Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Semin Cancer Biol ; 88: 81-95, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36526110

RESUMEN

Cancer vaccines and immune checkpoint inhibitors (ICIs) function at different stages of the cancer immune cycle due to their distinct mechanisms of action. Therapeutic cancer vaccines enhance the activation and infiltration of cytotoxic immune cells into the tumor microenvironment (TME), while ICIs, prevent and/or reverse the dysfunction of these immune cells. The efficacy of both classes of immunotherapy has been evaluated in monotherapy, but they have been met with several challenges. Although therapeutic cancer vaccines can activate anti-tumor immune responses, these responses are susceptible to attenuation by immunoregulatory molecules. Similarly, ICIs are ineffective in the absence of tumor-infiltrating lymphocytes (TILs). Further, ICIs are often associated with immune-related adverse effects that may limit quality of life and compliance. However, the combination of the improved immunogenicity afforded by cancer vaccines and restrained immunosuppression provided by immune checkpoint inhibitors may provide a suitable platform for therapeutic synergism. In this review, we revisit the history and various classifications of therapeutic cancer vaccines. We also provide a summary of the currently approved ICIs. Finally, we provide mechanistic insights into the synergism between ICIs and cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Calidad de Vida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Linfocitos Infiltrantes de Tumor , Inmunoterapia , Microambiente Tumoral
2.
Eur J Immunol ; 52(6): 936-945, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35304741

RESUMEN

COVID-19 vaccines prevent severe forms of the disease, but do not warrant complete protection against breakthrough infections. This could be due to suboptimal mucosal immunity at the site of virus entry, given that all currently approved vaccines are administered via the intramuscular route. In this study, we assessed humoral and cellular immune responses in BALB/c mice after intranasal and intramuscular immunization with adenoviral vector ChAdOx1-S expressing full-length Spike protein of SARS-CoV-2. We showed that both routes of vaccination induced a potent IgG antibody response, as well as robust neutralizing capacity, but intranasal vaccination elicited a superior IgA antibody titer in the sera and in the respiratory mucosa. Bronchoalveolar lavage from intranasally immunized mice efficiently neutralized SARS-CoV-2, which has not been the case in intramuscularly immunized group. Moreover, substantially higher percentages of epitope-specific CD8 T cells exhibiting a tissue resident phenotype were found in the lungs of intranasally immunized animals. Finally, both intranasal and intramuscular vaccination with ChAdOx1-S efficiently protected the mice after the challenge with recombinant herpesvirus expressing the Spike protein. Our results demonstrate that intranasal application of adenoviral vector ChAdOx1-S induces superior mucosal immunity and therefore could be a promising strategy for putting the COVID-19 pandemic under control.


Asunto(s)
COVID-19 , Vacunas Virales , Adenoviridae/genética , Administración Intranasal , Animales , Anticuerpos Antivirales , COVID-19/prevención & control , Vacunas contra la COVID-19 , Humanos , Inmunidad Celular , Inmunidad Mucosa , Ratones , Ratones Endogámicos BALB C , Pandemias/prevención & control , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Vacunación/métodos
3.
Int J Mol Sci ; 24(22)2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-38003300

RESUMEN

Herpesviruses are large DNA viruses that have long been used as powerful gene therapy tools. In recent years, the ability of herpesviruses to stimulate both innate and adaptive immune responses has led to their transition to various applications as vaccine vectors. This vaccinology branch is growing at an unprecedented and accelerated rate. To date, human herpesvirus-based vectors have been used in vaccines to combat a variety of infectious agents, including the Ebola virus, foot and mouth disease virus, and human immunodeficiency viruses. Additionally, these vectors are being tested as potential vaccines for cancer-associated antigens. Thanks to advances in recombinant DNA technology, immunology, and genomics, numerous steps in vaccine development have been greatly improved. A better understanding of herpesvirus biology and the interactions between these viruses and the host cells will undoubtedly foster the use of herpesvirus-based vaccine vectors in clinical settings. To overcome the existing drawbacks of these vectors, ongoing research is needed to further advance our knowledge of herpesvirus biology and to develop safer and more effective vaccine vectors. Advanced molecular virology and cell biology techniques must be used to better understand the mechanisms by which herpesviruses manipulate host cells and how viral gene expression is regulated during infection. In this review, we cover the underlying molecular structure of herpesviruses and the strategies used to engineer their genomes to optimize capacity and efficacy as vaccine vectors. Also, we assess the available data on the successful application of herpesvirus-based vaccines for combating diseases such as viral infections and the potential drawbacks and alternative approaches to surmount them.


Asunto(s)
Herpesviridae , Vacunas Virales , Virosis , Humanos , Herpesviridae/genética , Simplexvirus/genética , Vectores Genéticos/genética
4.
Immunology ; 164(2): 279-291, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34003499

RESUMEN

Cytomegalovirus (CMV) induction of large frequencies of highly functional memory T cells has attracted much interest in the utility of CMV-based vaccine vectors, with exciting preclinical data obtained in models of infectious diseases and cancer. However, pathogenesis of human CMV (HCMV) remains a concern. Attenuated CMV-based vectors, such as replication- or spread-deficient viruses, potentially offer an alternative to fully replicating vectors. However, it is not well understood how CMV attenuation impacts vector immunogenicity, particularly when administered via relevant routes of immunization such as the skin. Herein, we used the murine cytomegalovirus (MCMV) model to investigate the impact of vector attenuation on T-cell memory formation following subcutaneous administration. We found that the spread-deficient virus (ΔgL-MCMV) was impaired in its ability to induce memory CD8+ T cells reactive to some (M38, IE1) but not all (IE3) viral antigens. Impaired-memory T-cell development was associated with a preferential and pronounced loss of polyfunctional (IFN-γ+ TNF-α+ ) T cells and also reduced accumulation of TCF1+ T cells, and was not rescued by increasing the dose of replication-defective MCMV. Finally, whilst vector attenuation reduced dendritic cell (DC) recruitment to skin-draining lymph nodes, systematic depletion of multiple DC subsets during acute subcutaneous MCMV infection had a negligible impact on T-cell memory formation, implying that attenuated responses induced by replication-deficient vectors were likely not a consequence of impaired initial DC activation. Thus, overall, these data imply that the choice of antigen and/or cloning strategy of exogenous antigen in combination with the route of immunization may influence the ability of attenuated CMV vectors to induce robust functional T-cell memory.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Células Dendríticas/inmunología , Memoria Inmunológica/inmunología , Piel/inmunología , Replicación Viral/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/virología , Infecciones por Citomegalovirus/virología , Células Dendríticas/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Piel/virología
5.
Med Microbiol Immunol ; 208(3-4): 349-363, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30900089

RESUMEN

Cytomegaloviruses (CMVs) present singular features that are particularly advantageous for human vaccine development, a current medical need. Vaccines that induce neutralizing antibodies are among the most successful and efficacious available. However, chronic and persistent human infections, pathogens with high variability of exposed proteins, as well as tumors, highlight the need for developing novel vaccines inducing strong and long-lasting cellular immune responses mediated by effector or effector memory CD8+ cytotoxic T lymphocytes. CMVs induce the most potent CD8+ T lymphocyte response to a pathogen known in each of their hosts, maintain and even increase it for life for selected antigens, in what is known as the ever growing inflationary memory, and maintain an effector memory status due to recent and repeated antigen stimulation that endows these inflationary T lymphocytes with superior and faster protective potency. In addition to these CMV singularities, this family of viruses has two more common favorable features: they can superinfect an already infected host, which is needed in face of the high CMV prevalence, and they can harbor very large segments of foreign DNA at many different genomic sites. All these properties endow CMVs with a singular potential to be used as human vaccine vectors. Current developments with most of the recombinant CMV-based vaccine candidates that have been tested in animal models against clinically relevant viral and bacterial infections and for their use in tumor immunotherapy are reviewed herein. Since CMV vectors should be designed to avoid the risk of disease in immunocompromised individuals, special attention is also paid to attenuated vectors. Taken together, the results support the future use of CMV-based vaccine vectors to induce protective CD8+ T lymphocyte responses in humans, mainly against viral infections and as anti-tumor vaccines.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citomegalovirus/genética , Portadores de Fármacos , Vectores Genéticos , Vacunas Virales/inmunología , Descubrimiento de Drogas/métodos , Humanos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/genética
6.
J Biol Chem ; 292(10): 3958-3969, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28154190

RESUMEN

This Reflections article describes my early work on viral enzymes and the discovery of mRNA capping, how my training in medicine and biochemistry merged as I evolved into a virologist, the development of viruses as vaccine vectors, and how scientific and technological developments during the 1970s and beyond set the stage for the interrogation of nearly every step in the reproductive cycle of vaccinia virus (VACV), a large DNA virus with about 200 genes. The reader may view this article as a work in progress, because I remain actively engaged in research at the National Institutes of Health (NIH) notwithstanding 50 memorable years there.


Asunto(s)
Biología Molecular , Virus Vaccinia/fisiología , Vaccinia/prevención & control , Vaccinia/virología , Proteínas Virales/metabolismo , Replicación Viral , Humanos , Vaccinia/genética
7.
Malar J ; 16(1): 254, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28619071

RESUMEN

BACKGROUND: An effective malaria transmission-blocking vaccine (TBV) would be a major advance in the current efforts to eliminate and, ultimately, eradicate malaria. Antibodies against Plasmodium falciparum surface protein, Pfs25, are known to block parasite development in the mosquito vector. However, in initial clinical trials the limited immunogenicity of recombinant Pfs25 protein-in-adjuvant vaccines has been a challenge. METHODS: Novel human adenovirus type 5 (Ad5) vectors were used in heterologous prime boost vaccination strategies to augment the immune response against Pfs25. Specifically, an Ad5 vector that directs expression of full-length, membrane-bound Pfs25 was used as a priming immunization followed by a boost with Ad5 viral particles displaying only the Pfs25 epitope targeted by transmission-blocking antibodies 4B7 and 1D2 (Pfs25 aa 122-134) in hypervariable region 5 of the hexon capsid protein. RESULTS: This heterologous prime-boost vaccine strategy induced antibodies that significantly inhibit P. falciparum transmission to mosquitoes in a standard membrane-feeding assay. Further, immunized mice generated a robust anti-Pfs25 antibody response characterized by higher titer, higher relative avidity and a broader IgG subclass profile than observed with a homologous prime-boost with recombinant Pfs25/alum. CONCLUSION: The data suggest that focusing the immune response against defined epitopes displayed on the viral capsid is an effective strategy for transmission-blocking vaccine development.


Asunto(s)
Vacunas contra el Adenovirus/genética , Anticuerpos Antiprotozoarios/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Epítopos/química , Vectores Genéticos , Células HEK293 , Células HeLa , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Malaria Falciparum/transmisión , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/química
8.
J Appl Microbiol ; 123(2): 325-339, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28295939

RESUMEN

Gram-positive, nonpathogenic lactic acid bacteria (LAB) are considered to be promising candidates for the development of novel, safe production and delivery systems of heterologous proteins. Recombinant LAB strains were shown to elicit specific systemic and mucosal immune responses against selected antigens. For this reason, this group of bacteria is considered as a potential replacement of classical, often pathogenic, attenuated microbial carriers. Mucosal administration of recombinant LAB, especially via the best explored and universal oral route, offers many advantages in comparison to systemic inoculation, and is attractive from the immunological and practical point of view. Research aimed at designing efficient, mucosally applied vaccines in combination with improved immunization efficiency, monitoring of in vivo antigen production, determination of optimal dose for vaccination, strain selection and characterization is a priority in modern vaccinology. This paper summarizes and organizes the available knowledge on the application of LAB as live oral vaccine vectors. It constitutes a valuable source of general information for researchers interested in mucosal vaccine development and constructing LAB strains with vaccine potential.


Asunto(s)
Vectores Genéticos/genética , Lactobacillus/genética , Vacunas/genética , Animales , Expresión Génica , Vectores Genéticos/inmunología , Humanos , Inmunización , Lactobacillus/inmunología , Vacunas/administración & dosificación , Vacunas/inmunología
9.
Inmunologia ; 32(3): 102-116, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-32287712

RESUMEN

Vaccination is considered one of the most effective ways to control pathogens and prevent diseases in humans as well as in the veterinary field. Traditional vaccines against animal viral diseases are based on inactivated or attenuated viruses, but new subunit vaccines are gaining attention from researchers in animal vaccinology. Among these, virus-like particles (VLPs) represent one of the most appealing approaches opening up interesting frontiers in animal vaccines. VLPs are robust protein scaffolds exhibiting well-defined geometry and uniformity that mimic the overall structure of the native virions but lack the viral genome. They are often antigenically indistinguishable from the virus from which they were derived and present important advantages in terms of safety. VLPs can stimulate strong humoral and cellular immune responses and have been shown to exhibit self-adjuvanting abilities. In addition to their suitability as a vaccine for the homologous virus from which they are derived, VLPs can also be used as vectors for the multimeric presentation of foreign antigens. VLPs have therefore shown dramatic effectiveness as candidate vaccines; nevertheless, only one veterinary VLP-base vaccine is licensed. Here, we review and examine in detail the current status of VLPs as a vaccine strategy in the veterinary field, and discuss the potential advantages and challenges of this technology.


La vacunación constituye uno de los procedimientos más eficaces para controlar los patógenos y prevenir enfermedades tanto en seres humanos como en el campo veterinario. Las vacunas tradicionales frente a enfermedades animales se basan por lo general en la utilización de virus atenuados o inactivados. Sin embargo, las vacunas de subunidad están ganando terreno progresivamente en el campo de la sanidad animal. Entre ellas, las vacunas basadas en pseudopartículas virales o VLPs (por su nombre en inglés virus-like particles), representan una de las estrategias más atractivas actualmente en el campo de las vacunas para animales. Las VLPs son estructuras proteicas con una geometría y uniformidad muy definidas, que mimetizan la estructura de los virus nativos pero carecen de genoma viral. Por lo general son antigénicamente indistinguibles de los virus de los que proceden y su empleo como inmunógenos presenta importantes ventajas en términos de seguridad. Las VLPs pueden inducir una fuerte respuesta inmune, tanto humoral como celular, y se ha demostrado que poseen capacidad de actuar como adyuvantes (self-adjuvanting). Además de su idoneidad como vacunas frente al virus homólogo del cual proceden, las VLPs también se pueden utilizar como vectores para la presentación multimérica de antígenos heterólogos. Las VLPs han mostrado una elevada eficacia como candidatos vacunales, sin embargo, hasta el momento sólo una vacuna basada en VLPs ha sido autorizada y comercializada en el campo veterinario. En este trabajo se revisa el estado actual de las VLP empleadas como nuevas estrategias vacunales en el campo de la veterinaria, analizando las potenciales ventajas y desafíos que enfrenta esta tecnología.

10.
Pharmaceutics ; 15(3)2023 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-36986707

RESUMEN

Lentiviral vectors are among the most effective viral vectors for vaccination. In clear contrast to the reference adenoviral vectors, lentiviral vectors have a high potential for transducing dendritic cells in vivo. Within these cells, which are the most efficient at activating naive T cells, lentiviral vectors induce endogenous expression of transgenic antigens that directly access antigen presentation pathways without the need for external antigen capture or cross-presentation. Lentiviral vectors induce strong, robust, and long-lasting humoral, CD8+ T-cell immunity and effective protection against several infectious diseases. There is no pre-existing immunity to lentiviral vectors in the human population and the very low pro-inflammatory properties of these vectors pave the way for their use in mucosal vaccination. In this review, we have mainly summarized the immunological aspects of lentiviral vectors, their recent optimization to induce CD4+ T cells, and our recent data on lentiviral vector-based vaccination in preclinical models, including prophylaxis against flaviviruses, SARS-CoV-2, and Mycobacterium tuberculosis.

11.
Viruses ; 14(6)2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35746665

RESUMEN

The poultry industry is the largest source of meat and eggs for human consumption worldwide. However, viral outbreaks in farmed stock are a common occurrence and a major source of concern for the industry. Mortality and morbidity resulting from an outbreak can cause significant economic losses with subsequent detrimental impacts on the global food supply chain. Mass vaccination is one of the main strategies for controlling and preventing viral infection in poultry. The development of broadly protective vaccines against avian viral diseases will alleviate selection pressure on field virus strains and simplify vaccination regimens for commercial farms with overall savings in husbandry costs. With the increasing number of emerging and re-emerging viral infectious diseases in the poultry industry, there is an urgent need to understand the strategies for broadening the protective efficacy of the vaccines against distinct viral strains. The current review provides an overview of viral vaccines and vaccination regimens available for common avian viral infections, and strategies for developing safer and more efficacious viral vaccines for poultry.


Asunto(s)
Enfermedades de las Aves de Corral , Vacunas Virales , Virosis , Animales , Pollos , Humanos , Aves de Corral , Vacunación/veterinaria , Virosis/prevención & control , Virosis/veterinaria
12.
Front Immunol ; 12: 642316, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33936058

RESUMEN

The promise of tumor immunotherapy to significantly improve survival in patients who are refractory to long-standing therapies, such as chemotherapy and radiation, is now being realized. While immune checkpoint inhibitors that target PD-1 and CTLA-4 are leading the charge in clinical efficacy, there are a number of other promising tumor immunotherapies in advanced development such as Listeria-based vaccines. Due to its unique life cycle and ability to induce robust CTL responses, attenuated strains of Listeria monocytogenes (Lm) have been utilized as vaccine vectors targeting both infectious disease and cancer. In fact, preclinical studies in a multitude of cancer types have found Listeria-based vaccines to be highly effective at activating anti-tumor immunity and eradicating tumors. Several clinical trials have now recently reported their results, demonstrating promising efficacy against some cancers, and unique challenges. Development of the Lm-based immunotherapies continues with discovery of improved methods of attenuation, novel uses, and more effective combinatorial regimens. In this review, we provide a brief background of Listeria monocytogenes as a vaccine vector, discuss recent clinical experience with Listeria-based immunotherapies, and detail the advancements in development of improved Listeria-based vaccine platforms and in their utilization.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Listeria monocytogenes , Neoplasias/terapia , Animales , Vectores Genéticos , Humanos
13.
Vaccine ; 39(8): 1257-1264, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33518468

RESUMEN

Reverse genetically engineered recombinant lymphocytic choriomeningitis virus (rLCMV) is a novel vaccine vector platform. Here, we investigate the safety and efficacy of rLCMV in mice lacking a functional type I interferon system with high susceptibility to viral infections. Propagation-deficient rLCMV vector expressing ovalbumin as a model antigen is cleared from type I interferon receptor-deficient mice (Ifnar-/-) within seven days post vaccination. In Ifnar-/-, induction of vaccine antigen specific T cells is delayed compared to wild type animals. However, immunization of Ifnar-/- results in potent memory formation and generates multifunctional cytotoxic CD8+ T cells. Most importantly, Ifnar-/- vaccinated with rLCMV are protected from a challenge with the aggressive LCMV Clone 13. Our data provide evidence for an excellent safety profile with maintained efficacy in immunocompromised animals.


Asunto(s)
Virus de la Coriomeningitis Linfocítica , Receptor de Interferón alfa y beta , Vacunas Virales , Animales , Linfocitos T CD8-positivos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Vacunas Sintéticas/genética
14.
Yale J Biol Med ; 83(4): 217-22, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21165341

RESUMEN

As our scientific knowledge of bacteria grows, so does our ability to manipulate these bacteria to protect rather than infect mammalian hosts from a diverse group of diseases. The old axiom that the best way to protect from a disease is to get infected in the first place is not feasible in the face of the diverse group of pathogens that infect humans. Therefore, reprogramming bacteria to protect against diverse bacterial, viral, and parasitic diseases as well as cancer is a new reality in the field of vaccines.


Asunto(s)
Bacterias/inmunología , Vacunas/inmunología , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología , Animales , Bacterias/genética , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Vacunas/genética , Vacunas/metabolismo
15.
Curr Immunol Rev ; 15(1): 102-122, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452652

RESUMEN

Optimal protective immunity to HIV will likely require that plasma cells, memory B cells and memory T cells be stationed in mucosal tissues at portals of viral entry. Mucosal vaccine administration is more effective than parenteral vaccine delivery for this purpose. The challenge has been to achieve efficient vaccine uptake at mucosal surfaces, and to identify safe and effective adjuvants, especially for mucosally administered HIV envelope protein immunogens. Here, we discuss strategies used to deliver potential HIV vaccine candidates in the intestine, respiratory tract, and male and female genital tract of humans and nonhuman primates. We also review mucosal adjuvants, including Toll-like receptor agonists, which may adjuvant both mucosal humoral and cellular immune responses to HIV protein immunogens.

16.
Viruses ; 11(9)2019 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-31540123

RESUMEN

Vesicular stomatitis virus (VSV) represents a promising platform for developing oncolytic viruses, as well as vaccines against significant human pathogens. To safely control VSV infection in humans, small-molecule drugs that selectively inhibit VSV infection may be needed. Here, using a cell-based high-throughput screening assay followed by an in vitro transcription assay, compounds with a 7-hydroxy-6-methyl-3,4-dihydroquinolin-2(1H)-one structure and an aromatic group at position 4 (named vesiculopolins, VPIs) were identified as VSV RNA polymerase inhibitors. The most effective compound, VPI A, inhibited VSV-induced cytopathic effects and in vitro mRNA synthesis with micromolar to submicromolar 50% inhibitory concentrations. VPI A was found to inhibit terminal de novo initiation rather than elongation for leader RNA synthesis, but not mRNA capping, with the VSV L protein, suggesting that VPI A is targeted to the polymerase domain in the L protein. VPI A inhibited transcription of Chandipura virus, but not of human parainfluenza virus 3, suggesting that it specifically acts on vesiculoviral L proteins. These results suggest that VPIs may serve not only as molecular probes to elucidate the mechanisms of transcription of vesiculoviruses, but also as lead compounds to develop antiviral drugs against vesiculoviruses and other related rhabdoviruses.


Asunto(s)
Antivirales/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Transcripción Genética/efectos de los fármacos , Vesiculovirus/efectos de los fármacos , Vesiculovirus/genética , Animales , Línea Celular , Cricetinae , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , ARN Viral , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Virus de la Estomatitis Vesicular Indiana/genética , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos
17.
Hum Gene Ther ; 30(4): 429-445, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30351174

RESUMEN

Antigen delivery platforms based on engineered viruses or virus-like particles are currently developed as vaccines against infectious diseases. As the interaction of vaccines with dendritic cells (DCs) shapes the immunological response, we compared the interaction of a range of virus-based vectors and virus-like particles with DCs in a murine model of systemic administration and transcriptome analyses of splenic DCs. The transcriptome profiles of DCs separated the vaccine vectors into two distinct groups characterized by high- and low-magnitude differential gene expression, which strongly correlated with (1) the surface expression of costimulatory molecules CD40, CD83, and CD86 on DCs, and (2) antigen-specific T-cell responses. Pathway analysis using PANOGA (Pathway and Network-Oriented GWAS Analysis) revealed that the JAK/STAT pathway was significantly activated by both groups of vaccines. In contrast, the oxidative phosphorylation pathway was significantly downregulated only by the high-magnitude DC-stimulating vectors. A gene signature including exclusively chemokine-, cytokine-, and receptor-related genes revealed a vector-specific pattern. Overall, this in vivo DC stimulation model demonstrated a strong relationship between the levels of induced DC maturation and the intensity of T-cell-specific immune responses with a distinct cytokine/chemokine profile, metabolic shifting, and cell surface expression of maturation markers. It could represent an important tool for vaccine design.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Vectores Genéticos/genética , Fosforilación Oxidativa , Transcripción Genética , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología , Animales , Biomarcadores , Biología Computacional/métodos , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Anotación de Secuencia Molecular , Bazo/citología , Bazo/inmunología , Transcriptoma , Vacunas de Partículas Similares a Virus/administración & dosificación
18.
Virology ; 515: 150-157, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29288958

RESUMEN

The clinical use of many adenovirus vaccine vectors (AdVs) is limited by the presence of pre-existing antibodies in human populations, which prevent common AdVs from transducing cells and expressing immunogenic gene products. Rare serotype AdVs, such as HAdV-28D can bypass pre-existing immunity. However, rare AdVs stimulate high-levels of type I interferon (IFN), which suppresses antigenic gene expression and therefore limits immunogenicity. Recent studies identified Gas6 as a factor that connects enveloped viruses to host-cell receptor tyrosine kinases, in turn generating signaling cascades that antagonize type I IFN responses. We discovered that Gas6 bound to the fiber proteins of common AdV serotypes, such as HAdV-5C, with a higher affinity than rare HAd-28D fibers. AdV-associated Gas6 suppressed IFN production by common AdVs and enhanced long-term expression of AdV encoded genes. We hypothesize that rare AdV serotypes might be engineered to include Gas6 binding motifs, thereby generating novel vectors that are more effective.


Asunto(s)
Infecciones por Adenoviridae/metabolismo , Adenoviridae/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interferón beta/metabolismo , Adenoviridae/clasificación , Adenoviridae/genética , Adenoviridae/aislamiento & purificación , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/virología , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Interacciones Huésped-Patógeno , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Interferón beta/genética , Unión Proteica , Serogrupo
19.
Immunotherapy ; 10(16): 1349-1360, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30474481

RESUMEN

AIM: Lung cancer gene therapies require reagents to selectively transfect lung tumors after systemic administration. MATERIALS & METHODS: We created a reagent called NSCLC-NP by attaching a peptide with binding affinity for lung cancer to polyamidoamine dendrimers. The positively charged dendrimers electrostatically bind negatively charged nucleic acids, inhibit endogenous nucleases and transfect cells targeted by the attached peptide. RESULTS: In vitro, NSCLC-NP complexed to DNA plasmids bound and transfected three human lung cancer cell lines producing protein expression of the plasmid's gene. In vivo, systemically administered NSCLC-NP selectively transfected lung cancer cells growing in RAG1KO mice. CONCLUSION: The capability of NSCLC-NP to selectively transfect lung cancer allows its future use as a vehicle to implement human lung cancer gene therapy strategies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Dendrímeros/farmacología , Neoplasias Pulmonares , Plásmidos/farmacología , Transfección/métodos , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Dendrímeros/química , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Noqueados , Plásmidos/química
20.
Front Immunol ; 9: 1260, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29951052

RESUMEN

CD4+ T cell differentiation is influenced by a plethora of intrinsic and extrinsic factors, providing the immune system with the ability to tailor its response according to specific stimuli. Indeed, different classes of pathogens may induce a distinct balance of CD4+ T cell differentiation programmes. Here, we report an uncommonly strong bias toward follicular helper (Tfh) differentiation of CD4+ T cells reactive with a retroviral envelope glycoprotein model antigen, presented in its natural context during retroviral infection. Conversely, the response to the same antigen, presented in different immunization regimens, elicited a response typically balanced between Tfh and T helper 1 cells. Comprehensive quantitation of variables known to influence Tfh differentiation revealed the closest correlation with the strength of T cell receptor (TCR) signaling, leading to PD-1 expression, but not with surface TCR downregulation, irrespective of TCR clonotypic avidity. In contrast, strong TCR signaling leading to TCR downregulation and induction of LAG3 expression in high TCR avidity clonotypes restrained CD4+ T cell commitment and further differentiation. Finally, stunted Th1 differentiation, correlating with limited IL-2 availability in retroviral infection, provided permissive conditions for Tfh development, suggesting that Tfh differentiation is the default program of envelope-reactive CD4+ T cells.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones por Retroviridae/inmunología , Retroviridae/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citocinas/metabolismo , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Ratones Transgénicos , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/virología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA