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










Publication year range
1.
World J Stem Cells ; 15(6): 576-588, 2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37424954

ABSTRACT

Lung cancer is the major cause of cancer-related deaths worldwide, it has one of the lowest 5-year survival rate, mainly because it is diagnosed in the late stage of the disease. Lung cancer is classified into two groups, small cell lung cancer (SCLC) and non-SCLC (NSCLC). In turn, NSCLC is categorized into three distinct cell subtypes: Adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. NSCLC is the most common lung cancer, accounting for 85% of all lung cancers. Treatment for lung cancer is linked to the cell type and stage of the disease, involving chemotherapy, radiation therapy, and surgery. Despite improvements in therapeutic treatments, lung cancer patients show high rates of recurrence, metastasis, and resistance to chemotherapy. Lung stem cells (SCs) are undifferentiated cells capable of self-renewal and proliferation, are resistant to chemotherapy and radiotherapy and, due to their properties, could be involved in the development and progression of lung cancer. The presence of SCs in the lung tissue could be the reason why lung cancer is difficult to treat. The identification of lung cancer stem cells biomarkers is of interest for precision medicine using new therapeutic agents directed against these cell populations. In this review, we present the current knowledge on lung SCs and discuss their functional role in the initiation and progression of lung cancer, as well as their role in tumor resistance to chemotherapy.

2.
Biochim Biophys Acta Gen Subj ; 1866(12): 130239, 2022 12.
Article in English | MEDLINE | ID: mdl-36064072

ABSTRACT

Thyroid hormones (THs) play an essential role in the maintenance of cardiovascular homeostasis and are involved in the modulation of cardiac contractility, heart rate, diastolic function, systemic vascular resistance, and vasodilation. THs have actions on cardiovascular physiology through the activation or repression of target genes or the activation of intracellular signals through non-genomic mechanisms. Hyperthyroidism alters certain intracellular pathways involved in the preservation of the structure and functionality of the heart, causing relevant cardiovascular disorders. Reactive oxygen species (ROS) play an important role in the cardiovascular system, but the exacerbated increase in ROS caused by chronic hyperthyroidism together with regulation on the antioxidant system have been associated with the development of cardiovascular dysfunction. In this review, we analyze the role of THs-induced oxidative stress in the cellular and molecular changes that lead to cardiac dysfunction, as well as the effectiveness of antioxidant treatments in attenuating cardiac abnormalities developed during hyperthyroidism.


Subject(s)
Hyperthyroidism , Hypothyroidism , Humans , Hypothyroidism/complications , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Thyroid Hormones/metabolism , Hyperthyroidism/complications , Oxidative Stress , Cardiovascular Physiological Phenomena
3.
Endocrinol Diabetes Nutr (Engl Ed) ; 69(6): 409-417, 2022.
Article in English | MEDLINE | ID: mdl-35817546

ABSTRACT

BACKGROUND: Graves' disease is an autoimmune disorder characterised by excessive production of thyroid hormones, which induces increased cellular metabolism in most tissues and increased production of reactive oxygen species (ROS). The aim of this work was to analyse the effect of ROS on cell viability and the expression of catalase (CAT), glutathione peroxidase-1 (GPx-1), superoxide dismutase (SOD-1) and DNA methyltransferase-1 (DNMT-1) in peripheral blood mononuclear cells (PBMC) from patients with newly diagnosed Graves' disease or treated with methimazole. PATIENTS AND METHODS: For this study, women patients with newly diagnosed Graves' disease (n=18), treated with methimazole (n=6) and healthy subjects (n=15) were recruited. ROS were evaluated by flow cytometry, and the viability/apoptosis of PBMC was analysed by flow cytometry and fluorescence microscopy. Genomic expression of CAT, GPx-1, SOD-1 and DNMT-1 was quantified by real-time PCR. RESULTS: We found high levels of ROS and increased expression of CAT, GPx-1, SOD-1 and DNMT-1 in PBMC from patients with newly diagnosed Graves' disease. Methimazole treatment reversed these parameters. Cell viability was similar in all study groups. CONCLUSIONS: ROS induces the expression of CAT, GPx-1, and SOD-1. The activity of these enzymes may contribute to the protection of PBMC from the harmful effect of free radicals on cell viability. Increased expression of DNMT-1 may be associated with aberrant methylation patterns in immunoregulatory genes contributing to autoimmunity in Graves' disease.


Subject(s)
Graves Disease , Methimazole , DNA/metabolism , Female , Graves Disease/drug therapy , Humans , Leukocytes, Mononuclear/metabolism , Methimazole/pharmacology , Methimazole/therapeutic use , Methyltransferases/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
4.
Nat Prod Res ; 36(17): 4431-4434, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34569365

ABSTRACT

Most of the deaths from skin cancer are caused by melanoma, a malignancy in which STAT3 plays a crucial role. The inhibition of STAT3 is considered a potential target to induce cell death, tumor regression and metastasis inhibition. The objective of this work was to evaluate the activity of the aqueous extract of Larrea divaricata (Aq), a fraction rich in polyphenols (EA),and the isolated compound quercetin-3-methyl ether (Q3ME) on B16F10 melanoma cells. The effects of Aq, EA and Q3ME were assessed on B16F10 cells by determining the proliferation, viability, apoptosis induction and the expression and phosphorylation of STAT3. The phytochemical composition of the extracts was determined by High Performance Liquid Chromatography. Aq, EA and Q3ME presented antiproliferative activity on B6F10 cells through p-STAT3 inhibition and early and late apoptosis induction (EC50 EA= ≤0.1 µg/ml; Aq= 316 ± 30 µg/ml; Q3ME= <0.1 µg/ml). L. divaricata could be considered for the development of adjuvant phytotherapies in melanoma treatment.


Subject(s)
Larrea , Melanoma , Cell Line , Cell Line, Tumor , Humans , Larrea/chemistry , Melanoma/drug therapy , Plant Extracts/chemistry , Plant Extracts/pharmacology , Polyphenols/pharmacology , Water
5.
Endocr Relat Cancer ; 28(7): 403-418, 2021 05 31.
Article in English | MEDLINE | ID: mdl-33908371

ABSTRACT

The patient's hormonal context plays a crucial role in the outcome of cancer. However, the association between thyroid disease and breast cancer risk remains unclear. We evaluated the effect of thyroid status on breast cancer growth and dissemination in an immunocompetent mouse model. For this, hyperthyroid and hypothyroid Balb/c mice were orthotopically inoculated with triple-negative breast cancer 4T1 cells. Tumors from hyperthyroid mice showed an increased growth rate and an immunosuppressive tumor microenvironment, characterized by increased IL-10 levels and decreased percentage of activated cytotoxic T cells. On the other hand, delayed tumor growth in hypothyroid animals was associated with increased tumor infiltration of activated CD8+ cells and a high IFNγ/IL-10 ratio. Paradoxically, hypothyroid mice developed a higher number of lung metastasis than hyperthyroid animals. This was related to an increased secretion of tumor CCL2 and an immunosuppressive systemic environment, with increased proportion of regulatory T cells and IL-10 levels in spleens. A lower number of lung metastasis in hyperthyroid mice was related to the reduced presence of mesenchymal stem cells in tumors and metastatic sites. These animals also exhibited decreased percentages of regulatory T lymphocytes and myeloid-derived suppressor cells in spleens but increased activated CD8+ cells and the IFNγ/IL-10 ratio. Therefore, thyroid hormones modulate the cellular and cytokine content of the breast tumor microenvironment. A better understanding of the mechanisms involved in these effects could be a starting point for the discovery of new therapeutic targets for breast cancer.


Subject(s)
Breast Neoplasms , Hyperthyroidism , Hypothyroidism , Lung Neoplasms , Triple Negative Breast Neoplasms , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Interleukin-10/therapeutic use , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Tumor Microenvironment
6.
Oxid Med Cell Longev ; 2019: 7471890, 2019.
Article in English | MEDLINE | ID: mdl-31281590

ABSTRACT

Hyperthyroidism is an endocrine disorder characterized by excessive secretion of thyroid hormones T3 and T4. Thyroid hormones exert pleiotropic actions on numerous tissues and induce an overall increase in metabolism, with an increase in energy demand and oxygen consumption. Therefore, the purpose of this study was to investigate the effects of hyperthyroidism on the production of reactive oxygen species (ROS) in lymph node and spleen cells of euthyroid and hyperthyroid mice, analyzing antioxidant mechanisms involved in the restitution of the cellular redox state. For this, thirty female Balb/c (H-2d) mice were randomly divided into two groups: euthyroid (by treatment with placebo) and hyperthyroid (by treatment with 12 mg/l of T4 in drinking water for 30 days). We found a significant increase in ROS and an increase in the genomic and protein expression of the antioxidant enzymes catalase (CAT) and glutathione peroxidase-1 (GPx-1) in lymph node and spleen cells of hyperthyroid mice. In vitro treatment with H2O2 (250 µM) of the lymphoid cells of euthyroid mice increased the expression levels of CAT and GPx-1. The hyperthyroidism increased the phosphorylation levels of Nrf2 (nuclear factor erythroid 2-related factor) and the kinase activity of protein kinase C (PKC) and extracellular signal-regulated kinase (ERK). Additionally, we found an increase in the expression of the classic isoenzymes of PKCα, ß and γ. In conclusion, these results indicated that the increase in ROS found in the hyperthyroid state induces the antioxidant enzyme transcription through the activation of the Nrf-2 factor in lymphoid tissues. This shows the influence of hyperthyroidism on the regulation of the cellular antioxidant system.


Subject(s)
Catalase/genetics , Glutathione Peroxidase/genetics , Hyperthyroidism/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Superoxide Dismutase-1/genetics , Animals , Catalase/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glutathione Peroxidase/biosynthesis , Hyperthyroidism/blood , Hyperthyroidism/enzymology , Hyperthyroidism/genetics , Lymphoid Tissue/metabolism , Mice , Mice, Inbred BALB C , NF-E2-Related Factor 2/genetics , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase-1/biosynthesis , Thyrotropin/blood , Thyroxine/administration & dosage , Thyroxine/blood , Transcriptional Activation , Triiodothyronine/blood , Glutathione Peroxidase GPX1
7.
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
8.
Article in English | MEDLINE | ID: mdl-30814977

ABSTRACT

T-cell lymphomas (TCL) are a heterogeneous group of aggressive clinical lymphoproliferative disorders with considerable clinical, morphological, immunophenotypic, and genetic variation, including ~10-15% of all lymphoid neoplasms. Several evidences indicate an important role of the non-neoplastic microenvironment in promoting both tumor growth and dissemination in T cell malignancies. Thus, dysregulation of integrin expression and activity is associated with TCL survival and proliferation. We found that thyroid hormones acting via the integrin αvß3 receptor are crucial factors in tumor microenvironment (TME) affecting the pathophysiology of TCL cells. Specifically, TH-activated αvß3 integrin signaling promoted TCL proliferation and induced and an angiogenic program via the up-regulation of the vascular endothelial growth factor (VEGF). This was observed both on different TCL cell lines representing the different subtypes of human hematological malignancy, and in preclinical models of TCL tumors xenotransplanted in immunodeficient mice as well. Moreover, development of solid tumors by inoculation of murine TCLs in syngeneic hyperthyroid mice, showed increased tumor growth along with increased expression of cell cycle regulators. The genomic or pharmacological inhibition of integrin αvß3 decreased VEGF production, induced TCL cell death and decreased in vivo tumor growth and angiogenesis. Here, we review the non-genomic actions of THs on TCL regulation and their contribution to TCL development and evolution. These actions not only provide novel new insights on the endocrine modulation of TCL, but also provide a potential molecular target for its treatment.

9.
Pharmacol Res ; 109: 55-63, 2016 07.
Article in English | MEDLINE | ID: mdl-26855318

ABSTRACT

Thyroid hormones (THs) are important regulators of metabolism, differentiation and cell proliferation. They can modify the physiology of human and murine T cell lymphomas (TCL). These effects involve genomic mechanisms, mediated by specific nuclear receptors (TR), as well as nongenomic mechanisms, that lead to the activation of different signaling pathways through the activation of a membrane receptor, the integrin αvß3. Therefore, THs are able to induce the survival and growth of TCL. Specifically, the signaling induced by THs through the integrin αvß3 activates proliferative and angiogenic programs, mediated by the regulation of the vascular endothelial growth factor (VEGF). The genomic or pharmacologic inhibition of integrin αvß3 reduces the production of VEGF and induces cell death both in vitro and in xenograft models of human TCL. Here we review the mechanisms involved in the modulation of the physiology of TCL induced by THs, the analysis of the interaction between genomic and nongenomic actions of THs and their contribution to T cell lymphomagenesis. These actions of THs suggest a novel mechanism for the endocrine modulation of the physiopathology of TCL and they provide a potential molecular target for its treatment.


Subject(s)
Lymphoma, T-Cell/metabolism , Receptors, Thyroid Hormone/metabolism , Thyroid Hormones/metabolism , Animals , Genomics , Humans , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/genetics , T-Lymphocytes/metabolism
10.
Toxicol In Vitro ; 27(7): 2094-104, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23933437

ABSTRACT

Novel ß-lapachone analogs 2-phenyl-3,4-dihydro-2H-benzo[h]chromene-5,6-dione (NQ1), 2-p-tolyl-3,4-dihydro-2H-benzo[h]chromene-5,6-dione (NQ3) and 2-methyl-2-phenyl-3,4-dihydro-2H-benzo[h]chromene-5,6-dione (NQ7), which have trypanocidal activity, were assayed for cytotoxic effects on murine EL-4 T lymphoma cells. The NQs inhibited the proliferation of EL-4 cells at concentrations above 1µM. Nuclear staining of the EL-4 cells revealed chromatin condensation and a nuclear morphology compatible with the induction of apoptosis. Flow cytometry assays with annexin V-FITC and propidium iodide confirmed the cell death by apoptosis. Using electron paramagnetic resonance (EPR), a semiquinone radical was detected in EL-4 cells treated with NQs. In addition, a decrease in the GSH level in parallel with reactive oxygen species (ROS) production was observed. Preincubation with n-acetyl-l-cysteine (NAC) was able to reverse the inhibitory effects of the NQs on cell proliferation, indicating that ROS generation is involved in NQ-induced apoptosis. In addition, the NQs induced a decrease in the mitochondrial membrane potential and increased the proteolytic activation of caspases 9 and 3 and the cleavage of Poly (ADP-Ribose) Polymerase (PARP). In conclusion, these results indicate that redox cycling is induced by the NQs in the EL-4 cell line, with the generation of ROS and other free radicals that could inhibit cellular proliferation as a result of the induction of the intrinsic apoptosis pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lymphoma, T-Cell/drug therapy , Naphthoquinones/pharmacology , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects , Acetylcysteine/pharmacology , Animals , Antineoplastic Agents/antagonists & inhibitors , Benzopyrans/antagonists & inhibitors , Benzopyrans/pharmacology , Benzoquinones/metabolism , Cell Line, Tumor , Cell Nucleus Shape/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromatin Assembly and Disassembly/drug effects , Free Radical Scavengers/pharmacology , Glutathione/antagonists & inhibitors , Glutathione/metabolism , Kinetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Naphthoquinones/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Trypanocidal Agents/antagonists & inhibitors , Trypanocidal Agents/pharmacology
11.
Cancer Invest ; 28(2): 135-45, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19968502

ABSTRACT

D-limonene is a common monoterpene found in nature. Previously, it has been demonstrated that it has antiproliferative effects on a lymphoma cell line by increasing the nitric oxide (NO) level. In the present work this mechanism is analyzed by evaluating the participation of MAP38 and ERK pathways. Limonene increased the NO levels by inducing cell apoptosis by two mechanisms: through the production of H(2)O(2) and ERK pathway activation at low concentrations, and through the inhibition of farnesylation of proteins and O(2)- production at high concentrations. Both mechanisms were related to the increase in total nitrites, and the production of oxidative stress intermediates was involved.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cyclohexenes/pharmacology , Lymphoma/metabolism , Lymphoma/pathology , Nitric Oxide/metabolism , Oxidative Stress , Terpenes/pharmacology , Cell Line, Tumor , Humans , Hydrogen Peroxide/metabolism , Limonene , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Superoxides/metabolism , NF-kappaB-Inducing Kinase
12.
Int J Biochem Cell Biol ; 42(2): 273-81, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19914395

ABSTRACT

We previously showed that the antimicrobial peptide microcin J25 induced the over-production of reactive oxygen species with the concomitant release of cytochrome c from rat heart mitochondria via the opening of the mitochondrial permeability transition pore. Here, we were able to demonstrate that indeed, as a consequence of the oxidative burst, MccJ25 induces carbonylation of mitochondrial proteins, which may explain the irreversible inhibition of complex III and the partial inhibition of superoxide dismutase and catalase. Moreover, the peptide raised the levels of oxidized membrane lipids, which triggers the release of cytochrome c. From in silico analysis, we hypothesize that microcin would elicit these effects through interaction with heme c1 at mitochondrial complex III. On the other hand, under an excess of l-arginine, MccJ25 caused nitric oxide overproduction with no oxidative damage and a marked inhibition in oxygen consumption. Therefore, a beneficial anti-oxidative activity could be favored by the addition of l-arginine. Conversely, MccJ25 pro-oxidative-apoptotic effect can be unleashed in either an arginine-free medium or by suppressing the nitric oxide synthase activity.


Subject(s)
Bacteriocins/pharmacology , Cytochromes c/metabolism , Membrane Lipids/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Alkenes/chemistry , Alkenes/metabolism , Animals , Cytochromes c/chemistry , Enzyme Inhibitors/pharmacology , Mitochondria/enzymology , Mitochondrial Proteins/chemistry , Models, Molecular , Nitric Oxide/biosynthesis , Oxidation-Reduction , Protein Carbonylation/drug effects , Protein Conformation , Rats , Rats, Wistar
13.
Neuroimmunomodulation ; 15(1): 68-75, 2008.
Article in English | MEDLINE | ID: mdl-18667802

ABSTRACT

Autoimmune thyroid diseases (AITD) are the most common organ-specific autoimmune disorders affecting approximately 5% of the overall population. An aberrant interaction between abnormal thyrocytes, abnormal antigen-presenting cells and abnormal T cells forms the basis for the atypical autoimmune reaction targeting thyroid antigens. It was proposed that nongenetic (environmental and hormonal) factors play a crucial etiological role in AITD development, through altering immune-endocrine interactions. The most outstanding fact is that in genetically predisposed individuals, the disruption of these neuroendocrine-immune interactions by environmental factors results in thyroid autoimmune dysfunction. These interactions are able to incline the balance between Th1-Th2 immune response toward one side, resulting in a Th1-cell-mediated autoimmune reaction with thyrocyte destruction and hypothyroidism in Hashimoto's thyroiditis but to a hyperreactive Th2-mediated humoral response against TSH receptor with stimulatory antibodies leading to Graves' disease hyperthyroidism. In this review the main mechanisms involved are summarized. In this sense, the participation of stress-mediated activation of the sympathoadrenal system and hypothalamic-pituitary-adrenal axis, the hormonal changes occurring during pregnancy and postpartum acting on antigen-presenting cells and influencing, in this way, the balance of the immune status are shown to participate in AITD etiology. The possibility that altered levels of thyroid hormones during the course of the AITD may alter immune function is also discussed.


Subject(s)
Autoimmune Diseases/immunology , Endocrine System Diseases/immunology , Immune System/immunology , Thyroid Diseases/immunology , Animals , Autoimmune Diseases/physiopathology , Endocrine System Diseases/physiopathology , Humans , Immune System/physiopathology , Immunity, Cellular/immunology , Receptors, Thyrotropin/immunology , Stress, Psychological/immunology , Stress, Psychological/physiopathology , T-Lymphocytes/immunology , Thyroid Diseases/physiopathology , Thyroid Gland/immunology , Thyroid Gland/physiopathology
14.
Life Sci ; 79(21): 2043-8, 2006 Oct 19.
Article in English | MEDLINE | ID: mdl-16860346

ABSTRACT

Some coumarins possess enhancing effects on lymphocyte mitogen responsiveness. In this investigation, the activity of scopoletin, a coumarin that has been isolated from different plants and in this case specifically from T. cordata Mill., was evaluated. For this purpose, normal T lymphocytes and a hyperproliferative T lymphoma cell line were used. Scopoletin was found to exert a dual action on tumoral lymphocytes exhibiting both a cytostatic and a cytotoxic effect. These effects varied with the concentrations analysed and the time of cell incubation (EC(50): 251+/-15 microg/ml) and were associated to the induction of apoptosis. Scopoletin induced cell proliferation on normal T lymphocytes (Proliferation stimulation index: 1 microg/ml scopoletin: 1.26+/-0.1; 10 microg/ml scopoletin: 3+/-0.25; 100 microg/ml scopoletin: 1.86+/-0.08); this stimulatory action was found to be due to the interaction with kinase C (PKC) protein. These results indicate that scopoletin could be a potential antitumoral compound to be used for cancer treatment.


Subject(s)
Apoptosis/drug effects , Immunologic Factors/pharmacology , Scopoletin/pharmacology , T-Lymphocytes/drug effects , Animals , Apoptosis/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Concanavalin A/pharmacology , Dose-Response Relationship, Drug , Flowers/chemistry , Immunologic Factors/isolation & purification , Male , Mice , Mice, Inbred C3H , Scopoletin/isolation & purification , T-Lymphocytes/immunology , Tilia/chemistry
15.
J Endocrinol ; 189(1): 45-55, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16614380

ABSTRACT

Thyroid hormones play critical roles in differentiation, growth and metabolism, but their participation in immune system regulation has not been completely elucidated. Modulation of in vivo thyroid status was used to carry out an integrative analysis of the role of the hypothalamus-pituitary-thyroid (HPT) axis in T and B lymphocyte activity. The participation of the protein kinase C (PKC) signaling pathway and the release of some cytokines upon antigenic stimulation were analyzed. Lymphocytes from hyperthyroid mice displayed higher T-and B-cell mitogen-induced proliferation, and those from hypothyroid mice displayed lower T- and B-cell mitogen-induced proliferation, compared with euthyroid animals. Reversion of hypothyroid state by triiodothyronine (T3) administration recovered the proliferative responses. No differences were found in lymphoid subset balance. Both total PKC content and mitogen-induced PKC translocation were higher in T and B cells from hyperthyroid mice, and lower in cells from hypothyroid mice, compared with controls. Levels of thyroid-stimulating (TSH) and TSH-releasing (TRH) hormones were not directly related to lymphocyte proliferative responses. After immunization with sheep red blood cells (SRBCs) and re-stimulation, in vitro spleen cells from hyper- or hypothyroid mice showed, respectively, increased or decreased production of interleukin (IL)-2 and interferon (IFN)-gamma cytokines. Additionally, an increase in IL-6 and IFN-gamma levels was found in hyperthyroid cells after in vivo injection and in vitro re-stimulation with lipopolysaccharide (LPS). Our results show for the first time a thyroid hormone-mediated regulation of PKC content and of cytokine production in lymphocytes; this regulation could be involved in the altered responsiveness to mitogen-induced proliferation of T and B cells. The results also confirm the important role that these hormones play in regulating lymphocyte reactivity.


Subject(s)
Hypothalamus/immunology , Lymphocytes/immunology , Pituitary Gland/immunology , Protein Kinase C/immunology , Thyroid Gland/immunology , Animals , Antigens, CD/immunology , B-Lymphocytes/immunology , Cell Division/immunology , Cell Membrane/immunology , Cells, Cultured , Cytokines/immunology , Female , Hypothalamo-Hypophyseal System/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mitogens/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Thyroid Hormones/blood , Thyrotropin/blood
16.
Am J Physiol Cell Physiol ; 291(2): C327-36, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16495371

ABSTRACT

Regulation of cell proliferation by thyroid hormone (TH) has been demonstrated, but the effect of THs and the mechanisms involved in lymphocyte activity have not been elucidated. Differential expression of PKC isoenzymes and high nitric oxide synthase (NOS) activity have been described in tumor T lymphocytes. We have analyzed the direct actions of TH on normal T lymphocytes and BW5147 T lymphoma cells in relation to PKC and NOS activities. THs increased tumor and mitogen-induced normal T lymphocyte proliferation. PKC isoenzyme-selective blockers impaired these effects in both cell types, indicating the participation of Ca2+-dependent and -independent isoenzymes in normal and tumor cells, respectively. TH actions were blunted by extra- and intracellular Ca2+ blockers only in normal T lymphocytes, whereas NOS blockers impaired TH-induced proliferation in T lymphoma cells. Incubation for 24 h with TH induced a rise in total and membrane-associated PKC activities in both cell types and led to a rapid and transient effect only in tumor cells. THs increased atypical PKC-zeta expression in BW5147 cells and classical PKC isoenzymes in mitogen-stimulated normal T cells. TH augmented NOS activity and inducible NOS protein and gene expression only in tumor cells. Blockade of PKC and the atypical PKC-zeta isoform inhibited TH-mediated stimulation of inducible NOS and cell proliferation. These results show, for the first time, that differential intracellular signals are involved in TH modulation of lymphocyte physiology and pathophysiology.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Lymphoma, T-Cell/enzymology , Nitric Oxide Synthase Type II/metabolism , Protein Kinase C/metabolism , Thyroid Hormones/administration & dosage , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Mice
17.
Phytother Res ; 20(1): 34-40, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16397918

ABSTRACT

Aqueous extracts or infusions obtained from the flowers of Tilia species are widely used for the treatment of anxiety in folk medicine. In this work, the antiproliferative action of aqueous, dichloromethane and ethanol extracts obtained from Tilia cordata Mill. flowers on tumoral (BW 5147 lymphoma) and normal lymphocytes is described. Both extracts showed a selective action on tumoral cells, inducing apoptosis. In the case of normal T cells these extracts suppressed mitogen-induced proliferation without affecting viability, suggesting a suppressive but not cytotoxic effect. These effects were clearly concentration dependent. A coumarin (scopoletin), the main component in the dichloromethane extract presented antiproliferative action on BW 5147 cells, suggesting that it may be at least partly responsible for the activity displayed by this extract.


Subject(s)
Lymphocytes/drug effects , Phytotherapy , Plant Extracts/pharmacology , Scopoletin/pharmacology , Tilia/chemistry , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Flowers/chemistry , Lymphocytes/cytology , Lymphocytes/pathology , Male , Methylene Chloride , Mice , Mice, Inbred C3H , Necrosis/chemically induced , Plant Extracts/therapeutic use , Scopoletin/isolation & purification , Solvents
18.
Neuroimmunomodulation ; 12(2): 92-9, 2005.
Article in English | MEDLINE | ID: mdl-15785111

ABSTRACT

OBJECTIVE: The aim of this work was to analyze beta-adrenergic receptor (betaAR) regulation of T-lymphocyte proliferation in mice according to different thyroid hormone statuses. METHODS: T cells from eu-, hypo- (by propylthiouracil treatment) and hyperthyroid (by thyroxine, T4 administration) mice were purified and specific radioligand binding assays were performed. The effects of the beta-agonist isoproterenol (ISO) on intracellular levels of cyclic AMP (cAMP) were determined. Mitogen-induced T-cell proliferation was measured by [(3)H]-thymidine incorporation. Finally, protein kinase C (PKC) activity in cytosol and membrane fractions were determined using radiolabelled enzymatic substrates. RESULTS: Adecrease or a non-significant increase in betaAR number was found on T lymphocytes from hypo- and hyperthyroid mice compared to euthyroid controls. ISO stimulation of cAMP levels was lower in hypothyroid and higher in hyperthyroid T lymphocytes compared to controls. T-selective mitogen-induced proliferation was increased in T4-treated animals, but decreased in hypothyroid mice. During the peak of proliferation, downregulation of betaAR was observed in all animals. However, a higher or a lower decrease was observed in hyper- and hypothyroid T cells, respectively. In parallel, a higher translocation of PKC activity was observed in hyperthyroid cells, and a lower one was found in hypothyroid lymphocytes with respect to controls. CONCLUSIONS: These results indicate that intracellular signals triggered by mitogen activation, namely PKC, would be related to differential betaAR downregulation in T lymphocytes depending on the thyroid hormone status, contributing to the distinct proliferative responses found in hypo- or hyperthyroidism compared to the euthyroid state.


Subject(s)
Cell Proliferation/drug effects , Mitogens/pharmacology , Neuroimmunomodulation/immunology , Receptors, Adrenergic, beta/drug effects , T-Lymphocytes/metabolism , Thyroid Gland/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Cyclic AMP/metabolism , Down-Regulation/drug effects , Down-Regulation/immunology , Female , Hyperthyroidism/chemically induced , Hyperthyroidism/immunology , Hyperthyroidism/metabolism , Hypothyroidism/chemically induced , Hypothyroidism/immunology , Hypothyroidism/metabolism , Isoproterenol/pharmacology , Mice , Mice, Inbred BALB C , Neuroimmunomodulation/genetics , Propylthiouracil/pharmacology , Protein Kinase C/metabolism , Protein Transport/drug effects , Protein Transport/immunology , Receptor Aggregation/drug effects , Receptor Aggregation/immunology , Receptors, Adrenergic, beta/immunology , Receptors, Adrenergic, beta/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Thymidine/metabolism , Thyroid Gland/immunology , Thyroxine/pharmacology
19.
Nitric Oxide ; 8(2): 111-8, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12620374

ABSTRACT

Nitric oxide (NO)-derived from T lymphocytes in an autocrine fashion can modulate events in the cell. However, the exact role of NO on the control of lymphocyte growth is controversial since both stimulation and inhibition have been demonstrated. Nitric oxide synthase (NOS) activity in normal and tumor T lymphocyte proliferation was studied here. Resting normal T lymphocytes displayed low levels of NOS activity that were slightly increased upon mitogenic stimulation. In contrast, BW5147 T lymphoma cells displayed higher basal levels than normal T lymphocytes that were significantly augmented when induced to proliferate. This activity was slightly modified in the presence of the calcium chelator EGTA and was blocked by competitive and irreversible NOS inhibitors, as well as by selective blockers of iNOS. Furthermore, tumor but not normal cell proliferation was impaired by NOS and iNOS blockers, while a calcium blocker only affected normal cell growth. iNOS expression, both at the protein and at the mRNA levels, was demonstrated on growing BW5147 cells but not on arrested tumor or normal lymphocytes. The contribution of iNOS to sustained proliferation of tumor cells is discussed.


Subject(s)
Cell Line, Tumor/drug effects , Nitric Oxide Synthase/physiology , Animals , Calcium Channel Blockers/pharmacology , Cell Division , Cell Line, Tumor/enzymology , Cell Line, Tumor/pathology , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Lymphocyte Activation , Lymphoma , Mitogens/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
20.
Biochim Biophys Acta ; 1588(2): 179-88, 2002 Nov 20.
Article in English | MEDLINE | ID: mdl-12385783

ABSTRACT

Protein kinase C (PKC) is critical for T lymphocyte activation and proliferation, while nitric oxide synthase (NOS) may function both as an activator or inhibitor of T cell apoptosis. Both enzymatic activities were studied in T lymphoma cells in comparison to normal and activated T lymphocytes. Here we show a higher translocation of PKC in BW5147 lymphoma cells than in mitogen-stimulated T lymphocytes. Tumor cells overexpressed PKC zeta isoform, while high levels of the PKC beta isotype were found in mitogen-stimulated T lymphocytes. Moreover, tumoral T cells showed high NOS activity, almost undetectable in normal or stimulated T lymphocytes. PKC and NOS inhibitors or the intracellular delivery of an anti-PKC zeta antibody diminished both NO production and proliferation in tumor cells. These results suggest that atypical PKC zeta isoform expression and its association with NOS activity regulation would participate in the multistep process leading to BW5147 cell malignant transformation.


Subject(s)
Nitric Oxide Synthase/metabolism , Protein Kinase C/biosynthesis , Animals , Antibodies/pharmacology , Cell Division , Cell Survival , Isoenzymes/biosynthesis , Isoenzymes/immunology , Lymphoma, T-Cell , Mice , Mitogens , Protein Kinase C/immunology , Protein-Tyrosine Kinases/metabolism , Tumor Cells, Cultured , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...