45 results on '"Verhoye L"'
Search Results
2. Human skin-derived precusor cells: A potential source for cellular therapy of the liver
- Author
-
Kock, J.D., primary, Rodrigues, R.M., additional, Najar, M., additional, Verhoye, L., additional, Govaere, O., additional, Lootens, L., additional, Eenoo, P.V., additional, Sanders, N., additional, Lagneaux, L., additional, Leroux-Roels, G., additional, Roskams, T., additional, Meuleman, P., additional, Vanhaecke, T., additional, and Rogiers, V., additional
- Published
- 2018
- Full Text
- View/download PDF
3. Effect of hepatitis E virus infection on the human hepatic innate immune response in human liver chimeric mice
- Author
-
Sayed, I.M., primary, Verhoye, L., additional, Cocquerel, L., additional, Abravanel, F., additional, Foquet, L., additional, Montpellier, C., additional, Debing, Y., additional, Farhoudi, A., additional, Wychowski, C., additional, Dubuisson, J., additional, Leroux-Roels, G., additional, Neyts, J., additional, Izopet, J., additional, Michiels, T., additional, and Meuleman, P., additional
- Published
- 2017
- Full Text
- View/download PDF
4. Anti-CD81 but not anti-SR-BI blocks Plasmodium falciparum liver infection in a humanized mouse model
- Author
-
Foquet, L., Hermsen, C.C., Verhoye, L., Gemert, G.J.A. van, Cortese, R., Nicosia, A., Sauerwein, R.W., Leroux-Roels, G., Meuleman, P., Foquet, L., Hermsen, C.C., Verhoye, L., Gemert, G.J.A. van, Cortese, R., Nicosia, A., Sauerwein, R.W., Leroux-Roels, G., and Meuleman, P.
- Abstract
Item does not contain fulltext, OBJECTIVES: Plasmodium falciparum sporozoites, deposited in the skin by infected Anopheles mosquitoes taking a blood meal, cross the endothelium of skin capillaries and travel to the liver where they traverse Kupffer cells and hepatocytes to finally invade a small number of the latter. In hepatocytes, sporozoites replicate, differentiate and give rise to large numbers of merozoites that are released into the bloodstream where they invade red blood cells, thus initiating the symptomatic blood stage. Using in vitro systems and rodent models, it has been shown that the hepatocyte receptors CD81 and scavenger receptor type B class I (SR-BI) play a pivotal role during sporozoite invasion. We wanted to evaluate whether these two entry factors are genuine drug targets for the prevention of P. falciparum infection in humans. METHODS: Immunodeficient mice of which the liver is largely repopulated by human hepatocytes were treated with monoclonal antibodies blocking either CD81 or SR-BI 1 day prior to challenge with infected mosquitoes. P. falciparum infection of the liver was demonstrated using a qPCR assay. RESULTS: In human liver chimeric mice, an antibody directed against CD81 completely blocked P. falciparum sporozoite invasion while SR-BI-specific monoclonal antibodies did not influence in vivo infection. CONCLUSIONS: These observations confirm the role of CD81 in liver-stage malaria and question that of SR-BI. CD81 might be a valuable drug target for the prevention of malaria.
- Published
- 2015
5. FRI-122 - Human skin-derived precusor cells: A potential source for cellular therapy of the liver
- Author
-
Kock, J.D., Rodrigues, R.M., Najar, M., Verhoye, L., Govaere, O., Lootens, L., Eenoo, P.V., Sanders, N., Lagneaux, L., Leroux-Roels, G., Roskams, T., Meuleman, P., Vanhaecke, T., and Rogiers, V.
- Published
- 2018
- Full Text
- View/download PDF
6. G09 : Targeting a host-cell entry factor barricades antiviral resistant HCV variants from on-therapy breakthrough in human-liver mice
- Author
-
Vercauteren, K., primary, Brown, R., additional, Mesalam, A.A., additional, Doerrbecker, J., additional, Bhuju, S., additional, Geffers, R., additional, Van Den Eede, N., additional, Troise, F., additional, Verhoye, L., additional, Baumert, T., additional, Farhoudi, A., additional, Cortese, R., additional, Leroux-Roels, G., additional, Pietschmann, T., additional, Nicosia, A., additional, and Meuleman, P., additional
- Published
- 2015
- Full Text
- View/download PDF
7. 827 EXPERIMENTAL HCV VACCINE COMBINING ADENOVIRUS AND PROTEIN ELICITS POTENT T-CELL RESPONSES AND NEUTRALIZING ANTIBODIES IN RODENTS
- Author
-
Chmielewska, A., primary, Naddeo, M., additional, Ammendola, V., additional, Hu, K., additional, Verhoye, L., additional, Rappuoli, R., additional, Colloca, S., additional, Nicosia, A., additional, Cortese, R., additional, Leroux-Roels, G., additional, Meuleman, P., additional, Bienkowska-Szewczyk, K., additional, Balfe, P., additional, McKeating, J.A., additional, and Folgori, A., additional
- Published
- 2012
- Full Text
- View/download PDF
8. 861 THE LECTIN GRIFFITHSIN HAS ANTIVIRAL ACTIVITY AGAINST HEPATITIS C VIRUS IN VITRO AND IN VIVO
- Author
-
Meuleman, P., primary, Albecka, A., additional, Belouzard, S., additional, Vercauteren, K., additional, Verhoye, L., additional, Wychowski, C., additional, Leroux-Roels, G., additional, Palmer, K.E., additional, and Dubuisson, J., additional
- Published
- 2012
- Full Text
- View/download PDF
9. 101 HUMAN MONOCLONAL ANTIBODIES TARGETING SR-BI PREVENT HCV ENTRY AND VIRAL SPREAD IN VIVO
- Author
-
Meuleman, P., primary, Catanese, M.T., additional, Verhoye, L., additional, Desombere, I., additional, Jones, C.T., additional, Walter, H., additional, Grzyb, K., additional, Cortese, R., additional, Rice, C.M., additional, Leroux-Roels, G., additional, and Nicosia, A., additional
- Published
- 2011
- Full Text
- View/download PDF
10. Novel human SR-BI antibodies prevent infection and dissemination of HCV in vitro and in humanized mice
- Author
-
Arvind H. Patel, Krzysztof Lacek, Lieven Verhoye, Alfredo Nicosia, Riccardo Cortese, M. Naddeo, Samira Fafi-Kremer, Thomas F. Baumert, Marek Patryk Słowikowski, Koen Vercauteren, Katarzyna Grzyb, Antonella Folgori, Philip Meuleman, Geert Leroux-Roels, Immuno-Rhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Strasbourg (UNISTRA), Institut de Recherche sur les Maladies Virales et Hépatiques (IVH), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM), Lacek, K, Vercauteren, K, Grzyb, K, Naddeo, M, Verhoye, L, S?owikowski, Mp, Fafi Kremer, S, Patel, Ah, Baumert, Tf, Folgori, A, Leroux Roels, G, Cortese, R, Meuleman, P, and Nicosia, Alfredo
- Subjects
PHARMACOKINETICS ,medicine.medical_treatment ,Immunoglobulin Variable Region ,UPA-SCID MOUSE ,Hepacivirus ,Mice, SCID ,Liver transplantation ,HEPATITIS-C VIRUS ,medicine.disease_cause ,Liver disease ,Mice ,Cricetinae ,education.field_of_study ,Antibodies, Monoclonal ,Hep G2 Cells ,Scavenger Receptors, Class B ,LIVER-TRANSPLANT RECIPIENTS ,CELL ENTRY ,Viral hepatitis ,SCAVENGER RECEPTOR ,Genotype ,medicine.drug_class ,Hepatitis C virus ,Population ,Molecular Sequence Data ,Transplantation, Heterologous ,CHO Cells ,Biology ,Monoclonal antibody ,Virus ,NEUTRALIZING ANTIBODIES ,Peptide Library ,medicine ,Animals ,Humans ,Amino Acid Sequence ,education ,Transplantation Chimera ,Hepatology ,GENOTYPE 1 INFECTION ,Hepatitis C, Chronic ,medicine.disease ,Virology ,Transplantation ,Immunoglobulin G ,Immunology ,TELAPREVIR ,Hepatocytes ,HIGH-DENSITY-LIPOPROTEIN ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
BACKGROUND & AIMS: Hepatitis C virus (HCV)-induced end-stage liver disease is currently the major indication for liver transplantation in the Western world. After transplantation, the donor liver almost inevitably becomes infected by the circulating virus and disease progression is accelerated in immune suppressed transplant patients. The current standard therapy, based on pegylated interferon and ribavirin, induces severe side effects and is often ineffective in this population. Therefore, new strategies to prevent graft re-infection are urgently needed. We have previously shown that monoclonal antibodies (mAbs) against the HCV co-receptor scavenger receptor class B type I (SR-BI/Cla1) inhibit infection by different HCV genotypes in cell culture. METHODS: Using phage display libraries, we have generated a large set of novel human mAbs against SR-BI and evaluated their effectiveness in preventing HCV infection and direct cell-to-cell spread in vitro and in vivo using uPA-SCID mice with a humanized liver. RESULTS: Eleven human monoclonal antibodies were generated that specifically recognize SR-BI. Two antibodies, mAb8 and mAb151, displayed the highest binding and inhibitory properties and also interfered with direct cell-to-cell spread in vitro. Studies in humanized mice showed that both antibodies were capable of preventing HCV infection and could block intrahepatic spread and virus amplification when administered 3 days after infection. Interestingly, anti-SR-BI therapy was effective against an HCV variant that escaped the control of the adaptive immune response in a liver transplant patient. CONCLUSIONS: The anti-SR-BI mAbs generated in this study may represent novel therapeutic tools to prevent HCV re-infection of liver allografts.
- Published
- 2011
11. JFH1-based Core-NS2 genotype variants of HCV with genetic stability in vivo and in vitro: Important tools in the evaluation of virus neutralization.
- Author
-
Collignon L, Holmbeck K, Just A, Verhoye L, Velázquez-Moctezuma R, Fahnøe U, Carlsen THR, Law M, Prentoe J, Scheel TKH, Gottwein JM, Meuleman P, and Bukh J
- Subjects
- Animals, Humans, Mice, Antibodies, Neutralizing immunology, Neutralization Tests, Hepatitis C prevention & control, Hepatitis C virology, Hepatitis C immunology, Viral Nonstructural Proteins genetics, Viral Nonstructural Proteins immunology, Hepacivirus genetics, Hepacivirus immunology, Genotype
- Abstract
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., (Copyright © 2024 American Association for the Study of Liver Diseases.)
- Published
- 2024
- Full Text
- View/download PDF
12. Phosphatidylinositol 4-Kinase III Alpha Governs Cytoskeletal Organization for Invasiveness of Liver Cancer Cells.
- Author
-
Tran CS, Kersten J, Yan J, Breinig M, Huth T, Poth T, Colasanti O, Riedl T, Faure-Dupuy S, Diehl S, Verhoye L, Li TF, Lingemann M, Schult P, Ahlén G, Frelin L, Kühnel F, Vondran FWR, Breuhahn K, Meuleman P, Heikenwälder M, Schirmacher P, Bartenschlager R, Laketa V, Roessler S, Tschaharganeh DF, Sällberg M, and Lohmann V
- Subjects
- Humans, Animals, Mice, Phosphorylation, Hepacivirus, Cell Line, Tumor, Phosphotransferases (Alcohol Group Acceptor) metabolism, Phosphotransferases (Alcohol Group Acceptor) genetics, Hep G2 Cells, Hepatitis C pathology, Hepatitis C metabolism, Hepatitis C virology, RNA Interference, Liver Neoplasms pathology, Liver Neoplasms metabolism, Liver Neoplasms genetics, Carcinoma, Hepatocellular pathology, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular genetics, Cytoskeleton metabolism, Cytoskeleton pathology, Neoplasm Invasiveness, Paxillin metabolism, Signal Transduction, Cell Movement, Actin Depolymerizing Factors metabolism, Actin Depolymerizing Factors genetics
- Abstract
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., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Novel prime-boost immune-based therapy inhibiting both hepatitis B and D virus infections.
- Author
-
Burm R, Maravelia P, Ahlen G, Ciesek S, Caro Perez N, Pasetto A, Urban S, Van Houtte F, Verhoye L, Wedemeyer H, Johansson M, Frelin L, Sällberg M, and Meuleman P
- Subjects
- Humans, Mice, Animals, Rabbits, Hepatitis delta Antigens, Hepatitis B Surface Antigens, Hepatitis Delta Virus genetics, Hepatitis B virus genetics, Antibodies, Viral, Mice, Transgenic, Hepatitis B, Chronic prevention & control, Superinfection prevention & control, Hepatitis B prevention & control
- Abstract
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., Competing Interests: Competing interests: MS and LF are founders and shareholders of Svenska Vaccinfabriken, which holds the IP to the immunogens described in this manuscript. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
14. A human monoclonal antibody against HBsAg for the prevention and treatment of chronic HBV and HDV infection.
- Author
-
Burm R, Van Houtte F, Verhoye L, Mesalam AA, Ciesek S, Roingeard P, Wedemeyer H, Leroux-Roels G, and Meuleman P
- Abstract
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 IC
50 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., Competing Interests: Nothing to declare related to the content of this manuscript. Please refer to the accompanying ICMJE disclosure forms for further details., (© 2022 The Authors.)- Published
- 2022
- Full Text
- View/download PDF
15. A ribavirin-induced ORF2 single-nucleotide variant produces defective hepatitis E virus particles with immune decoy function.
- Author
-
Meister TL, Brüggemann Y, Nocke MK, Ulrich RG, Schuhenn J, Sutter K, Gömer A, Bader V, Winklhofer KF, Broering R, Verhoye L, Meuleman P, Vondran FWR, Camuzet C, Cocquerel L, Todt D, and Steinmann E
- Subjects
- Cell Line, Tumor, Hepatocytes virology, Humans, Neoplasm Recurrence, Local genetics, Nucleotides, RNA, Viral, Virus Replication, Hepatitis E virus genetics, Hepatitis E virus physiology, Ribavirin pharmacology, Viral Proteins genetics
- Abstract
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.
- Published
- 2022
- Full Text
- View/download PDF
16. Viral Interference of Hepatitis C and E Virus Replication in Novel Experimental Co-Infection Systems.
- Author
-
Burkard T, Proske N, Resner K, Collignon L, Knegendorf L, Friesland M, Verhoye L, Sayed IM, Brüggemann Y, Nocke MK, Behrendt P, Wedemeyer H, Meuleman P, Todt D, and Steinmann E
- Subjects
- Animals, Hepacivirus genetics, Mice, Viral Interference, Virus Replication, Coinfection, Hepatitis C, Hepatitis E virus genetics
- Abstract
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.
- Published
- 2022
- Full Text
- View/download PDF
17. Study of Hepatitis E Virus-4 Infection in Human Liver-Chimeric, Immunodeficient, and Immunocompetent Mice.
- Author
-
Collignon L, Verhoye L, Hakze-Van der Honing R, Van der Poel WHM, and Meuleman P
- Abstract
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., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Collignon, Verhoye, Hakze-Van der Honing, Van der Poel and Meuleman.)
- Published
- 2022
- Full Text
- View/download PDF
18. A Role for B Cells to Transmit Hepatitis C Virus Infection.
- Author
-
Desombere I, Van Houtte F, Farhoudi A, Verhoye L, Buysschaert C, Gijbels Y, Couvent S, Swinnen W, Van Vlierberghe H, Elewaut A, Magri A, Stamataki Z, Meuleman P, McKeating JA, and Leroux-Roels G
- Subjects
- Adult, Animals, Antibodies, Viral blood, Antibodies, Viral immunology, Broadly Neutralizing Antibodies blood, Broadly Neutralizing Antibodies immunology, Disease Models, Animal, Female, Hepacivirus immunology, Hepacivirus isolation & purification, Hepatitis C blood, Hepatitis C prevention & control, Hepatitis C virology, Humans, Liver pathology, Liver virology, Liver Transplantation, Male, Mice, Middle Aged, Serum virology, Transplantation Chimera, Vaccine Development methods, Viral Envelope Proteins immunology, Viral Hepatitis Vaccines therapeutic use, Young Adult, B-Lymphocytes virology, Hepacivirus pathogenicity, Hepatitis C transmission
- Abstract
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., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Desombere, Van Houtte, Farhoudi, Verhoye, Buysschaert, Gijbels, Couvent, Swinnen, Van Vlierberghe, Elewaut, Magri, Stamataki, Meuleman, McKeating and Leroux-Roels.)
- Published
- 2021
- Full Text
- View/download PDF
19. Blocking Entry of Hepatitis B and D Viruses to Hepatocytes as a Novel Immunotherapy for Treating Chronic Infections.
- Author
-
Maravelia P, Frelin L, Ni Y, Caro Pérez N, Ahlén G, Jagya N, Verch G, Verhoye L, Pater L, Johansson M, Pasetto A, Meuleman P, Urban S, and Sällberg M
- Subjects
- Animals, Female, Hepatitis B Vaccines, Hepatocytes drug effects, Humans, Immunotherapy methods, Mice, Mice, Inbred C57BL, Mice, SCID, Mice, Transgenic, Rabbits, Hepatitis B virus immunology, Hepatitis B, Chronic drug therapy, Hepatitis D, Chronic drug therapy, Hepatitis Delta Virus immunology, Virus Internalization drug effects
- Abstract
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., (© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America.)
- Published
- 2021
- Full Text
- View/download PDF
20. Liver-expressed Cd302 and Cr1l limit hepatitis C virus cross-species transmission to mice.
- Author
-
Brown RJP, Tegtmeyer B, Sheldon J, Khera T, Anggakusuma, Todt D, Vieyres G, Weller R, Joecks S, Zhang Y, Sake S, Bankwitz D, Welsch K, Ginkel C, Engelmann M, Gerold G, Steinmann E, Yuan Q, Ott M, Vondran FWR, Krey T, Ströh LJ, Miskey C, Ivics Z, Herder V, Baumgärtner W, Lauber C, Seifert M, Tarr AW, McClure CP, Randall G, Baktash Y, Ploss A, Thi VLD, Michailidis E, Saeed M, Verhoye L, Meuleman P, Goedecke N, Wirth D, Rice CM, and Pietschmann T
- Subjects
- Animals, Mice, Mice, Transgenic, Virus Internalization, Hepacivirus genetics, Hepatitis C
- Abstract
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., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2020
- Full Text
- View/download PDF
21. Inflammation Alters the Secretome and Immunomodulatory Properties of Human Skin-Derived Precursor Cells.
- Author
-
De Kock J, Rodrigues RM, Branson S, Verhoye L, Colemonts-Vroninks H, Rombaut M, Boeckmans J, Neuckermans J, Lequeue S, Buyl K, Merimi M, Moussa Agha D, De Boe V, Lagneaux L, Meuleman P, Vanhaecke T, and Najar M
- Subjects
- Animals, Cells, Cultured, Humans, Mice, Mice, SCID, Skin cytology, Cytokines metabolism, Immunomodulation immunology, Inflammation immunology, Skin metabolism
- Abstract
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.
- Published
- 2020
- Full Text
- View/download PDF
22. Robust hepatitis E virus infection and transcriptional response in human hepatocytes.
- Author
-
Todt D, Friesland M, Moeller N, Praditya D, Kinast V, Brüggemann Y, Knegendorf L, Burkard T, Steinmann J, Burm R, Verhoye L, Wahid A, Meister TL, Engelmann M, Pfankuche VM, Puff C, Vondran FWR, Baumgärtner W, Meuleman P, Behrendt P, and Steinmann E
- Subjects
- Animals, Antiviral Agents pharmacology, Carcinoma, Hepatocellular, Cell Culture Techniques, Cell Line, Tumor, Genotype, Hep G2 Cells, Hepatitis E virology, Hepatitis E virus drug effects, Humans, Liver Neoplasms drug therapy, Mice, RNA-Dependent RNA Polymerase genetics, RNA-Dependent RNA Polymerase metabolism, Replicon, Ribavirin metabolism, Swine, Viral Load, Virus Replication, Hepatitis E metabolism, Hepatitis E virus genetics, Hepatitis E virus physiology, Hepatocytes virology
- Abstract
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 10
5 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., Competing Interests: The authors declare no competing interest., (Copyright © 2020 the Author(s). Published by PNAS.)- Published
- 2020
- Full Text
- View/download PDF
23. Expansion, in vivo-ex vivo cycling, and genetic manipulation of primary human hepatocytes.
- Author
-
Michailidis E, Vercauteren K, Mancio-Silva L, Andrus L, Jahan C, Ricardo-Lax I, Zou C, Kabbani M, Park P, Quirk C, Pyrgaki C, Razooky B, Verhoye L, Zoluthkin I, Lu WY, Forbes SJ, Chiriboga L, Theise ND, Herzog RW, Suemizu H, Schneider WM, Shlomai A, Meuleman P, Bhatia SN, Rice CM, and de Jong YP
- Subjects
- Animals, Cell Transplantation, Chimera, Disease Models, Animal, Female, Genetic Therapy, Hepatitis B, Hepatitis B virus, Hepatocytes transplantation, Homeodomain Proteins genetics, Humans, Hydrolases genetics, Interleukin Receptor Common gamma Subunit genetics, Liver pathology, Liver Diseases pathology, Malaria, Male, Mice, Mice, Inbred NOD, Mice, Knockout, Plasmodium falciparum, Hepatocytes drug effects, Hepatocytes metabolism, Liver Diseases genetics, Pyrrolizidine Alkaloids pharmacology
- Abstract
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., Competing Interests: The authors declare no competing interest., (Copyright © 2020 the Author(s). Published by PNAS.)
- Published
- 2020
- Full Text
- View/download PDF
24. Hepatitis E virus prevalence in Flemish blood donors.
- Author
-
Vercouter AS, Van Houtte F, Verhoye L, González Fraile I, Blanco L, Compernolle V, and Meuleman P
- Subjects
- Adolescent, Adult, Aged, Belgium epidemiology, Female, Hepatitis Antibodies blood, Hepatitis E virus isolation & purification, Humans, Immunoglobulin G blood, Male, Middle Aged, Prevalence, RNA, Viral blood, Sequence Analysis, RNA, Young Adult, Blood Donors, Hepatitis E epidemiology
- Abstract
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%., (© 2019 John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
25. Hepatitis E Virus (HEV) Open Reading Frame 2 Antigen Kinetics in Human-Liver Chimeric Mice and Its Impact on HEV Diagnosis.
- Author
-
Sayed IM, Verhoye L, Montpellier C, Abravanel F, Izopet J, Cocquerel L, and Meuleman P
- Subjects
- Animals, Disease Models, Animal, Feces virology, Hepatitis E drug therapy, Hepatitis E immunology, Hepatitis E virus drug effects, Humans, Kinetics, Mice, Mice, SCID, RNA, Viral isolation & purification, Ribavirin, Hepatitis Antigens immunology, Hepatitis E diagnosis, Hepatitis E virus immunology, Liver virology, Viral Proteins immunology
- Abstract
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., (© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
26. The natural compound silvestrol inhibits hepatitis E virus (HEV) replication in vitro and in vivo.
- Author
-
Todt D, Moeller N, Praditya D, Kinast V, Friesland M, Engelmann M, Verhoye L, Sayed IM, Behrendt P, Dao Thi VL, Meuleman P, and Steinmann E
- Subjects
- Aglaia chemistry, Animals, Antiviral Agents administration & dosage, Antiviral Agents isolation & purification, Cells, Cultured, Disease Models, Animal, Drug Interactions, Feces virology, Hepatitis E virus growth & development, Humans, Mice, Ribavirin pharmacology, Triterpenes administration & dosage, Triterpenes isolation & purification, Viral Load, Antiviral Agents pharmacology, Hepatitis E drug therapy, Hepatitis E virus drug effects, Triterpenes pharmacology, Virus Replication drug effects
- Abstract
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., (Copyright © 2018 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
27. Development and characterization of a human monoclonal antibody targeting the N-terminal region of hepatitis C virus envelope glycoprotein E1.
- Author
-
Mesalam AA, Desombere I, Farhoudi A, Van Houtte F, Verhoye L, Ball J, Dubuisson J, Foung SKH, Patel AH, Persson MAA, Leroux-Roels G, and Meuleman P
- Subjects
- Antibodies, Monoclonal analysis, Antibodies, Neutralizing analysis, Epitope Mapping, Genotype, Hepacivirus chemistry, Hepacivirus genetics, Hepatitis C genetics, Humans, Viral Envelope Proteins chemistry, Viral Envelope Proteins genetics, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Hepacivirus immunology, Hepatitis C immunology, Viral Envelope Proteins immunology
- Abstract
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., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
28. A novel neutralizing human monoclonal antibody broadly abrogates hepatitis C virus infection in vitro and in vivo.
- Author
-
Desombere I, Mesalam AA, Urbanowicz RA, Van Houtte F, Verhoye L, Keck ZY, Farhoudi A, Vercauteren K, Weening KE, Baumert TF, Patel AH, Foung SKH, Ball J, Leroux-Roels G, and Meuleman P
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antibodies, Neutralizing immunology, Disease Models, Animal, Epitope Mapping, Epitopes genetics, Epitopes immunology, Genotype, Hepacivirus genetics, Hepacivirus immunology, Hepatitis C immunology, Hepatitis C prevention & control, Hepatitis C virology, Hepatitis C Antibodies immunology, Humans, Liver Transplantation, Mice, Mice, SCID, Mutation, Neutralization Tests, Structure-Activity Relationship, Viral Envelope Proteins genetics, Virus Internalization drug effects, Antibodies, Neutralizing pharmacology, Antibodies, Neutralizing therapeutic use, Hepacivirus drug effects, Hepatitis C drug therapy, Hepatitis C Antibodies pharmacology, Hepatitis C Antibodies therapeutic use, Viral Envelope Proteins immunology
- Abstract
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., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
29. Study of hepatitis E virus infection of genotype 1 and 3 in mice with humanised liver.
- Author
-
Sayed IM, Verhoye L, Cocquerel L, Abravanel F, Foquet L, Montpellier C, Debing Y, Farhoudi A, Wychowski C, Dubuisson J, Leroux-Roels G, Neyts J, Izopet J, Michiels T, and Meuleman P
- Subjects
- Animals, Antiviral Agents therapeutic use, Gene Expression, Genotype, Hepatitis E drug therapy, Hepatitis E genetics, Hepatocytes transplantation, Hepatocytes virology, Host-Pathogen Interactions, Humans, Mice, Ribavirin therapeutic use, Transplantation Chimera, Viral Load, Disease Models, Animal, Hepatitis E virology, Hepatitis E virus genetics, Liver chemistry, RNA, Viral analysis, Viral Proteins analysis
- Abstract
Objective: The hepatitis E virus (HEV) is responsible for approximately 20 million infections per year worldwide. Although most infected people can spontaneously clear an HEV infection, immune-compromised individuals may evolve towards chronicity. Chronic HEV infection can be cured using ribavirin, but viral isolates with low ribavirin sensitivity have recently been identified. Although some HEV isolates can be cultured in vitro, in vivo studies are essentially limited to primates and pigs. Since the use of these animals is hampered by financial, practical and/or ethical concerns, we evaluated if human liver chimeric mice could serve as an alternative., Design: Humanised mice were inoculated with different HEV-containing preparations., Results: Chronic HEV infection was observed after intrasplenic injection of cell culture-derived HEV, a filtered chimpanzee stool suspension and a patient-derived stool suspension. The viral load was significantly higher in the stool compared with the plasma. Overall, the viral titre in genotype 3-infected mice was lower than that in genotype 1-infected mice. Analysis of liver tissue of infected mice showed the presence of viral RNA and protein, and alterations in host gene expression. Intrasplenic injection of HEV-positive patient plasma and oral inoculation of filtered stool suspensions did not result in robust infection. Finally, we validated our model for the evaluation of novel antiviral compounds against HEV using ribavirin., Conclusions: Human liver chimeric mice can be infected with HEV of different genotypes. This small animal model will be a valuable tool for the in vivo study of HEV infection and the evaluation of novel antiviral molecules., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)
- Published
- 2017
- Full Text
- View/download PDF
30. Transmission of hepatitis E virus infection to human-liver chimeric FRG mice using patient plasma.
- Author
-
Sayed IM, Foquet L, Verhoye L, Abravanel F, Farhoudi A, Leroux-Roels G, Izopet J, and Meuleman P
- Subjects
- Animals, Blood Transfusion, DNA-Binding Proteins deficiency, Genotype, Hepatitis E virology, Hepatitis E virus genetics, Hepatitis E virus isolation & purification, Hepatitis E virus physiology, Humans, Interleukin Receptor Common gamma Subunit deficiency, Mice, Mice, SCID, Disease Models, Animal, Hepatitis E transmission, Hepatocytes virology, Liver virology, Plasma virology, RNA, Viral blood
- Abstract
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., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
31. Tracking HCV protease population diversity during transmission and susceptibility of founder populations to antiviral therapy.
- Author
-
Khera T, Todt D, Vercauteren K, McClure CP, Verhoye L, Farhoudi A, Bhuju S, Geffers R, Baumert TF, Steinmann E, Meuleman P, Pietschmann T, and Brown RJ
- Subjects
- Animals, Antiviral Agents therapeutic use, Disease Models, Animal, Evolution, Molecular, Genetic Variation, Genome, Viral, Genotype, Hepacivirus drug effects, Hepatitis C drug therapy, Hepatitis C virology, Humans, Mice, Mutation, Protease Inhibitors administration & dosage, Protease Inhibitors therapeutic use, Sequence Analysis, DNA, Serine Endopeptidases chemistry, Viral Nonstructural Proteins chemistry, Hepacivirus enzymology, Hepacivirus genetics, Hepatitis C transmission, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, Viral Nonstructural Proteins genetics, Viral Nonstructural Proteins metabolism
- Abstract
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., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
32. Successful Engraftment of Human Hepatocytes in uPA-SCID and FRG ® KO Mice.
- Author
-
Foquet L, Wilson EM, Verhoye L, Grompe M, Leroux-Roels G, Bial J, and Meuleman P
- Subjects
- Acetoacetates metabolism, Acetoacetates toxicity, Animals, Chemical and Drug Induced Liver Injury genetics, Disease Models, Animal, Humans, Hydrolases genetics, Liver cytology, Liver metabolism, Liver surgery, Mice, Mice, Knockout, Mice, SCID, Transplantation Chimera genetics, Urokinase-Type Plasminogen Activator genetics, Cell Transplantation methods, Chemical and Drug Induced Liver Injury surgery, Hepatocytes transplantation, Transplantation Chimera physiology
- Abstract
Mice with humanized chimeric liver are promising in vivo tools to evaluate the efficacy of novel compounds or vaccine induced antibodies directed against pathogens that infect the human liver. In addition they can be used to study the human-type metabolism of medicinal compounds and hepatotoxicity.
- Published
- 2017
- Full Text
- View/download PDF
33. Targeting a host-cell entry factor barricades antiviral-resistant HCV variants from on-therapy breakthrough in human-liver mice.
- Author
-
Vercauteren K, Brown RJ, Mesalam AA, Doerrbecker J, Bhuju S, Geffers R, Van Den Eede N, McClure CP, Troise F, Verhoye L, Baumert T, Farhoudi A, Cortese R, Ball JK, Leroux-Roels G, Pietschmann T, Nicosia A, and Meuleman P
- Subjects
- Amino Acid Substitution, Animals, Disease Models, Animal, Genotype, Hepacivirus drug effects, Hepatitis C, Chronic drug therapy, Liver drug effects, Mice, Mutation, Missense, Viral Nonstructural Proteins genetics, Antiviral Agents pharmacology, Drug Resistance, Viral genetics, Hepacivirus genetics, Protease Inhibitors pharmacology
- Abstract
Objective: Direct-acting antivirals (DAAs) inhibit hepatitis C virus (HCV) infection by targeting viral proteins that play essential roles in the replication process. However, selection of resistance-associated variants (RAVs) during DAA therapy has been a cause of therapeutic failure. In this study, we wished to address whether such RAVs could be controlled by the co-administration of host-targeting entry inhibitors that prevent intrahepatic viral spread., Design: We investigated the effect of adding an entry inhibitor (the anti-scavenger receptor class B type I mAb1671) to a DAA monotherapy (the protease inhibitor ciluprevir) in human-liver mice chronically infected with HCV of genotype 1b. Clinically relevant non-laboratory strains were used to achieve viraemia consisting of a cloud of related viral variants (quasispecies) and the emergence of RAVs was monitored at high resolution using next-generation sequencing., Results: HCV-infected human-liver mice receiving DAA monotherapy rapidly experienced on-therapy viral breakthrough. Deep sequencing of the HCV protease domain confirmed the manifestation of drug-resistant mutants upon viral rebound. In contrast, none of the mice treated with a combination of the DAA and the entry inhibitor experienced on-therapy viral breakthrough, despite detection of RAV emergence in some animals., Conclusions: This study provides preclinical in vivo evidence that addition of an entry inhibitor to an anti-HCV DAA regimen restricts the breakthrough of DAA-resistant viruses. Our approach is an excellent strategy to prevent therapeutic failure caused by on-therapy rebound of DAA-RAVs. Inclusion of an entry inhibitor to the newest DAA combination therapies may further increase response rates, especially in difficult-to-treat patient populations., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)
- Published
- 2016
- Full Text
- View/download PDF
34. HVR1-mediated antibody evasion of highly infectious in vivo adapted HCV in humanised mice.
- Author
-
Prentoe J, Verhoye L, Velázquez Moctezuma R, Buysschaert C, Farhoudi A, Wang R, Alter H, Meuleman P, and Bukh J
- Subjects
- Amino Acid Substitution genetics, Animals, Antibodies, Neutralizing blood, Disease Models, Animal, Genotype, Hepacivirus pathogenicity, Hepatitis C genetics, Hepatitis C immunology, Hepatitis C prevention & control, Hepatitis C virology, In Vitro Techniques, Mice, Viral Hepatitis Vaccines genetics, Viral Nonstructural Proteins genetics, Hepacivirus genetics, Mutation, Viral Proteins genetics
- Abstract
Objective: HCV is a major cause of chronic liver disease worldwide, but the role of neutralising antibodies (nAbs) in its natural history remains poorly defined. We analysed the in vivo role of hypervariable region 1 (HVR1) for HCV virion properties, including nAb susceptibility., Design: Analysis of HCV from human liver chimeric mice infected with cell-culture-derived prototype genotype 2a recombinant J6/JFH1 or HVR1-deleted variant J6/JFH1
ΔHVR1 identified adaptive mutations, which were analysed by reverse genetics in Huh7.5 and CD81-deficient S29 cells. The increased in vivo genomic stability of the adapted viruses facilitated ex vivo density analysis by ultracentrifugation and in vivo neutralisation experiments addressing the role of HVR1., Results: In vivo, J6/JFH1 and J6/JFH1ΔHVR1 depended on single substitutions within amino acids 867-876 in non-structural protein, NS2. The identified A876P-substitution resulted in a 4.7-fold increase in genomic stability. In vitro, NS2 substitutions enhanced infectivity 5-10-fold by increasing virus assembly. Mouse-derived mJ6/JFH1A876P and mJ6/JFH1ΔHVR1/A876P viruses displayed similar heterogeneous densities of 1.02-1.1 g/mL. Human liver chimeric mice loaded with heterologous patient H (genotype 1a) immunoglobulin had partial protection against mJ6/JFH1A876P and complete protection against mJ6/JFH1ΔHVR1/A876P . Interestingly, we identified a putative escape mutation, D476G, in mJ6/JFH1A876P . This mutation in hypervariable region 2 conferred 6.6-fold resistance against H06 IgG in vitro., Conclusions: The A876P-substitution bridges in vitro and in vivo studies using J6/JFH1-based recombinants. We provide the first in vivo evidence that HVR1 protects cross-genotype conserved HCV neutralisation epitopes, which advocates the possibility of using HVR1-deleted viruses as vaccine antigens to boost broadly reactive protective nAb responses., Competing Interests: Conflicts of Interest: None declared., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)- Published
- 2016
- Full Text
- View/download PDF
35. Monoclonal anti-envelope antibody AP33 protects humanized mice against a patient-derived hepatitis C virus challenge.
- Author
-
Desombere I, Fafi-Kremer S, Van Houtte F, Pessaux P, Farhoudi A, Heydmann L, Verhoye L, Cole S, McKeating JA, Leroux-Roels G, Baumert TF, Patel AH, and Meuleman P
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Epitopes, Hepatitis C Antibodies metabolism, Humans, Mice, Mice, Transgenic, Statistics, Nonparametric, Antibodies, Monoclonal pharmacology, Hepacivirus immunology, Hepatitis C drug therapy, Hepatitis C immunology, Hepatitis C Antibodies immunology, Viral Envelope Proteins immunology
- Abstract
Unlabelled: End-stage liver disease (ESLD) caused by hepatitis C virus (HCV) infection is a major indication for liver transplantation. However, immediately after transplantation, the liver graft of viremic patients universally becomes infected by circulating virus, resulting in accelerated liver disease progression. Currently available direct-acting antiviral therapies have reduced efficacy in patients with ESLD and prophylactic strategies to prevent HCV recurrence are still highly needed. In this study, we compared the ability of two broadly reactive monoclonal antibodies (mAbs), designated 3/11 and AP33, recognizing a distinct, but overlapping, epitope in the viral E2 glycoprotein to protect humanized mice from a patient-derived HCV challenge. Their neutralizing activity was assessed using the HCV pseudoparticles and cell-culture-derived HCV systems expressing multiple patient-derived envelopes and a human-liver chimeric mouse model. HCV RNA was readily detected in all control mice challenged with a patient-derived HCV genotype 1b isolate, whereas 3 of 4 AP33-treated mice were completely protected. In contrast, only one of four 3/11-treated mice remained HCV-RNA negative throughout the observation period, whereas the other 3 had a viral load that was indistinguishable from that in the control group. The increased in vivo efficacy of AP33 was in line with its higher affinity and neutralizing capacity observed in vitro., Conclusions: Although mAbs AP33 and 3/11 target the same region in E2, only mAb AP33 can efficiently protect from challenge with a heterologous HCV population in vivo. Given that mAb AP33 efficiently neutralizes viral variants that escaped the humoral immune response and reinfected the liver graft of transplant patients, it may be a valuable candidate to prevent HCV recurrence. In addition, our data are valuable for the design of a prophylactic vaccine., (© 2015 by the American Association for the Study of Liver Diseases.)
- Published
- 2016
- Full Text
- View/download PDF
36. Anti-CD81 but not anti-SR-BI blocks Plasmodium falciparum liver infection in a humanized mouse model.
- Author
-
Foquet L, Hermsen CC, Verhoye L, van Gemert GJ, Cortese R, Nicosia A, Sauerwein RW, Leroux-Roels G, and Meuleman P
- Subjects
- Animals, Anopheles parasitology, CD36 Antigens antagonists & inhibitors, Disease Models, Animal, Humans, Mice, SCID, Plasmodium falciparum growth & development, Liver parasitology, Malaria, Falciparum prevention & control, Plasmodium falciparum physiology, Tetraspanin 28 antagonists & inhibitors
- Abstract
Objectives: Plasmodium falciparum sporozoites, deposited in the skin by infected Anopheles mosquitoes taking a blood meal, cross the endothelium of skin capillaries and travel to the liver where they traverse Kupffer cells and hepatocytes to finally invade a small number of the latter. In hepatocytes, sporozoites replicate, differentiate and give rise to large numbers of merozoites that are released into the bloodstream where they invade red blood cells, thus initiating the symptomatic blood stage. Using in vitro systems and rodent models, it has been shown that the hepatocyte receptors CD81 and scavenger receptor type B class I (SR-BI) play a pivotal role during sporozoite invasion. We wanted to evaluate whether these two entry factors are genuine drug targets for the prevention of P. falciparum infection in humans., Methods: Immunodeficient mice of which the liver is largely repopulated by human hepatocytes were treated with monoclonal antibodies blocking either CD81 or SR-BI 1 day prior to challenge with infected mosquitoes. P. falciparum infection of the liver was demonstrated using a qPCR assay., Results: In human liver chimeric mice, an antibody directed against CD81 completely blocked P. falciparum sporozoite invasion while SR-BI-specific monoclonal antibodies did not influence in vivo infection., Conclusions: These observations confirm the role of CD81 in liver-stage malaria and question that of SR-BI. CD81 might be a valuable drug target for the prevention of malaria., (© The Author 2015. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
37. Combined adenovirus vector and hepatitis C virus envelope protein prime-boost regimen elicits T cell and neutralizing antibody immune responses.
- Author
-
Chmielewska AM, Naddeo M, Capone S, Ammendola V, Hu K, Meredith L, Verhoye L, Rychlowska M, Rappuoli R, Ulmer JB, Colloca S, Nicosia A, Cortese R, Leroux-Roels G, Balfe P, Bienkowska-Szewczyk K, Meuleman P, McKeating JA, and Folgori A
- Subjects
- Adenoviridae genetics, Adjuvants, Immunologic administration & dosage, Animals, Female, Genetic Vectors, Guinea Pigs, Hepacivirus genetics, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Polysorbates administration & dosage, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Squalene administration & dosage, Vaccination methods, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Viral Envelope Proteins genetics, Viral Vaccines administration & dosage, Viral Vaccines genetics, Antibodies, Neutralizing blood, Hepacivirus immunology, Hepatitis C Antibodies blood, T-Lymphocytes immunology, Viral Envelope Proteins immunology, Viral Vaccines immunology
- Abstract
Unlabelled: Despite the recent progress in the development of new antiviral agents, hepatitis C virus (HCV) infection remains a major global health problem, and there is a need for a preventive vaccine. We previously reported that adenoviral vectors expressing HCV nonstructural proteins elicit protective T cell responses in chimpanzees and were immunogenic in healthy volunteers. Furthermore, recombinant HCV E1E2 protein formulated with adjuvant MF59 induced protective antibody responses in chimpanzees and was immunogenic in humans. To develop an HCV vaccine capable of inducing both T cell and antibody responses, we constructed adenoviral vectors expressing full-length and truncated E1E2 envelope glycoproteins from HCV genotype 1b. Heterologous prime-boost immunization regimens with adenovirus and recombinant E1E2 glycoprotein (genotype 1a) plus MF59 were evaluated in mice and guinea pigs. Adenovirus prime and protein boost induced broad HCV-specific CD8+ and CD4+ T cell responses and functional Th1-type IgG responses. Immune sera neutralized luciferase reporter pseudoparticles expressing HCV envelope glycoproteins (HCVpp) and a diverse panel of recombinant cell culture-derived HCV (HCVcc) strains and limited cell-to-cell HCV transmission. This study demonstrated that combining adenovirus vector with protein antigen can induce strong antibody and T cell responses that surpass immune responses achieved by either vaccine alone., Importance: HCV infection is a major health problem. Despite the availability of new directly acting antiviral agents for treating chronic infection, an affordable preventive vaccine provides the best long-term goal for controlling the global epidemic. This report describes a new anti-HCV vaccine targeting the envelope viral proteins based on adenovirus vector and protein in adjuvant. Rodents primed with the adenovirus vaccine and boosted with the adjuvanted protein developed cross-neutralizing antibodies and potent T cell responses that surpassed immune responses achieved with either vaccine component alone. If combined with the adenovirus vaccine targeting the HCV NS antigens now under clinical testing, this new vaccine might lead to a stronger and broader immune response and to a more effective vaccine to prevent HCV infection. Importantly, the described approach represents a valuable strategy for other infectious diseases in which both T and B cell responses are essential for protection.
- Published
- 2014
- Full Text
- View/download PDF
38. Novel human SR-BI antibodies prevent infection and dissemination of HCV in vitro and in humanized mice.
- Author
-
Lacek K, Vercauteren K, Grzyb K, Naddeo M, Verhoye L, Słowikowski MP, Fafi-Kremer S, Patel AH, Baumert TF, Folgori A, Leroux-Roels G, Cortese R, Meuleman P, and Nicosia A
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal genetics, Antibodies, Monoclonal immunology, CHO Cells, Cricetinae, Genotype, Hep G2 Cells, Hepacivirus genetics, Hepacivirus growth & development, Hepatitis C, Chronic immunology, Humans, Immunoglobulin G genetics, Immunoglobulin G immunology, Immunoglobulin G pharmacology, Immunoglobulin Variable Region genetics, Immunoglobulin Variable Region immunology, Immunoglobulin Variable Region pharmacology, Mice, Mice, SCID, Molecular Sequence Data, Peptide Library, Transplantation Chimera, Transplantation, Heterologous, Antibodies, Monoclonal pharmacology, Hepacivirus immunology, Hepatitis C, Chronic prevention & control, Hepatocytes immunology, Hepatocytes transplantation, Hepatocytes virology, Scavenger Receptors, Class B immunology
- Abstract
Background & Aims: Hepatitis C virus (HCV)-induced end-stage liver disease is currently the major indication for liver transplantation in the Western world. After transplantation, the donor liver almost inevitably becomes infected by the circulating virus and disease progression is accelerated in immune suppressed transplant patients. The current standard therapy, based on pegylated interferon and ribavirin, induces severe side effects and is often ineffective in this population. Therefore, new strategies to prevent graft re-infection are urgently needed. We have previously shown that monoclonal antibodies (mAbs) against the HCV co-receptor scavenger receptor class B type I (SR-BI/Cla1) inhibit infection by different HCV genotypes in cell culture., Methods: Using phage display libraries, we have generated a large set of novel human mAbs against SR-BI and evaluated their effectiveness in preventing HCV infection and direct cell-to-cell spread in vitro and in vivo using uPA-SCID mice with a humanized liver., Results: Eleven human monoclonal antibodies were generated that specifically recognize SR-BI. Two antibodies, mAb8 and mAb151, displayed the highest binding and inhibitory properties and also interfered with direct cell-to-cell spread in vitro. Studies in humanized mice showed that both antibodies were capable of preventing HCV infection and could block intrahepatic spread and virus amplification when administered 3 days after infection. Interestingly, anti-SR-BI therapy was effective against an HCV variant that escaped the control of the adaptive immune response in a liver transplant patient., Conclusions: The anti-SR-BI mAbs generated in this study may represent novel therapeutic tools to prevent HCV re-infection of liver allografts., (Copyright © 2012 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
39. A human monoclonal antibody targeting scavenger receptor class B type I precludes hepatitis C virus infection and viral spread in vitro and in vivo.
- Author
-
Meuleman P, Catanese MT, Verhoye L, Desombere I, Farhoudi A, Jones CT, Sheahan T, Grzyb K, Cortese R, Rice CM, Leroux-Roels G, and Nicosia A
- Subjects
- Animals, Cell Line, Tumor, Chimera, Genotype, Humans, Liver Transplantation, Mice, Mice, SCID, Secondary Prevention, Antibodies, Monoclonal therapeutic use, CD36 Antigens immunology, Hepatitis C prevention & control
- Abstract
Unlabelled: Endstage liver disease caused by chronic hepatitis C virus (HCV) infection is the leading indication for liver transplantation in the Western world. However, immediate reinfection of the grafted donor liver by circulating virus is inevitable and liver disease progresses much faster than the original disease. Standard antiviral therapy is not well tolerated and usually ineffective in liver transplant patients, whereas anti-HCV immunotherapy is hampered by the extreme genetic diversity of the virus and its ability to spread by way of cell-cell contacts. We generated a human monoclonal antibody against scavenger receptor class B type I (SR-BI), monoclonal antibody (mAb)16-71, which can efficiently prevent infection of Huh-7.5 hepatoma cells and primary hepatocytes by cell-culture-derived HCV (HCVcc). Using an Huh7.5 coculture system we demonstrated that mAb16-71 interferes with direct cell-to-cell transmission of HCV. Finally we evaluated the in vivo efficacy of mAb16-71 in "human liver urokinase-type plasminogen activator, severe combined immune deficiency (uPA-SCID) mice" (chimeric mice). A 2-week anti-SR-BI therapy that was initiated 1 day before viral inoculation completely protected all chimeric mice from infection with serum-derived HCV of different genotypes. Moreover, a 9-day postexposure therapy that was initiated 3 days after viral inoculation (when viremia was already observed in the animals) suppressed the rapid viral spread observed in untreated control animals. After cessation of anti-SR-BI-specific antibody therapy, a rise of the viral load was observed., Conclusion: Using in vitro cell culture and human liver-chimeric mouse models, we show that a human mAb targeting the HCV coreceptor SR-BI completely prevents infection and intrahepatic spread of multiple HCV genotypes. This strategy may be an efficacious way to prevent infection of allografts following liver transplantation in chronic HCV patients, and may even hold promise for the prevention of virus rebound during or following antiviral therapy., (Copyright © 2011 American Association for the Study of Liver Diseases.)
- Published
- 2012
- Full Text
- View/download PDF
40. Griffithsin has antiviral activity against hepatitis C virus.
- Author
-
Meuleman P, Albecka A, Belouzard S, Vercauteren K, Verhoye L, Wychowski C, Leroux-Roels G, Palmer KE, and Dubuisson J
- Subjects
- Animals, Antiviral Agents therapeutic use, Cell Line, Cell Line, Tumor, Hepatitis C drug therapy, Humans, Immunoprecipitation, Mice, Mice, SCID, Plant Lectins, Protein Binding, Viral Proteins metabolism, Algal Proteins pharmacology, Antiviral Agents pharmacology, Hepacivirus drug effects, Lectins pharmacology
- Abstract
Hepatitis C virus (HCV)-infected patients undergoing liver transplantation universally experience rapid reinfection of their new liver graft. Current treatment protocols do not prevent graft reinfection and, in addition, an accelerated disease progression is observed. In the present study, we have evaluated a novel strategy to prevent HCV infection using a lectin, griffithsin (GRFT) that specifically binds N-linked high-mannose oligosaccharides that are present on the viral envelope. The antiviral effect of GRFT was evaluated in vitro using the HCV pseudoparticle (HCVpp) and HCV cell culture (HCVcc) systems. We show here that preincubation of HCVpp and HCVcc with GRFT prevents infection of Huh-7 hepatoma cells. Furthermore, GRFT interferes with direct cell-to-cell transmission of HCV. GRFT acts at an early phase of the viral life cycle by interfering in a genotype-independent fashion with the interaction between the viral envelope proteins and the viral receptor CD81. The capacity of GRFT to prevent infection in vivo was evaluated using uPA(+/+)-SCID mice (uPA stands for urokinase-type plasminogen activator) that harbor human primary hepatocytes in their liver (chimeric mice). In this proof-of-concept trial, we demonstrated that GRFT can mitigate HCV infection of chimeric mice. Treated animals that did become infected demonstrated a considerable delay in the kinetics of the viral infection. Our data demonstrate that GRFT can prevent HCV infection in vitro and mitigate HCV infection in vivo. GRFT treatment of chronically infected HCV patients undergoing liver transplantation may be a suitable strategy to prevent infection of the liver allograft.
- Published
- 2011
- Full Text
- View/download PDF
41. In vivo evaluation of the cross-genotype neutralizing activity of polyclonal antibodies against hepatitis C virus.
- Author
-
Meuleman P, Bukh J, Verhoye L, Farhoudi A, Vanwolleghem T, Wang RY, Desombere I, Alter H, Purcell RH, and Leroux-Roels G
- Subjects
- Amino Acid Sequence, Animals, Hepacivirus immunology, Hepatitis C, Chronic immunology, Humans, Mice, Mice, SCID, Transplantation Chimera immunology, Viral Envelope Proteins genetics, Viral Vaccines immunology, Antibodies, Neutralizing immunology, Hepatitis C Antibodies blood
- Abstract
Unlabelled: Control of hepatitis C virus (HCV) infection remains a huge challenge of global medical importance. Using a variety of in vitro approaches, neutralizing antibodies (nAbs) have been identified in patients with acute and chronic hepatitis C. The exact role these nAbs play in the resolution of acute HCV infection still remains elusive. We have previously shown that purified polyclonal antibodies isolated from plasma obtained in 2003 from a chronic HCV patient (Patient H) can protect human liver chimeric mice from a subsequent challenge with the autologous HCV strain isolated from Patient H in 1977 (H77). In this study we investigated whether polyclonal antibodies isolated from Patient H in 2006 (H06), which display high cross-genotype neutralizing activity in both the HCV pseudoparticle (HCVpp) and HCV cell culture (HCVcc) systems, were also able to prevent HCV infection of different genotypes (gt) in vivo. Following passive immunization with H06-antibodies, chimeric mice were challenged with the consensus strains H77C (gt1a), ED43 (gt4a), or HK6a (gt6a). In accordance with previous results, H06-antibodies prevented infection of chimeric mice with the autologous virus. However, the outcome of a homologous challenge is highly influenced by the amount of challenge virus injected. Depending on the viral genotype used, H06-antibodies were able to protect up to 50% of chimeric mice from a heterologous challenge. Animals in which the antibody pretreatment failed displayed a clear delay in the kinetics of viral infection. Sequence analysis of the recovered viruses did not suggest antibody-induced viral escape., Conclusion: Polyclonal anti-HCV antibodies isolated from a chronic HCV patient can protect against an in vivo challenge with different HCV genotypes. However, the in vivo protective efficacy of cross-genotype neutralizing antibodies was less than predicted by cell culture experiments., (Copyright © 2011 American Association for the Study of Liver Diseases.)
- Published
- 2011
- Full Text
- View/download PDF
42. Detection of mumps virus-specific memory B cells by transfer of peripheral blood mononuclear cells into immune-deficient mice.
- Author
-
Vandermeulen C, Verhoye L, Vaidya S, Clement F, Brown KE, Hoppenbrouwers K, and Leroux-Roels G
- Subjects
- Adolescent, Adult, Animals, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Viral analysis, Antibodies, Viral blood, Enzyme-Linked Immunosorbent Assay, Female, Humans, Measles-Mumps-Rubella Vaccine administration & dosage, Measles-Mumps-Rubella Vaccine immunology, Mice, Mice, Inbred NOD, Mice, SCID, Mumps immunology, Mumps prevention & control, Neutralization Tests, Spleen cytology, Spleen immunology, Vaccination, Young Adult, B-Lymphocytes immunology, Immunologic Memory immunology, Leukocytes, Mononuclear transplantation, Mumps virus immunology
- Abstract
Waning immunity to mumps after one or two doses of the measles, mumps and rubella (MMR) vaccine has been described. Using a human peripheral blood lymphocyte (PBL)-severe combined immunodeficiency (SCID) mouse model, MMR vaccine recipients with undetectable and high antibody titres against mumps were compared for the presence of circulating mumps-specific memory B cells. Peripheral blood mononuclear cells (PBMC) from six donors (three subjects with undetectable and three with high antibody titres against mumps) were injected into the spleens of non-obese diabetic (NOD)-SCID mice (three mice per subject). Mice were pretreated with TMbeta1 and total body irradiation to improve engraftment. In vivo production of human antibodies against mumps was evaluated in mouse plasma on days 7, 10 and 13 with a commercial enzyme-linked immunosorbent assay (ELISA), functional reduction neutralization test. Three donors had mumps antibody titres below the detection limit (titre < 230) and three had high antibody titres (range 5700-7300). None of the mice injected with PBMC from subjects with undetectable antibody titres showed detectable human antibody titres, despite the presence of cell-mediated immunity in two of the three donors. Seven out of nine mice injected with PBMC from subjects with high antibody titres acquired detectable antibody titres for mumps in their plasma. PBMC from vaccinees without detectable serum antibodies against mumps virus were unable to induce secretion of anti-mumps antibodies in the blood of recipient mice, whereas PBMC from vaccinees with high antibody titres were able to do so. This observation suggests that the frequency of mumps-specific memory B cells is very low in vaccinees with undetectable antibody titres. These individuals may therefore be at risk of developing mumps disease upon encounter with wild-type virus.
- Published
- 2010
- Full Text
- View/download PDF
43. Inhibition of replication of primary HIV-1 isolates in huPBL-NOD/Scid mice by antibodies from HIV-1 infected patients.
- Author
-
Steyaert S, Heyndrickx L, Verhoye L, Vermoesen T, Donners H, Fransen K, Van Wanzeele F, Vandergucht B, Vanham G, Leroux-Roels G, and Vanlandschoot P
- Subjects
- Adult, Aged, Animals, Antibodies isolation & purification, Antibodies therapeutic use, CD4 Lymphocyte Count, Disease Models, Animal, Female, HIV Infections prevention & control, HIV Infections therapy, HIV-1 isolation & purification, Humans, Immunization, Passive methods, Immunoglobulin G immunology, Immunoglobulin G isolation & purification, Immunoglobulin G therapeutic use, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear transplantation, Male, Mice, Mice, Inbred NOD, Mice, SCID, Middle Aged, Neutralization Tests, Viral Load, Antibodies immunology, HIV Infections immunology, HIV-1 immunology, Virus Replication immunology
- Abstract
Although a limited number of HIV-infected patients have broadly neutralizing antibodies, it has not been examined whether these antibodies can protect against infection with primary virus in vivo. Here we screened the plasma of 23 HIV-1-infected patients for broadly neutralizing antibodies. Purified antibodies from subjects with broad and more narrow responses were administered to huPBL-NOD/Scid mice that were subsequently challenged with primary viruses of clade A, B and CRF01_AE. Although we observed a lack of correlation between the data from the in vitro neutralization assay and the results from the passive immunization experiments, we report for the first time that antibodies from HIV-infected persons can inhibit replication of primary virus isolates in an animal model.
- Published
- 2007
- Full Text
- View/download PDF
44. The intraspleen huPBL NOD/SCID model to study the human HIV-specific antibody response selected in the course of natural infection.
- Author
-
Steyaert S, Verhoye L, Beirnaert E, Donners H, Fransen K, Heyndrickx L, Vanham G, Leroux-Roels G, and Vanlandschoot P
- Subjects
- Animals, B-Lymphocytes physiology, B-Lymphocytes virology, HIV Antibodies biosynthesis, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Spleen immunology, Viral Load, Antibody Specificity, B-Lymphocytes immunology, Drug Administration Routes, HIV Antibodies immunology, HIV Infections immunology, Models, Animal
- Abstract
The intrasplenic injection of human peripheral blood mononuclear cells (PBMCs) into severely immune deficient NOD/SCID mice, causes a massive and transient dominant expansion of human B cells in the spleen. This permits the easy isolation of human monoclonal antibodies specific for different antigens by a Kohler and Milstein-based method. Here we studied the human HIV-specific antibody response in the circulation of mice after intrasplenic transfer of PBMC from untreated HIV-infected patients with detectable to high viral load as well as from HAART-treated and from untreated patients, who kept an undetectable viral load (the latter referred to as "natural suppressors"). Excellent B cell expansion was obtained for all PBMC. High level replication of virus was observed after transfer of PBMC of untreated viremic patients only. A strong and multispecific HIV-specific antibody response was observed after transfer of PBMC of untreated viremic patients and natural suppressors. In contrast, only a weak and pauci-specific antibody response was detected in mice reconstituted with PBMC from successfully treated patients. Based on these observations we conclude that the use of the intraspleen mouse model confirmed a) the presence of HIV-specific circulating memory B cells in untreated patients and natural suppressors; b) the nearly complete absence of circulating memory B cells in patients receiving highly active antiretroviral therapy. Using the intraspleen model we generated large numbers of immortalized B cells and isolated two anti-p24 human monoclonal antibodies. We further conclude that the intraspleen huPBL NOD/SCID model is a small animal model useful for the analysis of the antibody response against HIV found in patients.
- Published
- 2007
- Full Text
- View/download PDF
45. Human B cell growth and differentiation in the spleen of immunodeficient mice.
- Author
-
Depraetere S, Verhoye L, Leclercq G, and Leroux-Roels G
- Subjects
- Animals, Antibodies, Monoclonal biosynthesis, B-Lymphocytes cytology, B-Lymphocytes metabolism, Cell Differentiation genetics, Cell Differentiation immunology, Cell Division genetics, Cell Division immunology, Cell Survival genetics, Cell Survival immunology, Epitopes, B-Lymphocyte immunology, Hepatitis B Antibodies biosynthesis, Hepatitis B Surface Antigens immunology, Hepatitis C Antibodies biosynthesis, Hepatitis C Antigens immunology, Humans, Hybridomas, Immunoglobulins biosynthesis, Immunophenotyping, Lymphocyte Transfusion, Mice, Mice, Inbred NOD, Mice, SCID, Plasma Cells cytology, Plasma Cells immunology, Plasma Cells metabolism, Plasma Cells pathology, Postoperative Period, Severe Combined Immunodeficiency genetics, Species Specificity, Spleen metabolism, Spleen pathology, Transplantation Conditioning methods, B-Lymphocytes immunology, B-Lymphocytes transplantation, Severe Combined Immunodeficiency immunology, Severe Combined Immunodeficiency pathology, Spleen immunology
- Abstract
Human mAbs (HumAbs) have therapeutic potential against infectious diseases and cancer. Heretofore, their production has been hampered by ethical constraints preventing the isolation of Ag-specific activated B cells by in vivo immunization. Alternatively, severe combined immune deficient (SCID) mice, transplanted i.p. with human (Hu)-PBLs, allow the in vivo stimulation of human Ab responses without the usual constraints. Unfortunately, human B cells only represent a minor fraction of the surviving graft, they are scattered all over the animal body, and thus are hard to isolate for subsequent immortalization procedures. To prevent this dispersion and to provide the human B cells with a niche for expansion and maturation, SCID mice were engrafted with Hu-PBL directly into the spleen. Simultaneously endogenous murine NK cell activity was depleted by treatment with an anti-mouse IL-2 receptor beta-chain Ab. During engraftment, human B lymphocytes became activated, divided intensely, and differentiated into plasmacytoid cells. In vivo exposure to a recall Ag after cell transfer induced expansion of Ag-specific B cell clones. One week after inoculation, human B cells were abundant in the spleen and could easily be recovered for fusion with a heteromyeloma line. This resulted in the formation of stable hybridoma cell lines that secreted Ag-specific HumAbs. Thus transplantation of human lymphoid cells in the spleens of immune deficient mice represents a model for the study of human T cell-dependent B cell activation and proves to be an excellent tool for the successful production of HumAbs.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.