Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 9(1): 5031, 2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30487575

RESUMEN

Mice engrafted with components of a human immune system have become widely-used models for studying aspects of human immunity and disease. However, a defined methodology to objectively measure and compare the quality of the human immune response in different models is lacking. Here, by taking advantage of the highly immunogenic live-attenuated yellow fever virus vaccine YFV-17D, we provide an in-depth comparison of immune responses in human vaccinees, conventional humanized mice, and second generation humanized mice. We demonstrate that selective expansion of human myeloid and natural killer cells promotes transcriptomic responses akin to those of human vaccinees. These enhanced transcriptomic profiles correlate with the development of an antigen-specific cellular and humoral response to YFV-17D. Altogether, our approach provides a robust scoring of the quality of the human immune response in humanized mice and highlights a rational path towards developing better pre-clinical models for studying the human immune response and disease.


Asunto(s)
Células Asesinas Naturales/metabolismo , Células Mieloides/metabolismo , Vacunas Atenuadas/inmunología , Vacuna contra la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/inmunología , Animales , Humanos , Células Asesinas Naturales/inmunología , Ratones , Células Mieloides/inmunología , Transcriptoma/genética , Vacuna contra la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/genética
2.
Sci Transl Med ; 10(447)2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29950446

RESUMEN

Chronic delta hepatitis, caused by hepatitis delta virus (HDV), is the most severe form of viral hepatitis, affecting at least 20 million hepatitis B virus (HBV)-infected patients worldwide. HDV/HBV co- or superinfections are major drivers for hepatocarcinogenesis. Antiviral treatments exist only for HBV and can only suppress but not cure infection. Development of more effective therapies has been impeded by the scarcity of suitable small-animal models. We created a transgenic (tg) mouse model for HDV expressing the functional receptor for HBV and HDV, the human sodium taurocholate cotransporting peptide NTCP. Both HBV and HDV entered hepatocytes in these mice in a glycoprotein-dependent manner, but one or more postentry blocks prevented HBV replication. In contrast, HDV persistently infected hNTCP tg mice coexpressing the HBV envelope, consistent with HDV dependency on the HBV surface antigen (HBsAg) for packaging and spread. In immunocompromised mice lacking functional B, T, and natural killer cells, viremia lasted at least 80 days but resolved within 14 days in immunocompetent animals, demonstrating that lymphocytes are critical for controlling HDV infection. Although acute HDV infection did not cause overt liver damage in this model, cell-intrinsic and cellular innate immune responses were induced. We further demonstrated that single and dual treatment with myrcludex B and lonafarnib efficiently suppressed viremia but failed to cure HDV infection at the doses tested. This small-animal model with inheritable susceptibility to HDV opens opportunities for studying viral pathogenesis and immune responses and for testing novel HDV therapeutics.


Asunto(s)
Hepatitis D/tratamiento farmacológico , Hepatitis D/virología , Virus de la Hepatitis Delta/fisiología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Quimioterapia Combinada , Genoma Viral , Glicoproteínas/metabolismo , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Hepatitis D/inmunología , Virus de la Hepatitis Delta/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunocompetencia , Lipopéptidos/farmacología , Lipopéptidos/uso terapéutico , Ratones Endogámicos C57BL , Ratones Transgénicos , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Piperidinas/farmacología , Piperidinas/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Simportadores/metabolismo , Transgenes , Viremia/tratamiento farmacológico , Viremia/patología
3.
Cell Host Microbe ; 23(4): 523-535.e5, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29649443

RESUMEN

Plasmodium infection begins with the bite of an anopheline mosquito, when sporozoites along with saliva are injected into a vertebrate host. The role of the host responses to mosquito saliva components in malaria remains unclear. We observed that antisera against Anopheles gambiae salivary glands partially protected mice from mosquito-borne Plasmodium infection. Specifically, antibodies to A. gambiae TRIO (AgTRIO), a mosquito salivary gland antigen, contributed to the protection. Mice administered AgTRIO antiserum showed lower Plasmodium liver burden and decreased parasitemia when exposed to infected mosquitoes. Active immunization with AgTRIO was also partially protective against Plasmodium berghei infection. A combination of AgTRIO antiserum and antibodies against Plasmodium circumsporozoite protein, a vaccine candidate, further decreased P. berghei infection. In humanized mice, AgTRIO antiserum afforded some protection against mosquito-transmitted Plasmodium falciparum. AgTRIO antiserum reduced the movement of sporozoites in the murine dermis. AgTRIO may serve as an arthropod-based target against Plasmodium to combat malaria.


Asunto(s)
Anopheles/inmunología , Inmunización Pasiva , Proteínas de Insectos/inmunología , Malaria/prevención & control , Proteínas y Péptidos Salivales/inmunología , Animales , Modelos Animales de Enfermedad , Proteínas de Insectos/administración & dosificación , Hígado/parasitología , Hígado/patología , Malaria/parasitología , Malaria/patología , Ratones , Carga de Parásitos , Parasitemia/parasitología , Parasitemia/prevención & control , Plasmodium berghei/inmunología , Plasmodium falciparum , Proteínas y Péptidos Salivales/administración & dosificación , Resultado del Tratamiento
4.
PLoS Pathog ; 14(3): e1006908, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29505618

RESUMEN

Amino-acid coevolution can be referred to mutational compensatory patterns preserving the function of a protein. Viral envelope glycoproteins, which mediate entry of enveloped viruses into their host cells, are shaped by coevolution signals that confer to viruses the plasticity to evade neutralizing antibodies without altering viral entry mechanisms. The functions and structures of the two envelope glycoproteins of the Hepatitis C Virus (HCV), E1 and E2, are poorly described. Especially, how these two proteins mediate the HCV fusion process between the viral and the cell membrane remains elusive. Here, as a proof of concept, we aimed to take advantage of an original coevolution method recently developed to shed light on the HCV fusion mechanism. When first applied to the well-characterized Dengue Virus (DENV) envelope glycoproteins, coevolution analysis was able to predict important structural features and rearrangements of these viral protein complexes. When applied to HCV E1E2, computational coevolution analysis predicted that E1 and E2 refold interdependently during fusion through rearrangements of the E2 Back Layer (BL). Consistently, a soluble BL-derived polypeptide inhibited HCV infection of hepatoma cell lines, primary human hepatocytes and humanized liver mice. We showed that this polypeptide specifically inhibited HCV fusogenic rearrangements, hence supporting the critical role of this domain during HCV fusion. By combining coevolution analysis and in vitro assays, we also uncovered functionally-significant coevolving signals between E1 and E2 BL/Stem regions that govern HCV fusion, demonstrating the accuracy of our coevolution predictions. Altogether, our work shed light on important structural features of the HCV fusion mechanism and contributes to advance our functional understanding of this process. This study also provides an important proof of concept that coevolution can be employed to explore viral protein mediated-processes, and can guide the development of innovative translational strategies against challenging human-tropic viruses.


Asunto(s)
Evolución Molecular , Hepacivirus/fisiología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Hepatitis C/metabolismo , Hepatitis C/patología , Hepatitis C/virología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Células Tumorales Cultivadas , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Replicación Viral
5.
mBio ; 8(4)2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28811340

RESUMEN

Yellow fever virus (YFV) is an arthropod-borne flavivirus, infecting ~200,000 people worldwide annually and causing about 30,000 deaths. The live attenuated vaccine strain, YFV-17D, has significantly contributed in controlling the global burden of yellow fever worldwide. However, the viral and host contributions to YFV-17D attenuation remain elusive. Type I interferon (IFN-α/ß) signaling and type II interferon (IFN-γ) signaling have been shown to be mutually supportive in controlling YFV-17D infection despite distinct mechanisms of action in viral infection. However, it remains unclear how type III IFN (IFN-λ) integrates into this antiviral system. Here, we report that while wild-type (WT) and IFN-λ receptor knockout (λR-/-) mice were largely resistant to YFV-17D, deficiency in type I IFN signaling resulted in robust infection. Although IFN-α/ß receptor knockout (α/ßR-/-) mice survived the infection, mice with combined deficiencies in both type I signaling and type III IFN signaling were hypersusceptible to YFV-17D and succumbed to the infection. Mortality was associated with viral neuroinvasion and increased permeability of the blood-brain barrier (BBB). α/ßR-/- λR-/- mice also exhibited distinct changes in the frequencies of multiple immune cell lineages, impaired T-cell activation, and severe perturbation of the proinflammatory cytokine balance. Taken together, our data highlight that type III IFN has critical immunomodulatory and neuroprotective functions that prevent viral neuroinvasion during active YFV-17D replication. Type III IFN thus likely represents a safeguard mechanism crucial for controlling YFV-17D infection and contributing to shaping vaccine immunogenicity.IMPORTANCE YFV-17D is a live attenuated flavivirus vaccine strain recognized as one of the most effective vaccines ever developed. However, the host and viral determinants governing YFV-17D attenuation and its potent immunogenicity are still unknown. Here, we analyzed the role of type III interferon (IFN)-mediated signaling, a host immune defense mechanism, in controlling YFV-17D infection and attenuation in different mouse models. We uncovered a critical role of type III IFN-mediated signaling in preserving the integrity of the blood-brain barrier and preventing viral brain invasion. Type III IFN also played a major role in regulating the induction of a potent but balanced immune response that prevented viral evasion of the host immune system. An improved understanding of the complex mechanisms regulating YFV-17D attenuation will provide insights into the key virus-host interactions that regulate host immune responses and infection outcomes as well as open novel avenues for the development of innovative vaccine strategies.


Asunto(s)
Interferones/inmunología , Interferones/metabolismo , Transducción de Señal/inmunología , Vacuna contra la Fiebre Amarilla/inmunología , Fiebre Amarilla/inmunología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/virología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inmunidad Innata , Interferones/genética , Ratones , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Vacunas Atenuadas/inmunología , Fiebre Amarilla/prevención & control , Fiebre Amarilla/virología , Virus de la Fiebre Amarilla/inmunología
6.
J Infect Dis ; 215(12): 1824-1831, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28398489

RESUMEN

Background: The global control of hepatitis C virus (HCV) infection remains a great burden, owing to the high prices and potential drug resistance of the new direct-acting antivirals (DAAs), as well as the risk of reinfection in DAA-cured patients. Thus, a prophylactic vaccine for HCV is of great importance. We previously reported that a single recombinant soluble E2 (sE2) vaccine produced in insect cells was able to induce broadly neutralizing antibodies (NAbs) and prevent HCV infection in mice. Here the sE2 vaccine was evaluated in non-human primates. Methods: Rhesus macaques were immunized with sE2 vaccine in combination with different adjuvants. Vaccine-induced NAbs in antisera were tested for neutralization activities against a panel of cell culture-derived HCV (HCVcc), while T-cell responses were evaluated in splenocytes, peripheral blood mononuclear cells, and hepatic lymphocytes. Results: sE2 is able to elicit NAbs against HCVcc harboring structural proteins from multiple HCV genotypes in rhesus macaques. Moreover, sE2-immunized macaques developed systemic and intrahepatic memory T cells specific for E2. A significant correlation between the sE2-specific immunoglobulin G titers and neutralization spectrum was observed, highlighting the essential role of sE2 immunogenicity on achieving broad NAbs. Conclusions: sE2 is a promising HCV vaccine candidate that warrants further preclinical and clinical development.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Hepacivirus/inmunología , Anticuerpos contra la Hepatitis C/sangre , Linfocitos T/inmunología , Vacunas contra Hepatitis Viral/inmunología , Adyuvantes Inmunológicos/uso terapéutico , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Genotipo , Hepacivirus/genética , Hepatitis C/prevención & control , Hepatitis C/virología , Humanos , Inmunización , Leucocitos Mononucleares/inmunología , Hígado/inmunología , Hígado/virología , Macaca mulatta , Masculino
7.
Nat Commun ; 8: 14781, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28290449

RESUMEN

Positive-sense RNA viruses pose increasing health and economic concerns worldwide. Our limited understanding of how these viruses interact with their host and how these processes lead to virulence and disease seriously hampers the development of anti-viral strategies. Here, we demonstrate the tracking of (+) and (-) sense viral RNA at single-cell resolution within complex subsets of the human and murine immune system in different mouse models. Our results provide insights into how a prototypic flavivirus, yellow fever virus (YFV-17D), differentially interacts with murine and human hematopoietic cells in these mouse models and how these dynamics influence distinct outcomes of infection. We detect (-) YFV-17D RNA in specific secondary lymphoid compartments and cell subsets not previously recognized as permissive for YFV replication, and we highlight potential virus-host interaction events that could be pivotal in regulating flavivirus virulence and attenuation.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , ARN Viral/metabolismo , Fiebre Amarilla/metabolismo , Virus de la Fiebre Amarilla/genética , Animales , Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Femenino , Flavivirus/genética , Citometría de Flujo , Células HEK293 , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Humanos , Masculino , Ratones , ARN Viral/inmunología , Análisis de la Célula Individual , Especificidad de la Especie , Trasplante Heterólogo , Fiebre Amarilla/inmunología , Fiebre Amarilla/virología
8.
Proc Natl Acad Sci U S A ; 114(5): 1147-1152, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096411

RESUMEN

Hepatitis E virus (HEV) is the leading cause of enterically transmitted viral hepatitis globally. Of HEV's three ORFs, the function of ORF3 has remained elusive. Here, we demonstrate that via homophilic interactions ORF3 forms multimeric complexes associated with intracellular endoplasmic reticulum (ER)-derived membranes. HEV ORF3 shares several structural features with class I viroporins, and the function of HEV ORF3 can be maintained by replacing it with the well-characterized viroporin influenza A virus (IAV) matrix-2 protein. ORF3's ion channel function is further evidenced by its ability to mediate ionic currents when expressed in Xenopus laevis oocytes. Furthermore, we identified several positions in ORF3 critical for its formation of multimeric complexes, ion channel activity, and, ultimately, release of infectious particles. Collectively, our data demonstrate a previously undescribed function of HEV ORF3 as a viroporin, which may serve as an attractive target in developing direct-acting antivirals.


Asunto(s)
Virus de la Hepatitis E/fisiología , Canales Iónicos/fisiología , Proteínas Virales/fisiología , Liberación del Virus/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Retículo Endoplásmico/metabolismo , Eliminación de Gen , Células HEK293 , Células Hep G2 , Humanos , Canales Iónicos/química , Transporte Iónico , Oocitos , Técnicas de Placa-Clamp , Dominios Proteicos , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Proteínas de la Matriz Viral/fisiología , Proteínas Virales/química , Proteínas Virales/genética , Replicación Viral , Xenopus laevis
9.
Virology ; 502: 63-72, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28006671

RESUMEN

There are ~350 million chronic carriers of hepatitis B (HBV). While a prophylactic vaccine and drug regimens to suppress viremia are available, chronic HBV infection is rarely cured. HBV's limited host tropism leads to a scarcity of susceptible small animal models and is a hurdle to developing curative therapies. Mice that support engraftment with human hepatoctyes have traditionally been generated through crosses of murine liver injury models to immunodeficient backgrounds. Here, we describe the disruption of fumarylacetoacetate hydrolase directly in the NOD Rag1-/- IL2RγNULL (NRG) background using zinc finger nucleases. The resultant human liver chimeric mice sustain persistent HBV viremia for >90 days. When treated with standard of care therapy, HBV DNA levels decrease below detection but rebound when drug suppression is released, mimicking treatment response observed in patients. Our study highlights the utility of directed gene targeting approaches in zygotes to create new humanized mouse models for human diseases.


Asunto(s)
Antivirales/administración & dosificación , Modelos Animales de Enfermedad , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B Crónica/tratamiento farmacológico , Animales , Femenino , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica/genética , Hepatitis B Crónica/metabolismo , Hepatitis B Crónica/virología , Hepatocitos/virología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Hígado/virología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Replicación Viral
10.
J Virol ; 91(4)2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27928007

RESUMEN

Hepatitis C virus (HCV) causes chronic infections in at least 150 million individuals worldwide. HCV has a narrow host range and robustly infects only humans and chimpanzees. The underlying mechanisms for this narrow host range are incompletely understood. At the level of entry, differences in the amino acid sequences between the human and mouse orthologues of two essential host factors, the tetraspanin CD81 and the tight junction protein occludin (OCLN), explain, at least in part, HCV's limited ability to enter mouse hepatocytes. We have previously shown that adenoviral or transgenic overexpression of human CD81 and OCLN facilitates HCV uptake into mouse hepatocytes in vitro and in vivo In efforts to refine these models, we constructed knock-in mice in which the second extracellular loops of CD81 and OCLN were replaced with the respective human sequences, which contain the determinants that are critical for HCV uptake. We demonstrate that the humanized CD81 and OCLN were expressed at physiological levels in a tissue-appropriate fashion. Mice bearing the humanized alleles formed normal tight junctions and did not exhibit any immunologic abnormalities, indicating that interactions with their physiological ligands were intact. HCV entry factor knock-in mice take up HCV with an efficiency similar to that in mice expressing HCV entry factors transgenically or adenovirally, demonstrating the utility of this model for studying HCV infection in vivo IMPORTANCE: At least 150 million individuals are chronically infected with hepatitis C virus (HCV). Chronic hepatitis C can result in progressive liver disease and liver cancer. New antiviral treatments can cure HCV in the majority of patients, but a vaccine remains elusive. To gain a better understanding of the processes culminating in liver failure and cancer and to prioritize vaccine candidates more efficiently, small-animal models are needed. Here, we describe the characterization of a new mouse model in which the parts of two host factors that are essential for HCV uptake, CD81 and occludin (OCLN), which differ between mice and humans, were humanized. We demonstrate that such minimally humanized mice develop normally, express the modified genes at physiological levels, and support HCV uptake. This model is of considerable utility for studying viral entry in the three-dimensional context of the liver and to test approaches aimed at preventing HCV entry.


Asunto(s)
Expresión Génica , Hepacivirus/fisiología , Hepatitis C/genética , Hepatitis C/virología , Ocludina/genética , Tetraspanina 28/genética , Internalización del Virus , Alelos , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Marcación de Gen , Genes Reporteros , Sitios Genéticos , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Transgénicos , Ocludina/metabolismo , Especificidad de Órganos/genética , Fenotipo , Tetraspanina 28/metabolismo , Transcripción Genética
11.
mBio ; 7(6)2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27834208

RESUMEN

Hepatitis C virus (HCV) species tropism is incompletely understood. We have previously shown that at the level of entry, human CD81 and occludin (OCLN) comprise the minimal set of human factors needed for viral uptake into murine cells. As an alternative approach to genetic humanization, species barriers can be overcome by adapting HCV to use the murine orthologues of these entry factors. We previously generated a murine tropic HCV (mtHCV or Jc1/mCD81) strain harboring three mutations within the viral envelope proteins that allowed productive entry into mouse cell lines. In this study, we aimed to characterize the ability of mtHCV to enter and infect mouse hepatocytes in vivo and in vitro Using a highly sensitive, Cre-activatable reporter, we demonstrate that mtHCV can enter mouse hepatocytes in vivo in the absence of any human cofactors. Viral entry still relied on expression of mouse CD81 and SCARB1 and was more efficient when mouse CD81 and OCLN were overexpressed. HCV entry could be significantly reduced in the presence of anti-HCV E2 specific antibodies, suggesting that uptake of mtHCV is dependent on viral glycoproteins. Despite mtHCV's ability to enter murine hepatocytes in vivo, we did not observe persistent infection, even in animals with severely blunted type I and III interferon signaling and impaired adaptive immune responses. Altogether, these results establish proof of concept that the barriers limiting HCV species tropism can be overcome by viral adaptation. However, additional viral adaptations will likely be needed to increase the robustness of a murine model system for hepatitis C. IMPORTANCE: At least 150 million individuals are chronically infected with HCV and are at risk of developing serious liver disease. Despite the advent of effective antiviral therapy, the frequency of chronic carriers has only marginally decreased. A major roadblock in developing a vaccine that would prevent transmission is the scarcity of animal models that are susceptible to HCV infection. It is poorly understood why HCV infects only humans and chimpanzees. To develop an animal model for hepatitis C, previous efforts focused on modifying the host environment of mice, for example, to render them more susceptible to HCV infection. Here, we attempted a complementary approach in which a laboratory-derived HCV variant was tested for its ability to infect mice. We demonstrate that this engineered HCV strain can enter mouse liver cells but does not replicate efficiently. Thus, additional adaptations are likely needed to construct a robust animal model for HCV.


Asunto(s)
Adaptación Fisiológica , Hepacivirus/fisiología , Hepatitis C/virología , Internalización del Virus , Inmunidad Adaptativa , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Modelos Animales de Enfermedad , Hepacivirus/genética , Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatocitos/inmunología , Hepatocitos/virología , Especificidad del Huésped , Humanos , Ratones , Ocludina/genética , Receptores Depuradores de Clase B/genética , Tetraspanina 28/genética , Tropismo Viral
12.
J Virol ; 90(23): 10486-10498, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27630242

RESUMEN

Hepatitis C virus (HCV) infection is a global health problem for which no vaccine is available. HCV has a highly heterogeneous RNA genome and can be classified into seven genotypes. Due to the high genetic and resultant antigenic variation among the genotypes, inducing antibodies capable of neutralizing most of the HCV genotypes by experimental vaccination has been challenging. Previous efforts focused on priming humoral immune responses with recombinant HCV envelope E2 protein produced in mammalian cells. Here, we report that a soluble form of HCV E2 (sE2) produced in insect cells possesses different glycosylation patterns and is more immunogenic, as evidenced by the induction of higher titers of broadly neutralizing antibodies (bNAbs) against cell culture-derived HCV (HCVcc) harboring structural proteins from a diverse array of HCV genotypes. We affirm that continuous and discontinuous epitopes of well-characterized bNAbs are conserved, suggesting that sE2 produced in insect cells is properly folded. In a genetically humanized mouse model, active immunization with sE2 efficiently protected against challenge with a heterologous HCV genotype. These data not only demonstrate that sE2 is a promising HCV vaccine candidate, but also highlight the importance of glycosylation patterns in developing subunit viral vaccines. IMPORTANCE: A prophylactic vaccine with high efficacy and low cost is urgently needed for global control of HCV infection. Induction of broadly neutralizing antibodies against most HCV genotypes has been challenging due to the antigenic diversity of the HCV genome. Here, we refined a high-yield subunit HCV vaccine that elicited broadly neutralizing antibody responses in preclinical trials. We found that soluble HCV E2 protein (sE2) produced in insect cells is distinctly glycosylated and is more immunogenic than sE2 produced in mammalian cells, suggesting that glycosylation patterns should be taken into consideration in efforts to generate antibody-based recombinant vaccines against HCV. We further showed that sE2 vaccination confers protection against HCV infection in a genetically humanized mouse model. Thus, our work identified a promising broadly protective HCV vaccine candidate that should be considered for further preclinical and clinical development.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Hepacivirus/inmunología , Anticuerpos contra la Hepatitis C/biosíntesis , Vacunas contra Hepatitis Viral/inmunología , Animales , Especificidad de Anticuerpos , Línea Celular , Drosophila , Femenino , Glicosilación , Hepacivirus/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Eliminación de Secuencia , Solubilidad , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Vacunas contra Hepatitis Viral/química , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/inmunología
13.
Methods Mol Biol ; 1438: 79-101, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27150085

RESUMEN

Human liver chimeric mice have become valuable tools for the study of human hepatotropic pathogens and for the investigation of metabolism and pharmacokinetics of novel drugs. The evolution of the underlying mouse models has been rapid in the past years. The diverse fields of applications of those model systems and their technical challenges will be discussed in this chapter.


Asunto(s)
Hepatocitos/trasplante , Hígado/citología , Quimera por Trasplante/crecimiento & desarrollo , Animales , Criopreservación , Modelos Animales de Enfermedad , Hepatocitos/citología , Humanos , Hígado/crecimiento & desarrollo , Regeneración Hepática , Ratones , Trasplante Heterólogo
14.
Hepatology ; 62(1): 57-67, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25820364

RESUMEN

UNLABELLED: At least 170 million people are chronically infected with hepatitis C virus (HCV). Owing to the narrow host range of HCV and restricted use of chimpanzees, there is currently no suitable animal model for HCV pathogenesis studies or the development of a HCV vaccine. To identify cellular determinants of interspecies transmission and establish a novel immunocompetent model system, we examined the ability of HCV to infect hepatocytes from a small nonhuman primate, the rhesus macaque (Macaca mulatta). We show that the rhesus orthologs of critical HCV entry factors support viral glycoprotein-dependent virion uptake. Primary hepatocytes from rhesus macaques are also permissive for HCV-RNA replication and particle production, which is enhanced when antiviral signaling is suppressed. We demonstrate that this may be owing to the diminished capacity of HCV to antagonize mitochondrial antiviral-signaling protein-dependent innate cellular defenses. To test the ability of HCV to establish persistent replication in vivo, we engrafted primary rhesus macaque hepatocytes into immunocompromised xenorecipients. Inoculation of resulting simian liver chimeric mice with either HCV genotype 1a or 2a resulted in HCV serum viremia for up to 10 weeks. CONCLUSION: Together, these data indicate that rhesus macaques may be a viable model for HCV and implicate host immunity as a potential species-specific barrier to HCV infection. We conclude that suppression of host immunity or further viral adaptation may allow robust HCV infection in rhesus macaques and creation of a new animal model for studies of HCV pathogenesis, lentivirus coinfection, and vaccine development.


Asunto(s)
Modelos Animales de Enfermedad , Hepacivirus/fisiología , Hepatitis C , Hepatocitos/virología , Interacciones Huésped-Patógeno , Animales , Humanos , Inmunidad Innata , Macaca mulatta , Ratones , Internalización del Virus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...