Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ype P. de Jong is active.

Publication


Featured researches published by Ype P. de Jong.


Nature | 2009

Human occludin is a hepatitis C virus entry factor required for infection of mouse cells

Alexander Ploss; Matthew J. Evans; V. Gaysinskaya; Maryline Panis; Hana You; Ype P. de Jong; Charles M. Rice

Hepatitis C virus (HCV) is a leading cause of liver disease worldwide. The development of much needed specific antiviral therapies and an effective vaccine has been hampered by the lack of a convenient small animal model. The determinants restricting HCV tropism to human and chimpanzee hosts are unknown. Replication of the viral RNA has been demonstrated in mouse cells, but these cells are not infectable with either lentiviral particles bearing HCV glycoproteins (HCVpp) or HCV produced in cell culture (HCVcc) (A.P., M.E. and C.M.R., unpublished observations), suggesting that there is a block at the level of entry. Here we show, using an iterative complementary DNA library screening approach, that human occludin (OCLN) is an essential HCV cell entry factor that is able to render murine cells infectable with HCVpp. Similarly, OCLN is required for the HCV-susceptibility of human cells, because its overexpression in uninfectable cells specifically enhanced HCVpp uptake, whereas its silencing in permissive cells impaired both HCVpp and HCVcc infection. In addition to OCLN, HCVpp infection of murine cells required expression of the previously identified HCV entry factors CD81 (ref. 4), scavenger receptor class B type I (SR-BI, also known as SCARB1) and claudin-1 (CLDN1). Although the mouse versions of SR-BI and CLDN1 function at least as well as the human proteins in promoting HCV entry, both OCLN and CD81 must be of human origin to allow efficient infection. The species-specific determinants of OCLN were mapped to its second extracellular loop. The identification of OCLN as a new HCV entry factor further highlights the importance of the tight junction complex in the viral entry process, and provides an important advance towards efforts to develop small animal models for HCV.


Cell Host & Microbe | 2009

Humanized mice for modeling human infectious disease: challenges, progress, and outlook.

Nicolas Legrand; Alexander Ploss; Rudi Balling; Pablo D. Becker; Chiara Borsotti; Nicolas Brezillon; Jennifer Debarry; Ype P. de Jong; Hongkui Deng; James P. Di Santo; Stephanie C. Eisenbarth; Elizabeth E. Eynon; Richard A. Flavell; Carlos A. Guzmán; Nicholas D. Huntington; Dina Kremsdorf; Michael P. Manns; Markus G. Manz; Jean-Jacques Mention; Michael Ott; Chozhavendan Rathinam; Charles M. Rice; Anthony Rongvaux; Sean Stevens; Hergen Spits; Helene Strick-Marchand; Hitoshi Takizawa; Anja U. van Lent; Chengyan Wang; Kees Weijer

Over 800 million people worldwide are infected with hepatitis viruses, human immunodeficiency virus (HIV), and malaria, resulting in more than 5 million deaths annually. Here we discuss the potential and challenges of humanized mouse models for developing effective and affordable therapies and vaccines, which are desperately needed to combat these diseases.


Cell | 2015

Hepatitis C virus RNA functionally sequesters miR-122

Joseph M. Luna; Troels K. H. Scheel; Tal Danino; Katharina S. Shaw; Aldo Mele; John J. Fak; Eiko Nishiuchi; Constantin N. Takacs; Maria Teresa Catanese; Ype P. de Jong; Ira M. Jacobson; Charles M. Rice; Robert B. Darnell

Hepatitis C virus (HCV) uniquely requires the liver-specific microRNA-122 for replication, yet global effects on endogenous miRNA targets during infection are unexplored. Here, high-throughput sequencing and crosslinking immunoprecipitation (HITS-CLIP) experiments of human Argonaute (AGO) during HCV infection showed robust AGO binding on the HCV 5UTR at known and predicted miR-122 sites. On the human transcriptome, we observed reduced AGO binding and functional mRNA de-repression of miR-122 targets during virus infection. This miR-122 sponge effect was relieved and redirected to miR-15 targets by swapping the miRNA tropism of the virus. Single-cell expression data from reporters containing miR-122 sites showed significant de-repression during HCV infection depending on expression level and site number. We describe a quantitative mathematical model of HCV-induced miR-122 sequestration and propose that such miR-122 inhibition by HCV RNA may result in global de-repression of host miR-122 targets, providing an environment fertile for the long-term oncogenic potential of HCV.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Modeling host interactions with hepatitis B virus using primary and induced pluripotent stem cell-derived hepatocellular systems

Amir Shlomai; Robert E. Schwartz; Vyas Ramanan; Ankit Bhatta; Ype P. de Jong; Sangeeta N. Bhatia; Charles M. Rice

Significance Major obstacles for using human hepatocytes to study hepatitis B virus (HBV) pathobiology are rapid loss of hepatocyte function after plating and the variability between hepatocyte donors. We show that micropatterning and coculturing of primary human hepatocytes with fibroblasts (MPCC format) maintains prolonged infection that is restricted by the innate immune response, and can be further boosted by suppression of this response. To address the problem of donor variability, we show that induced pluripotent stem cells (iPSC) differentiated into hepatocyte-like cells support HBV infection in a differentiation-dependent manner. Our study opens an avenue for using these systems to study virus–host interactions and test antiviral drugs, and suggests HBV permissiveness as a surrogate reporter to assess the degree of differentiation of candidate iPSC-derived hepatocyte-like cells. Hepatitis B virus (HBV) chronically infects 400 million people worldwide and is a leading driver of end-stage liver disease and liver cancer. Research into the biology and treatment of HBV requires an in vitro cell-culture system that supports the infection of human hepatocytes, and accurately recapitulates virus–host interactions. Here, we report that micropatterned cocultures of primary human hepatocytes with stromal cells (MPCCs) reliably support productive HBV infection, and infection can be enhanced by blocking elements of the hepatocyte innate immune response associated with the induction of IFN-stimulated genes. MPCCs maintain prolonged, productive infection and represent a facile platform for studying virus–host interactions and for developing antiviral interventions. Hepatocytes obtained from different human donors vary dramatically in their permissiveness to HBV infection, suggesting that factors—such as divergence in genetic susceptibility to infection—may influence infection in vitro. To establish a complementary, renewable system on an isogenic background in which candidate genetics can be interrogated, we show that inducible pluripotent stem cells differentiated into hepatocyte-like cells (iHeps) support HBV infection that can also be enhanced by blocking interferon-stimulated gene induction. Notably, the emergence of the capacity to support HBV transcriptional activity and initial permissiveness for infection are marked by distinct stages of iHep differentiation, suggesting that infection of iHeps can be used both to study HBV, and conversely to assess the degree of iHep differentiation. Our work demonstrates the utility of these infectious systems for studying HBV biology and the virus’ interactions with host hepatocyte genetics and physiology.


Science Translational Medicine | 2014

Broadly neutralizing antibodies abrogate established hepatitis C virus infection

Ype P. de Jong; Marcus Dorner; Michiel C. Mommersteeg; Jing W. Xiao; Alejandro B. Balazs; Justin B. Robbins; Benjamin Y. Winer; Sherif Gerges; Kevin Vega; Rachael N. Labitt; Bridget M. Donovan; Erick Giang; Anuradha Krishnan; Luis Chiriboga; Michael R. Charlton; Dennis R. Burton; David Baltimore; Mansun Law; Charles M. Rice; Alexander Ploss

HCV-specific neutralizing antibodies protect humanized mice from challenge and suppress established infections. Neutralizing Antibodies Take Down the HCV Establishment In most individuals infected with hepatitis C virus (HCV), the HCV sets up shop—establishing a long-term, chronic infection that damages the liver and can lead to cirrhosis or liver cancer. de Jong et al. now report that a trio of neutralizing antibodies not only can prevent infection but also can treat and maybe even cure already established infection in multiple animal models. The broadly neutralizing antibodies, which could block multiple genotypes of HCV, were delivered into the muscle by a virus—an adeno-associated vector that does not cause disease—resulting in prolonged expression of the antibodies. If these data hold true in people, this approach may provide a new tool for treating HCV infection. In most exposed individuals, hepatitis C virus (HCV) establishes a chronic infection; this long-term infection in turn contributes to the development of liver diseases such as cirrhosis and hepatocellular carcinoma. The role of antibodies directed against HCV in disease progression is poorly understood. Neutralizing antibodies (nAbs) can prevent HCV infection in vitro and in animal models. However, the effects of nAbs on an established HCV infection are unclear. We demonstrate that three broadly nAbs—AR3A, AR3B, and AR4A—delivered with adeno-associated viral vectors can confer protection against viral challenge in humanized mice. Furthermore, we provide evidence that nAbs can abrogate an ongoing HCV infection in primary hepatocyte cultures and in a human liver chimeric mouse model. These results showcase a therapeutic approach to interfere with HCV infection by exploiting a previously unappreciated need for HCV to continuously infect new hepatocytes to sustain a chronic infection.


Seminars in Cancer Biology | 2014

Virus associated malignancies: the role of viral hepatitis in hepatocellular carcinoma.

Amir Shlomai; Ype P. de Jong; Charles M. Rice

Hepatocellular carcinoma (HCC) is the third leading fatal cancer worldwide and its incidence continues to increase. Chronic viral hepatitis involving either hepatitis B virus (HBV) or hepatitis C virus (HCV) infection is the leading etiology for HCC, making HCC prevention a major goal of antiviral therapy. While recent clinical observations and translational research have enhanced our understanding of the molecular mechanisms driving the initiation and progression of HCC, much remains unknown. Current data indicates that HCC tumors are highly complex and heterogeneous resulting from the aberrant function of multiple molecular pathways. This complex biology is responsible, at least in part, for the absence of highly efficient target-directed therapies for this deadly cancer. Additionally, the direct or indirect effect of HBV and HCV infection on the development of HCC is still a contentious issue. Thus, the question remains whether viral hepatitis-associated HCC stems from virus-specific factors, and/or from a general mechanism involving inflammation and tissue regeneration. In this review we summarize general mechanisms implicated in HCC, emphasizing data generated by new technologies available today. We also highlight specific pathways by which HBV and HCV could be involved in HCC pathogenesis. However, improvements to current in vitro and in vivo systems for both viruses will be needed to rigorously define the temporal sequence and specific pathway dysregulations that drive the strong clinical link between chronic hepatitis virus infection and HCC.


Current Topics in Microbiology and Immunology | 2013

Animal Models for Hepatitis C

Eva Billerbeck; Ype P. de Jong; Marcus Dorner; Cynthia de la Fuente; Alexander Ploss

Hepatitis C remains a global epidemic. Approximately 3xa0% of the worlds population suffers from chronic hepatitis C, which is caused by hepatitis C virus (HCV)-a positive sense, single-stranded RNA virus of the Flaviviridae family. HCV has a high propensity for establishing a chronic infection. If untreated chronic HCV carriers can develop severe liver disease including fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Antiviral treatment is only partially effective, costly, and poorly tolerated. A prophylactic or therapeutic vaccine for HCV does not exist. Mechanistic studies of virus-host interactions, HCV immunity, and pathogenesis as well as the development of more effective therapies have been hampered by the lack of a suitable small animal model. Besides humans, chimpanzees are the only species that is naturally susceptible to HCV infection. While experimentation in these large primates has yielded valuable insights, ethical considerations, limited availability, genetic heterogeneity, and cost limit their utility. In search for more tractable small animal models, numerous experimental approaches have been taken to recapitulate parts of the viral life cycle and/or aspects of viral pathogenesis that will be discussed in this review. Exciting new models and improvements in established models hold promise to further elucidate our understanding of chronic HCV infection.


Journal of Clinical Investigation | 2010

New horizons for studying human hepatotropic infections

Ype P. de Jong; Charles M. Rice; Alexander Ploss

The liver serves as a target organ for several important pathogens, including hepatitis B and C viruses (HBV and HCV, respectively) and the human malaria parasites, all of which represent serious global health problems. Because these pathogens are restricted to human hepatocytes, research in small animals has been compromised by the frailty of the current mouse xenotransplantation models. In this issue of the JCI, Bissig et al. demonstrate robust HBV and HCV infection in a novel xenotransplantation model in which large numbers of immunodeficient mice with liver injury were engrafted with significant quantities of human hepatocytes. This technical advance paves the way for more widespread use of human liver chimeric mice and forms the basis for creating increasingly complex humanized mouse models that could prove useful for studying immunopathogenesis and vaccine development against hepatotropic pathogens.


Journal of Hepatology | 2014

HCV animal models and liver disease.

Koen Vercauteren; Ype P. de Jong; Philip Meuleman

The development and evaluation of effective therapies and vaccines for the hepatitis C virus (HCV) and the study of its interactions with the mammalian host have been hindered for a long time by the absence of suitable small animal models. Due to the narrow host tropism of HCV, the development of mice that can be robustly engrafted with human hepatocytes was a major breakthrough since they recapitulate the complete HCV life cycle. This model has been useful to investigate many aspects of the HCV life cycle, including antiviral interventions. However, studies of cellular immunity, immunopathogenesis and resulting liver diseases have been hampered by the lack of a small animal model with a functional immune system. In this review, we summarize the evolution of in vivo models for the study of HCV.


Molecular Therapy | 2016

Superior In vivo Transduction of Human Hepatocytes Using Engineered AAV3 Capsid

Koen Vercauteren; Brad E. Hoffman; Irene Zolotukhin; Geoffrey D. Keeler; Jing W. Xiao; Etiena Basner-Tschakarjan; Katherine A. High; Hildegund C. J. Ertl; Charles M. Rice; Arun Srivastava; Ype P. de Jong; Roland W. Herzog

Adeno-associated viral (AAV) vectors are currently being tested in multiple clinical trials for liver-directed gene transfer to treat the bleeding disorders hemophilia A and B and metabolic disorders. The optimal viral capsid for transduction of human hepatocytes has been under active investigation, but results across various models are inconsistent. We tested in vivo transduction in humanized mice. Methods to quantitate percent AAV transduced human and murine hepatocytes in chimeric livers were optimized using flow cytometry and confocal microscopy with image analysis. Distinct transduction efficiencies were noted following peripheral vein administration of a self-complementary vector expressing a gfp reporter gene. An engineered AAV3 capsid with two amino acid changes, S663V+T492V (AAV3-ST), showed best efficiency for human hepatocytes (~3-times, ~8-times, and ~80-times higher than for AAV9, AAV8, and AAV5, respectively). AAV5, 8, and 9 were more efficient in transducing murine than human hepatocytes. AAV8 yielded the highest transduction rate of murine hepatocytes, which was 19-times higher than that for human hepatocytes. In summary, our data show substantial differences among AAV serotypes in transduction of human and mouse hepatocytes, are the first to report on AAV5 in humanized mice, and support the use of AAV3-based vectors for human liver gene transfer.

Collaboration


Dive into the Ype P. de Jong's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amir Shlomai

Tel Aviv Sourasky Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ira M. Jacobson

Beth Israel Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge