Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Type of study
Language
Publication year range
2.
FASEB J ; 37(4): e22865, 2023 04.
Article in English | MEDLINE | ID: mdl-36934391

ABSTRACT

Hypothyroidism exerts deleterious effects on immunity, but the precise role of the hypothalamic-pituitary-thyroid (HPT) axis in immunoregulatory and tolerogenic programs is barely understood. Here, we investigated the mechanisms underlying hypothyroid-related immunosuppression by examining the regulatory role of components of the HPT axis. We first analyzed lymphocyte activity in mice overexpressing the TRH gene (Tg-Trh). T cells from Tg-Trh showed increased proliferation than wild-type (WT) euthyroid mice in response to polyclonal activation. The release of Th1 pro-inflammatory cytokines was also increased in Tg-Trh and TSH levels correlated with T-cell proliferation. To gain further mechanistic insights into hypothyroidism-related immunosuppression, we evaluated T-cell subpopulations in lymphoid tissues of hypothyroid and control mice. No differences were observed in CD3/CD19 or CD4/CD8 ratios between these strains. However, the frequency of regulatory T cells (Tregs) was significantly increased in hypothyroid mice, and not in Tg-Trh mice. Accordingly, in vitro Tregs differentiation was more pronounced in naïve T cells isolated from hypothyroid mice. Since Tregs overexpress galectin-1 (Gal-1) and mice lacking this lectin (Lgals1-/- ) show reduced Treg function, we investigated the involvement of this immunoregulatory lectin in the control of Tregs in settings of hypothyroidism. Increased T lymphocyte reactivity and reduced frequency of Tregs were found in hypothyroid Lgals1-/- mice when compared to hypothyroid WT animals. This effect was rescued by the addition of recombinant Gal-1. Finally, increased expression of Gal-1 was found in Tregs purified from hypothyroid WT mice compared with their euthyroid counterpart. Thus, a substantial increase in the frequency and activity of Gal-1-expressing Tregs underlies immunosuppression associated with hypothyroid conditions, with critical implications in immunopathology, metabolic disorders, and cancer.


Subject(s)
Hypothyroidism , Thyrotropin , Mice , Animals , Thyrotropin/metabolism , Thyrotropin-Releasing Hormone/pharmacology , T-Lymphocytes, Regulatory/metabolism , Galectin 1/genetics , Hypothyroidism/metabolism , Immunosuppression Therapy
3.
PLoS One ; 15(1): e0227047, 2020.
Article in English | MEDLINE | ID: mdl-31929548

ABSTRACT

Enterotoxin-based proteins are powerful manipulators of mucosal immunity. The A1 domain of heat-labile enterotoxin from E. coli, or LTA1, is a newer adjuvant from this family under investigation for intranasal vaccines. Although LTA1 has been examined in mouse vaccination studies, its ability to directly stimulate immune cells compared to related adjuvant proteins has not been well explored. Here, we perform the first rigorous examination of LTA1's effect on antigen presenting cells (APC) using a human monocyte cell line THP-1. To better understand LTA1's stimulatory effects, we compared it to dmLT, or LT-R192G/L211A, a related AB5 adjuvant in clinical trials for oral or parenteral vaccines. LTA1 and dmLT both activated APCs to upregulate MHC-II (HLA-DR), CD86, cytokine secretion (e.g., IL-1ß) and inflammasome activation. The effect of LTA1 on surface marker changes (e.g., MHC-II) was highly dose-dependent whereas dmLT exhibited high MHC-II expression regardless of dose. In contrast, cytokine secretion profiles were similar and dose-dependent after both LTA1 and dmLT treatment. Cellular activation by LTA1 was independent of ganglioside binding, as pre-treatment with purified GM1 blocked the effect of dmLT but not LTA1. Unexpectedly, while activation of the inflammasome and cytokine secretion by LTA1 or dmLT was blocked by the protein kinase A inhibitor H89 (similar to previous reports), these responses were not inhibited by a more specific PKA peptide inhibitor or antagonist; thus Indicating that a novel and unknown mechanism is responsible for inflammasome activation and cytokine secretion by LT proteins. Lastly, LTA1 stimulated a similar cytokine profile in primary human monocytes as it did in THP1 cells, including IL-1ß, IL-6, IL-8, MIP-1α, MIP-1ß, and TNFα. Thus, we report that LTA1 protein programs a dendritic cell-like phenotype in APCs similar to dmLT in a mechanism that is independent of PKA activation and GM1 binding and entry.


Subject(s)
Antigen-Presenting Cells/metabolism , Enterotoxins/pharmacology , Lipopolysaccharides/pharmacology , Teichoic Acids/pharmacology , Adjuvants, Immunologic , Antigen-Presenting Cells/drug effects , Cell Line , Cell Membrane Permeability , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases , Cytokines/drug effects , Cytokines/metabolism , Enterotoxins/immunology , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Lipopolysaccharides/immunology , Monocytes/pathology , THP-1 Cells , Teichoic Acids/immunology
4.
Endocrine ; 66(2): 266-277, 2019 11.
Article in English | MEDLINE | ID: mdl-31056723

ABSTRACT

PURPOSE: Hypothyroidism has been shown to induce immunosuppression and both the thyroid status and immunity are affected by zinc deficiency. However, the impact of hypothyroidism on zinc metabolism and its possible relationship with the immune status has not yet been deeply explored. Here, our aim was to study whether hypothyroidism may alter zinc metabolism and thus lead to the impairment of T lymphocyte activity. METHODS: Variations in the distribution of zinc in the body were evaluated in PTU-treated hypothyroid mice. The effects of hypothyroidism and zinc deficiency were studied on T lymphocyte proliferation after stimulation both in vitro and in vivo. For in vitro assays, thyroid hormone-free or zinc chelator (TPEN or DTPA)-supplemented media were used. For in vivo assays, lymphocyte activity was evaluated in cells from hypothyroid, T3-treated, and zinc-supplemented mice. RESULTS: Hypothyroid mice showed lower levels of zinc in femur and lymph nodes than controls. T3 and zinc supplementation reversed these effects. In vitro, both thyroid hormone and zinc deficiency led to a decreased response to mitogen stimulation. However, only zinc deficiency was able to induce lymphocyte apoptosis. Mitogen-stimulated T cells from hypothyroid mice showed impaired proliferation, accompanied by decreased activation of PKC and lower levels of p-ERK, effects that were reversed by T3 replacement or zinc supplementation. CONCLUSIONS: Our results show an important role of zinc deficiency in hypothyroid-mediated T-cell suppression and suggest the importance of evaluating zinc levels and restoring them when necessary to maintain an efficient immune response in hypothyroid patients.


Subject(s)
Cell Proliferation/physiology , Hypothyroidism/complications , T-Lymphocytes/metabolism , Zinc/deficiency , Animals , Apoptosis/physiology , Femur/metabolism , Hypothyroidism/metabolism , Lymph Nodes/metabolism , Lymphocyte Activation , Mice , Thyroid Gland/metabolism , Zinc/metabolism
5.
Cancer Biol Ther ; 16(1): 137-48, 2015.
Article in English | MEDLINE | ID: mdl-25482934

ABSTRACT

The radioprotective potential of histamine on healthy tissue has been previously demonstrated. The aims of this work were to investigate the combinatorial effect of histamine or its receptor ligands and gamma radiation in vitro on the radiobiological response of 2 breast cancer cell lines (MDA-MB-231 and MCF-7), to explore the potential molecular mechanisms of the radiosensitizing action and to evaluate the histamine-induced radiosensitization in vivo in a triple negative breast cancer model. Results indicate that histamine significantly increased the radiosensitivity of MDA-MB-231 and MCF-7 cells. This effect was mimicked by the H1R agonist 2-(3-(trifluoromethyl)phenyl)histamine and the H4R agonists (Clobenpropit and VUF8430) in MDA-MB-231 and MCF-7 cells, respectively. Histamine and its agonists enhanced radiation-induced oxidative DNA damage, DNA double-strand breaks, apoptosis and senescence. These effects were associated with increased production of reactive oxygen species, which correlated with the inhibition of catalase, glutathione peroxidase and superoxide dismutase activities in MDA-MB-231 cells. Histamine was able also to potentiate in vivo the anti-tumoral effect of radiation, increasing the exponential tumor doubling time. We conclude that histamine increased radiation response of breast cancer cells, suggesting that it could be used as a potential adjuvant to enhance the efficacy of radiotherapy.


Subject(s)
Breast Neoplasms/metabolism , Histamine/metabolism , Radiation Tolerance , Radiation, Ionizing , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Histamine/pharmacology , Humans , MCF-7 Cells , Oxidation-Reduction , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
6.
Biol Trace Elem Res ; 157(2): 138-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24363240

ABSTRACT

Scleroderma, sclerosis of the skin, is a severe autoimmune disease refractant to all kind of treatments. To study the in vivo effects of a combination of three oligoelements selenium (Se), zinc (Zn), and manganese (Mn) plus Lachesis muta venom (O-LM) on the bleomycin (BLM)-induced scleroderma mouse experimental model. C3H mice were randomly divided into four groups: control (phosphate-buffered saline (PBS)), O-LM, BLM, and BLM + O-LM. All administrations were performed subcutaneously into the back of mice. BLM was injected 5 days per week for three consecutive weeks and O-LM was administered simultaneously with BLM from the beginning of the experiments and lasted for 3 weeks after the final BLM or PBS injection (for O-LM and BLM + O-LM groups), when animals were sacrificed and histopathological, immunohistochemical, thiobarbituric acid reactive species (TBARS) evaluation, and autoantibodies detection were determined. O-LM significantly reduced BLM-induced enhanced dermal thickness (605 ± 47 vs. 956 ± 59 µm, P < 0.01), collagen deposition, and mast cells infiltration (43.1 ± 1.0 vs. 102 ± 14.1 mast cells, P < 0.05). O-LM administration significantly blocked BLM-induced oxidative damage and the enhanced immunoreactive fibroblasts for α-smooth muscle actin while reduced BLM-induced autoantibodies that strongly react mainly with skin and spleen. O-LM significantly reduced BLM-induced scleroderma through the modulation of antioxidant and immunological pathways.


Subject(s)
Crotalid Venoms/therapeutic use , Manganese/therapeutic use , Scleroderma, Systemic/drug therapy , Selenium/therapeutic use , Skin/drug effects , Zinc/therapeutic use , Animals , Antioxidants/metabolism , Autoantibodies/blood , Bleomycin/pharmacology , Cell Count , Cell Survival/drug effects , Crotalid Venoms/administration & dosage , Crotalid Venoms/toxicity , Disease Models, Animal , Drug Therapy, Combination , Manganese/administration & dosage , Manganese/toxicity , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Organ Specificity , Oxidative Stress/drug effects , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Selenium/administration & dosage , Selenium/toxicity , Skin/immunology , Skin/pathology , Zinc/administration & dosage , Zinc/toxicity
7.
J Cell Physiol ; 226(12): 3208-18, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21344381

ABSTRACT

Thyroid hormones (THs) exert a broad range of actions on development, growth, and cell differentiation by both genomic and nongenomic mechanisms. THs regulate lymphocyte function, but the participation of nongenomic actions is still unknown. Here the contribution of both genomic and nongenomic effects on TH-induced division of T cells was studied by using free and noncell permeable THs coupled to agarose (TH-ag). THs-ag led to cell division, but to a lesser extent than free hormones. THs induced nongenomically the rapid translocation of protein kinase C (PKC) ζ isoform to cell membranes, extracellular-signal-regulated kinases (ERK1/2) phosphorylation and nuclear factor-κB (NF-κB) activation. The signaling cascade include sphingomyelinases acting up-stream the activation of PKCζ isoform, while ERK and NF-κB are activated downstream this PKC isoenzyme. Both free and THs-ag increased the protein and mRNA levels of TH nuclear receptor TRα1, while only free hormones incremented the inducible NOS gene and protein levels as well as a calcium independent NOS activity. Both effects were blunted by PKCζ inhibition. These results indicate that THs, by triggering a nongenomic signaling cascade that involves Smases-mediated activation of PKCζ, lead to ERK 1/2 and NF-κB activation and to the genomic increase of TRs and the inducible nitric oxide synthase protein and mRNA levels, improving T lymphocyte proliferation. These finding not only contribute to the understanding of the mechanisms involved in TH modulation of lymphocyte physiology, but would also point out for the first time the interplay between genomic and nongenomic TH actions in T cells.


Subject(s)
Cell Proliferation , Nitric Oxide Synthase Type II/metabolism , T-Lymphocytes/enzymology , Thyroid Hormone Receptors alpha/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Enzymologic , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport , RNA, Messenger/metabolism , Signal Transduction , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes/drug effects , Thyroid Hormone Receptors alpha/genetics , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...