Network


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

Hotspot


Dive into the research topics where Marcus Dorner is active.

Publication


Featured researches published by Marcus Dorner.


Nature | 2012

HIV therapy by a combination of broadly neutralizing antibodies in humanized mice

Florian Klein; Ariel Halper-Stromberg; Joshua A. Horwitz; Henning Gruell; Johannes F. Scheid; Stylianos Bournazos; Hugo Mouquet; Linda Spatz; Ron Diskin; Alexander Abadir; Trinity Zang; Marcus Dorner; Eva Billerbeck; Rachael N. Labitt; Christian Gaebler; Paola M. Marcovecchio; Reha-Baris Incesu; Thomas R. Eisenreich; Paul D. Bieniasz; Michael S. Seaman; Pamela J. Bjorkman; Jeffrey V. Ravetch; Alexander Ploss; Michel C. Nussenzweig

Human antibodies to human immunodeficiency virus-1 (HIV-1) can neutralize a broad range of viral isolates in vitro and protect non-human primates against infection. Previous work showed that antibodies exert selective pressure on the virus but escape variants emerge within a short period of time. However, these experiments were performed before the recent discovery of more potent anti-HIV-1 antibodies and their improvement by structure-based design. Here we re-examine passive antibody transfer as a therapeutic modality in HIV-1-infected humanized mice. Although HIV-1 can escape from antibody monotherapy, combinations of broadly neutralizing antibodies can effectively control HIV-1 infection and suppress viral load to levels below detection. Moreover, in contrast to antiretroviral therapy, the longer half-life of antibodies led to control of viraemia for an average of 60 days after cessation of therapy. Thus, combinations of potent monoclonal antibodies can effectively control HIV-1 replication in humanized mice, and should be re-examined as a therapeutic modality in HIV-1-infected individuals.


Nature | 2011

A genetically humanized mouse model for hepatitis C virus infection

Marcus Dorner; Joshua A. Horwitz; Justin B. Robbins; Walter T. Barry; Qian Feng; Kathy Mu; Christopher T. Jones; John W. Schoggins; Maria Teresa Catanese; Dennis R. Burton; Mansun Law; Charles M. Rice; Alexander Ploss

Hepatitis C virus (HCV) remains a major medical problem. Antiviral treatment is only partially effective and a vaccine does not exist. Development of more effective therapies has been hampered by the lack of a suitable small animal model. Although xenotransplantation of immunodeficient mice with human hepatocytes has shown promise, these models are subject to important challenges. Building on the previous observation that CD81 and occludin comprise the minimal human factors required to render mouse cells permissive to HCV entry in vitro, we attempted murine humanization via a genetic approach. Here we show that expression of two human genes is sufficient to allow HCV infection of fully immunocompetent inbred mice. We establish a precedent for applying mouse genetics to dissect viral entry and validate the role of scavenger receptor type B class I for HCV uptake. We demonstrate that HCV can be blocked by passive immunization, as well as showing that a recombinant vaccinia virus vector induces humoral immunity and confers partial protection against heterologous challenge. This system recapitulates a portion of the HCV life cycle in an immunocompetent rodent for the first time, opening opportunities for studying viral pathogenesis and immunity and comprising an effective platform for testing HCV entry inhibitors in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Human broadly neutralizing antibodies to the envelope glycoprotein complex of hepatitis C virus

Erick Giang; Marcus Dorner; Jannick Prentoe; Marlène Dreux; Matthew J. Evans; Jens Bukh; Charles M. Rice; Alexander Ploss; Dennis R. Burton; Mansun Law

Hepatitis C virus (HCV) infects ∼2% of the worlds population. It is estimated that there are more than 500,000 new infections annually in Egypt, the country with the highest HCV prevalence. An effective vaccine would help control this expanding global health burden. HCV is highly variable, and an effective vaccine should target conserved T- and B-cell epitopes of the virus. Conserved B-cell epitopes overlapping the CD81 receptor-binding site (CD81bs) on the E2 viral envelope glycoprotein have been reported previously and provide promising vaccine targets. In this study, we isolated 73 human mAbs recognizing five distinct antigenic regions on the virus envelope glycoprotein complex E1E2 from an HCV-immune phage-display antibody library by using an exhaustive-panning strategy. Many of these mAbs were broadly neutralizing. In particular, the mAb AR4A, recognizing a discontinuous epitope outside the CD81bs on the E1E2 complex, has an exceptionally broad neutralizing activity toward diverse HCV genotypes and protects against heterologous HCV challenge in a small animal model. The mAb panel will be useful for the design and development of vaccine candidates to elicit broadly neutralizing antibodies to HCV.


Proceedings of the National Academy of Sciences of the United States of America | 2013

HIV-1 suppression and durable control by combining single broadly neutralizing antibodies and antiretroviral drugs in humanized mice

Joshua A. Horwitz; Ariel Halper-Stromberg; Hugo Mouquet; Alexander D. Gitlin; Anna Tretiakova; Thomas R. Eisenreich; Marine Malbec; Sophia Gravemann; Eva Billerbeck; Marcus Dorner; Hildegard Büning; Olivier Schwartz; Elena Knops; Rolf Kaiser; Michael S. Seaman; James M. Wilson; Charles M. Rice; Alexander Ploss; Pamela J. Bjorkman; Florian Klein; Michel C. Nussenzweig

Significance Treatment of HIV-1 infection in humans is achieved using combinations of highly effective antiretroviral therapy (ART) drugs to potently suppress viral replication and prevent the emergence of drug-resistant viruses. However, ART drugs must be taken indefinitely owing to rapid return of viremia upon termination of treatment. Highly potent broadly neutralizing antibodies (bNAbs) present a new potential therapeutic modality in the treatment of HIV-1 infection. Because of their comparatively longer half-lives relative to ART drugs and their ability to eliminate infected cells, bNAbs may alleviate some aspects of the lifelong treatment adherence burden of ART. Here we show that lowering the initial viral load with ART enables single bNAbs to effectively control an established HIV-1 infection in humanized mice. Effective control of HIV-1 infection in humans is achieved using combinations of antiretroviral therapy (ART) drugs. In humanized mice (hu-mice), control of viremia can be achieved using either ART or by immunotherapy using combinations of broadly neutralizing antibodies (bNAbs). Here we show that treatment of HIV-1–infected hu-mice with a combination of three highly potent bNAbs not only resulted in complete viremic control but also led to a reduction in cell-associated HIV-1 DNA. Moreover, lowering the initial viral load by coadministration of ART and immunotherapy enabled prolonged viremic control by a single bNAb after ART was withdrawn. Similarly, a single injection of adeno-associated virus directing expression of one bNAb produced durable viremic control after ART was terminated. We conclude that immunotherapy reduces plasma viral load and cell-associated HIV-1 DNA and that decreasing the initial viral load enables single bNAbs to control viremia in hu-mice.


Blood | 2011

Development of human CD4+FoxP3+ regulatory T cells in human stem cell factor–, granulocyte-macrophage colony-stimulating factor–, and interleukin-3–expressing NOD-SCID IL2Rγnull humanized mice

Eva Billerbeck; Walter T. Barry; Kathy Mu; Marcus Dorner; Charles M. Rice; Alexander Ploss

Human hematolymphoid mice have become valuable tools for the study of human hematopoiesis and uniquely human pathogens in vivo. Recent improvements in xenorecipient strains allow for long-term reconstitution with a human immune system. However, certain hematopoietic lineages, for example, the myeloid lineage, are underrepresented, possibly because of the limited cross-reactivity of murine and human cytokines. Therefore, we created a nonobese diabetic/severe combined immunodeficiency/interleukin-2 receptor-γ-null (NOD-SCID IL2Rγ(null)) mouse strain that expressed human stem cell factor, granulocyte-macrophage colony-stimulating factor, and interleukin-3, termed NSG-SGM3. Transplantation of CD34(+) human hematopoietic stem cells into NSG-SGM3 mice led to robust human hematopoietic reconstitution in blood, spleen, bone marrow, and liver. Human myeloid cell frequencies, specifically, myeloid dendritic cells, were elevated in the bone marrow of humanized NSG-SGM3 mice compared with nontransgenic NSG recipients. Most significant, however, was the increase in the CD4(+)FoxP3(+) regulatory T-cell population in all compartments analyzed. These CD4(+)FoxP3(+) regulatory T cells were functional, as evidenced by their ability to suppress T-cell proliferation. In conclusion, humanized NSG-SGM3 mice might serve as a useful model to study human regulatory T-cell development in vivo, but this unexpected lineage skewing also highlights the importance of adequate spatiotemporal expression of human cytokines for future xenorecipient strain development.


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.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Dengue reporter viruses reveal viral dynamics in interferon receptor-deficient mice and sensitivity to interferon effectors in vitro

John W. Schoggins; Marcus Dorner; Michael Feulner; Naoko Imanaka; Mary Y. Murphy; Alexander Ploss; Charles M. Rice

Dengue virus (DENV) is a global disease threat for which there are no approved antivirals or vaccines. Establishing state-of-the-art screening systems that rely on fluorescent or luminescent reporters may accelerate the development of anti-DENV therapeutics. However, relatively few reporter DENV platforms exist. Here, we show that DENV can be genetically engineered to express a green fluorescent protein or firefly luciferase. Reporter viruses are infectious in vitro and in vivo and are sensitive to antiviral compounds, neutralizing antibodies, and interferons. Bioluminescence imaging was used to follow the dynamics of DENV infection in mice and revealed that the virus localized predominantly to lymphoid and gut-associated tissues. The high-throughput potential of reporter DENV was demonstrated by screening a library of more than 350 IFN-stimulated genes for antiviral activity. Several antiviral effectors were identified, and they targeted DENV at two distinct life cycle steps. These viruses provide a powerful platform for applications ranging from validation of vaccine candidates to antiviral discovery.


Nature Medicine | 2013

Inflammatory Flt3l is essential to mobilize dendritic cells and for T cell responses during Plasmodium infection

Pierre Guermonprez; Julie Helft; Carla Claser; Stephanie Deroubaix; Henry Karanje; Anna Gazumyan; Guillaume Darasse-Jèze; Stephanie B. Telerman; Gaëlle Breton; Heidi A. Schreiber; Natalia Frias-Staheli; Eva Billerbeck; Marcus Dorner; Charles M. Rice; Alexander Ploss; Florian Klein; Melissa Swiecki; Marco Colonna; Alice O. Kamphorst; Matthew M. Meredith; Rachel E. Niec; Constantin N. Takacs; Fadi Mikhail; Aswin Hari; David Bosque; Tom Eisenreich; Miriam Merad; Yan Shi; Florent Ginhoux; Laurent Rénia

Innate sensing mechanisms trigger a variety of humoral and cellular events that are essential to adaptive immune responses. Here we describe an innate sensing pathway triggered by Plasmodium infection that regulates dendritic cell homeostasis and adaptive immunity through Flt3 ligand (Flt3l) release. Plasmodium-induced Flt3l release in mice requires Toll-like receptor (TLR) activation and type I interferon (IFN) production. We found that type I IFN supports the upregulation of xanthine dehydrogenase, which metabolizes the xanthine accumulating in infected erythrocytes to uric acid. Uric acid crystals trigger mast cells to release soluble Flt3l from a pre-synthesized membrane-associated precursor. During infection, Flt3l preferentially stimulates expansion of the CD8-α+ dendritic cell subset or its BDCA3+ human dendritic cell equivalent and has a substantial impact on the magnitude of T cell activation, mostly in the CD8+ compartment. Our findings highlight a new mechanism that regulates dendritic cell homeostasis and T cell responses to infection.


Cell Host & Microbe | 2014

Interferon lambda alleles predict innate antiviral immune responses and hepatitis C virus permissiveness.

Timothy Sheahan; Naoko Imanaka; Svetlana Marukian; Marcus Dorner; Peng Liu; Alexander Ploss; Charles M. Rice

Hepatitis C virus (HCV) infection can result in viral chronicity or clearance. Although host genetics and particularly genetic variation in the interferon lambda (IFNL) locus are associated with spontaneous HCV clearance and treatment success, the mechanisms guiding these clinical outcomes remain unknown. Using a laser capture microdissection-driven unbiased systems virology approach, we isolated and transcriptionally profiled HCV-infected and adjacent primary human hepatocytes (PHHs) approaching single-cell resolution. An innate antiviral immune signature dominated the transcriptional response but differed in magnitude and diversity between HCV-infected and adjacent cells. Molecular signatures associated with more effective antiviral control were determined by comparing donors with high and low infection frequencies. Cells from donors with clinically unfavorable IFNL genotypes were infected at a greater frequency and exhibited dampened antiviral and cell death responses. These data suggest that early virus-host interactions, particularly host genetics and induction of innate immunity, critically determine the outcome of HCV infection.


Immunology | 2009

Plasma cell toll-like receptor (TLR) expression differs from that of B cells, and plasma cell TLR triggering enhances immunoglobulin production

Marcus Dorner; Simone Brandt; Marianne Tinguely; Franziska Zucol; Jean-Pierre Bourquin; Christoph Berger; Michele Bernasconi; Roberto F. Speck; David Nadal

Toll‐like receptors (TLRs) are key receptors of the innate immune system and show cell subset‐specific expression. We investigated the messenger RNA (mRNA) expression of TLR genes in human haematopoietic stem cells (HSC), in naïve B cells, in memory B cells, in plasma cells from palatine tonsils and in plasma cells from peripheral blood. HSC and plasma cells showed unrestricted expression of TLR1–TLR9, in contrast to B cells which lacked TLR3, TLR4 and TLR8 but expressed mRNA of all other TLRs. We demonstrated, for the first time, that TLR triggering of terminally differentiated plasma cells augments immunoglobulin production. Thus, boosting the immediate antibody response by plasma cells upon pathogen recognition may point to a novel role of TLRs.

Collaboration


Dive into the Marcus Dorner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christoph Berger

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

David Nadal

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge