Network


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

Hotspot


Dive into the research topics where Thomas Calzascia is active.

Publication


Featured researches published by Thomas Calzascia.


Journal of Immunology | 2012

Different Adaptations of IgG Effector Function in Human and Nonhuman Primates and Implications for Therapeutic Antibody Treatment

Max Warncke; Thomas Calzascia; Michele Coulot; Nicole Balke; Ratiba Touil; Frank Kolbinger; Christoph Heusser

Safety of human therapeutic Abs is generally assessed in nonhuman primates. Whereas IgG1 shows identical FcγR interaction and effector function profile in both species, fundamental differences in the IgG2 and IgG4 Ab subclasses were found between the two species. Granulocytes, the main effector cells against IgG2- and IgG4-opsonized bacteria and parasites, do not express FcγRIIIb, but show higher levels of FcγRII in cynomolgus monkey. In humans, IgG2 and IgG4 adapted a silent Fc region with weak binding to FcγR and effector functions, whereas, in contrast, cynomolgus monkey IgG2 and IgG4 display strong effector function as well as differences in IgG4 Fab arm exchange. To balance this shift toward activation, the cynomolgus inhibitory FcγRIIb shows strongly increased affinity for IgG2. In view of these findings, in vitro and in vivo results for human IgG2 and IgG4 obtained in the cynomolgus monkey have to be cautiously interpreted, whereas effector function-related effects of human IgG1 Abs are expected to be predictable for humans.


Journal of Immunology | 2015

Deficiency of MALT1 Paracaspase Activity Results in Unbalanced Regulatory and Effector T and B Cell Responses Leading to Multiorgan Inflammation

Frédéric Bornancin; Florian Renner; Ratiba Touil; Heiko Sic; Yeter Kolb; Ismahane Touil-Allaoui; James Rush; Paul Smith; Marc Bigaud; Ursula Junker-Walker; Christoph Burkhart; Janet Dawson; Satoru Niwa; Andreas Katopodis; Barbara Nuesslein-Hildesheim; Gisbert Weckbecker; Gerhard Zenke; Bernd Kinzel; Elisabetta Traggiai; Dirk Brenner; Anne Brüstle; Michael St. Paul; Natasa Zamurovic; Kathleen McCoy; Antonius Rolink; Catherine H. Regnier; Tak W. Mak; Pamela S. Ohashi; Dhavalkumar D. Patel; Thomas Calzascia

The paracaspase MALT1 plays an important role in immune receptor-driven signaling pathways leading to NF-κB activation. MALT1 promotes signaling by acting as a scaffold, recruiting downstream signaling proteins, as well as by proteolytic cleavage of multiple substrates. However, the relative contributions of these two different activities to T and B cell function are not well understood. To investigate how MALT1 proteolytic activity contributes to overall immune cell regulation, we generated MALT1 protease-deficient mice (Malt1PD/PD) and compared their phenotype with that of MALT1 knockout animals (Malt1−/−). Malt1PD/PD mice displayed defects in multiple cell types including marginal zone B cells, B1 B cells, IL-10–producing B cells, regulatory T cells, and mature T and B cells. In general, immune defects were more pronounced in Malt1−/− animals. Both mouse lines showed abrogated B cell responses upon immunization with T-dependent and T-independent Ags. In vitro, inactivation of MALT1 protease activity caused reduced stimulation-induced T cell proliferation, impaired IL-2 and TNF-α production, as well as defective Th17 differentiation. Consequently, Malt1PD/PD mice were protected in a Th17-dependent experimental autoimmune encephalomyelitis model. Surprisingly, Malt1PD/PD animals developed a multiorgan inflammatory pathology, characterized by Th1 and Th2/0 responses and enhanced IgG1 and IgE levels, which was delayed by wild-type regulatory T cell reconstitution. We therefore propose that the pathology characterizing Malt1PD/PD animals arises from an immune imbalance featuring pathogenic Th1- and Th2/0-skewed effector responses and reduced immunosuppressive compartments. These data uncover a previously unappreciated key function of MALT1 protease activity in immune homeostasis and underline its relevance in human health and disease.


PLOS ONE | 2013

Partial Deficiency of Sphingosine-1-Phosphate Lyase Confers Protection in Experimental Autoimmune Encephalomyelitis

Andreas Billich; Thomas Baumruker; Christian Beerli; Marc Bigaud; Christian Bruns; Thomas Calzascia; Andrea Isken; Bernd Kinzel; Erika Loetscher; Barbara Metzler; Matthias Mueller; Barbara Nuesslein-Hildesheim; Bernadette Kleylein-Sohn

Background Sphingosine-1-phosphate (S1P) regulates the egress of T cells from lymphoid organs; levels of S1P in the tissues are controlled by S1P lyase (Sgpl1). Hence, Sgpl1 offers a target to block T cell-dependent inflammatory processes. However, the involvement of Sgpl1 in models of disease has not been fully elucidated yet, since Sgpl1 KO mice have a short life-span. Methodology We generated inducible Sgpl1 KO mice featuring partial reduction of Sgpl1 activity and analyzed them with respect to sphingolipid levels, T-cell distribution, and response in models of inflammation. Principal Findings The partially Sgpl1 deficient mice are viable but feature profound reduction of peripheral T cells, similar to the constitutive KO mice. While thymic T cell development in these mice appears normal, mature T cells are retained in thymus and lymph nodes, leading to reduced T cell numbers in spleen and blood, with a skewing towards increased proportions of memory T cells and T regulatory cells. The therapeutic relevance of Sgpl1 is demonstrated by the fact that the inducible KO mice are protected in experimental autoimmune encephalomyelitis (EAE). T cell immigration into the CNS was found to be profoundly reduced. Since S1P levels in the brain of the animals are unchanged, we conclude that protection in EAE is due to the peripheral effect on T cells, leading to reduced CNS immigration, rather than on local effects in the CNS. Significance The data suggest Sgpl1 as a novel therapeutic target for the treatment of multiple sclerosis.


Science Translational Medicine | 2016

Improved cancer immunotherapy by a CD25-mimobody conferring selectivity to human interleukin-2

Natalia Arenas-Ramirez; Chao Zou; Simone Popp; Daniel Zingg; Barbara Brannetti; Emmanuelle Wirth; Thomas Calzascia; Jiri Kovarik; Lukas Sommer; Gerhard Zenke; Janine Woytschak; Catherine H. Regnier; Andreas Katopodis; Onur Boyman

An antibody to human IL-2 phenocopies CD25 and improves IL-2–based cancer immunotherapy. Running interference Interleukin-2 (IL-2) binds to receptors on multiple different types of T cells. CD8 T cells, which can kill tumor cells, have IL-2 receptors with two subunits. When IL-2 binds to these, it promotes the T cells’ activation. In contrast, regulatory T cells dampen the antitumor immune response, and they express a different type of IL-2 receptor, which contains CD25 in addition to the other two subunits. CD25 binds IL-2 tightly but does not signal. To address this, Arenas-Ramirez et al. developed an anti–IL-2 antibody that can block CD25, such that delivering the antibody together with IL-2 allows IL-2 to bind specifically to the two-subunit IL-2 receptors and promote an antitumor immune response without interference from regulatory T cells. Interleukin-2 (IL-2) immunotherapy is an attractive approach in treating advanced cancer. However, by binding to its IL-2 receptor α (CD25) subunit, IL-2 exerts unwanted effects, including stimulation of immunosuppressive regulatory T cells (Tregs) and contribution to vascular leak syndrome. We used a rational approach to develop a monoclonal antibody to human IL-2, termed NARA1, which acts as a high-affinity CD25 mimic, thereby minimizing association of IL-2 with CD25. The structure of the IL-2–NARA1 complex revealed that NARA1 occupies the CD25 epitope of IL-2 and precisely overlaps with CD25. Association of NARA1 with IL-2 occurs with 10-fold higher affinity compared to CD25 and forms IL-2/NARA1 complexes, which, in vivo, preferentially stimulate CD8+ T cells while disfavoring CD25+ Tregs and improving the benefit–to–adverse effect ratio of IL-2. In two transplantable and one spontaneous metastatic melanoma model, IL-2/NARA1 complex immunotherapy resulted in efficient expansion of tumor-specific and polyclonal CD8+ T cells. These CD8+ T cells showed robust interferon-γ production and expressed low levels of exhaustion markers programmed cell death protein-1, lymphocyte activation gene-3, and T cell immunoglobulin and mucin domain-3. These effects resulted in potent anticancer immune responses and prolonged survival in the tumor models. Collectively, our data demonstrate that NARA1 acts as a CD25-mimobody that confers selectivity and increased potency to IL-2 and warrant further assessment of NARA1 as a therapeutic.


Immunology and Cell Biology | 2018

The T-cell fingerprint of MALT1 paracaspase revealed by selective inhibition

Maureen Bardet; Adeline Unterreiner; Claire Malinverni; Frédérique Lafossas; Corinne Vedrine; Danielle Boesch; Yeter Kolb; Daniel Kaiser; Anton Glück; Martin A. Schneider; Andreas Katopodis; Martin Renatus; Oliver Simic; Achim Schlapbach; Jean Quancard; Catherine H. Regnier; Guido Bold; Carole Pissot-Soldermann; José M. Carballido; Jiri Kovarik; Thomas Calzascia; Frédéric Bornancin

Mucosa‐associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is essential for immune responses triggered by antigen receptors but the contribution of its paracaspase activity is not fully understood. Here, we studied how MALT1 proteolytic function regulates T‐cell activation and fate after engagement of the T‐cell receptor pathway. We show that MLT‐827, a potent and selective MALT1 paracaspase inhibitor, does not prevent the initial phase of T‐cell activation, in contrast to the pan‐protein kinase C inhibitor AEB071. However, MLT‐827 strongly impacted cell expansion after activation. We demonstrate this is the consequence of profound inhibition of IL‐2 production as well as reduced expression of the IL‐2 receptor alpha subunit (CD25), resulting from defective canonical NF‐κB activation and accelerated mRNA turnover mechanisms. Accordingly, MLT‐827 revealed a unique transcriptional fingerprint of MALT1 protease activity, providing evidence for broad control of T‐cell signaling pathways. Altogether, this first report with a potent and selective inhibitor elucidates how MALT1 paracaspase activity integrates several T‐cell activation pathways and indirectly controls gamma‐chain receptor dependent survival, to impact on T‐cell expansion.


PLOS ONE | 2017

Two Antagonistic MALT1 Auto-Cleavage Mechanisms Reveal a Role for TRAF6 to Unleash MALT1 Activation

Stefanie Ginster; Maureen Bardet; Adeline Unterreiner; Claire Malinverni; Florian Renner; Stephen Lam; Felix Freuler; Bertran Gerrits; Johannes Voshol; Thomas Calzascia; Catherine H. Regnier; Martin Renatus; Rainer Nikolay; Laura Israël; Frédéric Bornancin

The paracaspase MALT1 has arginine-directed proteolytic activity triggered by engagement of immune receptors. Recruitment of MALT1 into activation complexes is required for MALT1 proteolytic function. Here, co-expression of MALT1 in HEK293 cells, either with activated CARD11 and BCL10 or with TRAF6, was used to explore the mechanism of MALT1 activation at the molecular level. This work identified a prominent self-cleavage site of MALT1 isoform A (MALT1A) at R781 (R770 in MALT1B) and revealed that TRAF6 can activate MALT1 independently of the CBM. Intramolecular cleavage at R781/R770 removes a C-terminal TRAF6-binding site in both MALT1 isoforms, leaving MALT1B devoid of the two key interaction sites with TRAF6. A previously identified auto-proteolysis site of MALT1 at R149 leads to deletion of the death-domain, thereby abolishing interaction with BCL10. By using MALT1 isoforms and cleaved fragments thereof, as well as TRAF6 WT and mutant forms, this work shows that TRAF6 induces N-terminal auto-proteolytic cleavage of MALT1 at R149 and accelerates MALT1 protein turnover. The MALT1 fragment generated by N-terminal self-cleavage at R149 was labile and displayed enhanced signaling properties that required an intact K644 residue, previously shown to be a site for mono-ubiquitination of MALT1. Conversely, C-terminal self-cleavage at R781/R770 hampered the ability for self-cleavage at R149 and stabilized MALT1 by hindering interaction with TRAF6. C-terminal self-cleavage had limited impact on MALT1A but severely reduced MALT1B proteolytic and signaling functions. It also abrogated NF-κB activation by N-terminally cleaved MALT1A. Altogether, this study provides further insights into mechanisms that regulate the scaffolding and activation cycle of MALT1. It also emphasizes the reduced functional capacity of MALT1B as compared to MALT1A.


Transplant Immunology | 2012

Vav1 GEF activity is required for T cell mediated allograft rejection.

Dirk Haubert; Jianping Li; Alexander Saveliev; Thomas Calzascia; Esther Sutter; Barbara Metzler; Daniel Kaiser; Victor L. J. Tybulewicz; Gisbert Weckbecker

The GDP exchange factor (GEF) Vav1 is a central signal transducer downstream of the T cell receptor and has been identified as a key factor for T cell activation in the context of allograft rejection. Vav1 has been shown to transduce signals both dependent and independent of its GEF function. The most promising approach to disrupt Vav1 activity by pharmacological inhibition would be to target its GEF function. However, the contribution of Vav1 GEF activity for allogeneic T cell activation has not been clarified yet. To address this question, we used knock-in mice bearing a mutated Vav1 with disrupted GEF activity but intact GEF-independent functions. T cells from these mice showed strongly reduced proliferation and activation in response to allogeneic stimulation. Furthermore, lack of Vav1 GEF activity strongly abrogated the in vivo expansion of T cells in a systemic graft-versus-host model. In a cardiac transplantation model, mice with disrupted Vav1 GEF activity show prolonged allograft survival. These findings demonstrate a strong requirement for Vav1 GEF activity for allogeneic T cell activation and graft rejection suggesting that disruption of Vav1 GEF activity alone is sufficient to induce significant immunosuppression.


Molecular Immunology | 2015

Highly reduced binding to high and low affinity mouse Fc gamma receptors by L234A/L235A and N297A Fc mutations engineered into mouse IgG2a

Elodie Arduin; Seguinde Arora; Paulo Roky Bamert; Torsten Kuiper; Simone Popp; Sabine Geisse; Roger Grau; Thomas Calzascia; Gerhard Zenke; Jiri Kovarik


Immunology Letters | 2017

Selective MALT1 paracaspase inhibition does not block TNF-α production downstream of TLR4 in myeloid cells

Adeline Unterreiner; Natacha Stoehr; Christine Huppertz; Thomas Calzascia; Christopher J. Farady; Frédéric Bornancin


Archive | 2017

ANTICUERPOS DIRIGIDOS A CD32B Y MÉTODOS DE USO DE LOS MISMOS

Ewert Stefan; Balke Nicole; Thomas Calzascia; Harris Alan; Heather Huet; Isnardi Isabelle; Lu Haihui; Wilson Nicholas; Xu Fangmin; Matthew John Meyer

Collaboration


Dive into the Thomas Calzascia's collaboration.

Researchain Logo
Decentralizing Knowledge