Network


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

Hotspot


Dive into the research topics where Sem Saeland is active.

Publication


Featured researches published by Sem Saeland.


Immunity | 2000

Langerin, a novel C-type lectin specific to Langerhans cells, is an endocytic receptor that induces the formation of Birbeck granules

Jenny Valladeau; Odile Ravel; Colette Dezutter-Dambuyant; Kevin W. Moore; Monique J. Kleijmeer; Ying Liu; Valérie Duvert-Frances; Claude Vincent; Daniel Schmitt; Jean Davoust; Christophe Caux; Serge Lebecque; Sem Saeland

We have identified a type II Ca2+-dependent lectin displaying mannose-binding specificity, exclusively expressed by Langerhans cells (LC), and named Langerin. LC are uniquely characterized by Birbeck granules (BG), which are organelles consisting of superimposed and zippered membranes. Here, we have shown that Langerin is constitutively associated with BG and that antibody to Langerin is internalized into these structures. Remarkably, transfection of Langerin cDNA into fibroblasts created a compact network of membrane structures with typical features of BG. Langerin is thus a potent inducer of membrane superimposition and zippering leading to BG formation. Our data suggest that induction of BG is a consequence of the antigen-capture function of Langerin, allowing routing into these organelles and providing access to a nonclassical antigen-processing pathway.


Journal of Immunology | 2001

The Dendritic Cell Populations of Mouse Lymph Nodes

Sandrine Henri; David Vremec; Arun T. Kamath; Jason Waithman; Stuart Williams; Christophe Benoist; Kim Burnham; Sem Saeland; Emanuela Handman; Ken Shortman

The dendritic cells (DC) of mouse lymph nodes (LN) were isolated, analyzed for surface markers, and compared with those of spleen. Low to moderate staining of LN DC for CD4 and low staining for CD8 was shown to be attributable to pickup of these markers from T cells. Excluding this artifact, five LN DC subsets could be delineated. They included the three populations found in spleen (CD4+8−DEC-205−, CD4−8−DEC-205−, CD4−8+DEC-205+), although the CD4-expressing DC were of low incidence. LN DC included two additional populations, characterized by relatively low expression of CD8 but moderate or high expression of DEC-205. Both appeared among the DC migrating out of skin into LN, but only one was restricted to skin-draining LN and was identified as the mature form of epidermal Langerhans cells (LC). The putative LC-derived DC displayed the following properties: large size; high levels of class II MHC, which persisted to some extent even in CIITA null mice; expression of very high levels of DEC-205 and of CD40; expression of many myeloid surface markers; and no expression of CD4 and only low to moderate expression of CD8. The putative LC-derived DC among skin emigrants and in LN also showed strong intracellular staining of langerin.


European Journal of Immunology | 1999

The monoclonal antibody DCGM4 recognizes Langerin, a protein specific of Langerhans cells, and is rapidly internalized from the cell surface

Jenny Valladeau; Valérie Duvert-Frances; Jean-Jacques Pin; Colette Dezutter-Dambuyant; Claude Vincent; Catherine Massacrier; Jérôme Vincent; Kozo Yoneda; Jacques Banchereau; Christophe Caux; Jean Davoust; Sem Saeland

We generated monoclonal antibody (mAb) DCGM4 by immunization with human dendritic cells (DC) from CD34+ progenitors cultured with granulocyte‐macrophage colony‐stimulating factor and TNF‐α. mAb DCGM4 was selected for its reactivity with a cell surface epitope present only on a subset of DC. Reactivity was strongly enhanced by the Langerhans cell (LC) differentiation factor TGF‐β and down‐regulated by CD40 ligation. mAb DCGM4 selectively stained LC, hence we propose that the antigen be termed Langerin. mAb DCGM4 also stained intracytoplasmically, but neither colocalized with MHC class II nor with lysosomal LAMP‐1 markers. Notably, mAb DCGM4 was rapidly internalized at 37 °C, but did not gain access to MHC class II compartments. Finally, Langerin was immunoprecipitated as a 40‐kDa protein with a pI of 5.2 – 5.5. mAb DCGM4 will be useful to further characterize Langerin, an LC‐restricted molecule involved in routing of cell surface material in immature DC.


Journal of Immunology | 2002

Identification of Mouse Langerin/CD207 in Langerhans Cells and Some Dendritic Cells of Lymphoid Tissues

Jenny Valladeau; Valérie Clair-Moninot; Colette Dezutter-Dambuyant; Jean-Jacques Pin; Adrien Kissenpfennig; Marie-Geneviève Mattei; Smina Ait-Yahia; Elizabeth E. M. Bates; Bernard Malissen; Franz Koch; François Fossiez; Nikolaus Romani; Serge Lebecque; Sem Saeland

Human (h)Langerin/CD207 is a C-type lectin of Langerhans cells (LC) that induces the formation of Birbeck granules (BG). In this study, we have cloned a cDNA-encoding mouse (m)Langerin. The predicted protein is 66% homologous to hLangerin with conservation of its particular features. The organization of human and mouse Langerin genes are similar, consisting of six exons, three of which encode the carbohydrate recognition domain. The mLangerin gene maps to chromosome 6D, syntenic to the human gene on chromosome 2p13. mLangerin protein, detected by a mAb as a 48-kDa species, is abundant in epidermal LC in situ and is down-regulated upon culture. A subset of cells also expresses mLangerin in bone marrow cultures supplemented with TGF-β. Notably, dendritic cells in thymic medulla are mLangerin-positive. By contrast, only scattered cells express mLangerin in lymph nodes and spleen. mLangerin mRNA is also detected in some nonlymphoid tissues (e.g., lung, liver, and heart). Similarly to hLangerin, a network of BG form upon transfection of mLangerin cDNA into fibroblasts. Interestingly, substitution of a conserved residue (Phe244 to Leu) within the carbohydrate recognition domain transforms the BG in transfectant cells into structures resembling cored tubules, previously described in mouse LC. Our findings should facilitate further characterization of mouse LC, and provide insight into a plasticity of dendritic cell organelles which may have important functional consequences.


Journal of Leukocyte Biology | 1999

RESPECTIVE INVOLVEMENT OF TGF-BETA AND IL-4 IN THE DEVELOPMENT OF LANGERHANS CELLS AND NON-LANGERHANS DENDRITIC CELLS FROM CD34+ PROGENITORS

Christophe Caux; Catherine Massacrier; Bertrand Dubois; Jenny Valladeau; Colette Dezutter-Dambuyant; Isabelle Durand; Daniel Schmitt; Sem Saeland

In vivo, dendritic cells (DC) form a network comprising different populations. In particular, Langerhans cells (LC) appear as a unique population of cells dependent on transforming growth factor β (TGF‐β) for its development. In this study, we show that endogenous TGF‐β is required for the development of both LC and non‐LC DC from CD34+ hematopoietic progenitor cells (HPC) through induction of DC progenitor proliferation and of CD1a+ and CD14+ DC precursor differentiation. We further demonstrate that addition of exogenous TGF‐β polarized the differentiation of CD34+ HPC toward LC through induction of differentiation of CD14+ DC precursors into E‐cadherin+, Lag+CD68−, and Factor XIIIa−LC, displaying typical Birbeck granules. LC generated from CD34+ HPC in the presence of exogenous TGF‐β displayed overlapping functions with CD1a+ precursor‐derived DC. In particular, unlike CD14+‐derived DC obtained in the absence of TGF‐β, they neither secreted interleukin‐10 (IL‐10) on CD40 triggering nor stimulated the differentiation of CD40‐activated naive B cells. Finally, IL‐4, when combined with granulocyte‐macrophage colony‐stimulating factor (GM‐CSF), induced TGF‐β‐independent development of non‐LC DC from CD34+ HPC. Similarly, the development of DC from monocytes with GM‐CSF and IL‐4 was TGF‐β independent. Collectively these results show that TGF‐β polarized CD34+ HPC differentiation toward LC, whereas IL‐4 induced non‐LC DC development independently of TGF‐β. J. Leukoc. Biol. 66: 781–791; 1999.


Journal of Immunology | 2001

Immature Human Dendritic Cells Express Asialoglycoprotein Receptor Isoforms for Efficient Receptor-Mediated Endocytosis

Jenny Valladeau; Valérie Duvert-Frances; Jean-Jacques Pin; Monique J. Kleijmeer; Smina Ait-Yahia; Odile Ravel; Claude Vincent; Felix Vega; Alison Helms; Dan Gorman; Sandra Zurawski; Gerard Zurawski; John Ford; Sem Saeland

In a search for genes expressed by dendritic cells (DC), we have cloned cDNAs encoding different forms of an asialoglycoprotein receptor (ASGPR). The DC-ASGPR represents long and short isoforms of human macrophage lectin, a Ca2+-dependent type II transmembrane lectin displaying considerable homology with the H1 and H2 subunits of the hepatic ASGPR. Immunoprecipitation from DC using an anti-DC-ASGPR mAb yielded a major 40-kDa protein with an isoelectric point of 8.2. DC-ASGPR mRNA was observed predominantly in immune tissues. Both isoforms were detected in DC and granulocytes, but not in T, B, or NK cells, or monocytes. DC-ASGPR species were restricted to the CD14-derived DC obtained from CD34+ progenitors, while absent from the CD1a-derived subset. Accordingly, both monocyte-derived DC and tonsillar interstitial-type DC expressed DC-ASGPR protein, while Langerhans-type cells did not. Furthermore, DC-ASGPR is a feature of immaturity, as expression was lost upon CD40 activation. In agreement with the presence of tyrosine-based and dileucine motifs in the intracytoplasmic domain, mAb against DC-ASGPR was rapidly internalized by DC at 37°C. Finally, intracellular DC-ASGPR was localized to early endosomes, suggesting that the receptor recycles to the cell surface following internalization of ligand. Our findings identify DC-ASGPR/human macrophage lectin as a feature of immature DC, and as another lectin important for the specialized Ag-capture function of DC.


Molecular and Cellular Biology | 2005

Disruption of the langerin/CD207 Gene Abolishes Birbeck Granules without a Marked Loss of Langerhans Cell Function

Adrien Kissenpfennig; Smina Ait-Yahia; Valérie Clair-Moninot; Hella Stössel; Edgar Badell; Yann Bordat; Joanne Pooley; Thierry Lang; Eric Prina; Isabelle Coste; Olivia Gresser; Toufic Renno; Nathalie Winter; Geneviève Milon; Ken Shortman; Nikolaus Romani; Serge Lebecque; Bernard Malissen; Sem Saeland; Patrice Douillard

ABSTRACT Langerin is a C-type lectin expressed by a subset of dendritic leukocytes, the Langerhans cells (LC). Langerin is a cell surface receptor that induces the formation of an LC-specific organelle, the Birbeck granule (BG). We generated a langerin − / − mouse on a C57BL/6 background which did not display any macroscopic aberrant development. In the absence of langerin, LC were detected in normal numbers in the epidermis but the cells lacked BG. LC of langerin − / − mice did not present other phenotypic alterations compared to wild-type littermates. Functionally, the langerin − / − LC were able to capture antigen, to migrate towards skin draining lymph nodes, and to undergo phenotypic maturation. In addition, langerin − / − mice were not impaired in their capacity to process native OVA protein for I-A b -restricted presentation to CD4+ T lymphocytes or for H-2K b -restricted cross-presentation to CD8+ T lymphocytes. langerin − / − mice inoculated with mannosylated or skin-tropic microorganisms did not display an altered pathogen susceptibility. Finally, chemical mutagenesis resulted in a similar rate of skin tumor development in langerin − / − and wild-type mice. Overall, our data indicate that langerin and BG are dispensable for a number of LC functions. The langerin − / − C57BL/6 mouse should be a valuable model for further functional exploration of langerin and the role of BG.


Journal of Immunology | 2006

Lymph Node Resident Rather Than Skin-Derived Dendritic Cells Initiate Specific T Cell Responses after Leishmania major Infection

Giandomenica Iezzi; Anja Fröhlich; Bettina Ernst; Franziska Ampenberger; Sem Saeland; Nicolas Glaichenhaus; Manfred Kopf

Langerhans cells have been thought to play a major role as APCs for induction of specific immune responses to Leishmania major. Although their requirement for control of infection has been challenged recently, it remains unclear whether they can transport Ag to lymph nodes and promote initiation of T cell responses. Moreover, the role of dermal dendritic cells (DCs), another population of skin DCs, has so far not been addressed. We have investigated the origin and characterized the cell population responsible for initial activation of L. major-specific T cells in susceptible and resistant mice. We found that Ag presentation in draining lymph nodes peaks as early as 24 h after infection and is mainly mediated by a population of CD11chighCD11bhighGr-1−CD8−langerin− DCs residing in lymph nodes and acquiring soluble Ags possibly drained through the conduit network. In contrast, skin-derived DCs, including Langerhans cells and dermal DCs, migrated poorly to lymph nodes and played a minor role in early T cell activation. Furthermore, prevention of migration through early removal of the infection site did not affect Ag presentation by CD11chigh CD11bhigh DCs and activation of Leishmania major-specific naive CD4+ T cells in vivo.


Immunologic Research | 2003

Langerin/CD207 sheds light on formation of birbeck granules and their possible function in langerhans cells

Jenny Valladeau; Colette Dezutter-Dambuyant; Sem Saeland

Langerhans cells (LCs) are immature dendritic cells of epidermis and epithelia, playing a sentinel role through their specialized function in antigen capture, and their capacity to migrate to secondary lymphoid tissue to initiate specific immunity. A unique feature of LCs is the presence of Birbeck granules (BGs), which are disks of two limiting membranes, separated by leaflets with periodic “zipperlike” striations. The recent identification of Lagering/CD207 has allowed researchers to decipher the mechanism of BG formation and approach an understanding of their function. Langerin is a type II lelctin with mannose specificity expressed by LCs in epidermis and epithelia. Remarkably, transfection of Langerin cDNA into fibroblasts creates a dense network of membrane structures with features typical of BGs. Furthermore, mutated and deleted forms of Langerin have been engineered to map the functional domains essential for BG formation. Langerin is a potent LC-specific regulator of membrane superimposition and zippering, representing a key molecule to trace LCs and to probe BG function.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Vaginal epithelial dendritic cells renew from bone marrow precursors

Norifumi Iijima; Melissa M. Linehan; Sem Saeland; Akiko Iwasaki

Dendritic cells (DCs) represent key professional antigen-presenting cells capable of initiating primary immune responses. A specialized subset of DCs, the Langerhans cells (LCs), are located in the stratified squamous epithelial layer of the skin and within the mucosal epithelial lining of the vaginal and oral cavities. The vaginal mucosa undergoes cyclic changes under the control of sex hormones, and the renewal characteristics of the vaginal epithelial DCs (VEDCs) remain unknown. Here, we examined the origin of VEDCs. In contrast to the skin epidermal LCs, the DCs in the epithelium of the vagina were found to be repopulated mainly by nonmonocyte bone-marrow-derived precursors, with a half-life of 13 days under steady-state conditions. Upon infection with HSV-2, the Gr-1hi monocytes were found to give rise to VEDCs. Furthermore, flow cytometric analysis of the VEDCs revealed the presence of at least three distinct populations, namely, CD11b+F4/80hi, CD11b+F4/80int, and CD11b−F4/80−. Importantly, these VEDC populations expressed CD207 at low levels and had a constitutively more activated phenotype compared with the skin LCs. Collectively, our results revealed mucosa-specific features of the VEDCs with respect to their phenotype, activation status, and homeostatic renewal potential.

Collaboration


Dive into the Sem Saeland's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nikolaus Romani

Innsbruck Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Ford

Vanderbilt University

View shared research outputs
Researchain Logo
Decentralizing Knowledge