Network


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

Hotspot


Dive into the research topics where Leonardo Scarpellino is active.

Publication


Featured researches published by Leonardo Scarpellino.


Nature Immunology | 2004

Cis association of Ly49A with MHC class I restricts natural killer cell inhibition

Marie-Agnès Doucey; Leonardo Scarpellino; Jacques Zimmer; Philippe Guillaume; Immanuel F. Luescher; Claude Bron; Werner Held

Natural killer (NK) cell function is negatively regulated by inhibitory receptors interacting with major histocompatibility complex class I molecules expressed on target cells. Here we show that the inhibitory Ly49A NK cell receptor not only binds to its H-2Dd ligand expressed on potential target cells (in trans) but also is constitutively associated with H-2Dd in cis (on the same cell). Cis association and trans interaction occur through the same binding site. Consequently, cis association restricts the number of Ly49A receptors available for binding of H-2Dd on target cells and reduces NK cell inhibition through Ly49A. By lowering the threshold at which NK cell activation exceeds NK cell inhibition, cis interaction allows optimal discrimination of normal and abnormal host cells.


Immunity | 2009

A Role for cis Interaction between the Inhibitory Ly49A Receptor and MHC Class I for Natural Killer Cell Education

Anick Chalifour; Leonardo Scarpellino; Jonathan Back; Petter Brodin; Estelle Devevre; Frédéric Gros; Frédéric Lévy; Georges Leclercq; Petter Höglund; Friedrich Beermann; Werner Held

Natural killer (NK) cells show enhanced functional competence when they express inhibitory receptors specific for inherited major histocompatibility complex class I (MHC-I) molecules. Current models imply that NK cell education requires an interaction of inhibitory receptors with MHC-I expressed on other cells. However, the inhibitory Ly49A receptor can also bind MHC-I ligand on the NK cell itself (in cis). Here we describe a Ly49A variant, which can engage MHC-I expressed on other cells but not in cis. Even though this variant inhibited NK cell effector function, it failed to educate NK cells. The association with MHC-I in cis sequestered wild-type Ly49A, and this was found to relieve NK cells from a suppressive effect of unengaged Ly49A. These data explain how inhibitory MHC-I receptors can facilitate NK cell activation. They dissociate classical inhibitory from educating functions of Ly49A and suggest that cis interaction of Ly49A is necessary for NK cell education.


Journal of Immunology | 2007

Interactions of Ly49 family receptors with MHC class I ligands in trans and cis

Leonardo Scarpellino; Franziska Oeschger; Philippe Guillaume; Jérôme D. Coudert; Frédéric Lévy; Georges Leclercq; Werner Held

The Ly49A NK cell receptor interacts with MHC class I (MHC-I) molecules on target cells and negatively regulates NK cell-mediated target cell lysis. We have recently shown that the MHC-I ligand-binding capacity of the Ly49A NK cell receptor is controlled by the NK cells’ own MHC-I. To see whether this property was unique to Ly49A, we have investigated the binding of soluble MHC-I multimers to the Ly49 family receptors expressed in MHC-I-deficient and -sufficient C57BL/6 mice. In this study, we confirm the binding of classical MHC-I to the inhibitory Ly49A, C and I receptors, and demonstrate that detectable MHC-I binding to MHC-I-deficient NK cells is exclusively mediated by these three receptors. We did not detect significant multimer binding to stably transfected or NK cell-expressed Ly49D, E, F, G, and H receptors. Yet, we identified the more distantly related Ly49B and Ly49Q, which are not expressed by NK cells, as two novel MHC-I receptors in mice. Furthermore, we show using MHC-I-sufficient mice that the NK cells’ own MHC-I significantly masks the Ly49A and Ly49C, but not the Ly49I receptor. Nevertheless, Ly49I was partly masked on transfected tumor cells, suggesting that the structure of Ly49I is compatible in principal with cis binding of MHC-I. Finally, masking of Ly49Q by cis MHC-I was minor, whereas masking of Ly49B was not detected. These data significantly extend the MHC-I specificity of Ly49 family receptors and show that the accessibility of most, but not all, MHC-I-binding Ly49 receptors is modulated by the expression of MHC-I in cis.


PLOS ONE | 2011

Fibroblastic Reticular Cells From Lymph Nodes Attenuate T Cell Expansion by Producing Nitric Oxide

Stefanie Siegert; Hsin-Ying Huang; Chen-Ying Yang; Leonardo Scarpellino; Lucie Carrié; Sarah Essex; Peter J. Nelson; Matthias Heikenwalder; Hans Acha-Orbea; Christopher D. Buckley; Benjamin J. Marsland; Dietmar Zehn; Sanjiv A. Luther

Adaptive immune responses are initiated when T cells encounter antigen on dendritic cells (DC) in T zones of secondary lymphoid organs. T zones contain a 3-dimensional scaffold of fibroblastic reticular cells (FRC) but currently it is unclear how FRC influence T cell activation. Here we report that FRC lines and ex vivo FRC inhibit T cell proliferation but not differentiation. FRC share this feature with fibroblasts from non-lymphoid tissues as well as mesenchymal stromal cells. We identified FRC as strong source of nitric oxide (NO) thereby directly dampening T cell expansion as well as reducing the T cell priming capacity of DC. The expression of inducible nitric oxide synthase (iNOS) was up-regulated in a subset of FRC by both DC-signals as well as interferon-γ produced by primed CD8+ T cells. Importantly, iNOS expression was induced during viral infection in vivo in both LN FRC and DC. As a consequence, the primary T cell response was found to be exaggerated in Inos −/− mice. Our findings highlight that in addition to their established positive roles in T cell responses FRC and DC cooperate in a negative feedback loop to attenuate T cell expansion during acute inflammation.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Trapping of naive lymphocytes triggers rapid growth and remodeling of the fibroblast network in reactive murine lymph nodes

Chen-Ying Yang; Tobias K. Vogt; Stéphanie Favre; Leonardo Scarpellino; Hsin-Ying Huang; Fabienne Tacchini-Cottier; Sanjiv A. Luther

Significance Lymph node swelling is a hallmark of adaptive immunity. Fibroblastic reticular cells form a fairly rigid scaffold throughout lymph nodes. They not only support organ structure and compartmentalization, but also guide lymphocyte trafficking. We describe how this rigid fibroblast network reacts to acute organ swelling. Rather than being disrupted or destroyed, the fibroblast network rapidly expands by proliferation and finally covers a much larger volume to accommodate many more lymphocytes. We identified naive lymphocyte trapping by innate triggers as an early fibroblast growth signal, with activated lymphocytes playing a role only in the later growth phase. Adaptive immunity is initiated in T-cell zones of secondary lymphoid organs. These zones are organized in a rigid 3D network of fibroblastic reticular cells (FRCs) that are a rich cytokine source. In response to lymph-borne antigens, draining lymph nodes (LNs) expand several folds in size, but the fate and role of the FRC network during immune response is not fully understood. Here we show that T-cell responses are accompanied by the rapid activation and growth of FRCs, leading to an expanded but similarly organized network of T-zone FRCs that maintains its vital function for lymphocyte trafficking and survival. In addition, new FRC-rich environments were observed in the expanded medullary cords. FRCs are activated within hours after the onset of inflammation in the periphery. Surprisingly, FRC expansion depends mainly on trapping of naïve lymphocytes that is induced by both migratory and resident dendritic cells. Inflammatory signals are not required as homeostatic T-cell proliferation was sufficient to trigger FRC expansion. Activated lymphocytes are also dispensable for this process, but can enhance the later growth phase. Thus, this study documents the surprising plasticity as well as the complex regulation of FRC networks allowing the rapid LN hyperplasia that is critical for mounting efficient adaptive immunity.


Immunity | 2009

Distinct Conformations of Ly49 Natural Killer Cell Receptors Mediate MHC Class I Recognition in Trans and Cis

Jonathan Back; Emilio L. Malchiodi; Sangwoo Cho; Leonardo Scarpellino; Pascal Schneider; Melissa C. Kerzic; Roy A. Mariuzza; Werner Held

Certain cell-surface receptors engage ligands expressed on juxtaposed cells and ligands on the same cell. The structural basis for trans versus cis binding is not known. Here, we showed that Ly49 natural killer (NK) cell receptors bound two MHC class I (MHC-I) molecules in trans when the two ligand-binding domains were backfolded onto the long stalk region. In contrast, dissociation of the ligand-binding domains from the stalk and their reorientation relative to the NK cell membrane allowed monovalent binding of MHC-I in cis. The distinct conformations (backfolded and extended) define the structural basis for cis-trans binding by Ly49 receptors and explain the divergent functional consequences of cis versus trans interactions. Further analyses identified specific stalk segments that were not required for MHC-I binding in trans but were essential for inhibitory receptor function. These data identify multiple distinct roles of stalk regions for receptor function.


Immunological Reviews | 1999

Interplays between mouse mammary tumor virus and the cellular and humoral immune response

Hans Acha-Orbea; Daniela Finke; Antoine Attinger; Sandie Schmid; Nathalie Wehrli; Sonia Vacheron; Ioannls Xenarios; Leonardo Scarpellino; Kai-Michael Toellner; Ian C. M. MacLennan; Sanjiv A. Luther

Summary: Mouse mammary tumor virus has developed strategies to exploit the immune response. It requires vigorous immune stimulation to achieve efficient infection. The infected antigen‐presenting cells present a viral superantigen on the cell surface which stimulates strong CD4‐mediated T‐cell help but CDS T‐cell responses are undetectable. Despite the high frequency of superantigen‐reactive T cells, the superantigen‐induced immune response is comparable to classical antigen responses in terms of T‐cell priming, T‐cell—B‐cell collaboration as well as follicular and extra‐follicular B‐cell differentiation. Induction of systemic anergy is observed, similar to classical antigen responses wbere antigen is administered systemically but does not influence the role of the superantigen‐reactive T cells in the maintenance of the chronic germinal center reaction. So far we have been unable to detect a cytotoxic T‐cell response to mouse mammary tumor virus peptide antigens or to the superantigen. This might yet represent another step in the viral infection strategy.


Journal of Immunology | 2003

Deregulated MHC class II transactivator expression leads to a strong Th2 bias in CD4+ T lymphocytes

Luc A. Otten; Fabienne Tacchini-Cottier; Michael Lohoff; Francesco Annunziato; Lorenzo Cosmi; Leonardo Scarpellino; Jacques A. Louis; Viktor Steimle; Walter Reith; Hans Acha-Orbea

The MHC class II (MHC-II) transactivator (CIITA) is the master transcriptional regulator of genes involved in MHC-II-restricted Ag presentation. Fine tuning of CIITA gene expression determines the cell type-specific expression of MHC-II genes. This regulation is achieved by the selective usage of multiple CIITA promoters. It has recently been suggested that CIITA also contributes to Th cell differentiation by suppressing IL-4 expression in Th1 cells. In this study, we show that endogenous CIITA is expressed at low levels in activated mouse T cells. Importantly CIITA is not regulated differentially in murine and human Th1 and Th2 cells. Ectopic expression of a CIITA transgene in multiple mouse cell types including T cells, does not interfere with normal development of CD4+ T cells. However, upon TCR activation the CIITA transgenic CD4+ T cells preferentially differentiate into IL-4-secreting Th2-type cells. These results imply that CIITA is not a direct Th1-specific repressor of the IL-4 gene and that tight control over the expression of CIITA and MHC-II is required to maintain the normal balance between Th1 and Th2 responses.


Immunogenetics | 1991

Nonobese diabetic and nonobese nondiabetic mice have unique MHC class II haplotypes

Hans Acha-Orbea; Leonardo Scarpellino

The highly polymorphic major histocompatibility complex (MHC) class II molecules function as regulators of the immune response. Recent studies have shown these molecules bind peptide fragments of self and foreign proteins. These complexes in turn are recognized by the highly specific T-cell receptors expressed by T lymphocytes. The polymorphic residues of these heterodimeric glycoproteins are responsible for the peptide fine specificity of MHC molecules. Over 50 A and E alleles have been defined in the mouse by serology and eDNA analysis. Nonobese diabetic (NOD) and nonobese nondiabetic (NON) mouse strains were derived from outbred JCLICR mice. During this breeding a diabetic animal was detected and the three inbred lines NOD, NON, and CTS were derived. NOD and NON lines were separated after the 6th brother sister mating and inbred as separate lines (Makino et al. 1980). NOD mice, contrary to NON mice, develop spontaneous insulin-dependent diabetes mellitus (FDDM). Serological studies indicated that NOD mice express the MHC haplotype: Ka-AN°O-E--Dband NON mice I(°-AN°LE+-I~. (Hattori et al. 1986; Acha-Orbea and McDevitt 1987; Ikegami et al. 1988; Todd et al. 1988). Although it was shown that NON mice express E antigens, the E haplotype of NON mice was not known, eDNA se-


Journal of Clinical Investigation | 2017

Fibroblastic niches prime T cell alloimmunity through Delta-like Notch ligands

Jooho Chung; Christen L. Ebens; Eric Perkey; Vedran Radojcic; Ute Koch; Leonardo Scarpellino; Alexander Tong; Frederick Allen; Sherri C. Wood; Jiane Feng; Ann Friedman; David Granadier; Ivy T. Tran; Qian Chai; Lucas Onder; Minhong Yan; Pavan Reddy; Bruce R. Blazar; Alex Y. Huang; Todd V. Brennan; D. Keith Bishop; Burkhard Ludewig; Christian W. Siebel; Freddy Radtke; Sanjiv A. Luther; Ivan Maillard

Alloimmune T cell responses induce graft-versus-host disease (GVHD), a serious complication of allogeneic bone marrow transplantation (allo-BMT). Although Notch signaling mediated by Delta-like 1/4 (DLL1/4) Notch ligands has emerged as a major regulator of GVHD pathogenesis, little is known about the timing of essential Notch signals and the cellular source of Notch ligands after allo-BMT. Here, we have shown that critical DLL1/4-mediated Notch signals are delivered to donor T cells during a short 48-hour window after transplantation in a mouse allo-BMT model. Stromal, but not hematopoietic, cells were the essential source of Notch ligands during in vivo priming of alloreactive T cells. GVHD could be prevented by selective inactivation of Dll1 and Dll4 in subsets of fibroblastic stromal cells that were derived from chemokine Ccl19-expressing host cells, including fibroblastic reticular cells and follicular dendritic cells. However, neither T cell recruitment into secondary lymphoid organs nor initial T cell activation was affected by Dll1/4 loss. Thus, we have uncovered a pathogenic function for fibroblastic stromal cells in alloimmune reactivity that can be dissociated from their homeostatic functions. Our results reveal what we believe to be a previously unrecognized Notch-mediated immunopathogenic role for stromal cell niches in secondary lymphoid organs after allo-BMT and define a framework of early cellular and molecular interactions that regulate T cell alloimmunity.

Collaboration


Dive into the Leonardo Scarpellino's collaboration.

Top Co-Authors

Avatar

Werner Held

University of Lausanne

View shared research outputs
Top Co-Authors

Avatar

Hans Acha-Orbea

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander N. Shakhov

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jonathan Back

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Gary A. Waanders

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jérôme D. Coudert

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge