Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 306
Filtrar
1.
J Virol ; 95(20): e0079021, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34346765

RESUMEN

Rabies is a fatal zoonosis that causes encephalitis in mammals, and vaccination is the most effective method to control and eliminate rabies. Virus-like vesicles (VLVs), which are characterized as infectious, self-propagating membrane-enveloped particles composed of only Semliki Forest virus (SFV) replicase and vesicular stomatitis virus glycoprotein (VSV-G), have been proven safe and efficient as vaccine candidates. However, previous studies showed that VLVs containing rabies virus glycoprotein (RABV-G) grew at relatively low titers in cells, impeding their potential use as a rabies vaccine. In this study, we constructed novel VLVs by transfection of a mutant SFV RNA replicon encoding RABV-G. We found that these VLVs could self-propagate efficiently in cell culture and could evolve to high titers (approximately 108 focus-forming units [FFU]/ml) by extensive passaging 25 times in BHK-21 cells. Furthermore, we found that the evolved amino acid changes in SFV nonstructural protein 1 (nsP1) at positions 470 and 482 was critical for this high-titer phenotype. Remarkably, VLVs could induce robust type I interferon (IFN) expression in BV2 cells and were highly sensitive to IFN-α. We found that direct inoculation of VLVs into the mouse brain caused reduced body weight loss, mortality, and neuroinflammation compared with the RABV vaccine strain. Finally, it could induce increased generation of germinal center (GC) B cells, plasma cells (PCs), and virus-neutralizing antibodies (VNAs), as well as provide protection against virulent RABV challenge in immunized mice. This study demonstrated that VLVs containing RABV-G could proliferate in cells and were highly evolvable, revealing the feasibility of developing an economic, safe, and efficacious rabies vaccine. IMPORTANCE VLVs have been shown to represent a more versatile and superior vaccine platform. In previous studies, VLVs containing the Semliki Forest virus replicase (SFV nsP1 to nsP4) and rabies virus glycoprotein (RABV-G) grew to relatively low titers in cells. In our study, we not only succeeded in generating VLVs that proliferate in cells and stably express RABV-G, but the VLVs that evolved grew to higher titers, reaching 108 FFU/ml. We also found that nucleic acid changes at positions 470 and 482 in nsP1 were vital for this high-titer phenotype. Moreover, the VLVs that evolved in our studies were highly attenuated in mice, induced potent immunity, and protected mice from lethal RABV infection. Collectively, our study showed that high titers of VLVs containing RABV-G were achieved, demonstrating that these VLVs could be an economical, safe, and efficacious rabies vaccine candidate.


Asunto(s)
Vacunas Antirrábicas/inmunología , Rabia/inmunología , Vacunación/métodos , Animales , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Femenino , Ingeniería Genética/métodos , Centro Germinal/inmunología , Glicoproteínas/genética , Inmunización/métodos , Masculino , Ratones , Ratones Endogámicos ICR , Rabia/metabolismo , Vacunas Antirrábicas/metabolismo , Vacunas Antirrábicas/farmacología , Virus de la Rabia/inmunología , Virus de los Bosques Semliki/inmunología , Vesiculovirus/genética , Proteínas Virales/genética
2.
Science ; 371(6528)2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33243852

RESUMEN

Inflammasomes function as intracellular sensors of pathogen infection or cellular perturbation and thereby play a central role in numerous diseases. Given the high abundance of NLRP1 in epithelial barrier tissues, we screened a diverse panel of viruses for inflammasome activation in keratinocytes. We identified Semliki Forest virus (SFV), a positive-strand RNA virus, as a potent activator of human but not murine NLRP1B. SFV replication and the associated formation of double-stranded (ds) RNA was required to engage the NLRP1 inflammasome. Moreover, delivery of long dsRNA was sufficient to trigger activation. Biochemical studies revealed that NLRP1 binds dsRNA through its leucine-rich repeat domain, resulting in its NACHT domain gaining adenosine triphosphatase activity. Altogether, these results establish human NLRP1 as a direct sensor for dsRNA and thus RNA virus infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Inflamasomas/inmunología , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Infecciones por Alphavirus/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/química , Células HEK293 , Humanos , Hidrólisis , Inflamasomas/metabolismo , Queratinocitos/inmunología , Queratinocitos/metabolismo , Queratinocitos/virología , Ratones , Ratones Transgénicos , Proteínas NLR , Dominios Proteicos , Virus de los Bosques Semliki/inmunología , Virus de los Bosques Semliki/fisiología , Replicación Viral
3.
Am J Trop Med Hyg ; 103(6): 2429-2437, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33124532

RESUMEN

Madariaga virus (MADV) has recently been associated with severe human disease in Panama, where the closely related Venezuelan equine encephalitis virus (VEEV) also circulates. In June 2017, a fatal MADV infection was confirmed in a community of Darien Province. We conducted a cross-sectional outbreak investigation with human and mosquito collections in July 2017, where sera were tested for alphavirus antibodies and viral RNA. In addition, by applying a catalytic, force-of-infection (FOI) statistical model to two serosurveys from Darien Province in 2012 and 2017, we investigated whether endemic or epidemic alphavirus transmission occurred historically. In 2017, MADV and VEEV IgM seroprevalences were 1.6% and 4.4%, respectively; IgG antibody prevalences were MADV: 13.2%, VEEV: 16.8%, Una virus (UNAV): 16.0%, and Mayaro virus: 1.1%. Active viral circulation was not detected. Evidence of MADV and UNAV infection was found near households, raising questions about its vectors and enzootic transmission cycles. Insomnia was associated with MADV and VEEV infections, depression symptoms were associated with MADV, and dizziness with VEEV and UNAV. Force-of-infection analyses suggest endemic alphavirus transmission historically, with recent increased human exposure to MADV and VEEV in Aruza and Mercadeo, respectively. The lack of additional neurological cases suggests that severe MADV and VEEV infections occur only rarely. Our results indicate that over the past five decades, alphavirus infections have occurred at low levels in eastern Panama, but that MADV and VEEV infections have recently increased-potentially during the past decade. Endemic infections and outbreaks of MADV and VEEV appear to differ spatially in some locations of eastern Panama.


Asunto(s)
Encefalomielitis Equina Oriental/epidemiología , Encefalomielitis Equina Venezolana/epidemiología , Agricultores/estadística & datos numéricos , Adolescente , Adulto , Distribución por Edad , Anciano , Anciano de 80 o más Años , Alphavirus/inmunología , Infecciones por Alphavirus/epidemiología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/fisiopatología , Animales , Anticuerpos Antivirales/inmunología , Fiebre Chikungunya/epidemiología , Fiebre Chikungunya/inmunología , Fiebre Chikungunya/fisiopatología , Virus Chikungunya/inmunología , Niño , Preescolar , Estudios Transversales , Depresión/fisiopatología , Mareo/fisiopatología , Virus de la Encefalitis Equina del Este/inmunología , Virus de la Encefalitis Equina Venezolana/inmunología , Encefalomielitis Equina Oriental/inmunología , Encefalomielitis Equina Oriental/fisiopatología , Encefalomielitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/fisiopatología , Enfermedades Endémicas , Epidemias , Fatiga/fisiopatología , Femenino , Vivienda/estadística & datos numéricos , Humanos , Inmunoglobulina G , Inmunoglobulina M , Masculino , Persona de Mediana Edad , Mosquitos Vectores/virología , Panamá/epidemiología , Virus de los Bosques Semliki/inmunología , Estudios Seroepidemiológicos , Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología , Adulto Joven
4.
Viruses ; 11(1)2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30669393

RESUMEN

BACKGROUND: Alphaviruses are transmitted by arthropod vectors and can be found worldwide. Alphaviruses of the Semliki Forest complex such as chikungunya virus (CHIKV), Mayaro virus (MAYV) or Ross River virus (RRV) cause acute febrile illness and long-lasting arthralgia in humans, which cannot be clinically discriminated from a dengue virus or Zika virus infection. Alphaviruses utilize a diverse array of mosquito vectors for transmission and spread. For instance, adaptation of CHIKV to transmission by Aedes albopictus has increased its spread and resulted in large outbreaks in the Indian Ocean islands. For many alphaviruses commercial diagnostic tests are not available or show cross-reactivity among alphaviruses. Climate change and globalization will increase the spread of alphaviruses and monitoring of infections is necessary and requires virus-specific methods. METHOD: We established an alphavirus neutralization assay in a 384-well format by using pseudotyped lentiviral vectors. RESULTS: MAYV-specific reactivity could be discriminated from CHIKV reactivity. Human plasma from blood donors infected with RRV could be clearly identified and did not cross-react with other alphaviruses. CONCLUSION: This safe and easy to use multiplex assay allows the discrimination of alphavirus-specific reactivity within a single assay and has potential for epidemiological surveillance. It might also be useful for the development of a pan-alphavirus vaccine.


Asunto(s)
Infecciones por Alphavirus/diagnóstico , Infecciones por Alphavirus/inmunología , Pruebas de Neutralización/métodos , Virus del Río Ross/inmunología , Animales , Virus Chikungunya/inmunología , Reacciones Cruzadas , Ensayo de Inmunoadsorción Enzimática , Humanos , Concentración 50 Inhibidora , Mosquitos Vectores/virología , Virus de los Bosques Semliki/inmunología , Sensibilidad y Especificidad
5.
Viruses ; 10(12)2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30563205

RESUMEN

Arthropod vectors control the replication of arboviruses through their innate antiviral immune responses. In particular, the RNA interference (RNAi) pathways are of notable significance for the control of viral infections. Although much has been done to understand the role of RNAi in vector populations, little is known about its importance in non-vector mosquito species. In this study, we investigated the presence of an RNAi response in Toxorhynchites amboinensis, which is a non-blood feeding species proposed as a biological control agent against pest mosquitoes. Using a derived cell line (TRA-171), we demonstrate that these mosquitoes possess a functional RNAi response that is active against a mosquito-borne alphavirus, Semliki Forest virus. As observed in vector mosquito species, small RNAs are produced that target viral sequences. The size and characteristics of these small RNAs indicate that both the siRNA and piRNA pathways are induced in response to infection. Taken together, this data suggests that Tx. amboinensis are able to control viral infections in a similar way to natural arbovirus vector mosquito species. Understanding their ability to manage arboviral infections will be advantageous when assessing these and similar species as biological control agents.


Asunto(s)
Culicidae/genética , Culicidae/virología , Interferencia de ARN , Virus de los Bosques Semliki/genética , Infecciones por Alphavirus/inmunología , Animales , Agentes de Control Biológico , Línea Celular , Culicidae/citología , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunidad Innata , Mosquitos Vectores/genética , Mosquitos Vectores/virología , ARN Interferente Pequeño/genética , Virus de los Bosques Semliki/inmunología , Replicación Viral
6.
Viruses ; 10(5)2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29783708

RESUMEN

Alphaviruses are mosquito-transmitted RNA viruses which generally cause acute disease including mild febrile illness, rash, arthralgia, myalgia and more severely, encephalitis. In the mouse, peripheral infection with Semliki Forest virus (SFV) results in encephalitis. With non-virulent strains, infectious virus is detectable in the brain, by standard infectivity assays, for around ten days. As we have shown previously, in severe combined immunodeficient (SCID) mice, infectious virus is detectable for months in the brain. Here we show that in MHC-II-/- mice, with no functional CD4 T-cells, infectious virus is also detectable in the brain for long periods. In contrast, in the brains of CD8-/- mice, virus RNA persists but infectious virus is not detectable. In SCID mice infected with SFV, repeated intraperitoneal administration of anti-SFV immune serum rapidly reduced the titer of infectious virus in the brain to undetectable, however virus RNA persisted. Repeated intraperitoneal passive transfer of immune serum resulted in maintenance of brain virus RNA, with no detectable infectious virus, for several weeks. When passive antibody transfer was stopped, antibody levels declined and infectious virus was again detectable in the brain. In aged immunocompetent mice, previously infected with SFV, immunosuppression of antibody responses many months after initial infection also resulted in renewed ability to detect infectious virus in the brain. In summary, antiviral antibodies control and determine whether infectious virus is detectable in the brain but immune responses cannot clear this infection from the brain. Functional virus RNA capable of generating infectious virus persists and if antibody levels decline, infectious virus is again detectable.


Asunto(s)
Encéfalo/virología , Encefalitis Viral/inmunología , Encefalitis Viral/virología , ARN Viral/inmunología , Virus de los Bosques Semliki/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Encéfalo/inmunología , Línea Celular , Cricetinae , Ciclofosfamida/farmacología , Sueros Inmunes/inmunología , Sueros Inmunes/aislamiento & purificación , Inmunidad Celular/efectos de los fármacos , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos , Ratones SCID , ARN Viral/genética , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/fisiología , Linfocitos T/inmunología , Ensayo de Placa Viral
7.
J Virol ; 91(6)2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28077641

RESUMEN

Recombinant vesicular stomatitis virus (VSV)-based chimeric viruses that include genes from other viruses show promise as vaccines and oncolytic viruses. However, the critical safety concern is the neurotropic nature conveyed by the VSV glycoprotein. VSVs that include the VSV glycoprotein (G) gene, even in most recombinant attenuated strains, can still show substantial adverse or lethal actions in the brain. Here, we test 4 chimeric viruses in the brain, including those in which glycoprotein genes from Nipah, chikungunya (CHIKV), and influenza H5N1 viruses were substituted for the VSV glycoprotein gene. We also test a virus-like vesicle (VLV) in which the VSV glycoprotein gene is expressed from a replicon encoding the nonstructural proteins of Semliki Forest virus. VSVΔG-CHIKV, VSVΔG-H5N1, and VLV were all safe in the adult mouse brain, as were VSVΔG viruses expressing either the Nipah F or G glycoprotein. In contrast, a complementing pair of VSVΔG viruses expressing Nipah G and F glycoproteins were lethal within the brain within a surprisingly short time frame of 2 days. Intranasal inoculation in postnatal day 14 mice with VSVΔG-CHIKV or VLV evoked no adverse response, whereas VSVΔG-H5N1 by this route was lethal in most mice. A key immune mechanism underlying the safety of VSVΔG-CHIKV, VSVΔG-H5N1, and VLV in the adult brain was the type I interferon response; all three viruses were lethal in the brains of adult mice lacking the interferon receptor, suggesting that the viruses can infect and replicate and spread in brain cells if not blocked by interferon-stimulated genes within the brain.IMPORTANCE Vesicular stomatitis virus (VSV) shows considerable promise both as a vaccine vector and as an oncolytic virus. The greatest limitation of VSV is that it is highly neurotropic and can be lethal within the brain. The neurotropism can be mostly attributed to the VSV G glycoprotein. Here, we test 4 chimeric viruses of VSV with glycoprotein genes from Nipah, chikungunya, and influenza viruses and nonstructural genes from Semliki Forest virus. Two of the four, VSVΔG-CHIKV and VLV, show substantially attenuated neurotropism and were safe in the healthy adult mouse brain. VSVΔG-H5N1 was safe in the adult brain but lethal in the younger brain. VSVΔG Nipah F+G was even more neurotropic than wild-type VSV, evoking a rapid lethal response in the adult brain. These results suggest that while chimeric VSVs show promise, each must be tested with both intranasal and intracranial administration to ensure the absence of lethal neurotropism.


Asunto(s)
Encéfalo/patología , Vesiculovirus/patogenicidad , Vacunas Virales/efectos adversos , Animales , Virus Chikungunya/genética , Virus Chikungunya/inmunología , Interferón Tipo I/metabolismo , Ratones , Virus Nipah/genética , Virus Nipah/inmunología , Orthomyxoviridae/genética , Orthomyxoviridae/inmunología , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/inmunología , Análisis de Supervivencia , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vesiculovirus/genética , Vesiculovirus/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
8.
Vet Microbiol ; 206: 10-20, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28069290

RESUMEN

Due to its impact on animal health and pig industry, classical swine fever (CSF) is still one of the most important viral diseases of pigs. To control the disease, safe and highly efficacious live attenuated vaccines exist for decades. These vaccines have usually outstanding efficacy and safety but lack differentiability of infected from vaccinated animals (DIVA or marker strategy). In contrast, the first generation of E2 subunit marker vaccines shows constraints in efficacy, application, and production. To overcome these limitations, new generations of marker vaccines are developed. A wide range of approaches have been tried including recombinant vaccines, recombinant inactivated vaccines or subunit vaccines, vector vaccines, and DNA/RNA vaccines. During the last years, especially attenuated deletion vaccines or chimeric constructs have shown potential. At present, especially two new constructs have been intensively tested, the adenovirus-delivered, Semliki Forest virus replicon-vectored marker vaccine candidate "rAdV-SFV-E2" and the pestivirus chimera "CP7_E2alf". The later was recently licensed by the European Medicines Agency. Under field conditions, all marker vaccines have to be accompanied by a potent test system. Particularly this point shows still weaknesses and it is important to embed vaccination in a well-established vaccination strategy and a suitable diagnostic workflow. In summary, conventional vaccines are a standard in terms of efficacy. However, only vaccines with DIVA will allow improved eradication strategies e.g. also under emergency vaccination conditions in free regions. To answer this demand, new generations of marker vaccines have been developed and add now to the tool box of CSF control.


Asunto(s)
Virus de la Fiebre Porcina Clásica/inmunología , Peste Porcina Clásica/prevención & control , Vacunación/veterinaria , Vacunas Virales/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Peste Porcina Clásica/virología , Vectores Genéticos , Replicón , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/inmunología , Porcinos , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/inmunología , Vacunas Marcadoras/inmunología
9.
J Virol ; 89(20): 10407-15, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26246574

RESUMEN

UNLABELLED: More than 500,000 people die each year from the liver diseases that result from chronic hepatitis B virus (HBV) infection. Therapeutic vaccines, which aim to elicit an immune response capable of controlling the virus, offer a potential new treatment strategy for chronic hepatitis B. Recently, an evolved, high-titer vaccine platform consisting of Semliki Forest virus RNA replicons that express the vesicular stomatitis virus glycoprotein (VSV G) has been described. This platform generates virus-like vesicles (VLVs) that contain VSV G but no other viral structural proteins. We report here that the evolved VLV vector engineered to additionally express the HBV middle surface envelope glycoprotein (MHBs) induces functional CD8 T cell responses in mice. These responses were greater in magnitude and broader in specificity than those obtained with other immunization strategies, including recombinant protein and DNA. Additionally, a single immunization with VLV-MHBs protected mice from HBV hydrodynamic challenge, and this protection correlated with the elicitation of a CD8 T cell recall response. In contrast to MHBs, a VLV expressing HBV core protein (HBcAg) neither induced a CD8 T cell response in mice nor protected against challenge. Finally, combining DNA and VLV-MHBs immunization led to induction of HBV-specific CD8 T cell responses in a transgenic mouse model of chronic HBV infection. The ability of VLV-MHBs to induce a multispecific T cell response capable of controlling HBV replication, and to generate immune responses in a tolerogenic model of chronic infection, indicates that VLV vaccine platforms may offer a unique strategy for HBV therapeutic vaccination. IMPORTANCE: HBV infection is associated with significant morbidity and mortality. Furthermore, treatments for chronic infection are suboptimal and rarely result in complete elimination of the virus. Therapeutic vaccines represent a unique approach to HBV treatment and have the potential to induce long-term control of infection. Recently, a virus-based vector system that combines the nonstructural proteins of Semliki Forest virus with the VSV glycoprotein has been described. In this study, we used this system to construct a novel HBV vaccine and demonstrated that the vaccine is capable of inducing virus-specific immune responses in mouse models of acute and chronic HBV replication. These findings highlight the potential of this new vaccine system and support the idea that highly immunogenic vaccines, such as viral vectors, may be useful in the treatment of chronic hepatitis B.


Asunto(s)
Vacunas contra Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/prevención & control , Inmunidad Celular/efectos de los fármacos , Vacunas de Partículas Similares a Virus/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Línea Celular , Cricetulus , Ensayo de Immunospot Ligado a Enzimas , Células Epiteliales/inmunología , Células Epiteliales/virología , Ingeniería Genética , Vectores Genéticos/química , Vectores Genéticos/inmunología , Glicoproteínas/genética , Glicoproteínas/inmunología , Antígenos del Núcleo de la Hepatitis B/genética , Antígenos del Núcleo de la Hepatitis B/inmunología , Vacunas contra Hepatitis B/administración & dosificación , Vacunas contra Hepatitis B/genética , Virus de la Hepatitis B/genética , Hepatitis B Crónica/genética , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Inmunización , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Replicación Viral/efectos de los fármacos
10.
PLoS One ; 9(10): e109678, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25360552

RESUMEN

Identification of transmitted/founder simian immunodeficiency virus (SIV) envelope sequences responsible for infection may prove critical for understanding HIV/SIV mucosal transmission. We used single genome amplification and phylogenetic analyses to characterize transmitted/founder SIVs both in the inoculum and in immunized-infected rhesus monkeys. Single genome amplification of the SIVsmE660 inoculum revealed a maximum diversity of 1.4%. We also noted that the consensus sequence of the challenge stock differed from the vaccine construct in 10 amino acids including 3 changes in the V4 loop. Viral env was prepared from rhesus plasma in 3 groups of 6 immunized with vesicular stomatitis virus (VSV) vectors and boosted with Semliki forest virus (SFV) replicons expressing (a) SIVsmE660 gag-env (b) SIVsmE660 gag-env plus rhesus GM-CSF and (c) control influenza hemagglutinin protein. Macaques were immunized twice with VSV-vectors and once with SFV vector and challenged intrarectally with 4000 TCID50. Single genome amplification characterized the infections of 2 unprotected animals in the gag-env immunized group, both of which had reduced acute plasma viral loads that ended as transient infections indicating partial immune control. Four of 6 rhesus were infected in the gag-env + GM-CSF group which demonstrated that GM-CSF abrogated protection. All 6 animals from the control group were infected having high plasma viral loads. We obtained 246 full-length envelope sequences from SIVsmE660 infected macaques at the peak of infection and determined the number of transmitted/founder variants per animal. Our analysis found that 2 of 2 gag-env vaccinated but infected macaques exhibited single but distinct virus envelope lineages whereas rhesus vaccinated with gag-env-GM-CSF or HA control exhibited both single and multiple env lineages. Because there were only 2 infected animals in the gag-env vaccinated rhesus compared to 10 infected rhesus in the other 2 groups, the significance of finding single env variants in the gag-env vaccinated group could not be established.


Asunto(s)
Macaca mulatta/virología , Filogenia , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Proteínas del Envoltorio Viral/genética , Animales , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen gag/genética , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inmunización , Macaca mulatta/inmunología , Datos de Secuencia Molecular , Vacunas contra el SIDAS/inmunología , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Estomatitis Vesicular/virología , Proteínas del Envoltorio Viral/inmunología , Carga Viral
11.
J Virol ; 88(17): 9553-67, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899190

RESUMEN

UNLABELLED: The encephalitic response to viral infection requires local chemokine production and the ensuing recruitment of immune and inflammatory leukocytes. Accordingly, chemokine receptors present themselves as plausible therapeutic targets for drugs aimed at limiting encephalitic responses. However, it remains unclear which chemokines are central to this process and whether leukocyte recruitment is important for limiting viral proliferation and survival in the brain or whether it is predominantly a driver of coincident inflammatory pathogenesis. Here we examine chemokine expression and leukocyte recruitment in the context of avirulent and virulent Semliki Forest virus (SFV) as well as West Nile virus infection and demonstrate rapid and robust expression of a variety of inflammatory CC and CXC chemokines in all models. On this basis, we define a chemokine axis involved in leukocyte recruitment to the encephalitic brain during SFV infection. CXCR3 is the most active; CCR2 is also active but less so, and CCR5 plays only a modest role in leukocyte recruitment. Importantly, inhibition of each of these receptors individually and the resulting suppression of leukocyte recruitment to the infected brain have no effect on viral titer or survival following infection with a virulent SFV strain. In contrast, simultaneous blockade of CXCR3 and CCR2 results in significantly reduced mortality in response to virulent SFV infection. In summary, therefore, our data provide an unprecedented level of insight into chemokine orchestration of leukocyte recruitment in viral encephalitis. Our data also highlight CXCR3 and CCR2 as possible therapeutic targets for limiting inflammatory damage in response to viral infection of the brain. IMPORTANCE: Brain inflammation (encephalitis) in response to viral infection can lead to severe illness and even death. This therefore represents an important clinical problem and one that requires the development of new therapeutic approaches. Central to the pathogenesis of encephalitis is the recruitment of inflammatory leukocytes to the infected brain, a process driven by members of the chemokine family. Here we provide an in-depth analysis of the chemokines involved in leukocyte recruitment to the virally infected brain and demonstrate that simultaneous blockade of two of these receptors, namely, CXCR3 and CCR2, does not alter viral titers within the brain but markedly reduces inflammatory leukocyte recruitment and enhances survival in a murine model of lethal viral encephalitis. Our results therefore highlight chemokine receptors as plausible therapeutic targets in treating viral encephalitis.


Asunto(s)
Infecciones por Alphavirus/inmunología , Quimiocinas/metabolismo , Encefalitis Viral/inmunología , Leucocitos/inmunología , Fiebre del Nilo Occidental/inmunología , Animales , Encéfalo/patología , Encéfalo/virología , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Receptores de Quimiocina/inmunología , Virus de los Bosques Semliki/inmunología , Análisis de Supervivencia , Carga Viral , Virus del Nilo Occidental/inmunología
12.
PLoS One ; 9(3): e90551, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24608380

RESUMEN

A DNA-based replicon vaccine derived from Semliki Forest virus, PSVK-shFcG-GM/B7.1 (Fig. 1a) was designed for tumor immunotherapy as previously constructed. The expression of the fusion tumor antigen (survivin and hCGß-CTP37) and adjuvant molecular protein (Granulocyte-Macrophage Colony-Stimulating Factor/ GM-CSF/B7.1) genes was confirmed by Immunofluorescence assay in vitro, and immunohistochemistry assay in vivo. In this paper, the immunological effect of this vaccine was determined using immunological assays as well as animal models. The results showed that this DNA vaccine induced both humoral and cellular immune responses in C57BL/6 mice after immunization, as evaluated by the ratio of CD4+/CD8+ cells and the release of IFN-γ. Furthermore, the vaccination of C57BL/6 mice with PSVK-shFcG-GM/B7.1 significantly delayed the in vivo growth of tumors in animal models (survivin+ and hCGß+ murine melanoma, B16) when compared to vaccination with the empty vector or the other control constructs (Fig. 1b). These data indicate that this type of replicative DNA vaccine could be developed as a promising approach for tumor immunotherapy. Meanwhile, these results provide a basis for further study in vaccine pharmacodynamics and pharmacology, and lay a solid foundation for clinical application.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Replicón/inmunología , Virus de los Bosques Semliki/inmunología , Vacunas de ADN/inmunología , Animales , Línea Celular Tumoral , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Ratones , Ratones Endogámicos C57BL
13.
PLoS One ; 8(6): e68458, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840854

RESUMEN

MHCII molecules are heterodimeric cell surface proteins composed of an α and ß chain. These molecules are almost exclusively expressed on thymic epithelium and antigen presenting cells (APCs) and play a central role in the development and function of CD4 T cells. Various MHC-II knockout mice have been generated including MHC-IIAα(-/-) (I-Aα(-/-)), MHC-IIAß(-/-) (I-ß(-/-)) and the double knockout (I-Aαxß(-/-)). Here we report a very striking observation, namely that alphaviruses including the avirulent strain of Semliki Forest virus (aSFV), which causes asymptomatic infection in wild-type C57BL6/J (B6) mice, causes a very acute and lethal infection in I-Aα(-/-), but not in I-ß(-/-) or I-Aαxß(-/-), mice. This susceptibility to aSFV is associated with high virus titres in muscle, spleen, liver, and brain compared to B6 mice. In addition, I-Aα(-/-) mice show intact IFN-I responses in terms of IFN-I serum levels and IFN-I receptor expression and function. Radiation bone marrow chimeras of B6 mice reconstituted with I-Aα(-/-) bone marrow expressed B6 phenotype, whereas radiation chimeras of I-Aα(-/-) mice reconstituted with B6 bone marrow expressed the phenotype of high viral susceptibility. Virus replication experiments both in vivo and in vitro showed enhanced virus growth in tissues and cell cultures derived form I-Aα(-/-) compared to B6 mice. This enhanced virus replication is evident for other alpha-, flavi- and poxviruses and may be of great benefit to producers of viral vaccines. In conclusion, I-Aα(-/-) mice exhibit a striking susceptibility to virus infections independent of their defective MHC-II expression. Detailed genetic analysis will be carried out to characterise the underlining genetic defects responsible for the observed phenomenon.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Síndromes de Inmunodeficiencia/inmunología , Replicación Viral/inmunología , Animales , Médula Ósea/inmunología , Médula Ósea/virología , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de los Bosques Semliki/inmunología , Células Vero , Replicación Viral/genética
14.
J Immunol ; 190(6): 2994-3004, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23401594

RESUMEN

Semliki Forest virus vectors expressing IL-12 (SFV-IL-12) were shown to induce potent antitumor responses against s.c. MC38 colon adenocarcinomas in immunocompetent mice. However, when MC38 tumors were implanted in liver, where colon tumors usually metastasize, SFV-IL-12 efficacy was significantly reduced. We reasoned that characterization of immune responses against intrahepatic tumors in responder and nonresponder animals could provide useful information for designing more potent antitumor strategies. Remarkably, SFV-IL-12 induced a high percentage of circulating tumor-specific CD8 T cells in all treated animals. Depletion studies showed that these cells were essential for SFV-IL-12 antitumor activity. However, in comparison with nonresponders, tumor-specific cells from responder mice acquired an effector-like phenotype significantly earlier, were recruited more efficiently to the liver, and, importantly, persisted for a longer period of time. All treated mice had high levels of functional specific CD8 T cells at 8 d posttreatment reflected by both in vivo killing and IFN-γ-production assays, but responder animals showed a more avid and persistent IFN-γ response. Interestingly, differences in immune responses between responders and nonresponders seemed to correlate with the immune status of the animals before treatment and were not due to the treatment itself. Mice that rejected tumors were protected against tumor rechallenge, indicating that sustained memory responses are required for an efficacious therapy. Interestingly, tumor-specific CD8 T cells of responder animals showed upregulation of IL-15Rα expression compared with nonresponders. These results suggest that SFV-IL-12 therapy could benefit from the use of strategies that could either upregulate IL-15Rα expression or activate this receptor.


Asunto(s)
Interleucina-12/biosíntesis , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Hepáticas Experimentales/prevención & control , Virus de los Bosques Semliki/inmunología , Virus de los Bosques Semliki/metabolismo , Adenocarcinoma/inmunología , Adenocarcinoma/prevención & control , Adenocarcinoma/virología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/prevención & control , Infecciones por Alphavirus/virología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular , Células Cultivadas , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/prevención & control , Neoplasias Colorrectales/virología , Cricetinae , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Vectores Genéticos/metabolismo , Interleucina-12/genética , Neoplasias Hepáticas Experimentales/virología , Ratones , Ratones Endogámicos C57BL
15.
J Virol ; 87(4): 2363-6, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221568

RESUMEN

Attenuated Semliki Forest virus (SFV) may be suitable for targeting malignant glioma due to its natural neurotropism, but its replication in brain tumor cells may be restricted by innate antiviral defenses. We attempted to facilitate SFV replication in glioma cells by combining it with vaccinia virus, which is capable of antagonizing such defenses. Surprisingly, we found parenchymal mouse brain tumors to be refractory to both viruses. Also, vaccinia virus appears to be sensitive to SFV-induced antiviral interference.


Asunto(s)
Glioma/terapia , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/inmunología , Virus Vaccinia/crecimiento & desarrollo , Virus Vaccinia/inmunología , Animales , Modelos Animales de Enfermedad , Ratones
16.
Brain Res ; 1488: 92-103, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23031637

RESUMEN

Remyelination is one of the elusive topics in treatment of multiple sclerosis (MS). Our previous studies have shown that Semliki Forest virus (SFV)-infected δ-knock-out (KO) mice did not exhibit the extensive remyelination, seen in wild type (WT) B6 mice, after viral clearance and demyelination. The Remyelination in SFV-infected WT mice started on day 15 and was completed by day 35 post-infection (pi), whereas the KO mice remained partially demyelinated through day 42 pi. Treatment with E2 peptide2 in incomplete Freund's adjuvant (IFA), resulted in higher antibody production and earlier remyelination in SFV-infected KO (day 28 pi), than WT mice. This finding suggested that anti-E2 peptide2 antibody could play a part in remyelination. In the current study, the effect of E2 peptide2 treatment was evaluated in the experimental autoimmune encephalomyelitis (EAE) model. Mice with established EAE were treated with E2 peptide2 in IFA to develop antibody. Treated EAE mice made significantly higher anti-E2 peptide2 antibody than untreated EAE group. Average clinical disease scores were significantly lower in peptide treated compared to untreated EAE mice. Furthermore, histopathological and immunohistochemical studies demonstrated increased remyelinating areas and higher number of activated oligodendrocytes and astrocytes, in treated compared to untreated EAE groups. Moreover, the anti-E2 peptide2 antibody showed higher binding to the myelinated areas of treated than untreated EAE mice. We conclude that treatment with, or antibody to, SFV E2 peptide2 triggers some mechanism that promotes remyelination.


Asunto(s)
Anticuerpos Antivirales/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Regeneración Nerviosa/efectos de los fármacos , Péptidos/inmunología , Virus de los Bosques Semliki/inmunología , Animales , Astrocitos/inmunología , Astrocitos/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Inmunización , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Vaina de Mielina/inmunología , Vaina de Mielina/patología , Glicoproteína Mielina-Oligodendrócito/inmunología , Regeneración Nerviosa/inmunología , Oligodendroglía/inmunología , Oligodendroglía/patología , Péptidos/farmacología , Médula Espinal/inmunología , Médula Espinal/patología
17.
Mol Ther ; 20(9): 1664-75, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22735380

RESUMEN

Intratumoral injection of Semliki Forest virus encoding interleukin-12 (SFV-IL-12) combines acute expression of IL-12 and stressful apoptosis of infected malignant cells. Agonist antibodies directed to costimulatory receptor CD137 (4-1BB) strongly amplify pre-existing cellular immune responses toward weak tumor antigens. In this study, we provide evidence for powerful synergistic effects of a combined strategy consisting of intratumoral injection of SFV-IL-12 and systemic delivery of agonist anti-CD137 monoclonal antibodies (mAbs), which was substantiated against poorly immunogenic B16 melanomas (B16-OVA and B16.F10) and TC-1 lung carcinomas. Effector CD8(ß)(+) T cells were sufficient to mediate complete tumor eradications. Accordingly, there was an intensely synergistic in vivo enhancement of cytotoxic T lymphocytes (CTL)-mediated immunity against the tumor antigens OVA and tyrosine-related protein-2 (TRP-2). This train of phenomena led to long-lasting tumor-specific immunity against rechallenge, attained transient control of the progression of concomitant tumor lesions that were not directly treated with SFV-IL-12 and caused autoimmune vitiligo. Importantly, we found that SFV-IL-12 intratumoral injection induces bright expression of CD137 on most tumor-infiltrating CD8(+) T lymphocytes, thereby providing more abundant targets for the action of the agonist antibody. This efficacious combinatorial immunotherapy strategy offers feasibility for clinical translation since anti-CD137 mAbs are already undergoing clinical trials and development of clinical-grade SFV-IL-12 vectors is in progress.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Carcinoma/terapia , Interleucina-12/inmunología , Neoplasias Pulmonares/terapia , Melanoma Experimental/terapia , Virus de los Bosques Semliki/inmunología , Neoplasias Cutáneas/terapia , Animales , Anticuerpos Monoclonales/administración & dosificación , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Carcinoma/inmunología , Carcinoma/mortalidad , Línea Celular Tumoral , Cricetinae , Expresión Génica/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Inmunoterapia/métodos , Inyecciones Intralesiones , Inyecciones Intravenosas , Interleucina-12/administración & dosificación , Interleucina-12/genética , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Melanoma Experimental/inmunología , Melanoma Experimental/mortalidad , Ratones , Virus de los Bosques Semliki/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Tasa de Supervivencia , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
18.
J Virol ; 86(8): 4082-90, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22318135

RESUMEN

Vaccination using "naked" DNA is a highly attractive strategy for induction of pathogen-specific immune responses; however, it has been only weakly immunogenic in humans. Previously, we constructed DNA-launched Semliki Forest virus replicons (DREP), which stimulate pattern recognition receptors and induce augmented immune responses. Also, in vivo electroporation was shown to enhance immune responses induced by conventional DNA vaccines. Here, we combine these two approaches and show that in vivo electroporation increases CD8(+) T cell responses induced by DREP and consequently decreases the DNA dose required to induce a response. The vaccines used in this study encode the multiclade HIV-1 T cell immunogen HIVconsv, which is currently being evaluated in clinical trials. Using intradermal delivery followed by electroporation, the DREP.HIVconsv DNA dose could be reduced to as low as 3.2 ng to elicit frequencies of HIV-1-specific CD8(+) T cells comparable to those induced by 1 µg of a conventional pTH.HIVconsv DNA vaccine, representing a 625-fold molar reduction in dose. Responses induced by both DREP.HIVconsv and pTH.HIVconsv were further increased by heterologous vaccine boosts employing modified vaccinia virus Ankara MVA.HIVconsv and attenuated chimpanzee adenovirus ChAdV63.HIVconsv. Using the same HIVconsv vaccines, the mouse observations were supported by an at least 20-fold-lower dose of DNA vaccine in rhesus macaques. These data point toward a strategy for overcoming the low immunogenicity of DNA vaccines in humans and strongly support further development of the DREP vaccine platform for clinical evaluation.


Asunto(s)
ADN Viral/inmunología , VIH-1/inmunología , Plásmidos/inmunología , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/inmunología , Linfocitos T/inmunología , Vacunas de ADN/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular , ADN Viral/genética , Electroporación , Femenino , Orden Génico , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Plásmidos/administración & dosificación , Plásmidos/genética , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
19.
J Neuroimmunol ; 235(1-2): 18-26, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21612829

RESUMEN

Semliki Forest Virus (SFV) encephalomyelitis has been used to study the pathogenesis of virus-induced demyelination and serves as a model for multiple sclerosis. SFV-infection of mice invariably leads to clinical weakness accompanied by CNS inflammation, viral clearance and primary demyelination by day 21 postinfection (pi), followed by recovery and remyelination by day 35 pi. We have applied this model to the examination of the effects of γδ T cells in antibody production and the pathogenesis of demyelinating lesions. SFV-infection of γδ T cell KO mice resulted in more severe clinical signs than in wild type (WT) B6 mice. SFV-infected WT and γδ KO mice both cleared virus by day 10 pi and inflammation was comparable. Demyelination also appeared to be similar in both groups except that KO mice did not exhibit extensive remyelination which was seen in WT mice by day 21. SFV-infected WT mice showed widespread remyelination by day 35 pi, whereas KO mice still displayed some demyelination through day 42 pi. Both WT and KO mice developed serum antibodies to SFV. However, the reactivity of WT sera with the SFV epitope, E2 T(h) peptide2, was significantly higher than in KO sera. Immunization with E2 T(h) peptide2 resulted in elevated antibody production to this peptide (p<0.05) and earlier remyelination (day 28 pi) in KO mice. Thus, our study has shown for the first time that immunization of SFV-infected γδ T cell KO mice with a viral peptide, E2 T(h) peptide2 led to enhanced recovery and repair of the CNS.


Asunto(s)
Infecciones por Alphavirus/inmunología , Anticuerpos Antivirales/biosíntesis , Enfermedades Desmielinizantes/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/fisiología , Virus de los Bosques Semliki/inmunología , Subgrupos de Linfocitos T/inmunología , Infecciones por Alphavirus/tratamiento farmacológico , Animales , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/virología , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/uso terapéutico , Femenino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Recuperación de la Función/inmunología , Subgrupos de Linfocitos T/virología
20.
Antivir Ther ; 16(2): 207-18, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21447870

RESUMEN

BACKGROUND: Regulatory T-cells (Treg) hamper immune responses elicited by cancer vaccines. Therefore, depletion of Treg is being used to improve the outcome of vaccinations. METHODS: We studied whether an alphavirus vector-based immunotherapeutic vaccine changes the number and/or activity of Treg and if Treg depletion improves the efficacy of this vaccine against tumours. The vaccine is based on a Semliki Forest virus (SFV). The recombinant SFV replicon particles encode a fusion protein of E6 and E7 from human papillomavirus (HPV) type 16 (SFVeE6,7). RESULTS: We demonstrated that SFVeE6,7 immunization did not change Treg levels and their suppressive activity. Depletion of Treg in mice, using the novel anti-folate receptor 4 antibody, did not enhance the immune response induced by SFVeE6,7 immunization. Both the priming and the proliferation phases of the HPV-specific response elicited with SFVeE6,7 were not affected by the immune-suppressive activity of Treg. Moreover, Treg depletion did not improve the therapeutic antitumour response of SFVeE6,7 in a murine tumour model. CONCLUSIONS: The efficacy of the SFVeE6,7 vaccine was not hampered by Treg. Therefore, SFVeE6,7 seems a very promising candidate for the treatment of HPV-induced disease, as it may not require additional immune interventions to modulate Treg activity.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vectores Genéticos/genética , Inmunización/métodos , Neoplasias/terapia , Recombinación Genética , Virus de los Bosques Semliki/genética , Linfocitos T Reguladores/inmunología , Alphavirus , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Activación de Linfocitos , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/inmunología , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Virus de los Bosques Semliki/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...