Network


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

Hotspot


Dive into the research topics where Anthony Rongvaux is active.

Publication


Featured researches published by Anthony Rongvaux.


Nature | 2011

Control of TH17 cells occurs in the small intestine

Enric Esplugues; Samuel Huber; Nicola Gagliani; Anja E. Hauser; Terrence Town; Yisong Y. Wan; William O’Connor; Anthony Rongvaux; Nico van Rooijen; Ann M. Haberman; Yoichiro Iwakura; Vijay K. Kuchroo; Jay K. Kolls; Jeffrey A. Bluestone; Kevan C. Herold; Richard A. Flavell

Interleukin (IL)-17-producing T helper cells (TH17) are a recently identified CD4+ T cell subset distinct from T helper type 1 (TH1) and T helper type 2 (TH2) cells. TH17 cells can drive antigen-specific autoimmune diseases and are considered the main population of pathogenic T cells driving experimental autoimmune encephalomyelitis (EAE), the mouse model for multiple sclerosis. The factors that are needed for the generation of TH17 cells have been well characterized. However, where and how the immune system controls TH17 cells in vivo remains unclear. Here, by using a model of tolerance induced by CD3-specific antibody, a model of sepsis and influenza A viral infection (H1N1), we show that pro-inflammatory TH17 cells can be redirected to and controlled in the small intestine. TH17-specific IL-17A secretion induced expression of the chemokine CCL20 in the small intestine, facilitating the migration of these cells specifically to the small intestine via the CCR6/CCL20 axis. Moreover, we found that TH17 cells are controlled by two different mechanisms in the small intestine: first, they are eliminated via the intestinal lumen; second, pro-inflammatory TH17 cells simultaneously acquire a regulatory phenotype with in vitro and in vivo immune-suppressive properties (rTH17). These results identify mechanisms limiting TH17 cell pathogenicity and implicate the gastrointestinal tract as a site for control of TH17 cells.


Nature | 2015

Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations

Kai Deng; Mihaela Pertea; Anthony Rongvaux; Leyao Wang; Christine M. Durand; Gabriel Ghiaur; Jun Lai; Holly McHugh; Haiping Hao; Hao Zhang; Joseph B. Margolick; Cagan Gurer; Andrew J. Murphy; David M. Valenzuela; George D. Yancopoulos; Steven G. Deeks; Till Strowig; Priti Kumar; Janet D. Siliciano; Richard A. Flavell; Liang Shan; Robert F. Siliciano

Despite antiretroviral therapy (ART), human immunodeficiency virus (HIV)-1 persists in a stable latent reservoir, primarily in resting memory CD4+ T cells. This reservoir presents a major barrier to the cure of HIV-1 infection. To purge the reservoir, pharmacological reactivation of latent HIV-1 has been proposed and tested both in vitro and in vivo. A key remaining question is whether virus-specific immune mechanisms, including cytotoxic T lymphocytes (CTLs), can clear infected cells in ART-treated patients after latency is reversed. Here we show that there is a striking all or none pattern for CTL escape mutations in HIV-1 Gag epitopes. Unless ART is started early, the vast majority (>98%) of latent viruses carry CTL escape mutations that render infected cells insensitive to CTLs directed at common epitopes. To solve this problem, we identified CTLs that could recognize epitopes from latent HIV-1 that were unmutated in every chronically infected patient tested. Upon stimulation, these CTLs eliminated target cells infected with autologous virus derived from the latent reservoir, both in vitro and in patient-derived humanized mice. The predominance of CTL-resistant viruses in the latent reservoir poses a major challenge to viral eradication. Our results demonstrate that chronically infected patients retain a broad-spectrum viral-specific CTL response and that appropriate boosting of this response may be required for the elimination of the latent reservoir.


Nature Biotechnology | 2014

Development and function of human innate immune cells in a humanized mouse model

Anthony Rongvaux; Tim Willinger; Jan Martinek; Till Strowig; Sofia V Gearty; Lino L. Teichmann; Yasuyuki Saito; Florentina Marches; Stephanie Halene; A. Karolina Palucka; Markus G. Manz; Richard A. Flavell

Mice repopulated with human hematopoietic cells are a powerful tool for the study of human hematopoiesis and immune function in vivo. However, existing humanized mouse models cannot support development of human innate immune cells, including myeloid cells and natural killer (NK) cells. Here we describe two mouse strains called MITRG and MISTRG, in which human versions of four genes encoding cytokines important for innate immune cell development are knocked into their respective mouse loci. The human cytokines support the development and function of monocytes, macrophages and NK cells derived from human fetal liver or adult CD34+ progenitor cells injected into the mice. Human macrophages infiltrated a human tumor xenograft in MITRG and MISTRG mice in a manner resembling that observed in tumors obtained from human patients. This humanized mouse model may be used to model the human immune system in scenarios of health and pathology, and may enable evaluation of therapeutic candidates in an in vivo setting relevant to human physiology.


Cell | 2014

Apoptotic Caspases Prevent the Induction of Type I Interferons by Mitochondrial DNA

Anthony Rongvaux; Ruaidhrí Jackson; Christian C. D. Harman; Tuo Li; A. Phillip West; Marcel R. de Zoete; Youtong Wu; Brian Yordy; Saquib A. Lakhani; Chia Yi Kuan; Tadatsugu Taniguchi; Gerald S. Shadel; Zhijian J. Chen; Akiko Iwasaki; Richard A. Flavell

The mechanism by which cells undergo death determines whether dying cells trigger inflammatory responses or remain immunologically silent. Mitochondria play a central role in the induction of cell death, as well as in immune signaling pathways. Here, we identify a mechanism by which mitochondria and downstream proapoptotic caspases regulate the activation of antiviral immunity. In the absence of active caspases, mitochondrial outer membrane permeabilization by Bax and Bak results in the expression of type I interferons (IFNs). This induction is mediated by mitochondrial DNA-dependent activation of the cGAS/STING pathway and results in the establishment of a potent state of viral resistance. Our results show that mitochondria have the capacity to simultaneously expose a cell-intrinsic inducer of the IFN response and to inactivate this response in a caspase-dependent manner. This mechanism provides a dual control, which determines whether mitochondria initiate an immunologically silent or a proinflammatory type of cell death.


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.


Annual Review of Immunology | 2013

Human Hemato-Lymphoid System Mice: Current Use and Future Potential for Medicine

Anthony Rongvaux; Hitoshi Takizawa; Till Strowig; Tim Willinger; Elizabeth E. Eynon; Richard A. Flavell; Markus G. Manz

To directly study complex human hemato-lymphoid system physiology and respective system-associated diseases in vivo, human-to-mouse xenotransplantation models for human blood and blood-forming cells and organs have been developed over the past three decades. We here review the fundamental requirements and the remarkable progress made over the past few years in improving these systems, the current major achievements reached by use of these models, and the future challenges to more closely model and study human health and disease and to achieve predictive preclinical testing of both prevention measures and potential new therapies.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Human thrombopoietin knockin mice efficiently support human hematopoiesis in vivo

Anthony Rongvaux; Tim Willinger; Hitoshi Takizawa; Chozhavendan Rathinam; Wojtek Auerbach; Andrew J. Murphy; David M. Valenzuela; George D. Yancopoulos; Elizabeth E. Eynon; Sean Stevens; Markus G. Manz; Richard A. Flavell

Hematopoietic stem cells (HSCs) both self-renew and give rise to all blood cells for the lifetime of an individual. Xenogeneic mouse models are broadly used to study human hematopoietic stem and progenitor cell biology in vivo. However, maintenance, differentiation, and function of human hematopoietic cells are suboptimal in these hosts. Thrombopoietin (TPO) has been demonstrated as a crucial cytokine supporting maintenance and self-renewal of HSCs. We generated RAG2−/−γc−/− mice in which we replaced the gene encoding mouse TPO by its human homolog. Homozygous humanization of TPO led to increased levels of human engraftment in the bone marrow of the hosts, and multilineage differentiation of hematopoietic cells was improved, with an increased ratio of myelomonocytic verus lymphoid lineages. Moreover, maintenance of human stem and progenitor cells was improved, as demonstrated by serial transplantation. Therefore, RAG2−/−γc−/− TPO-humanized mice represent a useful model to study human hematopoiesis in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Transgenic expression of human signal regulatory protein alpha in Rag2−/−γc−/− mice improves engraftment of human hematopoietic cells in humanized mice

Till Strowig; Anthony Rongvaux; Chozhavendan Rathinam; Hitoshi Takizawa; Chiara Borsotti; William M. Philbrick; Elizabeth E. Eynon; Markus G. Manz; Richard A. Flavell

Transplantation of human hematopoietic stem cells into severely immunocompromised newborn mice allows the development of a human hematopoietic and immune system in vivo. NOD/scid/γc−/− (NSG) and BALB/c Rag2−/−γc−/− mice are the most commonly used mouse strains for this purpose and a number of studies have demonstrated the high value of these model systems in areas spanning from basic to translational research. However, limited cross-reactivity of many murine cytokines on human cells and residual host immune function against the xenogeneic grafts results in defective development and maintenance of human cells in vivo. Whereas NSG mice have higher levels of absolute human engraftment than similar mice on a BALB/c background, they have a shorter lifespan and NOD ES cells are unsuitable for the complex genetic engineering that is required to improve human hematopoiesis and immune responses by transgenesis or knockin of human genes. We have generated mice that faithfully express a transgene of human signal regulatory protein alpha (SIRPa), a receptor that negatively regulates phagocytosis, in Rag2−/−γc−/− mice on a mixed 129/BALB/c background, which can easily be genetically engineered. These mice allow significantly increased engraftment and maintenance of human hematopoietic cells reaching levels comparable to NSG mice. Furthermore, we found improved functionality of the human immune system in these mice. In summary, hSIRPa-transgenic Rag2−/−γc−/− mice represent a unique mouse strain supporting high levels of human cell engraftment, which can easily be genetically manipulated.


Journal of Immunology | 2008

Nicotinamide Phosphoribosyl Transferase/Pre-B Cell Colony-Enhancing Factor/Visfatin Is Required for Lymphocyte Development and Cellular Resistance to Genotoxic Stress

Anthony Rongvaux; Mara Galli; Sébastien Denanglaire; Frédéric Van Gool; Pierre Luc Dreze; Claude Szpirer; Fabrice Bureau; Fabienne Andris; Oberdan Leo

Nicotinamide phosphoribosyl transferase (Nampt)/pre-B cell colony-enhancing factor (PBEF)/visfatin is a protein displaying multiple functional properties. Originally described as a cytokine-like protein able to regulate B cell development, apoptosis, and glucose metabolism, this protein also plays an important role in NAD biosynthesis. To gain insight into its physiological role, we have generated a mouse strain expressing a conditional Nampt allele. Lack of Nampt expression strongly affects development of both T and B lymphocytes. Analysis of hemizygous cells and in vitro cell lines expressing distinct levels of Nampt illustrates the critical role of this protein in regulating intracellular NAD levels. Consequently, a clear relationship was found between intracellular Nampt levels and cell death in response to the genotoxic agent MNNG (N-methyl-N′-nitro-N-nitrosoguanidine), confirming that this enzyme represents a key regulator of cell sensitivity to NAD-consuming stress secondary to poly(ADP-ribose) polymerases overactivation. By using mutant forms of this protein and a well-characterized pharmacological inhibitor (FK866), we unequivocally demonstrate that the ability of the Nampt to regulate cell viability during genotoxic stress requires its enzymatic activity. Collectively, these data demonstrate that Nampt participates in cellular resistance to genotoxic/oxidative stress, and it may confer to cells of the immune system the ability to survive during stressful situations such as inflammation.


Trends in Immunology | 2011

Improving human hemato-lymphoid-system mice by cytokine knock-in gene replacement

Tim Willinger; Anthony Rongvaux; Till Strowig; Markus G. Manz; Richard A. Flavell

Human hemato-lymphoid-system mice hold great promise for modeling and studying important human diseases in vivo, and to enable vaccine testing. Until now, several major limitations have restricted the utility of human hemato-lymphoid-system mice in translational research. Recently, however, significant advances have been made in improving these mice, based on the delivery of human cytokines to create a better environment for human cells in the mouse host. In this review, we discuss the various approaches with a particular focus on improving human hemato-lymphoid-system mice by human cytokine knock-in gene replacement.

Collaboration


Dive into the Anthony Rongvaux'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
Top Co-Authors

Avatar

Jorge Henao-Mejia

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Oberdan Leo

Université libre de Bruxelles

View shared research outputs
Researchain Logo
Decentralizing Knowledge