Network


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

Hotspot


Dive into the research topics where Martin Vaeth is active.

Publication


Featured researches published by Martin Vaeth.


Journal of Experimental Medicine | 2014

Follicular regulatory T cells control humoral autoimmunity via NFAT2-regulated CXCR5 expression

Martin Vaeth; Gerd Müller; Dennis Stauss; Lena Dietz; Stefan Klein-Hessling; Edgar Serfling; Martin Lipp; Ingolf Berberich; Friederike Berberich-Siebelt

T cell–specific NFAT2 deletion results in reduced CXCR5+ follicular regulatory T cells, leading to uncontrolled germinal center responses and humoral autoimmunity.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Dependence on nuclear factor of activated T-cells (NFAT) levels discriminates conventional T cells from Foxp3+ regulatory T cells.

Martin Vaeth; Ulrike Schliesser; Gerd Müller; Sonja Reissig; Kazuki Satoh; Helmut Jonuleit; Ari Waisman; Martin R. Müller; Edgar Serfling; Birgit Sawitzki; Friederike Berberich-Siebelt

Several lines of evidence suggest nuclear factor of activated T-cells (NFAT) to control regulatory T cells: thymus-derived naturally occurring regulatory T cells (nTreg) depend on calcium signals, the Foxp3 gene harbors several NFAT binding sites, and the Foxp3 (Fork head box P3) protein interacts with NFAT. Therefore, we investigated the impact of NFAT on Foxp3 expression. Indeed, the generation of peripherally induced Treg (iTreg) by TGF-β was highly dependent on NFAT expression because the ability of CD4+ T cells to differentiate into iTreg diminished markedly with the number of NFAT family members missing. It can be concluded that the expression of Foxp3 in TGF-β–induced iTreg depends on the threshold value of NFAT rather than on an individual member present. This is specific for iTreg development, because frequency of nTreg remained unaltered in mice lacking NFAT1, NFAT2, or NFAT4 alone or in combination. Different from expectation, however, the function of both nTreg and iTreg was independent on robust NFAT levels, reflected by less nuclear NFAT in nTreg and iTreg. Accordingly, absence of one or two NFAT members did not alter suppressor activity in vitro or during colitis and transplantation in vivo. This scenario emphasizes an inhibition of high NFAT activity as treatment for autoimmune diseases and in transplantation, selectively targeting the proinflammatory conventional T cells, while keeping Treg functional.


Journal of Experimental Medicine | 2011

NFATc1 affects mouse splenic B cell function by controlling the calcineurin--NFAT signaling network.

Sankar Bhattacharyya; Jolly Deb; Amiya K. Patra; Duong Anh Thuy Pham; Wen Chen; Martin Vaeth; Friederike Berberich-Siebelt; Stefan Klein-Hessling; Edward D. Lamperti; Kurt Reifenberg; Julia Jellusova; Astrid Schweizer; Lars Nitschke; Ellen Leich; Andreas Rosenwald; Cornelia Brunner; Swen Engelmann; Ursula Bommhardt; Andris Avots; Martin Müller; Eisaku Kondo; Edgar Serfling

Mouse B cells lacking NFATc1 exhibit defective proliferation, survival, isotype class switching, cytokine production, and T cell help.


Journal of Biological Chemistry | 2009

Sumoylation of the Transcription Factor NFATc1 Leads to Its Subnuclear Relocalization and Interleukin-2 Repression by Histone Deacetylase

Arnab Nayak; Judith Glöckner-Pagel; Martin Vaeth; Julia Schumann; Mathias Buttmann; Tobias Bopp; Edgar Schmitt; Edgar Serfling; Friederike Berberich-Siebelt

The family of NFAT (nuclear factor of activated T-cells) transcription factors plays an important role in cytokine gene regulation. In peripheral T-cells NFATc1 and -c2 are predominantly expressed. Because of different promoter and poly(A) site usage as well as alternative splicing events, NFATc1 is synthesized in multiple isoforms. The highly inducible NFATc1/A contains a relatively short C terminus, whereas the longer, constitutively expressed isoform NFATc1/C spans an extra C-terminal peptide of 246 amino acids. Interestingly, this NFATc1/C-specific terminus can be highly sumoylated. Upon sumoylation, NFATc1/C, but not the unsumoylated NFATc1/A, translocates to promyelocytic leukemia nuclear bodies. This leads to interaction with histone deacetylases followed by deacetylation of histones, which in turn induces transcriptionally inactive chromatin. As a consequence, expression of the NFATc1 target gene interleukin-2 is suppressed. These findings demonstrate that the modification by SUMO (small ubiquitin-like modifier) converts NFATc1 from an activator to a site-specific transcriptional repressor, revealing a novel regulatory mechanism for NFATc1 function.


European Journal of Immunology | 2012

Cyclic AMP underpins suppression by regulatory T cells

Josef Bodor; Tobias Bopp; Martin Vaeth; Matthias Klein; Edgar Serfling; Thomas Hünig; Christian Becker; Hansjörg Schild; Edgar Schmitt

Elevated levels of intracellular cyclic adenosine monophosphate (cAMP) in naturally occurring T regulatory (nTreg) cells play a key role in nTreg‐cell‐mediated suppression. Upon contact with nTreg cells, cAMP is transferred from nTreg cells into activated target CD4+ T cells and/or antigen‐presenting cells (APCs) via gap junctions to suppress CD4+ T‐cell function. cAMP facilitates the expression and nuclear function of a potent transcriptional inhibitor, inducible cAMP early repressor (ICER), resulting in ICER‐mediated suppression of interleukin‐2 (IL‐2). Furthermore, ICER inhibits transcription of nuclear factor of activated T cell c1/α (NFATc1/α) and forms inhibitory complexes with preexisting NFATc1/c2, thereby inhibiting NFAT‐driven transcription, including that of IL‐2. In addition to its suppressive effects mediated via ICER, cAMP can also modulate the levels of surface‐expressed cytotoxic T lymphocyte antigen‐4 (CTLA‐4) and its cognate B7 ligands on conventional CD4+ T cells and/or APCs, fine‐tuning suppression. These cAMP‐driven nTreg‐cell suppression mechanisms are the focus of this review.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Regulatory T cells facilitate the nuclear accumulation of inducible cAMP early repressor (ICER) and suppress nuclear factor of activated T cell c1 (NFATc1)

Martin Vaeth; Tea Gogishvili; Tobias Bopp; Matthias Klein; Friederike Berberich-Siebelt; Stefan Gattenloehner; Andris Avots; Tim Sparwasser; Nadine Grebe; Edgar Schmitt; Thomas Hünig; Edgar Serfling; Josef Bodor

Inducible cAMP early repressor (ICER) is a transcriptional repressor, which, because of alternate promoter use, is generated from the 3′ region of the cAMP response modulator (Crem) gene. Its expression and nuclear occurrence are elevated by high cAMP levels in naturally occurring regulatory T cells (nTregs). Using two mouse models, we demonstrate that nTregs control the cellular localization of ICER/CREM, and thereby inhibit IL-2 synthesis in conventional CD4+ T cells. Ablation of nTregs in depletion of regulatory T-cell (DEREG) mice resulted in cytosolic localization of ICER/CREM and increased IL-2 synthesis upon stimulation. Direct contacts between nTregs and conventional CD4+ T cells led to nuclear accumulation of ICER/CREM and suppression of IL-2 synthesis on administration of CD28 superagonistic (CD28SA) Ab. In a similar way, nTregs communicated with B cells and induced the cAMP-driven nuclear localization of ICER/CREM. High levels of ICER suppressed the induction of nuclear factor of activated T cell c1 (Nfatc1) gene in T cells whose inducible Nfatc1 P1 promoter bears two highly conserved cAMP-responsive elements to which ICER/CREM can bind. These findings suggest that nTregs suppress T-cell responses by the cAMP-dependent nuclear accumulation of ICER/CREM and inhibition of NFATc1 and IL-2 induction.


Journal of Experimental Medicine | 2016

Exogenous TNFR2 activation protects from acute GvHD via host T reg cell expansion.

Martin Chopra; Marlene Biehl; Tim Steinfatt; Andreas Brandl; Juliane Kums; Jorge Amich; Martin Vaeth; Janina Kuen; Rafaela Holtappels; Jürgen Podlech; Anja Mottok; Sabrina Kraus; Ana-Laura Jordán-Garrote; Carina A. Bäuerlein; Christian Brede; Eliana Ribechini; Andrea Fick; Axel Seher; Johannes Polz; Katja J. Ottmüller; Jeanette Baker; Hidekazu Nishikii; Miriam Ritz; Katharina Mattenheimer; Stefanie Schwinn; Thorsten Winter; Viktoria Schäfer; Sven Krappmann; Hermann Einsele; Thomas Müller

Activation of TNFR2 with a novel agonist expands T reg cells in vivo and protects allo-HCT recipients from acute GvHD while sparing antilymphoma and antiinfectious properties of transplanted donor T cells.


Cell Communication and Signaling | 2012

NFATc1/αA: The other Face of NFAT Factors in Lymphocytes

Edgar Serfling; Andris Avots; Stefan Klein-Hessling; Ronald Rudolf; Martin Vaeth; Friederike Berberich-Siebelt

In effector T and B cells immune receptor signals induce within minutes a rise of intracellular Ca++, the activation of the phosphatase calcineurin and the translocation of NFAT transcription factors from cytosol to nucleus. In addition to this first wave of NFAT activation, in a second step the occurrence of NFATc1/αA, a short isoform of NFATc1, is strongly induced. Upon primary stimulation of lymphocytes the induction of NFATc1/αA takes place during the G1 phase of cell cycle. Due to an auto-regulatory feedback circuit high levels of NFATc1/αA are kept constant during persistent immune receptor stimulation. Contrary to NFATc2 and further NFATc proteins which dampen lymphocyte proliferation, induce anergy and enhance activation induced cell death (AICD), NFATc1/αA supports antigen-mediated proliferation and protects lymphocytes against rapid AICD. Whereas high concentrations of NFATc1/αA can also lead to apoptosis, in collaboration with NF-κB-inducing co-stimulatory signals they support the survival of mature lymphocytes in late phases after their activation. However, if dysregulated, NFATc1/αA appears to contribute to lymphoma genesis and – as we assume – to further disorders of the lymphoid system. While the molecular details of NFATc1/αA action and its contribution to lymphoid disorders have to be investigated, NFATc1/αA differs in its generation and function markedly from all the other NFAT proteins which are expressed in lymphoid cells. Therefore, it represents a prime target for causal therapies of immune disorders in future.


Journal of Immunology | 2015

Ca2+ Signaling but Not Store-Operated Ca2+ Entry Is Required for the Function of Macrophages and Dendritic Cells

Martin Vaeth; Isabelle Zee; Axel R. Concepcion; Mate Maus; Patrick J. Shaw; Cynthia Portal-Celhay; Aleena Zahra; Lina Kozhaya; Carl Weidinger; Jennifer A. Philips; Derya Unutmaz; Stefan Feske

Store-operated Ca2+ entry (SOCE) through Ca2+ release–activated Ca2+ (CRAC) channels is essential for immunity to infection. CRAC channels are formed by ORAI1 proteins in the plasma membrane and activated by stromal interaction molecule (STIM)1 and STIM2 in the endoplasmic reticulum. Mutations in ORAI1 and STIM1 genes that abolish SOCE cause severe immunodeficiency with recurrent infections due to impaired T cell function. SOCE has also been observed in cells of the innate immune system such as macrophages and dendritic cells (DCs) and may provide Ca2+ signals required for their function. The specific role of SOCE in macrophage and DC function, as well as its contribution to innate immunity, however, is not well defined. We found that nonselective inhibition of Ca2+ signaling strongly impairs many effector functions of bone marrow–derived macrophages and bone marrow–derived DCs, including phagocytosis, inflammasome activation, and priming of T cells. Surprisingly, however, macrophages and DCs from mice with conditional deletion of Stim1 and Stim2 genes, and therefore complete inhibition of SOCE, showed no major functional defects. Their differentiation, FcR-dependent and -independent phagocytosis, phagolysosome fusion, cytokine production, NLRP3 inflammasome activation, and their ability to present Ags to activate T cells were preserved. Our findings demonstrate that STIM1, STIM2, and SOCE are dispensable for many critical effector functions of macrophages and DCs, which has important implications for CRAC channel inhibition as a therapeutic strategy to suppress pathogenic T cells while not interfering with myeloid cell functions required for innate immunity.


Journal of Immunology | 2012

Repression of Cyclic Adenosine Monophosphate Upregulation Disarms and Expands Human Regulatory T Cells

Matthias Klein; Martin Vaeth; Tobias Scheel; Stephan Grabbe; Ria Baumgrass; Friederike Berberich-Siebelt; Tobias Bopp; Edgar Schmitt; Christian Becker

The main molecular mechanism of human regulatory T cell (Treg)-mediated suppression has not been elucidated. We show in this study that cAMP represents a key regulator of human Treg function. Repression of cAMP production by inhibition of adenylate cyclase activity or augmentation of cAMP degradation through ectopic expression of a cAMP-degrading phosphodiesterase greatly reduces the suppressive activity of human Treg in vitro and in a humanized mouse model in vivo. Notably, cAMP repression additionally abrogates the anergic state of human Treg, accompanied by nuclear translocation of NFATc1 and induction of its short isoform NFATc1/αA. Treg expanded under cAMP repression, however, do not convert into effector T cells and regain their anergic state and suppressive activity upon proliferation. Together, these findings reveal the cAMP pathway as an attractive target for clinical intervention with Treg function.

Collaboration


Dive into the Martin Vaeth's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge