Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Virol ; 98(5): e0151623, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38567951

RESUMEN

The non-human primate (NHP) model (specifically rhesus and cynomolgus macaques) has facilitated our understanding of the pathogenic mechanisms of yellow fever (YF) disease and allowed the evaluation of the safety and efficacy of YF-17D vaccines. However, the accuracy of this model in mimicking vaccine-induced immunity in humans remains to be fully determined. We used a systems biology approach to compare hematological, biochemical, transcriptomic, and innate and antibody-mediated immune responses in cynomolgus macaques and human participants following YF-17D vaccination. Immune response progression in cynomolgus macaques followed a similar course as in adult humans but with a slightly earlier onset. Yellow fever virus neutralizing antibody responses occurred earlier in cynomolgus macaques [by Day 7[(D7)], but titers > 10 were reached in both species by D14 post-vaccination and were not significantly different by D28 [plaque reduction neutralization assay (PRNT)50 titers 3.6 Log vs 3.5 Log in cynomolgus macaques and human participants, respectively; P = 0.821]. Changes in neutrophils, NK cells, monocytes, and T- and B-cell frequencies were higher in cynomolgus macaques and persisted for 4 weeks versus less than 2 weeks in humans. Low levels of systemic inflammatory cytokines (IL-1RA, IL-8, MIP-1α, IP-10, MCP-1, or VEGF) were detected in either or both species but with no or only slight changes versus baseline. Similar changes in gene expression profiles were elicited in both species. These included enriched and up-regulated type I IFN-associated viral sensing, antiviral innate response, and dendritic cell activation pathways D3-D7 post-vaccination in both species. Hematological and blood biochemical parameters remained relatively unchanged versus baseline in both species. Low-level YF-17D viremia (RNAemia) was transiently detected in some cynomolgus macaques [28% (5/18)] but generally absent in humans [except one participant (5%; 1/20)].IMPORTANCECynomolgus macaques were confirmed as a valid surrogate model for replicating YF-17D vaccine-induced responses in humans and suggest a key role for type I IFN.


Asunto(s)
Macaca fascicularis , Modelos Animales , Vacuna contra la Fiebre Amarilla , Animales , Femenino , Humanos , Masculino , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Inmunidad Innata , Biología de Sistemas/métodos , Vacunación , Fiebre Amarilla/prevención & control , Fiebre Amarilla/inmunología , Fiebre Amarilla/virología , Vacuna contra la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/inmunología
2.
Front Microbiol ; 13: 932408, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36033843

RESUMEN

The fight against infectious diseases calls for the development of safe and effective vaccines that generate long-lasting protective immunity. In a few situations, vaccine-mediated immune responses may have led to exacerbated pathology upon subsequent infection with the pathogen targeted by the vaccine. Such vaccine-associated enhanced disease (VAED) has been reported, or at least suspected, in animal models, and in a few instances in humans, for vaccine candidates against the respiratory syncytial virus (RSV), measles virus (MV), dengue virus (DENV), HIV-1, simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), and the Middle East respiratory syndrome coronavirus (MERS-CoV). Although alleviated by clinical and epidemiological evidence, a number of concerns were also initially raised concerning the short- and long-term safety of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is causing the ongoing COVID-19 pandemic. Although the mechanisms leading to this phenomenon are not yet completely understood, the individual and/or collective role of antibody-dependent enhancement (ADE), complement-dependent enhancement, and cell-dependent enhancement have been highlighted. Here, we review mechanisms that may be associated with the risk of VAED, which are important to take into consideration, both in the assessment of vaccine safety and in finding ways to define models and immunization strategies that can alleviate such concerns.

3.
Front Immunol ; 12: 645210, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33959127

RESUMEN

Vaccination is one of the most efficient public healthcare measures to fight infectious diseases. Nevertheless, the immune mechanisms induced in vivo by vaccination are still unclear. The route of administration, an important vaccination parameter, can substantially modify the quality of the response. How the route of administration affects the generation and profile of immune responses is of major interest. Here, we aimed to extensively characterize the profiles of the innate and adaptive response to vaccination induced after intradermal, subcutaneous, or intramuscular administration with a modified vaccinia virus Ankara model vaccine in non-human primates. The adaptive response following subcutaneous immunization was clearly different from that following intradermal or intramuscular immunization. The subcutaneous route induced a higher level of neutralizing antibodies than the intradermal and intramuscular vaccination routes. In contrast, polyfunctional CD8+ T-cell responses were preferentially induced after intradermal or intramuscular injection. We observed the same dichotomy when analyzing the early molecular and cellular immune events, highlighting the recruitment of cell populations, such as CD8+ T lymphocytes and myeloid-derived suppressive cells, and the activation of key immunomodulatory gene pathways. These results demonstrate that the quality of the vaccine response induced by an attenuated vaccine is shaped by early and subtle modifications of the innate immune response. In this immunization context, the route of administration must be tailored to the desired type of protective immune response. This will be achieved through systems vaccinology and mathematical modeling, which will be critical for predicting the efficacy of the vaccination route for personalized medicine.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Células Supresoras de Origen Mieloide/inmunología , Vacunación , Virus Vaccinia/inmunología , Vaccinia/inmunología , Vacunas Virales/farmacología , Animales , Inyecciones Intradérmicas , Inyecciones Intramusculares , Macaca fascicularis , Masculino , Vacunas Atenuadas/farmacología
4.
Mol Immunol ; 135: 147-164, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33895579

RESUMEN

Respiratory pathogens represent a great burden for humanity and a potential source of new pandemics, as illustrated by the recent emergence of coronavirus disease 2019 (COVID-19). In recent decades, biotechnological advances have led to the development of numerous innovative therapeutic molecules and vaccine immunogens. However, we still lack effective treatments and vaccines against many respiratory pathogens. More than ever, there is a need for a fast, predictive, preclinical pipeline, to keep pace with emerging diseases. Animal models are key for the preclinical development of disease management strategies. The predictive value of these models depends on their ability to reproduce the features of the human disease, the mode of transmission of the infectious agent and the availability of technologies for monitoring infection. This review focuses on the use of non-human primates as relevant preclinical models for the development of prevention and treatment for human respiratory infections.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Modelos Animales de Enfermedad , SARS-CoV-2/inmunología , Animales , COVID-19/patología , COVID-19/prevención & control , Vacunas contra la COVID-19/uso terapéutico , Haplorrinos , Humanos
5.
Front Immunol ; 12: 612747, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33763063

RESUMEN

Most vaccines require multiple doses to induce long-lasting protective immunity in a high frequency of vaccines, and to ensure strong both individual and herd immunity. Repetitive immunogenic stimulations not only increase the intensity and durability of adaptive immunity, but also influence its quality. Several vaccine parameters are known to influence adaptive immune responses, including notably the number of immunizations, the delay between them, and the delivery sequence of different recombinant vaccine vectors. Furthermore, the initial effector innate immune response is key to activate and modulate B and T cell responses. Optimization of homologous and heterologous prime/boost vaccination strategies requires a thorough understanding of how vaccination history affects memory B and T cell characteristics. This requires deeper knowledge of how innate cells respond to multiple vaccine encounters. Here, we review how innate cells, more particularly those of the myeloid lineage, sense and respond differently to a 1st and a 2nd vaccine dose, both in an extrinsic and intrinsic manner. On one hand, the presence of primary specific antibodies and memory T cells, whose critical properties change with time after priming, provides a distinct environment for innate cells at the time of re-vaccination. On the other hand, innate cells themselves can exert enhanced intrinsic antimicrobial functions, long after initial stimulation, which is referred to as trained immunity. We discuss the potential of trained innate cells to be game-changers in prime/boost vaccine strategies. Their increased functionality in antigen uptake, antigen presentation, migration, and as cytokine producers, could indeed improve the restimulation of primary memory B and T cells and their differentiation into potent secondary memory cells in response to the boost. A better understanding of trained immunity mechanisms will be highly valuable for harnessing the full potential of trained innate cells, to optimize immunization strategies.


Asunto(s)
Inmunidad Adaptativa , Inmunización Secundaria , Vacunación , Vacunas/inmunología , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Humanos , Inmunidad Humoral , Inmunidad Innata , Esquemas de Inmunización , Inmunización Secundaria/métodos , Memoria Inmunológica , Linfocitos T/inmunología , Linfocitos T/metabolismo , Vacunación/métodos , Vacunas/administración & dosificación
6.
Front Immunol ; 12: 784813, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35058925

RESUMEN

Innate immunity modulates adaptive immunity and defines the magnitude, quality, and longevity of antigen-specific T- and B- cell immune memory. Various vaccine and administration factors influence the immune response to vaccination, including the route of vaccine delivery. We studied the dynamics of innate cell responses in blood using a preclinical model of non-human primates immunized with a live attenuated vaccinia virus, a recombinant Modified vaccinia virus Ankara (MVA) expressing a gag-pol-nef fusion of HIV-1, and mass cytometry. We previously showed that it induces a strong, early, and transient innate response, but also late phenotypic modifications of blood myeloid cells after two months when injected subcutaneously. Here, we show that the early innate effector cell responses and plasma inflammatory cytokine profiles differ between subcutaneous and intradermal vaccine injection. Additionally, we show that the intradermal administration fails to induce more highly activated/mature neutrophils long after immunization, in contrast to subcutaneous administration. Different batches of antibodies, staining protocols and generations of mass cytometers were used to generate the two datasets. Mass cytometry data were analyzed in parallel using the same analytical pipeline based on three successive clustering steps, including SPADE, and categorical heatmaps were compared using the Manhattan distance to measure the similarity between cell cluster phenotypes. Overall, we show that the vaccine per se is not sufficient for the late phenotypic modifications of innate myeloid cells, which are evocative of innate immune training. Its route of administration is also crucial, likely by influencing the early innate response, and systemic inflammation, and vaccine biodistribution.


Asunto(s)
Vacunas contra el SIDA , VIH-1 , Neutrófilos/inmunología , Virus Vaccinia , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Animales , Citocinas/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/genética , VIH-1/inmunología , Macaca fascicularis , Masculino , Virus Vaccinia/genética , Virus Vaccinia/inmunología
7.
J Immunol ; 204(12): 3375-3388, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32385135

RESUMEN

DNA vaccines delivered with electroporation (EP) have shown promising results in preclinical models and are evaluated in clinical trials. In this study, we aim to characterize early mechanisms occurring in the skin after intradermal injection and EP of the auxoGTUmultiSIV DNA vaccine in nonhuman primates. First, we show that EP acts as an adjuvant by enhancing local inflammation, notably via granulocytes, monocytes/macrophages, and CD1aint-expressing cell recruitment. EP also induced Langerhans cell maturation, illustrated by CD86, CD83, and HLA-DR upregulation and their migration out of the epidermis. Second, we demonstrate the crucial role of the DNA vaccine in soluble factors release, such as MCP-1 or IL-15. Transcriptomic analysis showed that EP played a major role in gene expression changes postvaccination. However, the DNA vaccine is required to strongly upregulate several genes involved in inflammatory responses (e.g., Saa4), cell migration (e.g., Ccl3, Ccl5, or Cxcl10), APC activation (e.g., Cd86), and IFN-inducible genes (e.g., Ifit3, Ifit5, Irf7, Isg15, orMx1), illustrating an antiviral response signature. Also, AIM-2, a cytosolic DNA sensor, appeared to be strongly upregulated only in the presence of the DNA vaccine and trends to positively correlate with several IFN-inducible genes, suggesting the potential role of AIM-2 in vaccine sensing and the subsequent innate response activation leading to strong adaptive T cell responses. Overall, these results demonstrate that a combined stimulation of the immune response, in which EP and the auxoGTUmultiSIV vaccine triggered different components of the innate immunity, led to strong and persistent cellular recall responses.


Asunto(s)
Inmunidad Celular/inmunología , Inmunidad Innata/inmunología , Piel/inmunología , Linfocitos T/inmunología , Vacunas de ADN/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Movimiento Celular/inmunología , Proteínas de Unión al ADN/inmunología , Electroporación/métodos , Epidermis/inmunología , Expresión Génica/inmunología , Perfilación de la Expresión Génica/métodos , Inflamación/inmunología , Interferones/inmunología , Interleucina-15/inmunología , Macaca fascicularis , Masculino , Regulación hacia Arriba/inmunología , Vacunación/métodos
8.
NPJ Vaccines ; 5(1): 24, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32218996

RESUMEN

Comprehending the mechanisms behind the impact of vaccine regimens on immunity is critical for improving vaccines. Indeed, the time-interval between immunizations may influence B and T cells, as well as innate responses. We compared two vaccine schedules using cynomolgus macaques immunized with an attenuated vaccinia virus. Two subcutaneous injections 2 weeks apart led to an impaired secondary antibody response and similar innate myeloid responses to both immunizations. In contrast, a delayed boost (2 months) improved the quality of the antibody response and involved more activated/mature innate cells, induced late after the prime and responding to the recall. The magnitude and quality of the secondary antibody response correlated with the abundance of these neutrophils, monocytes, and dendritic cells that were modified phenotypically and enriched prior to revaccination at 2 months, but not 2 weeks. These late phenotypic modifications were associated with an enhanced ex vivo cytokine production (including IL-12/23 and IL-1ß) by PBMCs short after the second immunization, linking phenotype and functions. This integrated analysis reveals a deep impact of the timing between immunizations, and highlights the importance of early but also late innate responses involving phenotypical changes, in shaping humoral immunity.

9.
J Leukoc Biol ; 105(5): 1055-1073, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30794328

RESUMEN

A better understanding of innate responses induced by vaccination is critical for designing optimal vaccines. Here, we studied the diversity and dynamics of the NK cell compartment after prime-boost immunization with the modified vaccinia virus Ankara using cynomolgus macaques as a model. Mass cytometry was used to deeply characterize blood NK cells. The NK cell subphenotype composition was modified by the prime. Certain phenotypic changes induced by the prime were maintained over time and, as a result, the NK cell composition prior to boost differed from that before prime. The key phenotypic signature that distinguished NK cells responding to the boost from those responding to the prime included stronger expression of several cytotoxic, homing, and adhesion molecules, suggesting that NK cells at recall were functionally distinct. Our data reveal potential priming or imprinting of NK cells after the first vaccine injection. This study provides novel insights into prime-boost vaccination protocols that could be used to optimize future vaccines.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , VIH/inmunología , Inmunización Secundaria/métodos , Células Asesinas Naturales/efectos de los fármacos , Virus Vaccinia/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Biomarcadores/metabolismo , Citocinas/genética , Citocinas/inmunología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Expresión Génica , Heterogeneidad Genética , Antígenos HLA/genética , Antígenos HLA/inmunología , Humanos , Esquemas de Inmunización , Inmunofenotipificación , Inyecciones Subcutáneas , Células Asesinas Naturales/clasificación , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Macaca fascicularis , Masculino , Vacunas Atenuadas
10.
Sci Rep ; 8(1): 15841, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30367171

RESUMEN

Recognition of nucleic acids by endosomal Toll-like receptors (TLR) is essential to combat pathogens, but requires strict control to limit inflammatory responses. The mechanisms governing this tight regulation are unclear. We found that single-stranded oligonucleotides (ssON) inhibit endocytic pathways used by cargo destined for TLR3/4/7 signaling endosomes. Both ssDNA and ssRNA conferred the endocytic inhibition, it was concentration dependent, and required a certain ssON length. The ssON-mediated inhibition modulated signaling downstream of TLRs that localized within the affected endosomal pathway. We further show that injection of ssON dampens dsRNA-mediated inflammatory responses in the skin of non-human primates. These studies reveal a regulatory role for extracellular ssON in the endocytic uptake of TLR ligands and provide a mechanistic explanation of their immunomodulation. The identified ssON-mediated interference of endocytosis (SOMIE) is a regulatory process that temporarily dampens TLR3/4/7 signaling, thereby averting excessive immune responses.


Asunto(s)
Clatrina/metabolismo , Endocitosis/efectos de los fármacos , Oligonucleótidos/farmacología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 7/metabolismo , Animales , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , ADN de Cadena Simple/farmacología , Endosomas/metabolismo , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Macaca fascicularis , Poli I-C/farmacología , Transducción de Señal/efectos de los fármacos , Piel/metabolismo , Piel/patología , Receptor Toll-Like 3/antagonistas & inhibidores , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 7/antagonistas & inhibidores
11.
Front Immunol ; 9: 870, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29922280

RESUMEN

New vaccine design approaches would be greatly facilitated by a better understanding of the early systemic changes, and those that occur at the site of injection, responsible for the installation of a durable and oriented protective response. We performed a detailed characterization of very early infection and host response events following the intradermal administration of the modified vaccinia virus Ankara as a live attenuated vaccine model in non-human primates. Integrated analysis of the data obtained from in vivo imaging, histology, flow cytometry, multiplex cytokine, and transcriptomic analysis using tools derived from systems biology, such as co-expression networks, showed a strong early local and systemic inflammatory response that peaked at 24 h, which was then progressively replaced by an adaptive response during the installation of the host response to the vaccine. Granulocytes, macrophages, and monocytoid cells were massively recruited during the local innate response in association with local productions of GM-CSF, IL-1ß, MIP1α, MIP1ß, and TNFα. We also observed a rapid and transient granulocyte recruitment and the release of IL-6 and IL-1RA, followed by a persistent phase involving inflammatory monocytes. This systemic inflammation was confirmed by molecular signatures, such as upregulations of IL-6 and TNF pathways and acute phase response signaling. Such comprehensive approaches improve our understanding of the spatiotemporal orchestration of vaccine-elicited immune response, in a live-attenuated vaccine model, and thus contribute to rational vaccine development.


Asunto(s)
Inmunidad Innata , Inmunogenicidad Vacunal , Virus Vaccinia/inmunología , Vaccinia/prevención & control , Vacunas Virales/inmunología , Animales , Biopsia , Células Cultivadas , Inyecciones Intradérmicas , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Macaca fascicularis , Masculino , Modelos Animales , Piel/inmunología , Piel/patología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vaccinia/sangre , Vaccinia/inmunología , Vaccinia/virología , Vacunas Virales/administración & dosificación
12.
Viruses ; 10(5)2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29772762

RESUMEN

Since 2005, Chikungunya virus (CHIKV) re-emerged and caused numerous outbreaks in the world, and finally, was introduced into the Americas in 2013. The lack of CHIKV-specific therapies has led to the use of non-specific drugs. Chloroquine, which is commonly used to treat febrile illnesses in the tropics, has been shown to inhibit CHIKV replication in vitro. To assess the in vivo effect of chloroquine, two complementary studies were performed: (i) a prophylactic study in a non-human primate model (NHP); and (ii) a curative study "CuraChik", which was performed during the Reunion Island outbreak in 2006 in a human cohort. Clinical, biological, and immunological data were compared between treated and placebo groups. Acute CHIKV infection was exacerbated in NHPs treated with prophylactic administration of chloroquine. These NHPs displayed a higher viremia and slower viral clearance (p < 0.003). Magnitude of viremia was correlated to the type I IFN response (Rho = 0.8, p < 0.001) and severe lymphopenia (Rho = 0.8, p < 0.0001), while treatment led to a delay in both CHIKV-specific cellular and IgM responses (p < 0.02 and p = 0.04, respectively). In humans, chloroquine treatment did not affect viremia or clinical parameters during the acute stage of the disease (D1 to D14), but affected the levels of C-reactive Protein (CRP), IFNα, IL-6, and MCP1 over time (D1 to D16). Importantly, no positive effect could be detected on prevalence of persistent arthralgia at Day 300. Although inhibitory in vitro, chloroquine as a prophylactic treatment in NHPs enhances CHIKV replication and delays cellular and humoral response. In patients, curative chloroquine treatment during the acute phase decreases the levels of key cytokines, and thus may delay adaptive immune responses, as observed in NHPs, without any suppressive effect on peripheral viral load.


Asunto(s)
Fiebre Chikungunya/inmunología , Virus Chikungunya/inmunología , Cloroquina/farmacología , Brotes de Enfermedades , Inmunidad/efectos de los fármacos , Macaca fascicularis/virología , Carga Viral/efectos de los fármacos , Animales , Células Cultivadas , Fiebre Chikungunya/tratamiento farmacológico , Fiebre Chikungunya/epidemiología , Fiebre Chikungunya/virología , Cloroquina/uso terapéutico , Estudios de Cohortes , Modelos Animales de Enfermedad , Fibroblastos/virología , Humanos , Macaca fascicularis/inmunología , Macrófagos/virología , Masculino , Reunión/epidemiología , Replicación Viral/efectos de los fármacos
13.
Sci Rep ; 8(1): 3087, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449630

RESUMEN

Understanding the innate immune response to vaccination is critical in vaccine design. Here, we studied blood innate myeloid cells after first and second immunization of cynomolgus macaques with the modified vaccinia virus Ankara. The inflammation at the injection site was moderate and resolved faster after the boost. The blood concentration of inflammation markers increased after both injections but was lower after the boost. The numbers of neutrophils, monocytes, and dendritic cells were transiently affected by vaccination, but without any major difference between prime and boost. However, phenotyping deeper those cells with mass cytometry unveiled their high phenotypic diversity with subsets responding differently after each injection, some enriched only after the primary injection and others only after the boost. Actually, the composition in subphenotype already differed just before the boost as compared to just before the prime. Multivariate analysis identified the key features that contributed to these differences. Cell subpopulations best characterizing the post-boost response were more activated, with a stronger expression of markers involved in phagocytosis, antigen presentation, costimulation, chemotaxis, and inflammation. This study revisits innate immunity by demonstrating that, like adaptive immunity, innate myeloid responses differ after one or two immunizations.


Asunto(s)
Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Virus Vaccinia/inmunología , Vacunas Virales/farmacología , Inmunidad Adaptativa/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos , Inmunidad Innata/inmunología , Inmunización Secundaria/métodos , Interferón gamma/inmunología , Interleucina-2/inmunología , Macaca fascicularis , Células Progenitoras Mieloides/inmunología , Vacunación/métodos , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología , Vacunas Virales/inmunología
14.
Sci Rep ; 7(1): 4122, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28646234

RESUMEN

In vivo electroporation (EP) is used to enhance the uptake of nucleic acids and its association with DNA vaccination greatly stimulates immune responses to vaccine antigens delivered through the skin. However, the effect of EP on cutaneous cell behavior, the dynamics of immune cell recruitment and local inflammatory factors, have not been fully described. Here, we show that intradermal DNA vaccination combined with EP extends antigen expression to the epidermis and the subcutaneous skin muscle in non-human primates. In vivo fibered confocal microscopy and dynamic ex vivo imaging revealed that EP promotes the mobility of Langerhans cells (LC) and their interactions with transfected cells prior to their migration from the epidermis. At the peak of vaccine expression, we detected antigen in damaged keratinocyte areas in the epidermis and we characterized recruited immune cells in the skin, the hypodermis and the subcutaneous muscle. EP alone was sufficient to induce the production of pro-inflammatory cytokines in the skin and significantly increased local concentrations of Transforming Growth Factor (TGF)-alpha and IL-12. Our results show the kinetics of inflammatory processes in response to EP of the skin, and reveal its potential as a vaccine adjuvant.


Asunto(s)
Adyuvantes Inmunológicos , Electroporación , Inmunización , Plásmidos/administración & dosificación , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos/genética , Antígenos/inmunología , Citocinas/metabolismo , Epidermis/inmunología , Epidermis/metabolismo , Expresión Génica , Inmunización/métodos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Inyecciones Intradérmicas , Queratinocitos/inmunología , Queratinocitos/metabolismo , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Macaca , Plásmidos/genética , Vacunación/métodos , Vacunas de ADN/genética
15.
Front Immunol ; 8: 63, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28203239

RESUMEN

Persistent B cell responses in mucosal tissues are crucial to control infection against sexually transmitted pathogens like human immunodeficiency virus 1 (HIV-1). The genital tract is a major site of infection by HIV. Sublingual (SL) immunization in mice was previously shown to generate HIV-specific B cell immunity that disseminates to the genital tract. We report here the immunogenicity in female cynomolgus macaques of a SL vaccine based on a modified gp41 polypeptide coupled to the cholera toxin B subunit designed to expose hidden epitopes and to improve mucosal retention. Combined SL/intramuscular (IM) immunization with such mucoadhesive gp41-based vaccine elicited mucosal HIV-specific IgG and IgA antibodies more efficiently than IM immunization alone. This strategy increased the number and duration of gp41-specific IgA secreting cells. Importantly, combined immunization improved the generation of functional antibodies 3 months after vaccination as detected in HIV-neutralizing assays. Therefore, SL immunization represents a promising vaccine strategy to block HIV-1 transmission.

16.
Eur J Immunol ; 46(3): 689-700, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26678013

RESUMEN

The development of new immunization strategies requires a better understanding of early molecular and cellular events occurring at the site of injection. The skin is particularly rich in immune cells and represents an attractive site for vaccine administration. Here, we specifically targeted vaccine antigens to epidermal Langerhans cells (LCs) using a fusion protein composed of HIV antigens and a monoclonal antibody targeting Langerin. We developed a fluorescence imaging approach to visualize, in vivo, the vaccine-targeted cells. Studies were performed in nonhuman primates (NHPs) because of their relevance as a model to assess human vaccines. We directly demonstrated that in NHPs, intradermally injected anti-Langerin-HIVGag specifically targets epidermal LCs and induces rapid changes in the LC network, including LC activation and migration out of the epidermis. Vaccine targeting of LCs significantly improved anti-HIV immune response without requirement of an adjuvant. Although the co-injection of the TLR-7/8 synthetic ligand, R-848 (resiquimod), with the vaccine, did not enhance significantly the antibody response, it stimulated recruitment of HLA-DR+ inflammatory cells to the site of immunization. This study allowed us to characterize the dynamics of early local events following the injection of a vaccine-targeted epidermal LCs and R-848.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antígenos CD/inmunología , Células de Langerhans/inmunología , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/inmunología , Vacunas/administración & dosificación , Animales , Anticuerpos Antivirales/sangre , Antígenos CD/administración & dosificación , Células Epidérmicas , Epidermis/inmunología , Proteína p24 del Núcleo del VIH/administración & dosificación , Proteína p24 del Núcleo del VIH/inmunología , Humanos , Imidazoles/administración & dosificación , Imidazoles/inmunología , Inyecciones Intradérmicas , Microscopía Intravital , Células de Langerhans/ultraestructura , Lectinas Tipo C/administración & dosificación , Macaca fascicularis , Lectinas de Unión a Manosa/administración & dosificación , Imagen Óptica , Vacunas/inmunología
17.
PLoS One ; 10(9): e0135513, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26407317

RESUMEN

Targeting dendritic cell-specific endocytic receptors using monoclonal antibodies fused to desired antigens is an approach widely used in vaccine development to enhance the poor immunogenicity of protein-based vaccines and to induce immune responses. Here, we engineered an anti-human DCIR recombinant antibody, which cross-reacts with the homologous cynomolgous macaque receptor and was fused via the heavy chain C-terminus to HIV Gagp24 protein (αDCIR.Gagp24). In vitro, αDCIR.Gagp24 expanded multifunctional antigen-specific memory CD4+ T cells recognizing multiple Gagp24 peptides from HIV-infected patient peripheral blood mononuclear cells. In non human primates, priming with αDCIR.Gagp24 without adjuvant elicited a strong anti-Gagp24 antibody response after the second immunization, while in the non-targeted HIV Gagp24 protein control groups the titers were weak. The presence of the double-stranded RNA poly(I:C) adjuvant significantly enhanced the anti-Gagp24 antibody response in all the groups and reduced the discrimination between the different vaccine groups. The avidity of the anti-Gagp24 antibody responses was similar with either αDCIR.Gagp24 or Gagp24 immunization, but increased from medium to high avidity in both groups when poly(I:C) was co-administered. This data provides a comparative analysis of DC-targeted and non-targeted proteins for their capacity to induce antigen-specific antibody responses in vivo. This study supports the further development of DCIR-based DC-targeting vaccines for protective durable antibody induction, especially in the absence of adjuvant.


Asunto(s)
Formación de Anticuerpos/inmunología , Proteína p24 del Núcleo del VIH/inmunología , VIH-1/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Lectinas Tipo C/inmunología , Glicoproteínas de Membrana/inmunología , Receptores Inmunológicos/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Afinidad de Anticuerpos/inmunología , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Epítopos de Linfocito T , Anticuerpos Anti-VIH/inmunología , Proteína p24 del Núcleo del VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Inmunización , Esquemas de Inmunización , Cadenas Pesadas de Inmunoglobulina/genética , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Macaca fascicularis , Masculino , Proteínas Recombinantes de Fusión/genética , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología
18.
J Immunol Methods ; 426: 42-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26219836

RESUMEN

The skin is a valuable target for vaccine delivery because it contains many immune cell populations, notably antigen presenting cells. Skin immune cells have been extensively described in mice and humans but not in non-human primates, which are pertinent models for immunological research in vaccination. The aim of this work was to describe immune cell populations in the epidermis, dermis and skin draining lymph nodes in cynomolgus macaques by a single 12-parameter flow cytometry protocol. Given that skin cells share several markers, we defined a gating strategy to identify accurately immune cells and to limit contamination of one immune cell population by another. The epidermis contained CD1a(+)CD1c(-) Langerhans cells (LCs), CD3(+) T cells and putative NK cells. The dermis contained CD1a(+)CD1c(-) cells, which were similar to LCs, CD1a(+)CD1c(+) dermal dendritic cells (DDCs), CD163(high)CD11b(+) resident macrophages, CD3(+) T cells and putative NK cells. The skin also contained CD66(+) polymorphonuclear cells in some animals. Thus, immune cell populations in the macaque are similar to those in humans despite some differences in phenotype. In skin draining lymph nodes, we identified migratory LCs, CD1a(+)CD1c(+) DDCs and macrophages. The simultaneous identification of these different immune cells with one panel of markers avoids the use of large amounts of precious sample and may improve the understanding of immune mechanisms in the skin after treatment or vaccination.


Asunto(s)
Dermis/inmunología , Epidermis/inmunología , Citometría de Flujo/métodos , Ganglios Linfáticos/inmunología , Animales , Biomarcadores/análisis , Movimiento Celular/inmunología , Dermis/citología , Células Epidérmicas , Células Asesinas Naturales/inmunología , Células de Langerhans/inmunología , Ganglios Linfáticos/citología , Macaca fascicularis , Macrófagos/inmunología , Neutrófilos/inmunología , Linfocitos T/inmunología
19.
Vaccine ; 33(20): 2354-9, 2015 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-25839103

RESUMEN

We evaluated the immunogenicity of a prime/boost vaccine strategy combining 5 lipopeptides (HIV-Lipo-5) and a recombinant modified vaccinia virus Ankara (rMVA-HIV) in cynomolgus macaques. Both of these vaccine components deliver HIV LAI Gag, Pol, and Nef antigens. Systemic and local safety was excellent in all groups. Immunization with HIV-Lipo-5 alone induced significant serum anti-HIV antibody titers which were not modified by rMVA-HIV immunization. However, induction of T-cell responses, as measured by IFNγ and IL-2 producing cells upon short-term stimulation with HIV peptide pools, required combined immunization with rMVA-HIV. Responses were preferentially observed against Gag antigen. Interestingly, HIV-Lipo-5 efficiently primed HIV induced T-cell responses upon the injection of rMVA-HIV, which may help to reduce the required number of vector injections. Our results provide a rationale for the use of a strategy involving HIV-Lipo-5 priming followed by rMVA-HIV booster immunization as a prophylactic or therapeutic vaccine approach against HIV infection and AIDS.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/inmunología , Anticuerpos Anti-VIH/sangre , Antígenos VIH/inmunología , Lipopéptidos/inmunología , Linfocitos T/inmunología , Virus Vaccinia/genética , Vacunas contra el SIDA/genética , Animales , Linfocitos T CD8-positivos/inmunología , Citocinas/sangre , Citocinas/inmunología , Ensayo de Immunospot Ligado a Enzimas , Anticuerpos Anti-VIH/inmunología , Antígenos VIH/administración & dosificación , Inmunización Secundaria , Interferón gamma/inmunología , Interleucina-2/inmunología , Macaca fascicularis , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
20.
J Immunol ; 193(5): 2416-26, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25057007

RESUMEN

Dendritic cells are major APCs that can efficiently prime immune responses. However, the roles of skin-resident Langerhans cells (LCs) in eliciting immune responses have not been fully understood. In this study, we demonstrate for the first time, to our knowledge, that LCs in cynomolgus macaque skin are capable of inducing antiviral-specific immune responses in vivo. Targeting HIV-Gag or influenza hemagglutinin Ags to skin LCs using recombinant fusion proteins of anti-Langerin Ab and Ags resulted in the induction of the viral Ag-specific responses. We further demonstrated that such Ag-specific immune responses elicited by skin LCs were greatly enhanced by TLR ligands, polyriboinosinic polyribocytidylic acid, and R848. These enhancements were not due to the direct actions of TLR ligands on LCs, but mainly dependent on TNF-α secreted from macrophages and neutrophils recruited to local tissues. Skin LC activation and migration out of the epidermis are associated with macrophage and neutrophil infiltration into the tissues. More importantly, blocking TNF-α abrogated the activation and migration of skin LCs. This study highlights that the cross-talk between innate immune cells in local tissues is an important component for the establishment of adaptive immunity. Understanding the importance of local immune networks will help us to design new and effective vaccines against microbial pathogens.


Asunto(s)
VIH-1/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/farmacología , Virus de la Influenza A/inmunología , Células de Langerhans/inmunología , Piel/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/farmacología , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/fisiología , Animales , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Imidazoles/farmacología , Macaca mulatta , Macrófagos/inmunología , Neutrófilos/inmunología , Poli I/farmacología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA