Network


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

Hotspot


Dive into the research topics where Rita Vicente is active.

Publication


Featured researches published by Rita Vicente.


Nature Reviews Rheumatology | 2016

Deregulation and therapeutic potential of microRNAs in arthritic diseases.

Rita Vicente; Danièle Noël; Yves-Marie Pers; Florence Apparailly; Christian Jorgensen

Epigenetic abnormalities are part of the pathogenetic alterations involved in the development of rheumatic disorders. In this context, the main musculoskeletal cell lineages, which are generated from the pool of mesenchymal stromal cells (MSCs), and the immune cells that participate in rheumatic diseases are deregulated. In this Review, we focus on microRNA (miRNA)-mediated regulatory pathways that control cell proliferation, drive the production of proinflammatory mediators and modulate bone remodelling. The main studies that identify miRNAs as regulators of immune cell fate, MSC differentiation and immunomodulatory properties — parameters that are altered in rheumatoid arthritis (RA) and osteoarthritis (OA) — are also discussed, with emphasis on the importance of miRNAs in the regulation of cellular machinery, extracellular matrix remodelling and cytokine release. A deeper understanding of the involvement of miRNAs in rheumatic diseases is needed before these regulatory pathways can be explored as therapeutic approaches for patients with RA or OA.


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.


Aging Cell | 2016

Cellular senescence impact on immune cell fate and function.

Rita Vicente; Anne-Laure Mausset-Bonnefont; Christian Jorgensen; Pascale Louis-Plence; Jean-Marc Brondello

Cellular senescence occurs not only in cultured fibroblasts, but also in undifferentiated and specialized cells from various tissues of all ages, in vitro and in vivo. Here, we review recent findings on the role of cellular senescence in immune cell fate decisions in macrophage polarization, natural killer cell phenotype, and following T‐lymphocyte activation. We also introduce the involvement of the onset of cellular senescence in some immune responses including T‐helper lymphocyte‐dependent tissue homeostatic functions and T‐regulatory cell‐dependent suppressive mechanisms. Altogether, these data propose that cellular senescence plays a wide‐reaching role as a homeostatic orchestrator.


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.


Methods of Molecular Biology | 2008

Lentiviral Transduction of Immune Cells

Louise Swainson; Cédric Mongellaz; Oumeya Adjali; Rita Vicente; Naomi Taylor

Gene transfer into mammalian cells has been of crucial importance for studies determining the role of specific genes in the differentiation and cell fate of various hematopoietic lineages. Until recently, the majority of these studies were performed in transformed cell lines due to difficulties in achieving levels of transfection of greater than 1-3% in primary hematopoietic cells. Vectors based on retrovirus and lentivirus backbones have revolutionized our ability to transfer genes into primary hematopoietic cells. These vectors have allowed extensive ex vivo and in vivo studies following introduction of a gene of interest and have been used clinically in individuals suffering from cancers, infections, and genetic diseases. Ex vivo lentiviral gene transfer can result in efficient transduction of progenitor cells (>80%) that can then be further differentiated into immune lineage cells including T, B, dendritic, or natural killer cells. Alternatively, differentiated immune cells can themselves be transduced ex vivo with lentiviral vectors. Here, we discuss optimization of technologies for human immunodeficiency virus (HIV)-based gene transfer into murine and human progenitor and immune cell lineages.


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.


Methods of Molecular Biology | 2009

In Vivo and Ex Vivo Gene Transfer in Thymocytes and Thymocyte Precursors

Oumeya Adjali; Amélie Montel-Hagen; Louise Swainson; Sophie Marty; Rita Vicente; Cédric Mongellaz; Chantal Jacquet; Valérie S. Zimmermann; Naomi Taylor

The thymus provides a specialized environment allowing the differentiation of T lymphocytes from bone marrow-derived progenitor cells. We and others have demonstrated that gene transfer into distinct thymocyte populations can be obtained, both in vivo and ex vivo, using lentiviral vectors. Here, we describe techniques for intrathymic lentiviral transduction in mice, using a surgical approach wherein the thoracic cavity is exposed as well as a significantly less invasive strategy wherein virions are directly injected through the skin. Moreover, thymocyte differentiation from murine and human progenitors is now feasible in vitro, under conditions wherein the Notch and IL-7 signaling pathways are activated. We describe methods allowing transduction of murine and human progenitors and their subsequent differentiation into more mature thymocytes. Conditions for lentiviral gene transfer into more differentiated human thymocyte subsets are also presented. Optimization of technologies for HIV-based gene transfer into murine and human thymocyte progenitors will advance strategies aimed at modulating T-cell differentiation and function in-vivo; approaches potentially targeting patients with genetic and acquired immunodeficiencies as well as immune-sensitive tumors. Furthermore, this technology will foster the progression of basic research aimed at elucidating molecular aspects of T-cell differentiation in mice and humans.


PLOS ONE | 2015

Systemic LPS Translocation Activates Cross-Presenting Dendritic Cells but Is Dispensable for the Breakdown of CD8 + T Cell Peripheral Tolerance in Irradiated Mice

Gabriel Espinosa-Carrasco; Marine Villard; Cecile Le Saout; Pascale Louis-Plence; Rita Vicente; Javier Hernandez

Lymphodepletion is currently used to enhance the efficacy of cytotoxic T lymphocyte adoptive transfer immunotherapy against cancer. This beneficial effect of conditioning regimens is due, at least in part, to promoting the breakdown of peripheral CD8+ T cell tolerance. Lymphodepletion by total body irradiation induces systemic translocation of commensal bacteria LPS from the gastrointestinal tract. Since LPS is a potent activator of the innate immune system, including antigen presenting dendritic cells, we hypothesized that LPS translocation could be required for the breakdown of peripheral tolerance observed in irradiated mice. To address this issue, we have treated irradiated mice with antibiotics in order to prevent LPS translocation and utilized them in T cell adoptive transfer experiments. Surprisingly, we found that despite of completely blocking LPS translocation into the bloodstream, antibiotic treatment did not prevent the breakdown of peripheral tolerance. Although irradiation induced the activation of cross-presenting CD8+ dendritic cells in the lymphoid tissue, LPS could not solely account for this effect. Activation of dendritic cells by mechanisms other than LPS translocation is sufficient to promote the differentiation of potentially autoreactive CD8+ T cells into effectors in irradiated mice. Our data indicate that LPS translocation is dispensable for the breakdown of CD8+ T cell tolerance in irradiated mice.


Annals of the Rheumatic Diseases | 2014

A8.26 Inducible IL-10 secreting CD49b +Treg cells as cell based-therapy for rheumatoid arthritis

Julie Quentin; Rita Vicente; Delphine Martire; Anne-Laure Mausset-Bonnefont; Christian Jorgensen; Pascale Louis-Plence

Background Adoptive transfer of regulatory T cells (Tregs) is a promising approach to restore tolerance in autoimmune diseases. However based on the heterogeneity of the Tregs, we need to precisely establish which Tregs will be able to dampen efficiently the immune response in the various settings. We previously showed the potential of CD49b+ Treg cells to protect and prevent an experimental model of arthritis. Nevertheless the optimal injection dose, the phenotype and the in vivo-suppressive mechanism of these Treg cells remain unknown. In our study, we investigated and compared the therapeutic potential of CD25+FoxP3+ and induced IL10-secreting CD49b+ Treg cells in an experimental model of arthritis, the collagen-induced arthritis (CIA). Materials and Methods IL-10 secreting CD49b+ Treg cells were generated in naïve mice following repetitive injections of immature DCs (iDCs). Treg purification was based on the negative selection of CD4 T cells isolated from spleen and liver of the iDC-vaccinated mice. Cell sorting was performed to obtain 98% pure CD49b+ or CD25+ Treg cells. Several doses of CD49b+ were intravenously (i.v.) injected at day 28 in established CIA. Clinical signs of arthritis were scored, as well as biological parameters such as the level of anti-bCII antibodies in sera and the cytokine profile of bCII specific T cells. Phenotypes of both Treg cells were compared as well as their suppressive activity in vitro and in vivo. Results Several doses of CD49+ Treg cells were tested in curative settings experiments. The dose of 105 CD49b+ or CD25+ cells reverse clinical symptoms of arthritis while interestingly, a lack of efficacy was observed after higher doses. In vitro suppressive experiments confirmed the similar efficiency of both populations and phenotype analyses of CD49b+ Treg cells showed expression profile of several Treg specific markers (LAP+, LAG+, CTLA-4high). Moreover, in an OVA-specific model of inflammation, we demonstrated the high impact of the CD49b Treg cells on the proliferation of effector cells in vivo. Conclusions Altogether, our results confirm the therapeutic potential of IL-10 secreting T cells in experimental model of arthritis in curative settings and unravel their mechanism of suppression.

Collaboration


Dive into the Rita Vicente's collaboration.

Top Co-Authors

Avatar

Naomi Taylor

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chantal Jacquet

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Valérie S. Zimmermann

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Julie Quentin

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar

Cédric Mongellaz

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Louise Swainson

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge