Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 30(7): 1352-1366, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37921808

RESUMO

PURPOSE: Patients with aggressive thyroid cancer are frequently failed by the central therapy of ablative radioiodide (RAI) uptake, due to reduced plasma membrane (PM) localization of the sodium/iodide symporter (NIS). We aimed to understand how NIS is endocytosed away from the PM of human thyroid cancer cells, and whether this was druggable in vivo. EXPERIMENTAL DESIGN: Informed by analysis of endocytic gene expression in patients with aggressive thyroid cancer, we used mutagenesis, NanoBiT interaction assays, cell surface biotinylation assays, RAI uptake, and NanoBRET to understand the mechanisms of NIS endocytosis in transformed cell lines and patient-derived human primary thyroid cells. Systemic drug responses were monitored via 99mTc pertechnetate gamma counting and gene expression in BALB/c mice. RESULTS: We identified an acidic dipeptide within the NIS C-terminus that mediates binding to the σ2 subunit of the Adaptor Protein 2 (AP2) heterotetramer. We discovered that the FDA-approved drug chloroquine (CQ) modulates NIS accumulation at the PM in a functional manner that is AP2 dependent. In vivo, CQ treatment of BALB/c mice significantly enhanced thyroidal uptake of 99mTc pertechnetate in combination with the histone deacetylase (HDAC) inhibitor vorinostat/SAHA, accompanied by increased thyroidal NIS mRNA. Bioinformatic analyses validated the clinical relevance of AP2 genes with disease-free survival in RAI-treated DTC, enabling construction of an AP2 gene-related risk score classifier for predicting recurrence. CONCLUSIONS: NIS internalization is specifically druggable in vivo. Our data, therefore, provide new translatable potential for improving RAI therapy using FDA-approved drugs in patients with aggressive thyroid cancer. See related commentary by Lechner and Brent, p. 1220.


Assuntos
Simportadores , Neoplasias da Glândula Tireoide , Camundongos , Animais , Humanos , Vorinostat/farmacologia , Pertecnetato Tc 99m de Sódio/metabolismo , Radioisótopos do Iodo/uso terapêutico , Neoplasias da Glândula Tireoide/diagnóstico por imagem , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/genética , Simportadores/genética , Simportadores/metabolismo , Inibidores de Histona Desacetilases , Linhagem Celular Tumoral
2.
Biochimie ; 197: 59-73, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35134457

RESUMO

Short linear motifs (SLiMs) are key to cell physiology mediating reversible protein-protein interactions. Precise identification of SLiMs remains a challenge, being the main drawback of most bioinformatic prediction tools, their low specificity (high number of false positives). An important, usually overlooked, aspect is the relation between SLiMs mutations and disease. The presence of variants in each residue position can be used to assess the relevance of the corresponding residue(s) for protein function, and its (in)tolerance to change. In the present work, we combined sequence variant information and structural analysis of the energetic impact of single amino acid substitution (SAS) in SLiM-Receptor complex structure, and showed that it improves prediction of true functional SLiMs. Our strategy is based on building a SAS tolerance matrix that shows, for each position, whether one of the possible 19 SAS is tolerated or not. Herein we present the MotSASi strategy and analyze in detail 3 SLiMs involved in intracellular protein trafficking (phospho-independent tyrosine-based motif (NPx[Y/F]), type 1 PDZ-binding motif ([S/T]x[V/I/L]COOH) and tryptophan-acidic motif ([L/M]xW[D/E])). Our results show that inclusion of variant and structure information improves both prediction of true SLiMs and rejection of false positives, while also allowing better classification of variants inside SLiMs, a result with a direct impact in clinical genomics.


Assuntos
Biologia Computacional , Genômica , Motivos de Aminoácidos , Sequência de Aminoácidos , Biologia Computacional/métodos , Nucleotídeos
3.
J Endocrinol ; 240(2): 229-241, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30400032

RESUMO

The molecular mechanisms underlying the ERα nuclear/cytoplasmic pool that modulates pituitary cell proliferation have been widely described, but it is still not clear how ERα is targeted to the plasma membrane. The aim of this study was to analyse ERα palmitoylation and the plasma membrane ERα (mERα) pool, and their participation in E2-triggered membrane-initiated signalling in normal and pituitary tumour cell growth. Cell cultures were prepared from anterior pituitaries of female Wistar rats and tumour GH3 cells, and treated with 10 nM of oestradiol (E2). The basal expression of ERα was higher in tumour GH3 than in normal pituitary cells. Full-length palmitoylated ERα was observed in normal and pituitary tumour cells, demonstrating that E2 stimulation increased both, ERα in plasma membrane and ERα and caveolin-1 interaction after short-term treatment. In addition, the Dhhc7 and Dhhc21 palmitoylases were negatively regulated after sustained stimulation of E2 for 3 h. Although the uptake of BrdU into the nucleus in normal pituitary cells was not modified by E2, a significant increase in the GH3 tumoural cell, as well as ERK1/2 activation, with this effect being mimicked by PPT, a selective antagonist of ERα. These proliferative effects were blocked by ICI 182780 and the global inhibitor of palmitoylation. These findings indicate that ERα palmitoylation modulated the mERα pool and consequently the ERK1/2 pathway, thereby contributing to pituitary tumour cell proliferation. These results suggest that the plasma membrane ERα pool might be related to the proliferative behaviour of prolactinoma and may be a marker of pituitary tumour growth.


Assuntos
Membrana Celular/metabolismo , Proliferação de Células , Receptor alfa de Estrogênio/metabolismo , Neoplasias Hipofisárias/metabolismo , Animais , Antineoplásicos Hormonais/farmacologia , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Estrogênios/farmacologia , Feminino , Fulvestranto/farmacologia , Expressão Gênica/efeitos dos fármacos , Lipoilação/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia , Ratos Wistar
4.
Front Immunol ; 9: 1980, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30233581

RESUMO

Neutrophils are major effectors of acute inflammation against infection and tissue damage, with ability to adapt their phenotype according to the microenvironment. Although sex hormones regulate adaptive immune cells, which explains sex differences in immunity and infection, little information is available about the effects of androgens on neutrophils. We therefore aimed to examine neutrophil recruitment and plasticity in androgen-dependent and -independent sites under androgen manipulation. By using a bacterial model of prostate inflammation, we showed that neutrophil recruitment was higher in testosterone-treated rats, with neutrophil accumulation being positively correlated to serum levels of testosterone and associated to stronger inflammatory signs and tissue damage. Testosterone also promoted LPS-induced neutrophil recruitment to the prostate, peritoneum, and liver sinusoids, as revealed by histopathology, flow cytometry, and intravital microscopy. Strikingly, neutrophils in presence of testosterone exhibited an impaired bactericidal ability and a reduced myeloperoxidase activity. This inefficient cellular profile was accompanied by high expression of the anti-inflammatory cytokines IL10 and TGFß1, which is compatible with the "N2-like" neutrophil phenotype previously reported in the tumor microenvironment. These data reveal an intriguing role for testosterone promoting inefficient, anti-inflammatory neutrophils that prolong bacterial inflammation, generating a pathogenic environment for several conditions. However, these immunomodulatory properties might be beneficially exploited in autoimmune and other non-bacterial diseases.


Assuntos
Androgênios/metabolismo , Infecções por Escherichia coli/imunologia , Neutrófilos/imunologia , Prostatite/imunologia , Testosterona/metabolismo , Infecções Urinárias/imunologia , Escherichia coli Uropatogênica/fisiologia , Androgênios/administração & dosagem , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Interleucina-10/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Ratos , Ratos Wistar , Testosterona/administração & dosagem , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral
5.
Mol Cancer Res ; 16(5): 833-845, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29523762

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Proto-Oncogênica c-ets-1/genética , Câncer Papilífero da Tireoide/genética , Câncer Papilífero da Tireoide/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteína Proto-Oncogênica c-ets-1/metabolismo , Ratos , Ratos Endogâmicos F344 , Transdução de Sinais , Câncer Papilífero da Tireoide/patologia , Receptor 4 Toll-Like/genética , Transfecção
6.
J Cell Physiol ; 232(10): 2806-2817, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27861881

RESUMO

Prostatic smooth muscle cells (pSMCs) differentiation is a key factor for prostatic homeostasis, with androgens exerting multiple effects on these cells. Here, we demonstrated that the myodifferentiator complex Srf/Myocd is up-regulated by testosterone in a dose-dependent manner in primary cultures of rat pSMCs, which was associated to the increase in Acta2, Cnn1, and Lmod1 expressions. Blocking Srf or Myocd by siRNAs inhibited the myodifferentiator effect of testosterone. While LPS led to a dedifferentiated phenotype in pSMCs, characterized by down-regulation of Srf/Myocd and smooth muscle cell (SMC)-restricted genes, endotoxin treatment on Myocd-overexpressing cells did not result in phenotypic alterations. Testosterone at a physiological dose was able to restore the muscular phenotype by normalizing Srf/Myocd expression in inflammation-induced dedifferentiated pSMCs. Moreover, the androgen reestablished the proliferation rate and IL-6 secretion increased by LPS. These results provide novel evidence regarding the myodifferentiating role of testosterone on SMCs by modulating Srf/Myocd. Thus, androgens preserve prostatic SMC phenotype, which is essential to maintain the normal structure and function of the prostate. J. Cell. Physiol. 232: 2806-2817, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Desdiferenciação Celular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Testosterona/farmacologia , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Actinas/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Proteínas dos Microfilamentos/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/genética , Fenótipo , Próstata , Interferência de RNA , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transativadores/genética , Fatores de Transcrição/genética , Transfecção , Calponinas
7.
Mol Cell Endocrinol ; 437: 292-301, 2016 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-27568464

RESUMO

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.


Assuntos
Aldeídos/farmacologia , Glândula Tireoide/citologia , Glândula Tireoide/fisiologia , Animais , Cálcio/metabolismo , Linhagem Celular , Desoxiglucose/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/metabolismo , Iodetos/metabolismo , Especificidade de Órgãos/genética , Regiões Promotoras Genéticas/genética , Ratos , Hormônios Tireóideos/biossíntese , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos
8.
J Gen Physiol ; 145(6): 555-63, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26009546

RESUMO

Prokaryotic and eukaryotic Na(+)-driven transporters couple the movement of one or more Na(+) ions down their electrochemical gradient to the active transport of a variety of solutes. When more than one Na(+) is involved, Na(+)-binding data are usually analyzed using the Hill equation with a non-integer exponent n. The results of this analysis are an overall Kd-like constant equal to the concentration of ligand that produces half saturation and n, a measure of cooperativity. This information is usually insufficient to provide the basis for mechanistic models. In the case of transport using two Na(+) ions, an n < 2 indicates that molecules with only one of the two sites occupied are present at low saturation. Here, we propose a new way of analyzing Na(+)-binding data for the case of two Na(+) ions that, by taking into account binding to individual sites, provides far more information than can be obtained by using the Hill equation with a non-integer coefficient: it yields pairs of possible values for the Na(+) affinities of the individual sites that can only vary within narrowly bounded ranges. To illustrate the advantages of the method, we present experimental scintillation proximity assay (SPA) data on binding of Na(+) to the Na(+)/I(-) symporter (NIS). SPA is a method widely used to study the binding of Na(+) to Na(+)-driven transporters. NIS is the key plasma membrane protein that mediates active I(-) transport in the thyroid gland, the first step in the biosynthesis of the thyroid hormones, of which iodine is an essential constituent. NIS activity is electrogenic, with a 2:1 Na(+)/I(-) transport stoichiometry. The formalism proposed here is general and can be used to analyze data on other proteins with two binding sites for the same substrate.


Assuntos
Membrana Celular/metabolismo , Modelos Biológicos , Sódio , Simportadores/metabolismo , Animais , Sítios de Ligação , Células HEK293 , Humanos , Cinética , Ligantes , Potenciais da Membrana , Ligação Proteica , Ratos , Transfecção
9.
Nat Commun ; 5: 3948, 2014 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-24888603

RESUMO

The Na(+)/I(-) symporter (NIS) mediates active I(-) transport--the first step in thyroid hormonogenesis--with a 2Na(+):1I(-) stoichiometry. NIS-mediated (131)I(-) treatment of thyroid cancer post-thyroidectomy is the most effective targeted internal radiation cancer treatment available. Here to uncover mechanistic information on NIS, we use statistical thermodynamics to obtain Kds and estimate the relative populations of the different NIS species during Na(+)/anion binding and transport. We show that, although the affinity of NIS for I(-) is low (Kd=224 µM), it increases when Na(+) is bound (Kd=22.4 µM). However, this Kd is still much higher than the submicromolar physiological I(-) concentration. To overcome this, NIS takes advantage of the extracellular Na(+) concentration and the pronounced increase in its own affinity for I(-) and for the second Na(+) elicited by binding of the first. Thus, at physiological Na(+) concentrations, ~79% of NIS molecules are occupied by two Na(+) ions and ready to bind and transport I(-).


Assuntos
Iodetos/metabolismo , Sódio/metabolismo , Simportadores/metabolismo , Animais , Linhagem Celular , Humanos , Ratos , Rênio/metabolismo , Termodinâmica
10.
J Cell Sci ; 126(Pt 15): 3305-13, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23690546

RESUMO

Na(+)/I(-) symporter (NIS)-mediated active accumulation of I(-) in thyrocytes is a key step in the biosynthesis of the iodine-containing thyroid hormones T3 and T4. Several NIS mutants have been identified as a cause of congenital I(-) transport defect (ITD), and their investigation has yielded valuable mechanistic information on NIS. Here we report novel findings derived from the thorough characterization of the ITD-causing mutation R124H, located in the second intracellular loop (IL-2). R124H NIS is incompletely glycosylated and colocalizes with endoplasmic reticulum (ER)-resident protein markers. As a result, R124H NIS is not targeted to the plasma membrane and therefore does not mediate any I(-) transport in transfected COS-7 cells. Strikingly, however, the mutant is intrinsically active, as revealed by its ability to mediate I(-) transport in membrane vesicles. Of all the amino acid substitutions we carried out at position 124 (K, D, E, A, W, N and Q), only Gln restored targeting of NIS to the plasma membrane and NIS activity, suggesting a key structural role for the δ-amino group of R124 in the transporter's maturation and cell surface targeting. Using our NIS homology model based on the structure of the Vibrio parahaemolyticus Na(+)/galactose symporter, we propose an interaction between the δ-amino group of either R or Q124 and the thiol group of C440, located in IL-6. We conclude that the interaction between IL-2 and IL-6 is critical for the local folding required for NIS maturation and plasma membrane trafficking.


Assuntos
Iodetos/metabolismo , Mutação , Simportadores/genética , Simportadores/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Membrana Celular/genética , Membrana Celular/metabolismo , Chlorocebus aethiops , Cães , Humanos , Células Madin Darby de Rim Canino , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Dobramento de Proteína , Transporte Proteico , Relação Estrutura-Atividade , Simportadores/química , Transfecção
11.
FASEB J ; 26(8): 3252-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22549510

RESUMO

The KCNQ1 α subunit and the KCNE2 ß subunit form a potassium channel in thyroid epithelial cells. Genetic disruption of KCNQ1-KCNE2 causes hypothyroidism in mice, resulting in cardiac hypertrophy, dwarfism, alopecia, and prenatal mortality. Here, we investigated the mechanistic requirement for KCNQ1-KCNE2 in thyroid hormone biosynthesis, utilizing whole-animal dynamic positron emission tomography. The KCNQ1-specific antagonist (-)-[3R,4S]-chromanol 293B (C293B) significantly impaired thyroid cell I(-) uptake, which is mediated by the Na(+)/I(-) symporter (NIS), in vivo (dSUV/dt: vehicle, 0.028 ± 0.004 min(-1); 10 mg/kg C293B, 0.009 ± 0.006 min(-1)) and in vitro (EC(50): 99 ± 10 µM C293B). Na(+)-dependent nicotinate uptake by SMCT, however, was unaffected. Kcne2 deletion did not alter the balance of free vs. thyroglobulin-bound I(-) in the thyroid (distinguished using ClO(4)(-), a competitive inhibitor of NIS), indicating that KCNQ1-KCNE2 is not required for Duox/TPO-mediated I(-) organification. However, Kcne2 deletion doubled the rate of free I(-) efflux from the thyroid following ClO(4)(-) injection, a NIS-independent process. Thus, KCNQ1-KCNE2 is necessary for adequate thyroid cell I(-) uptake, the most likely explanation being that it is prerequisite for adequate NIS activity.


Assuntos
Iodetos/metabolismo , Canal de Potássio KCNQ1/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Glândula Tireoide/metabolismo , Animais , Células COS , Chlorocebus aethiops , Humanos , Hipotireoidismo/genética , Canal de Potássio KCNQ1/antagonistas & inibidores , Canal de Potássio KCNQ1/genética , Camundongos , Tomografia por Emissão de Pósitrons , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Simportadores/antagonistas & inibidores , Simportadores/metabolismo , Glândula Tireoide/efeitos dos fármacos
12.
J Biol Chem ; 285(13): 9569-9582, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20018842

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Animais , Células Dendríticas/metabolismo , Ativação Enzimática , Feminino , Immunoblotting , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Tri-Iodotironina/metabolismo
13.
Prostate ; 69(13): 1387-97, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19496069

RESUMO

BACKGROUND: Chronic inflammation has been postulated to be an important driving force to prostate carcinoma. Toll-like receptors (TLRs) compose a family of receptors mainly expressed on immune cells. Recently, functional TLRs have been shown to be also expressed in numerous cancer cells, but their significance has only recently begun to be explored. The purpose of this study was to investigate the putative role of TLR4 expression in prostate carcinoma. METHODS: To determine if there is an association between TLR4 expression and the malignancy of the tumor, 35 prostate carcinoma samples showing different Gleason grades were analyzed by immunohistochemistry. Also, to explore the functionality of the receptors expressed on the epithelium, we analyzed the type of cytokine response elicited and the signaling pathways involved after TLR4 triggering in the human prostate adenocarcinoma cell line, DU-145. RESULTS: TLR4 is expressed in the normal prostate gland in both stroma and epithelium. TLR4 expression significantly drops to negative values as the Gleason grade augments in both, stroma and epithelium. Moreover, DU-145 cells also exhibit TLR4 expression and respond to TLR4 agonists, activating the transcription factor NF-kappaB and increasing the expression of pro-inflammatory mediators. Inhibition of the molecular adaptors MyD88 and MAL by overexpression of dominant-negative mutants diminished LPS-induced activation of NF-kappaB, showing that DU-145 cells activate the NF-kappaB through MyD88-dependent signaling pathways. CONCLUSIONS: We hypothesize that TLR4 in prostate cells could synergize with innate immune cells contributing to an eventual inflammatory process, which in genetically prone individuals could promote carcinogenesis. Prostate 69: 1387-1397, 2009. (c) 2009 Wiley-Liss, Inc.


Assuntos
Adenocarcinoma/imunologia , Próstata/fisiologia , Neoplasias da Próstata/imunologia , Prostatite/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/fisiopatologia , Linhagem Celular Tumoral , Quimiocinas/genética , Citocinas/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Próstata/patologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , Prostatite/patologia , Prostatite/fisiopatologia , Índice de Gravidade de Doença , Transdução de Sinais/imunologia , Regulação para Cima/imunologia
14.
J Endocrinol ; 192(3): 627-37, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17332530

RESUMO

Nitric oxide (NO) is a free radical that mediates a wide array of cell functions. It is generated from l-arginine by NO-synthase (NOS). Expression of NOS isoforms has been demonstrated in thyroid cells. Previous reports indicated that NO donors induce dedifferentiation in thyrocytes. However, the functional significance of endogenous thyrocyte-produced NO has not been explored. This work aimed to study the influence of endogenous NO on parameters of thyroid cell function and differentiation in FRTL-5 cells. We observed that treatment with the NOS inhibitor, Nomega-nitro-L-arginine methyl ester (L-NAME), increased the TSH-stimulated iodide uptake. The TSH-induced sodium iodide symporter (NIS) and thyroglobulin (TG) mRNA expressions were increased after incubation with L-NAME. In transient transfection assays, TSH-stimulated transcriptional activities of NIS and TG promoters were increased by L-NAME. An increment of the TSH-stimulated cell proliferation was observed after NOS inhibition. Similar results were obtained when the action of another NOS inhibitor, N(g)-monomethyl-L-arginine, was analysed for most of these studies. The production of NO, which was not detectable in basal conditions, was increased by TSH. Our data provide strong evidence that endogenous NO could act as a negative signal for TSH-stimulated iodide uptake and thyroid-specific gene expression as well as proliferation in thyrocytes. These findings reveal a possible new inhibitory pathway in the regulation of thyroid cell function.


Assuntos
Iodetos/metabolismo , Óxido Nítrico/metabolismo , Glândula Tireoide/metabolismo , Animais , Northern Blotting , Carbazóis/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Expressão Gênica , Indóis/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/análise , Óxido Nítrico Sintase/antagonistas & inibidores , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simportadores/genética , Simportadores/metabolismo , Tireoglobulina/genética , Tireoglobulina/metabolismo , Tireotropina/farmacologia , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA