Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Gastroenterology ; 167(3): 522-537, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38636680

RESUMEN

BACKGROUND & AIMS: High expression of phosphatidylinositol 4-kinase III alpha (PI4KIIIα) correlates with poor survival rates in patients with hepatocellular carcinoma. In addition, hepatitis C virus (HCV) infections activate PI4KIIIα and contribute to hepatocellular carcinoma progression. We aimed at mechanistically understanding the impact of PI4KIIIα on the progression of liver cancer and the potential contribution of HCV in this process. METHODS: Several hepatic cell culture and mouse models were used to study the functional importance of PI4KIIIα on liver pathogenesis. Antibody arrays, gene silencing, and PI4KIIIα-specific inhibitor were applied to identify the involved signaling pathways. The contribution of HCV was examined by using HCV infection or overexpression of its nonstructural protein. RESULTS: High PI4KIIIα expression and/or activity induced cytoskeletal rearrangements via increased phosphorylation of paxillin and cofilin. This led to morphologic alterations and higher migratory and invasive properties of liver cancer cells. We further identified the liver-specific lipid kinase phosphatidylinositol 3-kinase C2 domain-containing subunit gamma (PIK3C2γ) working downstream of PI4KIIIα in regulation of the cytoskeleton. PIK3C2γ generates plasma membrane phosphatidylinositol 3,4-bisphosphate-enriched, invadopodia-like structures that regulate cytoskeletal reorganization by promoting Akt2 phosphorylation. CONCLUSIONS: PI4KIIIα regulates cytoskeleton organization via PIK3C2γ/Akt2/paxillin-cofilin to favor migration and invasion of liver cancer cells. These findings provide mechanistic insight into the contribution of PI4KIIIα and HCV to the progression of liver cancer and identify promising targets for therapeutic intervention.


Asunto(s)
Factores Despolimerizantes de la Actina , Carcinoma Hepatocelular , Movimiento Celular , Citoesqueleto , Neoplasias Hepáticas , Invasividad Neoplásica , Paxillin , Transducción de Señal , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Humanos , Animales , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Citoesqueleto/metabolismo , Citoesqueleto/patología , Paxillin/metabolismo , Ratones , Factores Despolimerizantes de la Actina/metabolismo , Factores Despolimerizantes de la Actina/genética , Fosforilación , Hepacivirus , Línea Celular Tumoral , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Células Hep G2 , Hepatitis C/patología , Hepatitis C/metabolismo , Hepatitis C/virología , Interferencia de ARN
2.
Hepatology ; 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38652584

RESUMEN

BACKGROUND AND AIMS: HCV infection continues to be a major global health burden despite effective antiviral treatments. The urgent need for a protective vaccine is hindered by the scarcity of suitable HCV-permissive animal models tractable in vaccination and challenge studies. Currently, only antibody neutralization studies in infectious cell culture systems or studies of protection by passive immunization of human liver chimeric mice offer the possibility to evaluate the effect of vaccine-induced antibodies. However, differences between culture-permissive and in vivo-permissive viruses make it a challenge to compare analyses between platforms. To address this problem, we aimed at developing genotype-specific virus variants with genetic stability both in vitro and in vivo. APPROACH AND RESULTS: We demonstrated infection of human liver chimeric mice with cell culture-adapted HCV JFH1-based Core-NS2 recombinants of genotype 1-6, with a panel of 10 virus strains used extensively in neutralization and receptor studies. Clonal re-engineering of mouse-selected mutations resulted in virus variants with robust replication both in Huh7.5 cells and human liver chimeric mice, with genetic stability. Furthermore, we showed that, overall, these virus variants have similar in vitro neutralization profiles as their parent strains and demonstrated their use for in vivo neutralization studies. CONCLUSIONS: These mouse-selected HCV recombinants enable the triage of new vaccine-relevant antibodies in vitro and further allow characterization of protection from infection in vivo using identical viruses in human liver chimeric mice. As such, these viruses will serve as important resources in testing novel antibodies and can thus guide strategies to develop an efficient protective vaccine against HCV infection.

3.
JHEP Rep ; 5(3): 100646, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36748051

RESUMEN

Background & Aims: Elimination of chronic HBV/HDV infection remains a major global health challenge. Targeting excessive hepatitis B surface antigen (HBsAg) release may provide an interesting window of opportunity to break immune tolerance and to achieve a functional cure using additional antivirals. Methods: We evaluated a HBsAg-specific human monoclonal antibody, as part of either a prophylactic or therapeutic strategy, against HBV/HDV infection in cell culture models and in human-liver chimeric mice. To assess prophylactic efficacy, mice were passively immunized prior to infection with HBV or HBV/HDV (coinfection and superinfection setting). Therapeutic efficacy was assessed in HBV and HBV/HDV-coinfected mice receiving 4 weeks of treatment. Viral parameters (HBV DNA, HDV RNA and HBsAg) were assessed in mouse plasma. Results: The antibody could effectively prevent HBV/HDV infection in a dose-dependent manner with IC50 values of ∼3.5 ng/ml. Passive immunization showed complete protection of mice from both HBV and HBV/HDV coinfection. Moreover, HDV superinfection was either completely prevented or at least attenuated in HBV-infected mice. Finally, antibody treatment in mice with established HBV/HDV infection resulted in a significant decline in viremia and a concomitant drop in on-treatment HBsAg, with a moderate viral rebound following treatment cessation. Conclusion: We present data on a valuable antibody candidate that could complement other antivirals in strategies aimed at achieving functional cure of chronic HBV and HDV infection. Impact and implications: Patients chronically infected with HBV may eventually develop liver cancer and are at great risk of being superinfected with HDV, which worsens and accelerates disease progression. Unfortunately, current treatments can rarely eliminate both viruses from chronically infected patients. In this study, we present data on a novel antibody that is able to prevent chronic HBV/HDV infection in a mouse model with a humanized liver. Moreover, antibody treatment of HBV/HDV-infected mice strongly diminishes viral loads during therapy. This antibody is a valuable candidate for further clinical development.

4.
Gut ; 72(6): 1186-1195, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35977815

RESUMEN

OBJECTIVE: Chronic HBV/HDV infections are a major cause of liver cancer. Current treatments can only rarely eliminate HBV and HDV. Our previously developed preS1-HDAg immunotherapy could induce neutralising antibodies to HBV in vivo and raise HBV/HDV-specific T-cells. Here, we further investigate if a heterologous prime-boost strategy can circumvent T-cell tolerance and preclude HDV superinfection in vivo. DESIGN: A DNA prime-protein boost strategy was evaluated for immunogenicity in mice and rabbits. Its ability to circumvent T-cell tolerance was assessed in immunocompetent hepatitis B surface antigen (HBsAg)-transgenic mice. Neutralisation of HBV and HDV was evaluated both in vitro and in immunodeficient human-liver chimeric mice upon adoptive transfer. RESULTS: The prime-boost strategy elicits robust HBV/HDV-specific T-cells and preS1-antibodies that can effectively prevent HBV and HDV (co-)infection in vitro and in vivo. In a mouse model representing the chronic HBsAg carrier state, active immunisation primes high levels of preS1-antibodies and HDAg-specific T-cells. Moreover, transfer of vaccine-induced antibodies completely protects HBV-infected human-liver chimeric mice from HDV superinfection. CONCLUSION: The herein described preS1-HDAg immunotherapy is shown to be immunogenic and vaccine-induced antibodies are highly effective at preventing HBV and HDV (super)infection both in vitro and in vivo. Our vaccine can complement current and future therapies for the control of chronic HBV and HDV infection.


Asunto(s)
Hepatitis B Crónica , Hepatitis B , Sobreinfección , Humanos , Ratones , Animales , Conejos , Antígenos de Hepatitis delta , Antígenos de Superficie de la Hepatitis B , Hepatitis B Crónica/prevención & control , Sobreinfección/prevención & control , Virus de la Hepatitis Delta/genética , Hepatitis B/prevención & control , Virus de la Hepatitis B/genética , Anticuerpos Antivirales , Ratones Transgénicos
5.
Proc Natl Acad Sci U S A ; 119(34): e2202653119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969792

RESUMEN

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and is the leading cause of enterically transmitted viral hepatitis worldwide. Ribavirin (RBV) is currently the only treatment option for many patients; however, cases of treatment failures or posttreatment relapses have been frequently reported. RBV therapy was shown to be associated with an increase in HEV genome heterogeneity and the emergence of distinct HEV variants. In this study, we analyzed the impact of eight patient-derived open reading frame 2 (ORF2) single-nucleotide variants (SNVs), which occurred under RBV treatment, on the replication cycle and pathogenesis of HEV. The parental HEV strain and seven ORF2 variants showed comparable levels of RNA replication in human hepatoma cells and primary human hepatocytes. However, a P79S ORF2 variant demonstrated reduced RNA copy numbers released in the supernatant and an impairment in the production of infectious particles. Biophysical and biochemical characterization revealed that this SNV caused defective, smaller HEV particles with a loss of infectiousness. Furthermore, the P79S variant displayed an altered subcellular distribution of the ORF2 protein and was able to interfere with antibody-mediated neutralization of HEV in a competition assay. In conclusion, an SNV in the HEV ORF2 could be identified that resulted in altered virus particles that were noninfectious in vitro and in vivo, but could potentially serve as immune decoys. These findings provide insights in understanding the biology of circulating HEV variants and may guide development of personalized antiviral strategies in the future.


Asunto(s)
Virus de la Hepatitis E , Ribavirina , Proteínas Virales , Línea Celular Tumoral , Virus de la Hepatitis E/genética , Virus de la Hepatitis E/fisiología , Hepatocitos/virología , Humanos , Recurrencia Local de Neoplasia/genética , Nucleótidos , ARN Viral , Ribavirina/farmacología , Proteínas Virales/genética , Replicación Viral
6.
Cells ; 11(6)2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35326378

RESUMEN

BACKGROUND: Hepatitis C virus (HCV) constitutes a global health problem, while hepatitis E virus (HEV) is the major cause of acute viral hepatitis globally. HCV/HEV co-infections have been poorly characterized, as they are hampered by the lack of robust HEV cell culture systems. This study developed experimental models to study HCV/HEV co-infections and investigate viral interference in cells and humanized mice. METHODS: We used state-of-the art human hepatocytes tissue culture models to assess HEV and HCV replication in co- or super-transfection settings. Findings were confirmed by co- and super-infection experiments in human hepatocytes and in vivo in human liver chimeric mice. RESULTS: HEV was inhibited by concurrent HCV replication in human hepatocytes. This exclusion phenotype was linked to the protease activity of HCV. These findings were corroborated by the fact that in HEV on HCV super-infected mice, HEV viral loads were reduced in individual mice. Similarly, HCV on HEV super-infected mice showed reduced HCV viral loads. CONCLUSION: Direct interference of both viruses with HCV NS3/4A as the determinant was observed. In vivo, we detected reduced replication of both viruses after super-infection in individual mice. These findings provide new insights into the pathogenesis of HCV-HEV co-infections and should contribute to its clinical management in the future.


Asunto(s)
Coinfección , Hepatitis C , Virus de la Hepatitis E , Animales , Hepacivirus/genética , Virus de la Hepatitis E/genética , Ratones , Interferencia Viral , Replicación Viral
7.
Front Microbiol ; 13: 819877, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35295314

RESUMEN

The hepatitis E virus (HEV) is responsible for 20 million infections worldwide per year. Although, HEV infection is mostly self-limiting, immunocompromised individuals may evolve toward chronicity. The lack of an efficient small animal model has hampered the study of HEV and the discovery of anti-HEV therapies. Furthermore, new HEV strains, infectious to humans, are being discovered. Human liver-chimeric mice have greatly aided in the understanding of HEV, but only two genotypes (HEV-1 and HEV-3) have been studied in this model. Moreover, the immunodeficient nature of this mouse model does not allow full investigation of the virus and all aspects of its interaction with the host. Recent studies have shown the susceptibility of regular and nude Balb/c mice to a HEV-4 strain (KM01). This model should allow the investigation of the interplay between HEV and the adaptive immune system of its host, and potential immune-mediated complications. Here, we assess the susceptibility of human liver-chimeric and non-humanised mice to a different HEV-4 strain (BeSW67HEV4-2008). We report that humanised mice could be readily infected with this isolate, resulting in an infection pattern comparable to HEV-3 infection. Despite these results and in contrast to KM01, non-humanised mice were not susceptible to infection with this viral strain. Further investigation, using other HEV-4 isolates, is needed to conclusively determine HEV-4 tropism and mouse susceptibility.

8.
Front Immunol ; 12: 775098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975862

RESUMEN

Hepatitis C virus (HCV) is highly variable and transmits through infected blood to establish a chronic liver infection in the majority of patients. Our knowledge on the infectivity of clinical HCV strains is hampered by the lack of in vitro cell culture systems that support efficient viral replication. We and others have reported that HCV can associate with and infect immune cells and may thereby evade host immune surveillance and elimination. To evaluate whether B cells play a role in HCV transmission, we assessed the ability of B cells and sera from recent (<2 years) or chronic (≥ 2 years) HCV patients to infect humanized liver chimeric mice. HCV was transmitted by B cells from chronic infected patients whereas the sera were non-infectious. In contrast, B cells from recently infected patients failed to transmit HCV to the mice, whereas all serum samples were infectious. We observed an association between circulating anti-glycoprotein E1E2 antibodies and B cell HCV transmission. Taken together, our studies provide evidence for HCV transmission by B cells, findings that have clinical implications for prophylactic and therapeutic antibody-based vaccine design.


Asunto(s)
Linfocitos B/virología , Hepacivirus/patogenicidad , Hepatitis C/transmisión , Adulto , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Anticuerpos ampliamente neutralizantes/sangre , Anticuerpos ampliamente neutralizantes/inmunología , Modelos Animales de Enfermedad , Femenino , Hepacivirus/inmunología , Hepacivirus/aislamiento & purificación , Hepatitis C/sangre , Hepatitis C/prevención & control , Hepatitis C/virología , Humanos , Hígado/patología , Hígado/virología , Trasplante de Hígado , Masculino , Ratones , Persona de Mediana Edad , Suero/virología , Quimera por Trasplante , Desarrollo de Vacunas/métodos , Proteínas del Envoltorio Viral/inmunología , Vacunas contra Hepatitis Viral/uso terapéutico , Adulto Joven
9.
J Infect Dis ; 223(1): 128-138, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-31994701

RESUMEN

BACKGROUND: Chronic hepatitis B and D virus (HBV/HDV) infections can cause cancer. Current HBV therapy using nucleoside analogues (NAs) is life-long and reduces but does not eliminate the risk of cancer. A hallmark of chronic hepatitis B is a dysfunctional HBV-specific T-cell response. We therefore designed an immunotherapy driven by naive healthy T cells specific for the HDV antigen (HDAg) to bypass the need for HBV-specific T cells in order to prime PreS1-specific T cells and PreS1 antibodies blocking HBV entry. METHODS: Ten combinations of PreS1 and/or HDAg sequences were evaluated for induction of PreS1 antibodies and HBV- and HDV-specific T cells in vitro and in vivo. Neutralization of HBV by PreS1-specific murine and rabbit antibodies was evaluated in cell culture, and rabbit anti-PreS1 were tested for neutralization of HBV in mice repopulated with human hepatocytes. RESULTS: The best vaccine candidate induced T cells to PreS1 and HDAg, and PreS1 antibodies blocking HBV entry in vitro. Importantly, adoptive transfer of PreS1 antibodies prevented, or modulated, HBV infection after a subsequent challenge in humanized mice. CONCLUSIONS: We here describe a novel immunotherapy for chronic HBV/HDV that targets viral entry to complement NAs and coming therapies inhibiting viral maturation.


Asunto(s)
Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis D Crónica/tratamiento farmacológico , Virus de la Hepatitis Delta/inmunología , Internalización del Virus/efectos de los fármacos , Animales , Femenino , Vacunas contra Hepatitis B , Hepatocitos/efectos de los fármacos , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Conejos
10.
Sci Adv ; 6(45)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33148654

RESUMEN

Hepatitis C virus (HCV) has no animal reservoir, infecting only humans. To investigate species barrier determinants limiting infection of rodents, murine liver complementary DNA library screening was performed, identifying transmembrane proteins Cd302 and Cr1l as potent restrictors of HCV propagation. Combined ectopic expression in human hepatoma cells impeded HCV uptake and cooperatively mediated transcriptional dysregulation of a noncanonical program of immunity genes. Murine hepatocyte expression of both factors was constitutive and not interferon inducible, while differences in liver expression and the ability to restrict HCV were observed between the murine orthologs and their human counterparts. Genetic ablation of endogenous Cd302 expression in human HCV entry factor transgenic mice increased hepatocyte permissiveness for an adapted HCV strain and dysregulated expression of metabolic process and host defense genes. These findings highlight human-mouse differences in liver-intrinsic antiviral immunity and facilitate the development of next-generation murine models for preclinical testing of HCV vaccine candidates.


Asunto(s)
Hepacivirus , Hepatitis C , Animales , Hepacivirus/genética , Ratones , Ratones Transgénicos , Internalización del Virus
11.
Cells ; 9(4)2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32276503

RESUMEN

Human skin-derived precursors (SKP) represent a group of somatic stem/precursor cells that reside in dermal skin throughout life that harbor clinical potential. SKP have a high self-renewal capacity, the ability to differentiate into multiple cell types and low immunogenicity, rendering them key candidates for allogeneic cell-based, off-the-shelf therapy. However, potential clinical application of allogeneic SKP requires that these cells retain their therapeutic properties under all circumstances and, in particular, in the presence of an inflammation state. Therefore, in this study, we investigated the impact of pro-inflammatory stimulation on the secretome and immunosuppressive properties of SKP. We demonstrated that pro-inflammatory stimulation of SKP significantly changes their expression and the secretion profile of chemo/cytokines and growth factors. Most importantly, we observed that pro-inflammatory stimulated SKP were still able to suppress the graft-versus-host response when cotransplanted with human PBMC in severe-combined immune deficient (SCID) mice, albeit to a much lesser extent than unstimulated SKP. Altogether, this study demonstrates that an inflammatory microenvironment has a significant impact on the immunological properties of SKP. These alterations need to be taken into account when developing allogeneic SKP-based therapies.


Asunto(s)
Citocinas/metabolismo , Inmunomodulación/inmunología , Inflamación/inmunología , Piel/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Ratones SCID , Piel/citología
12.
Proc Natl Acad Sci U S A ; 117(3): 1731-1741, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31896581

RESUMEN

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and the leading cause for acute viral hepatitis worldwide. The virus is classified as a member of the genus Orthohepevirus A within the Hepeviridae family. Due to the absence of a robust cell culture model for HEV infection, the analysis of the viral life cycle, the development of effective antivirals and a vaccine is severely limited. In this study, we established a protocol based on the HEV genotype 3 p6 (Kernow C-1) and the human hepatoma cell lines HepG2 and HepG2/C3A with different media conditions to produce intracellular HEV cell culture-derived particles (HEVcc) with viral titers between 105 and 106 FFU/mL. Viral titers could be further enhanced by an HEV variant harboring a mutation in the RNA-dependent RNA polymerase. These HEVcc particles were characterized in density gradients and allowed the trans-complementation of subgenomic reporter HEV replicons. In addition, in vitro produced intracellular-derived particles were infectious in liver-humanized mice with high RNA copy numbers detectable in serum and feces. Efficient infection of primary human and swine hepatocytes using the developed protocol could be observed and was inhibited by ribavirin. Finally, RNA sequencing studies of HEV-infected primary human hepatocytes demonstrated a temporally structured transcriptional defense response. In conclusion, this robust cell culture model of HEV infection provides a powerful tool for studying viral-host interactions that should facilitate the discovery of antiviral drugs for this important zoonotic pathogen.


Asunto(s)
Virus de la Hepatitis E/genética , Virus de la Hepatitis E/fisiología , Hepatitis E/metabolismo , Hepatocitos/virología , Animales , Antivirales/farmacología , Carcinoma Hepatocelular , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Genotipo , Células Hep G2 , Hepatitis E/virología , Virus de la Hepatitis E/efectos de los fármacos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Replicón , Ribavirina/metabolismo , Porcinos , Carga Viral , Replicación Viral
13.
Proc Natl Acad Sci U S A ; 117(3): 1678-1688, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31915293

RESUMEN

Primary human hepatocytes (PHHs) are an essential tool for modeling drug metabolism and liver disease. However, variable plating efficiencies, short lifespan in culture, and resistance to genetic manipulation have limited their use. Here, we show that the pyrrolizidine alkaloid retrorsine improves PHH repopulation of chimeric mice on average 10-fold and rescues the ability of even poorly plateable donor hepatocytes to provide cells for subsequent ex vivo cultures. These mouse-passaged (mp) PHH cultures overcome the marked donor-to-donor variability of cryopreserved PHH and remain functional for months as demonstrated by metabolic assays and infection with hepatitis B virus and Plasmodium falciparum mpPHH can be efficiently genetically modified in culture, mobilized, and then recultured as spheroids or retransplanted to create highly humanized mice that carry a genetically altered hepatocyte graft. Together, these advances provide flexible tools for the study of human liver disease and evaluation of hepatocyte-targeted gene therapy approaches.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatopatías/genética , Alcaloides de Pirrolicidina/farmacología , Animales , Trasplante de Células , Quimera , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Hepatitis B , Virus de la Hepatitis B , Hepatocitos/trasplante , Proteínas de Homeodominio/genética , Humanos , Hidrolasas/genética , Subunidad gamma Común de Receptores de Interleucina/genética , Hígado/patología , Hepatopatías/patología , Malaria , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Plasmodium falciparum
14.
J Viral Hepat ; 26(10): 1218-1223, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31194897

RESUMEN

Transmission of hepatitis E virus (HEV) through transfusion of blood components has already been reported in several European countries. Here, we assessed the HEV prevalence in Flemish blood donors. This study is of importance in order to assess the risk of HEV transmission through blood transfusion. We analysed 38 137 blood donation samples that were collected by the Red Cross Flanders during the period May-June 2015. All samples were screened for the presence of HEV RNA and a selection for HEV-specific IgM/IgG. After pooling per 6, 11 pools reacted positive during RNA screening. Reactive pools were deconstructed, and individual samples were retested. After deconstruction, seven samples were confirmed as HEV RNA positive. Serological screening of the confirmed RNA-positive samples showed that six out of these seven samples were HEV IgM positive, of which three donors were also IgG positive. Serological screening was also performed on the samples that constituted the four initially HEV RNA reactive pools where RNA positivity was not confirmed on the individual level. In three pools, we found indirect evidence of recent HEV exposure. Within 356 randomly selected samples, 31 donations were HEV IgG positive. Here we show that at least 1:5448 of blood donations in Flanders may originate from donors that are actively infected with HEV. Upon transfusion, these donations may pose a major threat towards patients at risk. Finally, a serological analysis showed that the anti-HEV IgG prevalence in Flemish blood donors is 8.71%.


Asunto(s)
Donantes de Sangre , Hepatitis E/epidemiología , Adolescente , Adulto , Anciano , Bélgica/epidemiología , Femenino , Anticuerpos Antihepatitis/sangre , Virus de la Hepatitis E/aislamiento & purificación , Humanos , Inmunoglobulina G/sangre , Masculino , Persona de Mediana Edad , Prevalencia , ARN Viral/sangre , Análisis de Secuencia de ARN , Adulto Joven
15.
J Infect Dis ; 220(5): 811-819, 2019 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-31001628

RESUMEN

BACKGROUND: Hepatitis E virus infection (HEV) is an emerging problem in developed countries. Diagnosis of HEV infection is based on the detection of HEV-specific antibodies, viral RNA, and/or antigen (Ag). Humanized mice were previously reported as a model for the study of HEV infection, but published data were focused on the quantification of viral RNA. However, the kinetics of HEV Ag expression during infection remains poorly understood. METHODS: Plasma specimens and suspensions of fecal specimens from HEV-infected and ribavirin-treated humanized mice were analyzed using HEV antigen-specific enzyme-linked immunosorbent assay, reverse transcription-quantitative polymerase chain reaction analysis, density gradient analysis, and Western blotting. RESULT: Open reading frame 2 (ORF2) Ag was detected in both plasma and stool from HEV-infected mice, and levels increased over time. Contrary to HEV RNA, ORF2 Ag levels were higher in mouse plasma than in stool. Interestingly, ORF2 was detected in plasma from mice that tested negative for HEV RNA in plasma but positive for HEV RNA in stool and was detected after viral clearance in mice that were treated with ribavirin. Plasma density gradient analysis revealed the presence of the noninfectious glycosylated form of ORF2. CONCLUSION: ORF2 Ag can be used as a marker of active HEV infection and for assessment of the effect of antiviral therapy, especially when fecal samples are not available or molecular diagnostic tests are not accessible.


Asunto(s)
Antígenos de la Hepatitis/inmunología , Virus de la Hepatitis E/inmunología , Hepatitis E/diagnóstico , Hígado/virología , Proteínas Virales/inmunología , Animales , Modelos Animales de Enfermedad , Heces/virología , Hepatitis E/tratamiento farmacológico , Hepatitis E/inmunología , Virus de la Hepatitis E/efectos de los fármacos , Humanos , Cinética , Ratones , Ratones SCID , ARN Viral/aislamiento & purificación , Ribavirina
16.
Antiviral Res ; 157: 151-158, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30036559

RESUMEN

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and a member of the genus Orthohepevirus in the family Hepeviridae. HEV infections are the common cause of acute hepatitis but can also take chronic courses. Ribavirin is the treatment of choice for most patients and type I interferon (IFN) has been evaluated in a few infected transplantation patients in vivo. However, no effective and specific treatments against HEV infections are currently available. In this study, we evaluated the natural compound silvestrol, isolated from the plant Aglaia foveolata, and known for its specific inhibition of the DEAD-box RNA helicase eIF4A in state-of-the-art HEV experimental model systems. Silvestrol blocked HEV replication of different subgenomic replicons in a dose-dependent manner at low nanomolar concentrations and acted additive to ribavirin (RBV). In addition, HEV p6-based full length replication and production of infectious particles was reduced in the presence of silvestrol. A pangenotypic effect of the compound was further demonstrated with primary isolates from four different human genotypes in HEV infection experiments of hepatocyte-like cells derived from human embryonic and induced pluripotent stem cells. In vivo, HEV RNA levels rapidly declined in the feces of treated mice while no effect was observed in the vehicle treated control animals. In conclusion, silvestrol could be identified as pangenotypic HEV replication inhibitor in vitro with additive effect to RBV and further demonstrated high potency in vivo. The compound therefore may be considered in future treatment strategies of chronic hepatitis E in immunocompromised patients.


Asunto(s)
Antivirales/farmacología , Virus de la Hepatitis E/efectos de los fármacos , Hepatitis E/tratamiento farmacológico , Triterpenos/farmacología , Replicación Viral/efectos de los fármacos , Aglaia/química , Animales , Antivirales/administración & dosificación , Antivirales/aislamiento & purificación , Células Cultivadas , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Heces/virología , Virus de la Hepatitis E/crecimiento & desarrollo , Humanos , Ratones , Ribavirina/farmacología , Triterpenos/administración & dosificación , Triterpenos/aislamiento & purificación , Carga Viral
17.
Virology ; 514: 30-41, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29128754

RESUMEN

Monoclonal antibodies (mAbs) targeting the hepatitis C virus (HCV) envelope have been raised mainly against envelope protein 2 (E2), while the antigenic epitopes of envelope protein 1 (E1) are not fully identified. Here we describe the detailed characterization of a human mAb, designated A6, generated from an HCV genotype 1b infected patient. ELISA results showed reactivity of mAb A6 to full-length HCV E1E2 of genotypes 1a, 1b and 2a. Epitope mapping identified a region spanning amino acids 230-239 within the N-terminal region of E1 as critical for binding. Antibody binding to this epitope was not conformation dependent. Neutralization assays showed that mAb A6 lacks neutralizing capacity and does not interfere with the activity of known neutralizing antibodies. In summary, mAb A6 is an important tool to study the structure and function of E1 within the viral envelope, a crucial step in the development of an effective prophylactic HCV vaccine.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Hepacivirus/inmunología , Hepatitis C/inmunología , Proteínas del Envoltorio Viral/inmunología , Anticuerpos Monoclonales/análisis , Anticuerpos Neutralizantes/análisis , Mapeo Epitopo , Genotipo , Hepacivirus/química , Hepacivirus/genética , Hepatitis C/genética , Humanos , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
18.
Antiviral Res ; 148: 53-64, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29074219

RESUMEN

Infections with hepatitis C virus (HCV) represent a worldwide health burden and a prophylactic vaccine is still not available. Liver transplantation (LT) is often the only option for patients with HCV-induced end-stage liver disease. However, immediately after transplantation, the liver graft becomes infected by circulating virus, resulting in accelerated progression of liver disease. Although the efficacy of HCV treatment using direct-acting antivirals has improved significantly, immune compromised LT-patients and patients with advanced liver disease remain difficult to treat. As an alternative approach, interfering with viral entry could prevent infection of the donor liver. We generated a human monoclonal antibody (mAb), designated 2A5, which targets the HCV envelope. The neutralizing activity of mAb 2A5 was assessed using multiple prototype and patient-derived HCV pseudoparticles (HCVpp), cell culture produced HCV (HCVcc), and a human-liver chimeric mouse model. Neutralization levels observed for mAb 2A5 were generally high and mostly superior to those obtained with AP33, a well-characterized HCV-neutralizing monoclonal antibody. Using humanized mice, complete protection was observed after genotype 1a and 4a HCV challenge, while only partial protection was achieved using gt1b and 6a isolates. Epitope mapping revealed that mAb 2A5 binding is conformation-dependent and identified the E2-region spanning amino acids 434 to 446 (epitope II) as the predominant contact domain. CONCLUSION: mAb 2A5 shows potent anti-HCV neutralizing activity both in vitro and in vivo and could hence represent a valuable candidate to prevent HCV recurrence in LT-patients. In addition, the detailed identification of the neutralizing epitope can be applied for the design of prophylactic HCV vaccines.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Hepacivirus/efectos de los fármacos , Anticuerpos contra la Hepatitis C/farmacología , Anticuerpos contra la Hepatitis C/uso terapéutico , Hepatitis C/tratamiento farmacológico , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Modelos Animales de Enfermedad , Mapeo Epitopo , Epítopos/genética , Epítopos/inmunología , Genotipo , Hepacivirus/genética , Hepacivirus/inmunología , Hepatitis C/inmunología , Hepatitis C/prevención & control , Hepatitis C/virología , Anticuerpos contra la Hepatitis C/inmunología , Humanos , Trasplante de Hígado , Ratones , Ratones SCID , Mutación , Pruebas de Neutralización , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/genética , Internalización del Virus/efectos de los fármacos
19.
Antiviral Res ; 141: 150-154, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28232247

RESUMEN

Hepatitis E virus (HEV) is considered as an important pathogen in developing countries but there is growing evidence of its increasing significance and prevalence in the Western world. Although most acute HEV infections resolve spontaneously, chronicity has been observed in immunocompromised patients. The study of HEV has been hampered by the absence of practical animal models. Because the in vivo study of HEV was essentially limited to primates and pigs we recently established the human-liver chimeric uPA-SCID mouse model as a useful tool to study HEV infection. Because the humanized FRG mouse model, another type of mouse with humanized liver, is more easily accessible to the scientific community, we investigated its susceptibility to HEV infection. FRG mice were transplanted with human hepatocytes and challenged with different HEV genotypes using different routes of exposure. Our data clearly shows that the humanized FRG mouse is an alternative animal model for the study HEV infection. As observed in the uPA-SCID model, controlled oral inoculation did not lead to active infection. However, intrasplenic injection of genotype 3-infected patient plasma did result into persistent infection. Although the efficiency of transmission was low, this observation corroborates previously published case reports of blood transfusion-associated HEV transmission.


Asunto(s)
Modelos Animales de Enfermedad , Hepatitis E/transmisión , Hepatocitos/virología , Hígado/virología , Plasma/virología , ARN Viral/sangre , Animales , Transfusión Sanguínea , Proteínas de Unión al ADN/deficiencia , Genotipo , Hepatitis E/virología , Virus de la Hepatitis E/genética , Virus de la Hepatitis E/aislamiento & purificación , Virus de la Hepatitis E/fisiología , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Ratones , Ratones SCID
20.
Antiviral Res ; 139: 129-137, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28062191

RESUMEN

Due to the highly restricted species-tropism of Hepatitis C virus (HCV) a limited number of animal models exist for pre-clinical evaluation of vaccines and antiviral compounds. The human-liver chimeric mouse model allows heterologous challenge with clinically relevant strains derived from patients. However, to date, the transmission and longitudinal evolution of founder viral populations in this model have not been characterized in-depth using state-of-the-art sequencing technologies. Focusing on NS3 protease encoding region of the viral genome, mutant spectra in a donor inoculum and individual recipient mice were determined via Illumina sequencing and compared, to determine the effects of transmission on founder viral population complexity. In all transmissions, a genetic bottleneck was observed, although diverse viral populations were transmitted in each case. A low frequency cloud of mutations (<1%) was detectable in the donor inoculum and recipient mice, with single nucleotide variants (SNVs) > 1% restricted to a subset of nucleotides. The population of SNVs >1% was reduced upon transmission while the low frequency SNV cloud remained stable. Fixation of multiple identical synonymous substitutions was apparent in independent transmissions, and no evidence for reversion of T-cell epitopes was observed. In addition, susceptibility of founder populations to antiviral therapy was assessed. Animals were treated with protease inhibitor (PI) monotherapy to track resistance associated substitution (RAS) emergence. Longitudinal analyses revealed a decline in population diversity under therapy, with no detectable RAS >1% prior to therapy commencement. Despite inoculation from a common source and identical therapeutic regimens, unique RAS emergence profiles were identified in different hosts prior to and during therapeutic failure, with complex mutational signatures at protease residues 155, 156 and 168 detected. Together these analyses track viral population complexity at high-resolution in the human-liver chimeric mouse model post-transmission and under therapeutic intervention, revealing novel insights into the evolutionary processes which shape viral protease population composition at various critical stages of the viral life-cycle.


Asunto(s)
Hepacivirus/enzimología , Hepacivirus/genética , Hepatitis C/transmisión , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Animales , Antivirales/uso terapéutico , Modelos Animales de Enfermedad , Evolución Molecular , Variación Genética , Genoma Viral , Genotipo , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Hepatitis C/virología , Humanos , Ratones , Mutación , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/uso terapéutico , Análisis de Secuencia de ADN , Serina Endopeptidasas/química , Proteínas no Estructurales Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA