Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Immunol ; 53(8): e2250353, 2023 08.
Article in English | MEDLINE | ID: mdl-37179252

ABSTRACT

Unraveling the immune signatures in rheumatoid arthritis (RA) patients receiving various treatment regimens can aid in comprehending the immune mechanisms' role in treatment efficacy and side effects. Given the critical role of cellular immunity in RA pathogenesis, we sought to identify T-cell profiles characterizing RA patients under specific treatments. We compared 75 immunophenotypic and biochemical variables in healthy donors (HD) and RA patients, including those receiving different treatments as well as treatment-free patients. Additionally, we conducted in vitro experiments to evaluate the direct effect of tofacitinib on purified naïve and memory CD4+ and CD8+ T cells. Multivariate analysis revealed that tofacitinib-treated patients segregated from HD at the expense of T-cell activation, differentiation, and effector function-related variables. Additionally, tofacitinib led to an accumulation of peripheral senescent memory CD4+ and CD8+ T cells. In vitro, tofacitinib impaired the activation, proliferation, and effector molecules expression and triggered senescence pathways in T-cell subsets upon TCR-engagement, with the most significant impact on memory CD8+ T cells. Our findings suggest that tofacitinib may activate immunosenescence pathways while simultaneously inhibiting effector functions in T cells, both effects likely contributing to the high clinical success and reported side effects of this JAK inhibitor in RA.


Subject(s)
Arthritis, Rheumatoid , CD8-Positive T-Lymphocytes , Humans , CD4-Positive T-Lymphocytes , Arthritis, Rheumatoid/drug therapy , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
2.
J Endocrinol ; 232(2): 337-350, 2017 02.
Article in English | MEDLINE | ID: mdl-28052998

ABSTRACT

We reported thyroid hormone (TH) receptor expression in murine dendritic cells (DCs) and 3,5,3'-triiodothyronine (T3)-dependent stimulation of DC maturation and ability to develop a Th1-type adaptive response. Moreover, an increased DC capacity to promote antigen-specific cytotoxic T-cell activity, exploited in a DC-based antitumor vaccination protocol, was revealed. However, putative effects of the main circulating TH, l-thyroxine (T4) and the mechanisms of TH transport and metabolism at DC level, crucial events for TH action at target cell level, were not known. Herein, we show that T4 did not reproduce those registered T3-dependent effects, finding that may reflect a homoeostatic control to prevent unspecific systemic activation of DCs. Besides, DCs express MCT10 and LAT2 TH transporters, and these cells mainly transport T3 with a favored involvement of MCT10 as its inhibition almost prevented T3 saturable uptake mechanism and reduced T3-induced IL-12 production. In turn, DCs express iodothyronine deiodonases type 2 and 3 (D2, D3) and exhibit both enzymatic activities with a prevalence towards TH inactivation. Moreover, T3 increased MCT10 and LAT2 expression and T3 efflux from DCs but not T3 uptake, whereas it induced a robust induction of D3 with a parallel slight reduction in D2. These findings disclose pivotal events involved in the mechanism of action of THs on DCs, providing valuable tools for manipulating the immunogenic potential of these cells. Furthermore, they broaden the knowledge of the TH mechanism of action at the immune system network.


Subject(s)
Dendritic Cells/metabolism , Receptors, Thyroid Hormone/metabolism , Triiodothyronine/metabolism , Animals , Biological Transport/physiology , Female , Homeostasis/physiology , Iodide Peroxidase/metabolism , Mice
3.
Cancer Res ; 75(7): 1265-74, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25672979

ABSTRACT

Bidirectional cross-talk between the neuroendocrine and immune systems orchestrates immune responses in both physiologic and pathologic settings. In this study, we provide in vivo evidence of a critical role for the thyroid hormone triiodothyronine (T3) in controlling the maturation and antitumor functions of dendritic cells (DC). We used a thyroid hormone receptor (TR) ß mutant mouse (TRßPV) to establish the relevance of the T3-TRß system in vivo. In this model, TRß signaling endowed DCs with the ability to stimulate antigen-specific cytotoxic T-cell responses during tumor development. T3 binding to TRß increased DC viability and augmented DC migration to lymph nodes. Moreover, T3 stimulated the ability of DCs to cross-present antigens and to stimulate cytotoxic T-cell responses. In a B16-OVA mouse model of melanoma, vaccination with T3-stimulated DCs inhibited tumor growth and prolonged host survival, in part by promoting the generation of IFNγ-producing CD8(+) T cells. Overall, our results establish an adjuvant effect of T3-TRß signaling in DCs, suggesting an immediately translatable method to empower DC vaccination approaches for cancer immunotherapy.


Subject(s)
Dendritic Cells/immunology , Melanoma, Experimental/immunology , Thyroid Hormone Receptors beta/metabolism , Triiodothyronine/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Cell Survival , Cross-Priming , Cytotoxicity, Immunologic , Female , Immunotherapy , Lymph Nodes/immunology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Transgenic
4.
Steroids ; 77(1-2): 67-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22056479

ABSTRACT

Glucocorticoids (GCs) are widely used as anti-inflammatory and immunosuppressive agents. Several studies have indicated the important role of dendritic cells (DCs), highly specialized antigen-presenting and immunomodulatory cells, in GC-mediated suppression of adaptive immune responses. Recently, we demonstrated that triiodothyronine (T3) has potent immunostimulatory effects on bone marrow-derived mouse DCs through a mechanism involving T3 binding to cytosolic thyroid hormone receptor (TR) ß1, rapid and sustained Akt activation and IL-12 production. Here we explored the impact of GCs on T3-mediated DC maturation and function and the intracellular events underlying these effects. Dexamethasone (Dex), a synthetic GC, potently inhibited T3-induced stimulation of DCs by preventing the augmented expression of maturation markers and the enhanced IL-12 secretion through mechanisms involving the GC receptor. These effects were accompanied by increased IL-10 levels following exposure of T3-conditioned DCs to Dex. Accordingly, Dex inhibited the immunostimulatory capacity of T3-matured DCs on naive T-cell proliferation and IFN-γ production while increased IL-10 synthesis by allogeneic T cell cultures. A mechanistic analysis revealed the ability of Dex to dampen T3 responses through modulation of Akt phosphorylation and cytoplasmic-nuclear shuttling of nuclear factor-κB (NF-κB). In addition, Dex decreased TRß1 expression in both immature and T3-maturated DCs through mechanisms involving the GC receptor. Thus GCs, which are increased during the resolution of inflammatory responses, counteract the immunostimulatory effects of T3 on DCs and their ability to polarize adaptive immune responses toward a T helper (Th)-1-type through mechanisms involving, at least in part, NF-κB- and TRß1-dependent pathways. Our data provide an alternative mechanism for the anti-inflammatory effects of GCs with critical implications in immunopathology at the cross-roads of the immune-endocrine circuits.


Subject(s)
Adaptive Immunity/drug effects , Dendritic Cells/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Receptors, Glucocorticoid/metabolism , Receptors, Thyroid Hormone/metabolism , Triiodothyronine/pharmacology , Adjuvants, Immunologic/antagonists & inhibitors , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Biomarkers/analysis , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Glucocorticoid/immunology , Receptors, Thyroid Hormone/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Triiodothyronine/antagonists & inhibitors , Triiodothyronine/metabolism
5.
Clin Endocrinol (Oxf) ; 74(3): 346-53, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21091751

ABSTRACT

OBJECTIVE: Idiopathic short stature (ISS) describes short children with normal GH secretion. Although GH treatment increases their heights, growth response to the therapy differs among patients. Thyroid hormones (TH) are essential for longitudinal growth acting mainly through TH receptors (TR) α and ß. We have previously reported that GH treatment reduced peripheral TH action in Turner Syndrome by TR down-regulation. The aims of the study were to assess the effect of GH treatment to ISS on peripheral TH action and the correlation between thyroid status and growth response to the therapy. SUBJECTS, DESIGN AND MEASUREMENTS: Eighteen normal (control) and twenty-five ISS children were enrolled and evaluated before and after 12 months of life time (control) or 12 months of GH therapy (ISS). Fasting blood was used for serum biochemical evaluations, peripheral blood mononuclear cells for TR mRNA determination by QRT-PCR and growth parameters by standard methods. RESULTS: GH treatment modified neither TR mRNA levels nor serum markers of TH action in ISS evaluated as a whole group. However, the individual change in TRß mRNA levels correlated to the change in sex hormone-binding globulin (SHBG) levels after GH therapy. The growth response to GH correlated positively with the change in TRα mRNA level and negatively with that in TRß mRNA, TSH and SHBG levels. The change in each TR mRNA isoform after GH treatment correlated negatively with its own basal level. CONCLUSIONS: GH therapy induced individual changes in TR expression in ISS that correlated with their growth response. The basal TR mRNA level could predetermine the change in TR expression and therefore the sensitivity to GH treatment.


Subject(s)
Growth Disorders/blood , Growth Disorders/drug therapy , Human Growth Hormone/therapeutic use , Child , Growth Disorders/genetics , Humans , Immunoassay/methods , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor Binding Proteins/blood , Insulin-Like Growth Factor I/metabolism , Osteocalcin/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sex Hormone-Binding Globulin/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormones/blood , Thyrotropin/blood , Time Factors , Treatment Outcome
6.
J Biol Chem ; 285(13): 9569-9582, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20018842

ABSTRACT

Despite considerable progress in our understanding of the interplay between immune and endocrine systems, the role of thyroid hormones and their receptors in the control of adaptive immunity is still uncertain. Here, we investigated the role of thyroid hormone receptor (TR) beta(1) signaling in modulating dendritic cell (DC) physiology and the intracellular mechanisms underlying these immunoregulatory effects. Exposure of DCs to triiodothyronine (T(3)) resulted in a rapid and sustained increase in Akt phosphorylation independently of phosphatidylinositol 3-kinase activation, which was essential for supporting T(3)-induced DC maturation and interleukin (IL)-12 production. This effect was dependent on intact TR beta(1) signaling as small interfering RNA-mediated silencing of TR beta(1) expression prevented T(3)-induced DC maturation and IL-12 secretion as well as Akt activation and I kappaB-epsilon degradation. In turn, T(3) up-regulated TR beta(1) expression through mechanisms involving NF-kappaB, suggesting an autocrine regulatory loop to control hormone-dependent TR beta(1) signaling. These findings were confirmed by chromatin immunoprecipitation analysis, which disclosed a new functional NF-kappaB consensus site in the promoter region of the TRB1 gene. Thus, a T(3)-induced NF-kappaB-dependent mechanism controls TR beta(1) expression, which in turn signals DCs to promote maturation and function via an Akt-dependent but PI3K-independent pathway. These results underscore a novel unrecognized target that regulates DC maturation and function with critical implications in immunopathology at the cross-roads of the immune-endocrine circuits.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thyroid Hormone Receptors beta/metabolism , Animals , Dendritic Cells/metabolism , Enzyme Activation , Female , Immunoblotting , Interleukin-12/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , Triiodothyronine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...