Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Front Immunol ; 12: 826882, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1674339

RESUMEN

Swine enteric coronaviruses (SECoVs) including porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), and porcine deltacoronavirus (PDCoV), account for the majority of lethal watery diarrhea in neonatal pigs and pose significant economic and public health burdens in the world. While the three SECoVs primarily infect intestinal epithelia in vivo and cause similar clinical signs, there are evident discrepancies in their cellular tropism and pathogenicity. However, the underlying mechanisms to cause the differences remain unclear. Herein, we employed porcine enteroids that are a physiologically relevant model of the intestine to assess the host epithelial responses following infection with the three SECoVs (PEDV, TGEV, and PDCoV). Although SECoVs replicated similarly in jejunal enteroids, a parallel comparison of transcriptomics datasets uncovered that PEDV and TGEV infection induced similar transcriptional profiles and exhibited a more pronounced response with more differentially expressed genes (DEGs) in jejunal enteroids compared with PDCoV infection. Notably, TGEV and PDCoV induced high levels of type I and III IFNs and IFN-stimulated gene (ISG) responses, while PEDV displayed a delayed peak and elicited a much lesser extent of IFN responses. Furthermore, TGEV and PDCoV instead of PEDV elicited a substantial upregulation of antigen-presentation genes and T cell-recruiting chemokines in enteroids. Mechanistically, we demonstrated that IFNs treatment markedly elevated the expression of NOD-like receptor (NLR) family NLRC5 and major histocompatibility complex class I (MHC-I) molecules. Together, our results indicate unique and common viral strategies for manipulating the global IFN responses and antigen presentation utilized by SECoVs, which help us a better understanding of host-SECoVs interactions.


Asunto(s)
Presentación de Antígeno/inmunología , Infecciones por Coronavirus/veterinaria , Regulación de la Expresión Génica , Interferones/metabolismo , Virus de la Diarrea Epidémica Porcina/fisiología , Enfermedades de los Porcinos/etiología , Enfermedades de los Porcinos/metabolismo , Animales , Gastroenteritis Porcina Transmisible/etiología , Gastroenteritis Porcina Transmisible/metabolismo , Gastroenteritis Porcina Transmisible/patología , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Porcinos , Enfermedades de los Porcinos/patología , Virus de la Gastroenteritis Transmisible
2.
Int J Mol Sci ; 22(24)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1572495

RESUMEN

The pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has upended healthcare systems and economies around the world. Rapid understanding of the structural biology and pathogenesis of SARS-CoV-2 has allowed the development of emergency use or FDA-approved vaccines and various candidate vaccines. Among the recently developed SARS-CoV-2 candidate vaccines, natural protein-based nanoparticles well suited for multivalent antigen presentation and enhanced immune stimulation to elicit potent humoral and cellular immune responses are currently being investigated. This mini-review presents recent innovations in protein-based nanoparticle vaccines against SARS-CoV-2. The design and strategy of displaying antigenic domains, including spike protein, receptor-binding domain (RBD), and other domains on the surface of various protein-based nanoparticles and the performance of the developed nanoparticle-based vaccines are highlighted. In the final part of this review, we summarize and discuss recent advances in clinical trials and provide an outlook on protein-based nanoparticle vaccines.


Asunto(s)
Vacunas contra la COVID-19/inmunología , Nanopartículas/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Presentación de Antígeno/inmunología , COVID-19/inmunología , Vacunas contra la COVID-19/farmacología , Humanos , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Vacunas/inmunología
3.
PLoS Pathog ; 17(12): e1010085, 2021 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1559373

RESUMEN

Regulatory T (Treg) cells, which constitute about 5-10% of CD4+T cells expressing Foxp3 transcription factor and CD25(IL-2 receptor α chain), are key regulators in controlling immunological self-tolerance and various immune responses. However, how Treg cells control antigen-specific immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains unclear. In this study, we examined the effect of transient breakdown of the immunological tolerance induced by Treg-cell depletion on adaptive immune responses against administered SARS-CoV-2 antigen, spike protein 1 (S1). Notably, without the use of adjuvants, transient Treg-cell depletion in mice induced anti-S1 antibodies that neutralized authentic SARS-CoV-2, follicular helper T cell formation and S1-binding germinal center B cell responses, but prevented the onset of developing autoimmune diseases. To further clarify the mechanisms, we investigated maturation of dendritic cells (DCs), which is essential to initiate antigen-specific immunity. We found that the transient Treg-cell depletion resulted in maturation of both migratory and resident DCs in draining lymph nodes that captured S1-antigen. Moreover, we observed S1-specific CD4+ T cells and CD8+ T cells with interferon-γ production. Thus, captured S1 was successfully presented by DCs, including cross-presentation to CD8+ T cells. These data indicate that transient Treg-cell depletion in the absence of adjuvants induces maturation of antigen-presenting DCs and succeeds in generating antigen-specific humoral and cellular immunity against emerging SARS-CoV-2 antigens. Finally, we showed that SARS-CoV-2 antigen-specific immune responses induced by transient Treg-cell depletion in the absence of adjuvants were compatible with those induced with an effective adjuvant, polyriboinosinic:polyribocytidyl acid (poly IC) and that the combination of transient Treg-cell depletion with poly IC induced potent responses. These findings highlight the capacity for manipulating Treg cells to induce protective adaptive immunity to SARS-CoV-2 with activating antigen-presenting DCs, which may improve the efficacy of ongoing vaccine therapies and help enhance responses to emerging SARS-CoV-2 variants.


Asunto(s)
Inmunidad Adaptativa/inmunología , Antígenos Virales/inmunología , COVID-19/inmunología , Factores de Transcripción Forkhead/inmunología , SARS-CoV-2/inmunología , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , COVID-19/virología , Chlorocebus aethiops , Células Dendríticas/inmunología , Femenino , Centro Germinal/inmunología , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Linfocitos T Reguladores/inmunología , Células Vero
4.
J Biol Chem ; 297(5): 101329, 2021 11.
Artículo en Inglés | MEDLINE | ID: covidwho-1474696

RESUMEN

Population genetic variability in immune system genes can often underlie variability in immune responses to pathogens. Cytotoxic T-lymphocytes are emerging as critical determinants of both severe acute respiratory syndrome coronavirus 2 infection severity and long-term immunity, after either recovery or vaccination. A hallmark of coronavirus disease 2019 is its highly variable severity and breadth of immune responses between individuals. To address the underlying mechanisms behind this phenomenon, we analyzed the proteolytic processing of S1 spike glycoprotein precursor antigenic peptides across ten common allotypes of endoplasmic reticulum aminopeptidase 1 (ERAP1), a polymorphic intracellular enzyme that can regulate cytotoxic T-lymphocyte responses by generating or destroying antigenic peptides. We utilized a systematic proteomic approach that allows the concurrent analysis of hundreds of trimming reactions in parallel, thus better emulating antigen processing in the cell. While all ERAP1 allotypes were capable of producing optimal ligands for major histocompatibility complex class I molecules, including known severe acute respiratory syndrome coronavirus 2 epitopes, they presented significant differences in peptide sequences produced, suggesting allotype-dependent sequence biases. Allotype 10, previously suggested to be enzymatically deficient, was rather found to be functionally distinct from other allotypes. Our findings suggest that common ERAP1 allotypes can be a major source of heterogeneity in antigen processing and through this mechanism contribute to variable immune responses in coronavirus disease 2019.


Asunto(s)
Aminopeptidasas/inmunología , Antígenos Virales/inmunología , Alotipos de Inmunoglobulinas/inmunología , Antígenos de Histocompatibilidad Menor/inmunología , Péptidos/inmunología , SARS-CoV-2/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Aminopeptidasas/química , Presentación de Antígeno/inmunología , Humanos , Antígenos de Histocompatibilidad Menor/química , Péptidos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/química
5.
Front Immunol ; 12: 697840, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1359188

RESUMEN

Monocytes are antigen-presenting cells (APCs) that play diverse roles in promoting or regulating inflammatory responses, but their role in T cell stimulation is not well defined. In inflammatory conditions, monocytes frequently show increased expression of CD169/Siglec-1, a type-I interferon (IFN-I)-regulated protein. However, little is known about the phenotype and function of these CD169+ monocytes. Here, we have investigated the phenotype of human CD169+ monocytes in different diseases, their capacity to activate CD8+ T cells, and the potential for a targeted-vaccination approach. Using spectral flow cytometry, we detected CD169 expression by CD14+ CD16- classical and CD14+ CD16+ intermediate monocytes and unbiased analysis showed that they were distinct from dendritic cells, including the recently described CD14-expressing DC3. CD169+ monocytes expressed higher levels of co-stimulatory and HLA molecules, suggesting an increased activation state. IFNα treatment highly upregulated CD169 expression on CD14+ monocytes and boosted their capacity to cross-present antigen to CD8+ T cells. Furthermore, we observed CD169+ monocytes in virally-infected patients, including in the blood and bronchoalveolar lavage fluid of COVID-19 patients, as well as in the blood of patients with different types of cancers. Finally, we evaluated two CD169-targeting nanovaccine platforms, antibody-based and liposome-based, and we showed that CD169+ monocytes efficiently presented tumor-associated peptides gp100 and WT1 to antigen-specific CD8+ T cells. In conclusion, our data indicate that CD169+ monocytes are activated monocytes with enhanced CD8+ T cell stimulatory capacity and that they emerge as an interesting target in nanovaccine strategies, because of their presence in health and different diseases.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Monocitos/inmunología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , COVID-19/inmunología , Carcinoma Ductal Pancreático/inmunología , Células Cultivadas , Citometría de Flujo , Humanos , Gripe Humana/inmunología , Interferón-alfa/farmacología , Receptores de Lipopolisacáridos/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pancreáticas/inmunología , SARS-CoV-2/inmunología
6.
Sci Adv ; 7(32)2021 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1343936

RESUMEN

Host antibody responses are pivotal for providing protection against infectious agents. We have pioneered a new class of self-assembling micelles based on pentablock copolymers that enhance antibody responses while providing a low inflammatory environment compared to traditional adjuvants. This type of "just-right" immune response is critical in the rational design of vaccines for older adults. Here, we report on the mechanism of enhancement of antibody responses by pentablock copolymer micelles, which act as scaffolds for antigen presentation to B cells and cross-link B cell receptors, unlike other micelle-forming synthetic block copolymers. We exploited this unique mechanism and developed these scaffolds as a platform technology to produce antibodies in vitro. We show that this novel approach can be used to generate laboratory-scale quantities of therapeutic antibodies against multiple antigens, including those associated with SARS-CoV-2 and Yersinia pestis, further expanding the value of these nanomaterials to rapidly develop countermeasures against infectious diseases.


Asunto(s)
Formación de Anticuerpos , Presentación de Antígeno/inmunología , Reactivos de Enlaces Cruzados/química , Receptores de Antígenos de Linfocitos B/química , Proteínas Recombinantes de Fusión/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Yersinia pestis/inmunología , Adyuvantes Inmunológicos , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Polímeros/química , Receptores de Antígenos de Linfocitos B/metabolismo
7.
Cell ; 184(15): 3962-3980.e17, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: covidwho-1252549

RESUMEN

T cell-mediated immunity plays an important role in controlling SARS-CoV-2 infection, but the repertoire of naturally processed and presented viral epitopes on class I human leukocyte antigen (HLA-I) remains uncharacterized. Here, we report the first HLA-I immunopeptidome of SARS-CoV-2 in two cell lines at different times post infection using mass spectrometry. We found HLA-I peptides derived not only from canonical open reading frames (ORFs) but also from internal out-of-frame ORFs in spike and nucleocapsid not captured by current vaccines. Some peptides from out-of-frame ORFs elicited T cell responses in a humanized mouse model and individuals with COVID-19 that exceeded responses to canonical peptides, including some of the strongest epitopes reported to date. Whole-proteome analysis of infected cells revealed that early expressed viral proteins contribute more to HLA-I presentation and immunogenicity. These biological insights, as well as the discovery of out-of-frame ORF epitopes, will facilitate selection of peptides for immune monitoring and vaccine development.


Asunto(s)
Epítopos de Linfocito T/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Sistemas de Lectura Abierta/genética , Péptidos/inmunología , Proteoma/inmunología , SARS-CoV-2/inmunología , Células A549 , Alelos , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/inmunología , COVID-19/inmunología , COVID-19/virología , Femenino , Células HEK293 , Humanos , Cinética , Masculino , Ratones , Péptidos/química , Linfocitos T/inmunología
8.
Nat Cell Biol ; 23(5): 538-551, 2021 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1223094

RESUMEN

COVID-19 can lead to life-threatening respiratory failure, with increased inflammatory mediators and viral load. Here, we perform single-cell RNA-sequencing to establish a high-resolution map of blood antigen-presenting cells (APCs) in 15 patients with moderate or severe COVID-19 pneumonia, at day 1 and day 4 post admission to intensive care unit or pulmonology department, as well as in 4 healthy donors. We generated a unique dataset of 81,643 APCs, including monocytes and rare dendritic cell (DC) subsets. We uncovered multi-process defects in antiviral immune defence in specific APCs from patients with severe disease: (1) increased pro-apoptotic pathways in plasmacytoid DCs (pDCs, key effectors of antiviral immunity), (2) a decrease of the innate sensors TLR9 and DHX36 in pDCs and CLEC9a+ DCs, respectively, (3) downregulation of antiviral interferon-stimulated genes in monocyte subsets and (4) a decrease of major histocompatibility complex (MHC) class II-related genes and MHC class II transactivator activity in cDC1c+ DCs, suggesting viral inhibition of antigen presentation. These novel mechanisms may explain patient aggravation and suggest strategies to restore the defective immune defence.


Asunto(s)
Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Antígenos Virales/inmunología , Antivirales/inmunología , COVID-19/sangre , COVID-19/inmunología , Células Dendríticas/inmunología , Humanos , Monocitos/inmunología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos
9.
Front Immunol ; 11: 595950, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-1110296

RESUMEN

In recent years, the success of immunotherapy targeting immunoregulatory receptors (immune checkpoints) in cancer have generated enthusiastic support to target these receptors in a wide range of other immune related diseases. While the overwhelming focus has been on blockade of these inhibitory pathways to augment immunity, agonistic triggering via these receptors offers the promise of dampening pathogenic inflammatory responses. V-domain Ig suppressor of T cell activation (VISTA) has emerged as an immunoregulatory receptor with constitutive expression on both the T cell and myeloid compartments, and whose agonistic targeting has proven a unique avenue relative to other checkpoint pathways to suppress pathologies mediated by the innate arm of the immune system. VISTA agonistic targeting profoundly changes the phenotype of human monocytes towards an anti-inflammatory cell state, as highlighted by striking suppression of the canonical markers CD14 and Fcγr3a (CD16), and the almost complete suppression of both the interferon I (IFN-I) and antigen presentation pathways. The insights from these very recent studies highlight the impact of VISTA agonistic targeting of myeloid cells, and its potential therapeutic implications in the settings of hyperinflammatory responses such as cytokine storms, driven by dysregulated immune responses to viral infections (with a focus on COVID-19) and autoimmune diseases. Collectively, these findings suggest that the VISTA pathway plays a conserved, non-redundant role in myeloid cell function.


Asunto(s)
Antígenos B7/agonistas , COVID-19/patología , Síndrome de Liberación de Citoquinas/prevención & control , Células Mieloides/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos B7/antagonistas & inhibidores , Antígenos B7/inmunología , Linfocitos T CD4-Positivos/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Humanos , Inmunoterapia , Interferón Tipo I/antagonistas & inhibidores , Receptores de Lipopolisacáridos/antagonistas & inhibidores , Activación de Linfocitos/inmunología , Ratones , Receptores de IgG/antagonistas & inhibidores , SARS-CoV-2/inmunología
10.
Am J Physiol Lung Cell Mol Physiol ; 320(1): L84-L98, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: covidwho-910283

RESUMEN

Coronavirus disease 2019 (COVID-19), driven by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was declared a global pandemic in March 2020. Pathogenic T cells and inflammatory monocytes are regarded as the central drivers of the cytokine storm associated with the severity of COVID-19. In this study, we explored the characteristic peripheral cellular profiles of patients with COVID-19 in both acute and convalescent phases by single-cell mass cytometry (CyTOF). Using a combination of algorithm-guided data analyses, we identified peripheral immune cell subsets in COVID-19 and revealed CD4+ T-cell depletion, T-cell differentiation, plasma cell expansion, and the reduced antigen presentation capacity of innate immunity. Notably, COVID-19 induces a dysregulation in the balance of monocyte populations by the expansion of the monocyte subsets. Collectively, our results represent a high-dimensional, single-cell profile of the peripheral immune response to SARS-CoV-2 infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , COVID-19/inmunología , Leucocitos Mononucleares/inmunología , Monocitos/inmunología , SARS-CoV-2/inmunología , Adulto , Anciano , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/citología , COVID-19/patología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Citocinas/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/citología , Depleción Linfocítica , Masculino , Persona de Mediana Edad , Monocitos/citología , Células Plasmáticas/citología , Análisis de la Célula Individual
11.
Front Immunol ; 11: 1949, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-732902

RESUMEN

After the 1918 flu pandemic, the world is again facing a similar situation. However, the advancement in medical science has made it possible to identify that the novel infectious agent is from the coronavirus family. Rapid genome sequencing by various groups helped in identifying the structure and function of the virus, its immunogenicity in diverse populations, and potential preventive measures. Coronavirus attacks the respiratory system, causing pneumonia and lymphopenia in infected individuals. Viral components like spike and nucleocapsid proteins trigger an immune response in the host to eliminate the virus. These viral antigens can be either recognized by the B cells or presented by MHC complexes to the T cells, resulting in antibody production, increased cytokine secretion, and cytolytic activity in the acute phase of infection. Genetic polymorphism in MHC enables it to present some of the T cell epitopes very well over the other MHC alleles. The association of MHC alleles and its downregulated expression has been correlated with disease severity against influenza and coronaviruses. Studies have reported that infected individuals can, after recovery, induce strong protective responses by generating a memory T-cell pool against SARS-CoV and MERS-CoV. These memory T cells were not persistent in the long term and, upon reactivation, caused local damage due to cross-reactivity. So far, the reports suggest that SARS-CoV-2, which is highly contagious, shows related symptoms in three different stages and develops an exhaustive T-cell pool at higher loads of viral infection. As there are no specific treatments available for this novel coronavirus, numerous small molecular drugs that are being used for the treatment of diseases like SARS, MERS, HIV, ebola, malaria, and tuberculosis are being given to COVID-19 patients, and clinical trials for many such drugs have already begun. A classical immunotherapy of convalescent plasma transfusion from recovered patients has also been initiated for the neutralization of viremia in terminally ill COVID-19 patients. Due to the limitations of plasma transfusion, researchers are now focusing on developing neutralizing antibodies against virus particles along with immuno-modulation of cytokines like IL-6, Type I interferons (IFNs), and TNF-α that could help in combating the infection. This review highlights the similarities of the coronaviruses that caused SARS and MERS to the novel SARS-CoV-2 in relation to their pathogenicity and immunogenicity and also focuses on various treatment strategies that could be employed for curing COVID-19.


Asunto(s)
Betacoronavirus/genética , Infecciones por Coronavirus/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Neumonía Viral/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Animales , Presentación de Antígeno/inmunología , Antivirales/uso terapéutico , Betacoronavirus/química , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Citocinas/biosíntesis , Genoma Viral , Humanos , Evasión Inmune , Inmunización Pasiva/métodos , Ratones , Coronavirus del Síndrome Respiratorio de Oriente Medio/química , Pandemias , Filogenia , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/virología , Linfocitos T/inmunología , Replicación Viral
12.
J Exp Med ; 217(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: covidwho-726090

RESUMEN

Type I interferons (IFN-I) are a major antiviral defense and are critical for the activation of the adaptive immune system. However, early viral clearance by IFN-I could limit antigen availability, which could in turn impinge upon the priming of the adaptive immune system. In this study, we hypothesized that transient IFN-I blockade could increase antigen presentation after acute viral infection. To test this hypothesis, we infected mice with viruses coadministered with a single dose of IFN-I receptor-blocking antibody to induce a short-term blockade of the IFN-I pathway. This resulted in a transient "spike" in antigen levels, followed by rapid antigen clearance. Interestingly, short-term IFN-I blockade after coronavirus, flavivirus, rhabdovirus, or arenavirus infection induced a long-lasting enhancement of immunological memory that conferred improved protection upon subsequent reinfections. Short-term IFN-I blockade also improved the efficacy of viral vaccines. These findings demonstrate a novel mechanism by which IFN-I regulate immunological memory and provide insights for rational vaccine design.


Asunto(s)
Inmunogenicidad Vacunal/inmunología , Interferón Tipo I/antagonistas & inhibidores , Interferón-alfa/inmunología , Receptor de Interferón alfa y beta/inmunología , Vacunas Virales/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Bloqueadores/farmacología , Anticuerpos Antivirales/inmunología , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Expresión Génica/inmunología , Células HEK293 , Humanos , Memoria Inmunológica , Interferón-alfa/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Transfección , Infección por el Virus Zika/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA