Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Thyroid ; 31(2): 299-314, 2021 02.
Article in English | MEDLINE | ID: mdl-32935630

ABSTRACT

Background: The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcription factor is a key regulator of cell survival, proliferation, and gene expression. Although activation of NF-κB signaling in thyroid follicular cells after thyrotropin (TSH) receptor (TSHR) engagement has been reported, the downstream signaling leading to NF-κB activation remains unexplored. Here, we sought to elucidate the mechanisms that regulate NF-κB signaling activation in response to TSH stimulation. Methods: Fisher rat-derived thyroid cell lines and primary cultures of NF-κB essential modulator (NEMO)-deficient mice thyrocytes were used as models. Signaling pathways leading to the activation of NF-κB were investigated by using chemical inhibitors and phospho-specific antibodies. Luciferase reporter gene assays and site-directed mutagenesis were used to monitor NF-κB-dependent gene transcriptional activity and the expression of thyroid differentiation markers was assessed by reverse transcription quantitative polymerase chain reaction and Western blot, respectively. Chromatin immunoprecipitation (ChIP) was carried out to investigate NF-κB subunit p65 DNA binding, and small interfering RNA (siRNA)-mediated gene knockdown approaches were used for studying gene function. Results: Using thyroid cell lines, we observed that TSH treatment leads to protein kinase C (PKC)-mediated canonical NF-κB p65 subunit nuclear expression. Moreover, TSH stimulation phosphorylated the kinase TAK-1, and its knockdown abolished TSH-induced NF-κB transcriptional activity. TSH induced the transcriptional activity of the NF-κB subunit p65 in a protein kinase A (PKA)-dependent phosphorylation at Ser-276. In addition, p65 phosphorylation at Ser-276 induced acetyl transferase p300 recruitment, leading to its acetylation on Lys-310 and thereby enhancing its transcriptional activity. Evaluation of the role played by NF-κB in thyroid physiology demonstrated that the canonical NF-κB inhibitor BAY 11-7082 reduced TSH-induced expression of thyroid differentiation markers. The involvement of NF-κB signaling in thyroid physiology was confirmed by assessing the TSH-induced gene expression in primary cultures of NEMO-deficient mice thyrocytes. ChIP and the knockdown experiments revealed that p65 is a nuclear effector of TSH actions, inducing the transcripcional expression of thyroid differentiation markers. Conclusions: Taken together, our results point to NF-κB being a pivotal mediator in the TSH-induced thyroid follicular cell differentiation, a relevant finding with potential physiological and pathophysiological implications.


Subject(s)
Cell Differentiation/drug effects , Thyroid Gland/drug effects , Thyrotropin/pharmacology , Transcription Factor RelA/metabolism , Acetylation , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice, Knockout , Phosphorylation , Protein Kinase C/metabolism , Rats, Inbred F344 , Signal Transduction , Thyroid Gland/metabolism , Transcription Factor RelA/genetics , p300-CBP Transcription Factors/metabolism
2.
Mol Cancer Res ; 16(5): 833-845, 2018 05.
Article in English | MEDLINE | ID: mdl-29523762

ABSTRACT

Emerging evidence suggests that unregulated Toll-like receptor (TLR) signaling promotes tumor survival signals, thus favoring tumor progression. Here, the mechanism underlying TLR4 overexpression in papillary thyroid carcinomas (PTC) mainly harboring the BRAFV600E mutation was studied. TLR4 was overexpressed in PTC compared with nonneoplastic thyroid tissue. Moreover, paired clinical specimens of primary PTC and its lymph node metastasis showed a significant upregulation of TLR4 levels in the metastatic tissues. In agreement, conditional BRAFV600E expression in normal rat thyroid cells and mouse thyroid tissue upregulated TLR4 expression levels. Furthermore, functional TLR4 expression was demonstrated in PTC cells by increased NF-κB transcriptional activity in response to the exogenous TLR4-agonist lipopolysaccharide. Of note, The Cancer Genome Atlas data analysis revealed that BRAFV600E-positive tumors with high TLR4 expression were associated with shorter disease-free survival. Transcriptomic data analysis indicated a positive correlation between TLR4 expression levels and MAPK/ERK signaling activation. Consistently, chemical blockade of MAPK/ERK signaling abrogated BRAFV600E-induced TLR4 expression. A detailed study of the TLR4 promoter revealed a critical MAPK/ERK-sensitive Ets-binding site involved in BRAFV600E responsiveness. Subsequent investigation revealed that the Ets-binding factor ETS1 is critical for BRAFV600E-induced MAPK/ERK signaling-dependent TLR4 gene expression. Together, these data indicate that functional TLR4 overexpression in PTCs is a consequence of thyroid tumor-oncogenic driver dysregulation of MAPK/ERK/ETS1 signaling.Implications: Considering the participation of aberrant NF-κB signaling activation in the promotion of thyroid tumor growth and the association of high TLR4 expression with more aggressive tumors, this study suggests a prooncogenic potential of TLR4 downstream signaling in thyroid tumorigenesis. Mol Cancer Res; 16(5); 833-45. ©2018 AACR.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Protein c-ets-1/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/metabolism , Toll-Like Receptor 4/metabolism , Animals , Female , Humans , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Rats , Rats, Inbred F344 , Signal Transduction , Thyroid Cancer, Papillary/pathology , Toll-Like Receptor 4/genetics , Transfection
3.
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
4.
Mol Cell Endocrinol ; 437: 292-301, 2016 12 05.
Article in English | MEDLINE | ID: mdl-27568464

ABSTRACT

Iodide has direct effects on thyroid function. Several iodinated lipids are biosynthesized by the thyroid and they were postulated as intermediaries in the action of iodide. Among them, 2-iodohexadecanal (2-IHDA) has been identified and proposed to play a role in thyroid autoregulation. The aim of this study was to compare the effect of iodide and 2-IHDA on thyroid cell physiology. For this purpose, FRTL-5 thyroid cells were incubated with the two compounds during 24 or 48 h and several thyroid parameters were evaluated such as: iodide uptake, intracellular calcium and H2O2 levels. To further explore the molecular mechanism involved in 2-IHDA action, transcript and protein levels of genes involved in thyroid hormone biosynthesis, as well as the transcriptional expression of these genes were evaluated in the presence of iodide and 2-IHDA. The results obtained indicate that 2-IHDA reproduces the action of excess iodide on the "Wolff-Chaikoff" effect as well as on thyroid specific genes transcription supporting its role in thyroid autoregulation.


Subject(s)
Aldehydes/pharmacology , Thyroid Gland/cytology , Thyroid Gland/physiology , Animals , Calcium/metabolism , Cell Line , Deoxyglucose/metabolism , Gene Expression Regulation/drug effects , Hydrogen Peroxide/metabolism , Iodides/metabolism , Organ Specificity/genetics , Promoter Regions, Genetic/genetics , Rats , Thyroid Hormones/biosynthesis , Transcription Factors/metabolism , Transcription, Genetic/drug effects
5.
Mol Cell Endocrinol ; 420: 105-15, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26610751

ABSTRACT

Thyroid peroxidase (TPO) is essential for thyroid hormone synthesis mediating the covalent incorporation of iodine into tyrosine residues of thyroglobulin process known as organification. Thyroid-stimulating hormone (TSH) via cAMP signaling is the main hormonal regulator of TPO gene expression. In thyroid cells, TSH-stimulated nitric oxide (NO) production inhibits TSH-induced thyroid-specific gene expression, suggesting a potential autocrine role of NO in modulating thyroid function. Indeed, NO donors downregulate TSH-induced iodide accumulation and organification in thyroid cells. Here, using FRTL-5 thyroid cells as model, we obtained insights into the molecular mechanism underlying the inhibitory effects of NO on iodide organification. We demonstrated that NO donors inhibited TSH-stimulated TPO expression by inducing a cyclic guanosine monophosphate-dependent protein kinase-mediated transcriptional repression of the TPO gene. Moreover, we characterized the FoxE1 binding site Z as mediator of the NO-inhibited TPO expression. Mechanistically, we demonstrated that NO decreases TSH-induced FoxE1 expression, thus repressing the transcripcional activation of TPO gene. Taken together, we provide novel evidence reinforcing the inhibitory role of NO on thyroid cell function, an observation of potential pathophysiological relevance associated with human thyroid pathologies that come along with changes in the NO production.


Subject(s)
Forkhead Transcription Factors/metabolism , Iodide Peroxidase/metabolism , Nitric Oxide/metabolism , Thyrotropin/pharmacology , Animals , Cattle , Cell Line , Cyclic AMP/pharmacology , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Down-Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Iodide Peroxidase/genetics , Nitric Oxide Donors/pharmacology , Nitrites/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Rats , Signal Transduction/drug effects , Transcription, Genetic/drug effects
6.
Endocrinology ; 156(12): 4741-54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26587909

ABSTRACT

Nitric oxide (NO) is a ubiquitous signaling molecule involved in a wide variety of cellular physiological processes. In thyroid cells, NO-synthase III-endogenously produced NO reduces TSH-stimulated thyroid-specific gene expression, suggesting a potential autocrine role of NO in modulating thyroid function. Further studies indicate that NO induces thyroid dedifferentiation, because NO donors repress TSH-stimulated iodide (I(-)) uptake. Here, we investigated the molecular mechanism underlying the NO-inhibited Na(+)/I(-) symporter (NIS)-mediated I(-) uptake in thyroid cells. We showed that NO donors reduce I(-) uptake in a concentration-dependent manner, which correlates with decreased NIS protein expression. NO-reduced I(-) uptake results from transcriptional repression of NIS gene rather than posttranslational modifications reducing functional NIS expression at the plasma membrane. We observed that NO donors repress TSH-induced NIS gene expression by reducing the transcriptional activity of the nuclear factor-κB subunit p65. NO-promoted p65 S-nitrosylation reduces p65-mediated transactivation of the NIS promoter in response to TSH stimulation. Overall, our data are consistent with the notion that NO plays a role as an inhibitory signal to counterbalance TSH-stimulated nuclear factor-κB activation, thus modulating thyroid hormone biosynthesis.


Subject(s)
Gene Expression Regulation/drug effects , Iodine/metabolism , Nitric Oxide Donors/pharmacology , RNA, Messenger/drug effects , Symporters/drug effects , Thyroid Gland/drug effects , Thyrotropin/metabolism , Transcription Factor RelA/drug effects , Transcriptional Activation/drug effects , Animals , Autocrine Communication , Cell Line , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitroprusside/pharmacology , Promoter Regions, Genetic , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , S-Nitrosoglutathione/pharmacology , Spermine/analogs & derivatives , Spermine/pharmacology , Symporters/genetics , Thyroid Gland/cytology , Thyroid Gland/metabolism , Transcription Factor RelA/metabolism
7.
PLoS One ; 10(6): e0130144, 2015.
Article in English | MEDLINE | ID: mdl-26091289

ABSTRACT

In this work, we have investigated the involvement of DmCatD, a cathepsin D-like peptidase, and acid phosphatase in the process of follicular atresia of Dipetalogaster maxima, a hematophagous insect vector of Chagas' disease. For the studies, fat bodies, ovaries and hemolymph were sampled from anautogenous females at representative days of the reproductive cycle: pre-vitellogenesis, vitellogenesis as well as early and late atresia. Real time PCR (qPCR) and western blot assays showed that DmCatD was expressed in fat bodies and ovaries at all reproductive stages, being the expression of its active form significantly higher at the atretic stages. In hemolymph samples, only the immunoreactive band compatible with pro-DmCatD was observed by western blot. Acid phosphatase activity in ovarian tissues significantly increased during follicular atresia in comparison to pre-vitellogenesis and vitellogenesis. A further enzyme characterization with inhibitors showed that the high levels of acid phosphatase activity in atretic ovaries corresponded mainly to a tyrosine phosphatase. Immunofluorescence assays demonstrated that DmCatD and tyrosine phosphatase were associated with yolk bodies in vitellogenic follicles, while in atretic stages they displayed a different cellular distribution. DmCatD and tyrosine phosphatase partially co-localized with vitellin. Moreover, their interaction was supported by FRET analysis. In vitro assays using homogenates of atretic ovaries as the enzyme source and enzyme inhibitors demonstrated that DmCatD, together with a tyrosine phosphatase, were necessary to promote the degradation of vitellin. Taken together, the results strongly suggested that both acid hydrolases play a central role in early vitellin proteolysis during the process of follicular atresia.


Subject(s)
Acid Phosphatase/physiology , Cathepsin D/physiology , Follicular Atresia , Hemiptera/enzymology , Insect Proteins/physiology , Insect Vectors/enzymology , Acid Phosphatase/chemistry , Animals , Cathepsin D/chemistry , Chagas Disease/parasitology , Fat Body/enzymology , Female , Gene Expression , Hemiptera/parasitology , Hemolymph/enzymology , Humans , Hydrogen-Ion Concentration , Insect Proteins/chemistry , Insect Vectors/parasitology , MCF-7 Cells , Male , Organ Specificity , Ovary/enzymology , Proteolysis , Trypanosoma cruzi/physiology , Vitellins/chemistry , Vitellins/metabolism
8.
Endocrinology ; 153(12): 6114-25, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23064013

ABSTRACT

Thyroid peroxidase (TPO), a tissue-specific enzyme expressed in differentiated thyroid follicular cells, is a major antigen that has been linked to autoimmune thyroid disease. We have previously reported the functional expression of the lipopolysaccharide (LPS) receptor Toll-like receptor 4 on thyroid follicular cells. Here we investigated the effect of LPS in TPO expression and analyzed the mechanisms involved. We found a dose-dependent enhancement of TSH-induced TPO expression in response to LPS stimulation. EMSAs demonstrated that LPS treatment increased thyroid transcription factor-1 and -2 binding to the B and Z regions of TPO promoter, respectively. Moreover, LPS increased TSH-stimulated TPO promoter activity. Using bioinformatic analysis, we identified a conserved binding site for transcription nuclear factor-κB (NF-κB) in the TPO promoter. Chemical inhibition of NF-κB signaling and site-directed mutagenesis of the identified κB-cis-acting element abolished LPS stimulation. Furthermore, chromatin immunoprecipitation assays confirmed that TPO constitutes a novel NF-κB p65 subunit target gene in response to LPS. Additionally, our results indicate that p65 phosphorylation of serine 536 constitutes an essential step in the p65-dependent, LPS-induced transcriptional expression of TPO. In conclusion, here we demonstrated that LPS increases TPO expression, suggesting a novel mechanism involved in the regulation of a major thyroid autoantigen. Our results provide new insights into the potential effects of infectious processes on thyroid homeostasis.


Subject(s)
Gene Expression Regulation , Iodide Peroxidase/biosynthesis , Lipopolysaccharides/metabolism , NF-kappa B/metabolism , Animals , Binding Sites , Cell Line , Chromatin Immunoprecipitation , Computational Biology/methods , Humans , Mice , Mutagenesis, Site-Directed , Phosphorylation , Promoter Regions, Genetic , Rats , Serine/chemistry , Toll-Like Receptor 4/metabolism
9.
J Clin Endocrinol Metab ; 96(7): E1100-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21565787

ABSTRACT

CONTEXT: Iodide transport defect (ITD) is an autosomal recessive disorder caused by impaired Na(+)/I(-) symporter (NIS)-mediated active iodide accumulation into thyroid follicular cells. Clinical manifestations comprise a variable degree of congenital hypothyroidism and goiter, and low to absent radioiodide uptake, as determined by thyroid scintigraphy. Hereditary molecular defects in NIS have been shown to cause ITD. OBJECTIVE: Our objective was to perform molecular studies on NIS in a patient with congenital hypothyroidism presenting a clinical ITD phenotype. DESIGN: The genomic DNA encoding NIS was sequenced, and an in vitro functional study of a newly identified NIS mutation was performed. RESULTS: The analysis revealed the presence of an undescribed homozygous C to T transition at nucleotide -54 (-54C>T) located in the 5'-untranslated region in the NIS sequence. Functional studies in vitro demonstrated that the mutation was associated with a substantial decrease in iodide uptake when transfected into Cos-7 cells. The mutation severely impaired NIS protein expression, although NIS mRNA levels remained similar to those in cells transfected with wild-type NIS, suggesting a translational deficiency elicited by the mutation. Polysome profile analysis demonstrated reduced levels of polyribosomes-associated mutant NIS mRNA, consistent with reduced translation efficiency. CONCLUSIONS: We described a novel mutation in the 5'-untranslated region of the NIS gene in a newborn with congenital hypothyroidism bearing a clinical ITD phenotype. Functional evaluation of the molecular mechanism responsible for impaired NIS-mediated iodide concentration in thyroid cells indicated that the identified mutation reduces NIS translation efficiency with a subsequent decrease in protein expression and function.


Subject(s)
5' Untranslated Regions , Congenital Hypothyroidism/genetics , Mutation , Symporters/genetics , Humans , Infant, Newborn , Male
10.
Mol Endocrinol ; 24(9): 1846-62, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20667985

ABSTRACT

The Gram-negative bacterial endotoxin lipopolysaccharide (LPS) elicits a variety of biological responses. Na(+)/I(-) symporter (NIS)-mediated iodide uptake is the main rate-limiting step in thyroid hormonogenesis. We have recently reported that LPS stimulates TSH-induced iodide uptake. Here, we further analyzed the molecular mechanism involved in the LPS-induced NIS expression in Fisher rat thyroid cell line 5 (FRTL-5) thyroid cells. We observed an increase in TSH-induced NIS mRNA expression in a dose-dependent manner upon LPS treatment. LPS enhanced the TSH-stimulated NIS promoter activity denoting the NIS-upstream enhancer region (NUE) as responsible for the stimulatory effects. We characterized a novel putative conserved kappaB site for the transcription factor nuclear factor-kappaB (NF-kappaB) within the NUE region. NUE contains two binding sites for the transcription factor paired box 8 (Pax8), main regulator of NIS transcription. A physical interaction was observed between the NF-kappaB p65 subunit and paired box 8 (Pax8), which appears to be responsible for the synergic effect displayed by these transcription factors on NIS gene transcription. Moreover, functional blockage of NF-kappaB signaling and site-directed mutagenesis of the kappaB cis-acting element abrogated LPS stimulation. Silencing expression of p65 confirmed its participation as an effector of LPS-induced NIS stimulation. Furthermore, chromatin immunoprecipitation corroborated that NIS is a novel target gene for p65 transactivation in response to LPS. Moreover, we were able to corroborate the LPS-stimulatory effect on thyroid cells in vivo in LPS-treated rats, supporting that thyrocytes are capable of responding to systemic infections. In conclusion, our results reveal a new mechanism involving p65 in the LPS-induced NIS expression, denoting a novel aspect in thyroid cell differentiation.


Subject(s)
Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Paired Box Transcription Factors/metabolism , Symporters/genetics , Transcription Factor RelA/metabolism , Animals , Base Sequence , Binding Sites , Enhancer Elements, Genetic/genetics , Gene Silencing/drug effects , Humans , Molecular Sequence Data , PAX8 Transcription Factor , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Symporters/metabolism , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyrotropin/pharmacology , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...