CoV-2 infection were prospectively followed alongside medical staff and biobank samples from winter 2018/2019. An in-house enzyme-linked immunosorbent assay utilizing recombinant receptor-binding domain (RBD) of the SARS-CoV-2 spike protein was developed and compared to three commercially available enzyme-linked immunosorbent assays (ELISAs) targeting the nucleoprotein (N), the S1 domain of the spike protein (S1), and a lateral flow immunoassay (LFI) based on full-length spike protein. Neutralization assays with live SARS-CoV-2 were performed. RESULTS: One thousand four hundred and seventy-seven individuals were included comprising 112 SARS-CoV-2 positives (defined as a positive real-time PCR result; prevalence 7.6%). IgG seroconversion occurred between day 0 and day 21. While the ELISAs showed sensitivities of 88.4% for RBD, 89.3% for S1, and 72.9% for N protein, the specificity was above 94% for all tests. Out of 54 SARS-CoV-2 positive individuals, 96.3% showed full neutralization of live SARS-CoV-2 at serum dilutions ≥ 1:16, while none of the 6 SARS-CoV-2-negative sera revealed neutralizing activity. CONCLUSIONS: ELISAs targeting RBD and S1 protein of SARS-CoV-2 are promising immunoassays which shall be further evaluated in studies verifying diagnostic accuracy and protective immunity against SARS-CoV-2.


Asunto(s)
Anticuerpos Antivirales/sangre , Prueba Serológica para COVID-19/métodos , SARS-CoV-2/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
17.
Semin Immunol ; 34: 123-132, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28887001

RESUMEN

Virus-like particles (VLPs) have made giant strides in the field of vaccinology over the last three decades. VLPs constitute versatile tools in vaccine development due to their favourable immunological characteristics such as their size, repetitive surface geometry, ability to induce both innate and adaptive immune responses as well as being safe templates with favourable economics. Several VLP-based vaccines are commercially available including vaccines against Human Papilloma Virus (HPV) such as Cervarix®, Gardasil® & Gardasil9® and Hepatitis B Virus (HBV) including the 3rd generation Sci-B-Vac™. In addition, the first licensed malaria-VLP-based vaccine Mosquirix™ has been recently approved by the European regulators. Several other VLP-based vaccines are currently undergoing preclinical and clinical development. This review summarizes some of the major findings and recent advances in VLP-based vaccine development and technologies and outlines general principles that may be harnessed for induction of targeted immune responses.


Asunto(s)
Virus de la Hepatitis B/inmunología , Papillomaviridae/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Vacunas Virales/inmunología , Virosis/inmunología , Inmunidad Adaptativa , Animales , Proteínas de la Cápside , Vacunas contra Hepatitis B , Vacuna Tetravalente Recombinante contra el Virus del Papiloma Humano Tipos 6, 11 , 16, 18 , Humanos , Inmunidad Innata , Vacunas contra Papillomavirus , Vacunas de Partículas Similares a Virus/química
18.
J Allergy Clin Immunol ; 145(1): 301-311.e4, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31437490

RESUMEN

BACKGROUND: Type I hypersensitivity is mediated by allergen-specific IgE, which sensitizes the high-affinity IgE receptor FcεRI on mast cells and basophils and drives allergic inflammation upon secondary allergen contact. CD23/FcεRII, the low-affinity receptor for IgE, is constitutively expressed on B cells and has been shown to regulate immune responses. Simultaneous binding of IgE to FcεRI and CD23 is blocked by reciprocal allosteric inhibition, suggesting that the 2 receptors exert distinct roles in IgE handling. OBJECTIVE: We aimed to study how free IgE versus precomplexed IgE-allergen immune complexes (IgE-ICs) target the 2 IgE receptors FcεRI and CD23, and we investigated the functional implications of the 2 pathways. METHODS: We performed binding and activation assays with human cells in vitro and IgE pharmacokinetics and anaphylaxis experiments in vivo. RESULTS: We demonstrate that FcεRI preferentially binds free IgE and CD23 preferentially binds IgE-ICs. We further show that those different binding properties directly translate to distinct biological functions: free IgE initiated allergic inflammation through FcεRI on allergic effector cells, while IgE-ICs were noninflammatory because of reduced FcεRI binding and enhanced CD23-dependent serum clearance. CONCLUSION: We propose that IgE-ICs are noninflammatory through reduced engagement by FcεRI but increased targeting of the CD23 pathway.


Asunto(s)
Alérgenos/inmunología , Anafilaxia/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Inmunoglobulina E/inmunología , Lectinas Tipo C/inmunología , Receptores de IgE/inmunología , Transducción de Señal/inmunología , Alérgenos/genética , Anafilaxia/genética , Anafilaxia/patología , Animales , Complejo Antígeno-Anticuerpo/genética , Humanos , Lectinas Tipo C/genética , Ratones , Ratones Noqueados , Receptores de IgE/genética , Transducción de Señal/genética
19.
J Allergy Clin Immunol ; 145(4): 1240-1253.e3, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31866435

RESUMEN

BACKGROUND: Peanut allergy is a severe and increasingly frequent disease with high medical, psychosocial, and economic burden for affected patients and wider society. A causal, safe, and effective therapy is not yet available. OBJECTIVE: We sought to develop an immunogenic, protective, and nonreactogenic vaccine candidate against peanut allergy based on virus-like particles (VLPs) coupled to single peanut allergens. METHODS: To generate vaccine candidates, extracts of roasted peanut (Ara R) or the single allergens Ara h 1 or Ara h 2 were coupled to immunologically optimized Cucumber Mosaic Virus-derived VLPs (CuMVtt). BALB/c mice were sensitized intraperitoneally with peanut extract absorbed to alum. Immunotherapy consisted of a single subcutaneous injection of CuMVtt coupled to Ara R, Ara h 1, or Ara h 2. RESULTS: The vaccines CuMVtt-Ara R, CuMVtt-Ara h 1, and CuMVtt-Ara h 2 protected peanut-sensitized mice against anaphylaxis after intravenous challenge with the whole peanut extract. Vaccines did not cause allergic reactions in sensitized mice. CuMVtt-Ara h 1 was able to induce specific IgG antibodies, diminished local reactions after skin prick tests, and reduced the infiltration of the gastrointestinal tract by eosinophils and mast cells after oral challenge with peanut. The ability of CuMVtt-Ara h 1 to protect against challenge with the whole extract was mediated by IgG, as shown via passive IgG transfer. FcγRIIb was required for protection, indicating that immune complexes with single allergens were able to block the allergic response against the whole extract, consisting of a complex allergen mixture. CONCLUSIONS: Our data suggest that vaccination using single peanut allergens displayed on CuMVtt may represent a novel therapy against peanut allergy with a favorable safety profile.


Asunto(s)
Antígenos de Plantas/genética , Desensibilización Inmunológica/métodos , Proteínas de la Membrana/genética , Hipersensibilidad al Cacahuete/terapia , Proteínas de Plantas/genética , Vacunas/genética , Virión/genética , Animales , Antígenos de Plantas/inmunología , Arachis/genética , Cucumovirus/genética , Ingeniería Genética , Humanos , Epítopos Inmunodominantes/inmunología , Inmunoglobulina E/metabolismo , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas de Plantas/inmunología , Receptores de IgG/metabolismo , Vacunas/inmunología , Virión/inmunología
20.
Int J Mol Sci ; 22(9)2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33925597

RESUMEN

Iron is a critical metal for several vital biological processes. Most of the body's iron is bound to hemoglobin in erythrocytes. Iron from senescent red blood cells is recycled by macrophages in the spleen, liver and bone marrow. Dietary iron is taken up by the divalent metal transporter 1 (DMT1) in enterocytes and transported to portal blood via ferroportin (FPN), where it is bound to transferrin and taken up by hepatocytes, macrophages and bone marrow cells via transferrin receptor 1 (TfR1). While most of the physiologically active iron is bound hemoglobin, the major storage of most iron occurs in the liver in a ferritin-bound fashion. In response to an increased iron load, hepatocytes secrete the peptide hormone hepcidin, which binds to and induces internalization and degradation of the iron transporter FPN, thus controlling the amount of iron released from the cells into the blood. This review summarizes the key mechanisms and players involved in cellular and systemic iron regulation.


Asunto(s)
Hierro/metabolismo , Hierro/fisiología , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Enterocitos/metabolismo , Ferritinas/metabolismo , Hemoglobinas/metabolismo , Hepatocitos/metabolismo , Humanos , Hierro de la Dieta/metabolismo , Hígado/metabolismo , Receptores de Transferrina/metabolismo , Bazo/metabolismo , Transferrina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA