Network


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

Hotspot


Dive into the research topics where Maria Teresa Pallotta is active.

Publication


Featured researches published by Maria Teresa Pallotta.


Nature Immunology | 2011

Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells

Maria Teresa Pallotta; Ciriana Orabona; Claudia Volpi; Carmine Vacca; Maria Laura Belladonna; Roberta Bianchi; Giuseppe Servillo; Cinzia Brunacci; Mario Calvitti; Silvio Bicciato; Emilia Maria Cristina Mazza; Louis Boon; Fabio Grassi; Maria C. Fioretti; Francesca Fallarino; Paolo Puccetti; Ursula Grohmann

Regulation of tryptophan metabolism by indoleamine 2,3-dioxygenase (IDO) in dendritic cells (DCs) is a highly versatile modulator of immunity. In inflammation, interferon-γ is the main inducer of IDO for the prevention of hyperinflammatory responses, yet IDO is also responsible for self-tolerance effects in the longer term. Here we show that treatment of mouse plasmacytoid DCs (pDCs) with transforming growth factor-β (TGF-β) conferred regulatory effects on IDO that were mechanistically separable from its enzymic activity. We found that IDO was involved in intracellular signaling events responsible for the self-amplification and maintenance of a stably regulatory phenotype in pDCs. Thus, IDO has a tonic, nonenzymic function that contributes to TGF-β-driven tolerance in noninflammatory contexts.


Nature | 2014

Aryl hydrocarbon receptor control of a disease tolerance defence pathway

Alban Bessede; Marco Gargaro; Maria Teresa Pallotta; Davide Matino; Giuseppe Servillo; Cinzia Brunacci; Silvio Bicciato; Emilia Maria Cristina Mazza; Antonio Macchiarulo; Carmine Vacca; Rossana G. Iannitti; Luciana Tissi; Claudia Volpi; Maria Laura Belladonna; Ciriana Orabona; Roberta Bianchi; Tobias V. Lanz; Michael Platten; Maria Agnese Della Fazia; Danilo Piobbico; Teresa Zelante; Hiroshi Funakoshi; Toshikazu Nakamura; David Gilot; Michael S. Denison; Gilles J. Guillemin; James B. DuHadaway; George C. Prendergast; Richard Metz; Michel Geffard

Disease tolerance is the ability of the host to reduce the effect of infection on host fitness. Analysis of disease tolerance pathways could provide new approaches for treating infections and other inflammatory diseases. Typically, an initial exposure to bacterial lipopolysaccharide (LPS) induces a state of refractoriness to further LPS challenge (endotoxin tolerance). We found that a first exposure of mice to LPS activated the ligand-operated transcription factor aryl hydrocarbon receptor (AhR) and the hepatic enzyme tryptophan 2,3-dioxygenase, which provided an activating ligand to the former, to downregulate early inflammatory gene expression. However, on LPS rechallenge, AhR engaged in long-term regulation of systemic inflammation only in the presence of indoleamine 2,3-dioxygenase 1 (IDO1). AhR-complex-associated Src kinase activity promoted IDO1 phosphorylation and signalling ability. The resulting endotoxin-tolerant state was found to protect mice against immunopathology in Gram-negative and Gram-positive infections, pointing to a role for AhR in contributing to host fitness.


Journal of Immunology | 2008

Cutting Edge: Autocrine TGF-β Sustains Default Tolerogenesis by IDO-Competent Dendritic Cells

Maria Laura Belladonna; Claudia Volpi; Roberta Bianchi; Carmine Vacca; Ciriana Orabona; Maria Teresa Pallotta; Louis Boon; Stefania Gizzi; Maria C. Fioretti; Ursula Grohmann; Paolo Puccetti

CD8− and CD8+ dendritic cells (DCs) are distinct subsets of mouse splenic accessory cells with opposite but flexible programs of Ag presentation, leading to immunogenic and tolerogenic responses, respectively. In this study, we show that the default tolerogenic function of CD8+ DCs relies on autocrine TGF-β, which sustains the activation of IDO in response to environmental stimuli. CD8− DCs do not produce TGF-β, yet externally added TGF-β induces IDO and turns those cells from immunogenic into tolerogenic cells. The acquisition of a suppressive phenotype by CD8− DCs correlates with activation of the PI3K/Akt and noncanonical NF-κB pathways. These data are the first to link TGF-β signaling with IDO in controlling spontaneous tolerogenesis by DCs.


Proceedings of the National Academy of Sciences of the United States of America | 2008

SOCS3 drives proteasomal degradation of indoleamine 2,3-dioxygenase (IDO) and antagonizes IDO-dependent tolerogenesis

Ciriana Orabona; Maria Teresa Pallotta; Claudia Volpi; Francesca Fallarino; Carmine Vacca; Roberta Bianchi; Maria Laura Belladonna; Maria C. Fioretti; Ursula Grohmann; Paolo Puccetti

Despite their common ability to activate intracellular signaling through CD80/CD86 molecules, cytotoxic T lymphocyte antigen 4 (CTLA-4)-Ig and CD28-Ig bias the downstream response in opposite directions, the latter promoting immunity, and CTLA-4-Ig tolerance, in dendritic cells (DCs) with opposite but flexible programs of antigen presentation. Nevertheless, in the absence of suppressor of cytokine signaling 3 (SOCS3), CD28-Ig—and the associated, dominant IL-6 response—become immunosuppressive and mimic the effect of CTLA-4-Ig, including a high functional expression of the tolerogenic enzyme indoleamine 2,3-dioxygenase (IDO). Here we show that forced SOCS3 expression antagonized CTLA-4-Ig activity in a proteasome-dependent fashion. Unrecognized by previous studies, IDO appeared to possess two tyrosine residues within two distinct putative immunoreceptor tyrosine-based inhibitory motifs, VPY115CEL and LLY253EGV. We found that SOCS3—known to interact with phosphotyrosine-containing peptides and be selectively induced by CD28-Ig/IL-6—would bind IDO and target the IDO/SOCS3 complex for ubiquitination and subsequent proteasomal degradation. This event accounted for the ability of CD28-Ig and IL-6 to convert otherwise tolerogenic, IDO-competent DCs into immunogenic cells. Thus onset of immunity in response to antigen within an early inflammatory context requires that IDO be degraded in tolerogenic DCs. In addition to identifying SOCS3 as a candidate signature for mouse DC subsets programmed to direct immunity, this study demonstrates that IDO undergoes regulatory proteolysis in response to immunogenic stimuli.


Transplantation | 2007

Immunosuppression via tryptophan catabolism: the role of kynurenine pathway enzymes.

Maria Laura Belladonna; Paolo Puccetti; Ciriana Orabona; Francesca Fallarino; Carmine Vacca; Claudia Volpi; Stefania Gizzi; Maria Teresa Pallotta; Maria C. Fioretti; Ursula Grohmann

Tryptophan catabolism occurring in dendritic cells (DCs) and initiated by indoleamine 2,3-dioxygenase (IDO) is an emerging major mechanism of peripheral tolerance. Here we provide evidence that: 1) tryptophan conversion to kynurenines is activated in DCs by cytotoxic T lymphocyte antigen 4, both in a soluble form or anchored to the regulatory T cell (Treg) membrane; 2) an increased IDO-dependent tolerogenesis correlates with the inhibition of DAP12 functions, an adapter molecule associated with activating receptors; 3) a tolerogenic phenotype can be acquired by DCs lacking functional IDO through the paracrine production of kynurenines by IDO-competent DCs; 4) the suppressive effect of Treg generated in a microenvironment with low tryptophan concentration and a mixture of kynurenines can protect mice in an experimental model of fulminant diabetes. Altogether, these data indicate that, in addition to tryptophan starvation induced by IDO activity, the paracrine production of kynurenines by enzymes downstream of IDO can also contribute to tolerogenesis in DCs, independently of tryptophan deprivation.


Nature Communications | 2013

High doses of CpG oligodeoxynucleotides stimulate a tolerogenic TLR9–TRIF pathway

Claudia Volpi; Francesca Fallarino; Maria Teresa Pallotta; Roberta Bianchi; Carmine Vacca; Maria Laura Belladonna; Ciriana Orabona; Antonella De Luca; Louis Boon; Luigina Romani; Ursula Grohmann; Paolo Puccetti

CpG-rich oligodeoxynucleotides activate the immune system, leading to innate and acquired immune responses. The immune-stimulatory effects of CpG-rich oligodeoxynucleotides are being exploited as a therapeutic approach. Here we show that at high doses, CpG-rich oligodeoxynucleotides promote an opposite, tolerogenic response in mouse plasmacytoid dendritic cells in vivo and in a human in vitro model. Unveiling a previously undescribed role for TRIF and TRAF6 proteins in Toll-like receptor 9 (TLR9) signalling, we demonstrate that physical association of TLR9, TRIF and TRAF6 leads to activation of noncanonical NF-κB signalling and the induction of IRF3- and TGF-β-dependent immune-suppressive tryptophan catabolism. In vivo, the TLR9-TRIF circuit--but not MyD88 signalling--was required for CpG protection against allergic inflammation. Our findings may be relevant to an increased understanding of the complexity of Toll-like receptor signalling and optimal exploitation of CpG-rich oligodeoxynucleotides as immune modulators.


Molecular Medicine | 2012

Different Partners, Opposite Outcomes: A New Perspective of the Immunobiology of Indoleamine 2,3-Dioxygenase

Ciriana Orabona; Maria Teresa Pallotta; Ursula Grohmann

Indoleamine 2,3-dioxygenase (IDO), a metabolic enzyme that catalyzes tryptophan conversion into kynurenines, is a crucial regulator of immunity. Altered IDO activity is often associated with pathology, including neoplasia and autoimmunity. IDO is highly expressed in dendritic cells (DCs) that exploit the enzyme’s activity and the production of tryptophan catabolites to regulate immune responses by acting on several cell types, including T lymphocytes, of which they promote a regulatory phenotype. IDO also contains immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that, once bound by distinct molecular partners, will either promote degradation or initiate signaling activity and self-maintenance of the enzyme. We here discuss how ITIM-dependent molecular events can affect the functional plasticity of IDO by modifying the protein half-life and its enzymic and nonenzymic functions.


Immunity | 2017

A Relay Pathway between Arginine and Tryptophan Metabolism Confers Immunosuppressive Properties on Dendritic Cells

Giada Mondanelli; Roberta Bianchi; Maria Teresa Pallotta; Ciriana Orabona; Elisa Albini; Alberta Iacono; Maria Laura Belladonna; Carmine Vacca; Francesca Fallarino; Antonio Macchiarulo; Stefano Ugel; Vincenzo Bronte; Federica Gevi; Lello Zolla; Auke Verhaar; Maikel P. Peppelenbosch; Emilia Maria Cristina Mazza; Silvio Bicciato; Yasmina Laouar; Laura Santambrogio; Paolo Puccetti; Claudia Volpi; Ursula Grohmann

SUMMARY Arginase 1 (Arg1) and indoleamine 2,3‐dioxygenase 1 (IDO1) are immunoregulatory enzymes catalyzing the degradation of l‐arginine and l‐tryptophan, respectively, resulting in local amino acid deprivation. In addition, unlike Arg1, IDO1 is also endowed with non‐enzymatic signaling activity in dendritic cells (DCs). Despite considerable knowledge of their individual biology, no integrated functions of Arg1 and IDO1 have been reported yet. We found that IDO1 phosphorylation and consequent activation of IDO1 signaling in DCs was strictly dependent on prior expression of Arg1 and Arg1‐dependent production of polyamines. Polyamines, either produced by DCs or released by bystander Arg1+ myeloid‐derived suppressor cells, conditioned DCs toward an IDO1‐dependent, immunosuppressive phenotype via activation of the Src kinase, which has IDO1‐phosphorylating activity. Thus our data indicate that Arg1 and IDO1 are linked by an entwined pathway in immunometabolism and that their joint modulation could represent an important target for effective immunotherapy in several disease settings. HighlightsDendritic cells (DCs) can co‐express Arg1 and IDO1 immunosuppressive enzymesArg1 activity is required for IDO1 induction by TGF‐&bgr; in DCsSpermidine, a downstream Arg1 product, but not arginine starvation, induces IDO1 in DCsArg1+ myeloid derived suppressor cells (MDSCs) can render DCs immunosuppressive via IDO1 &NA; Arginase 1 (Arg1) and indoleamine 2,3‐dioxygenase 1 (IDO1) are immunosuppressive enzymes known to operate in distinct immune cells. Mondanelli and colleagues demonstrate that Arg1 and IDO1 cooperate in conferring long‐term, immunosuppressive effects to dendritic cells.


Journal of Cellular and Molecular Medicine | 2014

Forced IDO1 expression in dendritic cells restores immunoregulatory signalling in autoimmune diabetes.

Maria Teresa Pallotta; Ciriana Orabona; Roberta Bianchi; Carmine Vacca; Francesca Fallarino; Maria Laura Belladonna; Claudia Volpi; Giada Mondanelli; Marco Gargaro; Massimo Allegrucci; Vincenzo Nicola Talesa; Paolo Puccetti; Ursula Grohmann

Indoleamine 2,3‐dioxygenase (IDO1), a tryptophan catabolizing enzyme, is recognized as an authentic regulator of immunity in several physiopathologic conditions. We have recently demonstrated that IDO1 does not merely degrade tryptophan and produce immunoregulatory kynurenines, but it also acts as a signal‐transducing molecule, independently of its enzymic function. IDO1 signalling activity is triggered in plasmacytoid dendritic cells (pDCs) by transforming growth factor‐β (TGF‐β), an event that requires the non‐canonical NF‐κB pathway and induces long‐lasting IDO1 expression and autocrine TGF‐β production in a positive feedback loop, thus sustaining a stably regulatory phenotype in pDCs. IDO1 expression and catalytic function are defective in pDCs from non‐obese diabetic (NOD) mice, a prototypic model of autoimmune diabetes. In the present study, we found that TGF‐β failed to activate IDO1 signalling function as well as up‐regulate IDO1 expression in NOD pDCs. Moreover, TGF‐β‐treated pDCs failed to exert immunosuppressive properties in vivo. Nevertheless, transfection of NOD pDCs with Ido1 prior to TGF‐β treatment resulted in activation of the Ido1 promoter and induction of non‐canonical NF‐κB and TGF‐β, as well as decreased production of the pro‐inflammatory cytokines, interleukin 6 (IL‐6) and tumour necrosis factor‐α (TNF‐α). Overexpression of IDO1 in TGF‐β‐treated NOD pDCs also resulted in pDC ability to suppress the in vivo presentation of a pancreatic β‐cell auto‐antigen. Thus, our data suggest that a correction of IDO1 expression may restore its dual function and thus represent a proper therapeutic manoeuvre in this autoimmune setting.


Journal of Immunology | 2013

Topical Application of Soluble CD83 Induces IDO-Mediated Immune Modulation, Increases Foxp3+ T Cells, and Prolongs Allogeneic Corneal Graft Survival

Felix Bock; Susanne Rössner; Jasmine Onderka; Matthias Lechmann; Maria Teresa Pallotta; Francesca Fallarino; Louis Boon; Charles Nicolette; Mark A. DeBenedette; Irina Tcherepanova; Ursula Grohmann; Alexander Steinkasserer; Claus Cursiefen; Elisabeth Zinser

Modulation of immune responses is one of the main research aims in transplant immunology. In this study, we investigate the local immunomodulatory properties of soluble CD83 (sCD83) at the graft-host interface using the high-risk corneal transplantation model. In this model, which mimics the inflammatory status and the preexisting vascularization of high-risk patients undergoing corneal transplantation, allogeneic donor corneas are transplanted onto sCD83-treated recipient animals. This model allows the direct and precise application of the immune modulator at the transplantation side. Interestingly, sCD83 was able to prolong graft survival after systemic application as well as after topical application, which is therapeutically more relevant. The therapeutic effect was accompanied by an increase in the frequency of regulatory T cells and was mediated by the immune-regulatory enzyme IDO and TGF-β. In vitro, sCD83 induced long-term IDO expression in both conventional and plasmacytoid dendritic cells via autocrine or paracrine production of TGF-β, a cytokine previously shown to be an essential mediator of IDO-dependent, long-term tolerance. These findings open new treatment avenues for local immune modulation after organ and tissue transplantation.

Collaboration


Dive into the Maria Teresa Pallotta'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