Network


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

Hotspot


Dive into the research topics where Valérie S. Zimmermann is active.

Publication


Featured researches published by Valérie S. Zimmermann.


Nature | 2016

Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites.

François Gerbe; Emmanuelle Sidot; Danielle J. Smyth; Makoto Ohmoto; Ichiro Matsumoto; Valérie Dardalhon; Pierre Cesses; Laure Garnier; Marie Pouzolles; Bénédicte Brulin; Marco Bruschi; Yvonne Harcus; Valérie S. Zimmermann; Naomi Taylor; Rick M. Maizels; Philippe Jay

Helminth parasitic infections are a major global health and social burden. The host defence against helminths such as Nippostrongylus brasiliensis is orchestrated by type 2 cell-mediated immunity. Induction of type 2 cytokines, including interleukins (IL) IL-4 and IL-13, induce goblet cell hyperplasia with mucus production, ultimately resulting in worm expulsion. However, the mechanisms underlying the initiation of type 2 responses remain incompletely understood. Here we show that tuft cells, a rare epithelial cell type in the steady-state intestinal epithelium, are responsible for initiating type 2 responses to parasites by a cytokine-mediated cellular relay. Tuft cells have a Th2-related gene expression signature and we demonstrate that they undergo a rapid and extensive IL-4Rα-dependent amplification following infection with helminth parasites, owing to direct differentiation of epithelial crypt progenitor cells. We find that the Pou2f3 gene is essential for tuft cell specification. Pou2f3−/− mice lack intestinal tuft cells and have defective mucosal type 2 responses to helminth infection; goblet cell hyperplasia is abrogated and worm expulsion is compromised. Notably, IL-4Rα signalling is sufficient to induce expansion of the tuft cell lineage, and ectopic stimulation of this signalling cascade obviates the need for tuft cells in the epithelial cell remodelling of the intestine. Moreover, tuft cells secrete IL-25, thereby regulating type 2 immune responses. Our data reveal a novel function of intestinal epithelial tuft cells and demonstrate a cellular relay required for initiating mucosal type 2 immunity to helminth infection.


Cell Stem Cell | 2014

Glucose and Glutamine Metabolism Regulate Human Hematopoietic Stem Cell Lineage Specification

Leal Oburoglu; Saverio Tardito; Vanessa Fritz; Stéphanie C. de Barros; Peggy Merida; Marco Craveiro; João I. Mamede; Gaspard Cretenet; Cédric Mongellaz; Xiuli An; Dorota Klysz; Jawida Touhami; Myriam Boyer-Clavel; Jean-Luc Battini; Valérie Dardalhon; Valérie S. Zimmermann; Narla Mohandas; Eyal Gottlieb; Marc Sitbon; Sandrina Kinet; Naomi Taylor

The metabolic state of quiescent hematopoietic stem cells (HSCs) is an important regulator of self-renewal, but it is unclear whether or how metabolic parameters contribute to HSC lineage specification and commitment. Here, we show that the commitment of human and murine HSCs to the erythroid lineage is dependent upon glutamine metabolism. HSCs require the ASCT2 glutamine transporter and active glutamine metabolism for erythroid specification. Blocking this pathway diverts EPO-stimulated HSCs to differentiate into myelomonocytic fates, altering in vivo HSC responses and erythroid commitment under stress conditions such as hemolytic anemia. Mechanistically, erythroid specification of HSCs requires glutamine-dependent de novo nucleotide biosynthesis. Exogenous nucleosides rescue erythroid commitment of human HSCs under conditions of limited glutamine catabolism, and glucose-stimulated nucleotide biosynthesis further enhances erythroid specification. Thus, the availability of glutamine and glucose to provide fuel for nucleotide biosynthesis regulates HSC lineage commitment under conditions of metabolic stress.


Seminars in Immunology | 2010

Molecular and cellular basis of T cell lineage commitment

Rita Vicente; Louise Swainson; Sophie Marty-Grès; Stéphanie C. de Barros; Sandrina Kinet; Valérie S. Zimmermann; Naomi Taylor

The thymus forms as an alymphoid thymic primordium with T cell differentiation requiring the seeding of this anlage. This review will focus on the characteristics of the hematopoietic progenitors which colonize the thymus and their subsequent commitment/differentiation, both in mice and men. Within the thymus, the interplay between Notch1 and IL-7 signals is crucial for the orchestration of T cell development, but the precise requirements for these factors in murine and human thympoeisis are not synonymous. Recent advances in our understanding of the mechanisms regulating precursor entry and their maintenance in the thymus will also be presented.


Blood | 2010

Intrathymic transplantation of bone marrow–derived progenitors provides long-term thymopoiesis

Rita Vicente; Oumeya Adjali; Chantal Jacquet; Valérie S. Zimmermann; Naomi Taylor

The sustained differentiation of T cells in the thymus cannot be maintained by resident intrathymic (IT) precursors and requires that progenitors be replenished from the bone marrow (BM). In patients with severe combined immunodeficiency (SCID) treated by hematopoietic stem cell transplantation, late T-cell differentiation defects are thought to be due to an insufficient entry of donor BM progenitors into the thymus. Indeed, we find that the intravenous injection of BM progenitors into nonconditioned zeta-chain-associated protein kinase 70 (ZAP-70)-deficient mice with SCID supports short- but not long-term thymopoiesis. Remarkably, we now show that the IT administration of these progenitors produces a significant level of donor-derived thymopoiesis for more than 6 months after transplantation. In contrast to physiologic thymopoiesis, long-term donor thymopoiesis was not due to the continued recruitment of progenitors from the BM. Rather, IT transplantation resulted in the unique generation of a large population of early c-Kit(high) donor precursors within the thymus. These ZAP-70-deficient mice that received an IT transplant had a significantly increased prothymocyte niche compared with their untreated counterparts; this phenotype was associated with the generation of a medulla. Thus, IT administration of BM progenitors results in the filling of an expanded precursor niche and may represent a strategy for enhancing T-cell differentiation in patients with SCID.


Molecular Therapy | 2009

Efficient Intrathymic Gene Transfer Following In Situ Administration of a rAAV Serotype 8 Vector in Mice and Nonhuman Primates

Aurélie Moreau; Rita Vicente; Laurence Dubreil; Oumeya Adjali; Guillaume Podevin; Chantal Jacquet; Jack Yves Deschamps; David Klatzmann; Yan Cherel; Naomi Taylor; Philippe Moullier; Valérie S. Zimmermann

The thymus is the primary site of T-cell development and plays a key role in the induction of self-tolerance. We previously showed that the intrathymic (i.t.) injection of a transgene-expressing lentiviral vector (LV) in mice can result in the correction of a T cell-specific genetic defect. Nevertheless, the efficiency of thymocyte transduction did not exceed 0.1-0.3% and we were unable to detect any thymus transduction in macaques. As such, we initiated studies to assess the capacity of recombinant adeno-associated virus (rAAV) vectors to transduce murine and primate thymic cells. In vivo administration of AAV serotype 2-derived single-stranded AAV (ssAAV) and self-complementary AAV (scAAV) vectors pseudotyped with capsid proteins of serotypes 1, 2, 4, 5, and 8 demonstrated that murine thymus transduction was significantly enhanced by scAAV2/8. Transgene expression was detected in 5% of thymocytes and, notably, transduced cells represented 1% of peripheral T lymphocytes. Moreover, i.t. administration of scAAV2/8 particles in macaques, by endoscopic-mediated guidance, resulted in significant gene transfer. Thus, in healthy animals, where thymic gene transfer does not provide a selective advantage, scAAV2/8 is a unique tool promoting the in situ transduction of thymocytes with the subsequent export of gene-modified lymphocytes to the periphery.


Blood | 2013

Intrathymic progenitor cell transplantation across histocompatibility barriers results in the persistence of early thymic progenitors and T-cell differentiation

Stéphanie C. de Barros; Rita Vicente; Karim Chebli; Chantal Jacquet; Valérie S. Zimmermann; Naomi Taylor

Donor hematopoietic stem cells (HSCs) can correct T-cell deficiencies in patients with severe combined immunodeficiency by replacing resident thymus cells. However, as those progenitors that naturally migrate to the thymus are not capable of supporting long-term thymopoiesis, a successful transplant is thought to require the ongoing migration of donor progenitors. We previously showed that the forced intrathymic administration of histocompatible HSCs can sustain long-term thymopoiesis in ZAP-70-immunodeficient mice. However, it is not known whether T-cell reconstitution across histocompatibility barriers is modulated by intrathymic vs intravenous administration of HSCs. In the absence of conditioning, long-term thymopoiesis by semiallogeneic progenitors was detected in mice transplanted via the intrathymic, but not the intravenous, route. In intrathymic-transplanted mice, ongoing thymopoiesis was associated with a 10-fold higher level of early thymic progenitors (ETPs). The enhanced reconstitution capacity of these intrathymic-derived ETPs was corroborated by their significantly augmented myeloid lineage potential compared with endogenous ETPs. Notably, though, myeloablative conditioning resulted in a reduced expansion of intrathymic-administered donor ETPs. Thus, in the absence of conditioning, the forced thymic entry of HSCs results in a sustained T-cell development across histocompatibility barriers, highlighting the capacity of the thymus to support cells with long-term renewal potential.


Stem Cells | 2013

Concise Review: Hematopoietic Stem Cell Transplantation: Targeting the Thymus†‡§

Stéphanie C. de Barros; Valérie S. Zimmermann; Naomi Taylor

Allogeneic hematopoietic stem cell (HSC) transplantation can cure patients suffering from diverse genetic and acquired diseases as well as cancers. Nevertheless, under conditions where T‐cell reconstitution is critical, the entry of donor progenitors into the thymus remains a major bottleneck. It is assumed that following the intravenous injection of HSC, they first home to the BM. More committed progenitors can then be exported to the thymus in response to a myriad of signals regulating thymus seeding. Notably although, the thymus is not continually receptive to the import of hematopoietic progenitors. Furthermore, as stem cells with self‐renewing capacity do not take up residence in the thymus under physiological conditions, the periodic colonization of the thymus is essential for the sustained differentiation of T lymphocytes. As such, we and others have invested significant efforts into exploring avenues that might foster a long‐term thymus‐autonomous differentiation. Here, we review strategic approaches that have resulted in long‐term T‐cell differentiation in immunodeficient (SCID) mice, even across histocompatibility barriers. These include the forced thymic entry of BM precursors by their direct intrathymic injection as well as the transplantation of neonatal thymi. The capacity of the thymus to support hematopoietic progenitors with renewal potential will hopefully promote the development of new therapeutic strategies aimed at enhancing T‐cell differentiation in patients undergoing HSC transplantation. STEM Cells2013;31:1245–1251


Nature | 2018

Single-cell mapping of the thymic stroma identifies IL-25-producing tuft epithelial cells

Chamutal Bornstein; Shir Nevo; Amir Giladi; Noam Kadouri; Marie Pouzolles; François Gerbe; Eyal David; Alice Machado; Anna Chuprin; Beáta Tóth; Ori Goldberg; Shalev Itzkovitz; Naomi Taylor; Philippe Jay; Valérie S. Zimmermann; Jakub Abramson; Ido Amit

T cell development and selection are coordinated in the thymus by a specialized niche of diverse stromal populations1–3. Although much progress has been made over the years in identifying the functions of the different cell types of the thymic stromal compartment, there is no comprehensive characterization of their diversity and heterogeneity. Here we combined massively parallel single-cell RNA-sequencing4,5, spatial mapping, chromatin profiling and gene targeting to characterize de novo the entire stromal compartment of the mouse thymus. We identified dozens of cell states, with thymic epithelial cells (TECs) showing the highest degree of heterogeneity. Our analysis highlights four major medullary TEC (mTEC I–IV) populations, with distinct molecular functions, epigenetic landscapes and lineage regulators. Specifically, mTEC IV constitutes a new and highly divergent TEC lineage with molecular characteristics of the gut chemosensory epithelial tuft cells. Mice deficient in Pou2f3, a master regulator of tuft cells, have complete and specific depletion of mTEC IV cells, which results in increased levels of thymus-resident type-2 innate lymphoid cells. Overall, our study provides a comprehensive characterization of the thymic stroma and identifies a new tuft-like TEC population, which is critical for shaping the immune niche in the thymus.A comprehensive characterization of the thymic stroma identifies a tuft-cell-like thymic epithelial cell population that is critical for shaping the immune niche in the thymus.


Science Signaling | 2017

Resveratrol stimulates the metabolic reprogramming of human CD4+ T cells to enhance effector function

Marco Craveiro; Gaspard Cretenet; Cédric Mongellaz; Matias Mi; Olivier Caron; Maria C. Pedroso de Lima; Valérie S. Zimmermann; Eric Solary; Valérie Dardalhon; Vjekoslav Dulic; Naomi Taylor

Resveratrol alters human T cell responsiveness to antigens, coupling a replication stress response to metabolic reprogramming. Resveratrol and lymphocyte responses Resveratrol is a polyphenolic plant compound, which has attracted much interest as a pharmacological agent because of its potential therapeutic effects against cancer, aging, and inflammation. However, many studies have produced conflicting evidence of the effects of resveratrol in different contexts. Craveiro et al. found that high doses of resveratrol inhibited the responses of human CD4+ T cells to antigens. However, low doses of the drug reprogrammed the metabolism of the cells to make them more responsive to antigens and produce increased amounts of the inflammatory cytokine interferon-γ. These data suggest that the use of resveratrol to treat various pathologies should be carefully assessed, especially in an autoimmune setting. The polyphenol resveratrol activates the deacetylase Sirt1, resulting in various antioxidant, chemoprotectant, neuroprotective, cardioprotective, and anti-inflammatory properties. We found that at high concentrations of resveratrol, human CD4+ T cells showed defective antigen receptor signaling and arrest at the G1 stage of the cell cycle, whereas at low concentrations, cells were readily activated and exhibited enhanced Sirt1 deacetylase activity. Nevertheless, low-dose resveratrol rapidly stimulated genotoxic stress in the T cells, which resulted in engagement of a DNA damage response pathway that depended on the kinase ATR [ataxia telangiectasia–mutated (ATM) and Rad3-related], but not ATM, and subsequently in premitotic cell cycle arrest. The concomitant activation of p53 was coupled to the expression of gene products that regulate cell metabolism, leading to a metabolic reprogramming that was characterized by decreased glycolysis, increased glutamine consumption, and a shift to oxidative phosphorylation. These alterations in the bioenergetic homeostasis of CD4+ T cells resulted in enhanced effector function, with both naïve and memory CD4+ T cells secreting increased amounts of the inflammatory cytokine interferon-γ. Thus, our data highlight the wide range of metabolic adaptations that CD4+ T lymphocytes undergo in response to genomic stress.


Current Opinion in Hematology | 2016

Hematopoietic stem cell lineage specification.

Marie Pouzolles; Leal Oburoglu; Naomi Taylor; Valérie S. Zimmermann

Purpose of reviewHematopoietic stem cells (HSCs) possess two fundamental characteristics, the capacity for self-renewal and the sustained production of all blood cell lineages. The fine balance between HSC expansion and lineage specification is dynamically regulated by the interplay between external and internal stimuli. This review introduces recent advances in the roles played by the stem cell niche, regulatory transcriptional networks, and metabolic pathways in governing HSC self-renewal, commitment, and lineage differentiation. We will further focus on discoveries made by studying hematopoiesis at single-cell resolution. Recent findingsHSCs require the support of an interactive milieu with their physical position within the perivascular niche dynamically regulating HSC behavior. In these microenvironments, transcription factor networks and nutrient-mediated regulation of energy resources, signaling pathways, and epigenetic status govern HSC quiescence and differentiation. Once HSCs begin their lineage specification, single-cell analyses show that they do not become oligopotent but rather, differentiate directly into committed unipotent progenitors. SummaryThe diversity of transcriptional networks and metabolic pathways in HSCs and their downstream progeny allows a high level of plasticity in blood differentiation. The intricate interactions between these pathways, within the perivascular niche, broaden the specification of HSCs in pathological and stressed conditions.

Collaboration


Dive into the Valérie S. Zimmermann's collaboration.

Top Co-Authors

Avatar

Naomi Taylor

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Rita Vicente

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar

Stéphanie C. de Barros

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Marie Pouzolles

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar

Chantal Jacquet

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Valérie Dardalhon

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Marco Craveiro

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cédric Mongellaz

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Gaspard Cretenet

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge