Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Endocr J ; 69(11): 1285-1293, 2022 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-36171093

RESUMO

The thyrotropin receptor (TSHR) plays critical roles in thyroid growth and function and in the pathogenesis of several thyroid diseases including Graves' hyperthyroidism and ophthalmopathy, non-autoimmune hyperthyroidism and thyroid cancer. Several low-molecular weight compounds (LMWCs) and anti-TSHR monoclonal antibodies (mAbs) with receptor antagonistic and inverse agonistic activities have been reported. The former binds to the pocket formed by the receptor transmembrane bundle, and the latter to the extracellular TSH binding site. Both are effective inhibitors of TSH/thyroid stimulating antibody-stimulated cAMP and/or hyaluronic acid production in TSHR-expressing cells. Anti-insulin-like growth factor 1 inhibitors are also found to inhibit TSHR signaling. Each agent has advantages and disadvantages; for example, mAbs have a higher affinity and longer half-life but are more costly than LMWCs. At present, mAbs appear most promising, yet the development of more efficacious LMWCs is desirable. These agents are anticipated to be efficacious not only for the above-mentioned diseases but also for resistance to thyroid hormone and have utility for thyroid cancer radionuclide scintigraphy/therapy as a new theranostic.


Assuntos
Hipertireoidismo , Receptores da Tireotropina , Doenças da Glândula Tireoide , Neoplasias da Glândula Tireoide , Humanos , Anticorpos Monoclonais/uso terapêutico , Autoanticorpos , Imunoglobulinas Estimuladoras da Glândula Tireoide , Receptores Acoplados a Proteínas G , Receptores da Tireotropina/antagonistas & inibidores , Doenças da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Tireotropina
2.
Endocr J ; 69(7): 847-862, 2022 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-35197412

RESUMO

Numerous studies have examined the role of autophagy in thyroid cancer treatment; however there are discrepancies among the reported data, with some showing the pro-survival and others the anti-survival effects of autophagy. These discrepant results appear to be at least in part due to insufficient analyses or data misinterpretation as well as improper assessments of autophagic activity. Therefore, the present study re-evaluated the regulation of autophagic activity by various anticancer modalities and examined the role of autophagy in thyroid cancer treatment in three thyroid cancer cell lines (TPC1, ACT1 and KTC1). The immunofluorescence and DalGreen findings demonstrated that cisplatin, irradiation and sorafenib were all autophagy inducers as previously reported, but, unlike previous studies using thyroid cancer cells, doxorubicin acted as an inhibitor. KTC1 cells are unique because they only responded to cisplatin. The efficacy of anticancer therapeutics was significantly higher in chloroquine or 3-methyladenine-treated autophagy-defective cells than in autophagy-competent cells, thereby indicating the pro-survival effect of autophagy induced by anticancer therapeutics, which is partly due to inhibition of apoptosis. Thus, the present findings relating to several anticancer therapeutics and three thyroid cancer cell lines demonstrate the pro-survival effect of autophagy in thyroid cancer treatment. Although the present study only involved cell lines, it provides evidence for the beneficial combination of the anticancer therapeutic modalities with autophagy inhibitors, and proposes that autophagy inhibitors may serve as a possible adjunctive therapy for thyroid cancer.


Assuntos
Antineoplásicos , Neoplasias da Glândula Tireoide , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Autofagia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Humanos , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/metabolismo
3.
Cancer Sci ; 111(8): 2814-2823, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32458504

RESUMO

Oncocytic cell tumor of the thyroid is composed of large polygonal cells with eosinophilic cytoplasm that is rich in mitochondria. These tumors frequently have the mutations in mitochondrial DNA encoding the mitochondrial electron transport system complex I. However, the mechanism for accumulation of abnormal mitochondria is unknown. A noncanonical mitophagy system has recently been identified, and mitochondria-eating protein (MIEAP) plays a key role in this system. We therefore hypothesized that accumulation of abnormal mitochondria could be attributed to defective MIEAP expression in these tumors. We first show that MIEAP was expressed in all the conventional thyroid follicular adenomas (FAs)/adenomatous goiters (AGs) but not in oncocytic FAs/AGs; its expression was defective not only in oncocytic thyroid cancers but also in the majority of conventional thyroid cancers. Expression of MIEAP was not correlated with methylation status of the 5'-UTR of the gene. Our functional analysis showed that exogenously induced MIEAP, but not PARK2, reduced the amounts of abnormal mitochondria, as indicated by decreased reactive oxygen species levels, mitochondrial DNA / nuclear DNA ratios, and cytoplasmic acidification. Therefore, together with previous studies showing that impaired mitochondrial function triggers compensatory mitochondrial biogenesis that causes an increase in the amounts of mitochondria, we conclude that, in oncocytic cell tumors of the thyroid, increased abnormal mitochondria cannot be efficiently eliminated because of a loss of MIEAP expression, ie impaired MIEAP-mediated noncanonical mitophagy.


Assuntos
Adenoma Oxífilo/patologia , Proteínas Mitocondriais/metabolismo , Células Oxífilas/patologia , Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/patologia , Adenoma Oxífilo/cirurgia , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Mitocôndrias/patologia , Mitofagia , Células Oxífilas/citologia , Estudos Retrospectivos , Glândula Tireoide/citologia , Glândula Tireoide/cirurgia , Neoplasias da Glândula Tireoide/cirurgia , Tireoidectomia , Ubiquitina-Proteína Ligases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Exp Cell Res ; 370(2): 699-707, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30053445

RESUMO

Cancer stem cells (CSCs), a small fraction of a tumor mass, are proposed to be highly crucial for cancer initiation, recurrence and metastasis. We have recently found that aldehyde dehydrogenase (ALDH) 1A3 is a CSC marker in some thyroid cancer cell lines, whose functional activity is, however, not relevant for thyroid cancer stemness. Since previous studies on malignancies in other organs suggest that intracellular reactive oxygen species (ROS) might be a functional and targetable CSC marker, the present study was conducted to elucidate the significance of ROS as a functional CSC marker in thyroid cancer cell lines. We first found that ROS levels controlled spherogenicity; that is, ROSlow cells were more spherogenic than ROShigh cells. However, unlike typical CSCs in other cancers, CSC-like ROSlow cells in thyroid cancer cells were plastic and were not accompanied by de-differentiation status (i.e., expression of stemness markers/thyroid-specific transcription factors) or chemo-/radio-resistance. The lower levels of ROS were functionally critical because a forced increase in ROS levels by L-buthionine-S,R-sulfoximine, an inhibitor of glutathione (GSH) synthesis, and irradiation suppressed spherogenicity. ROS levels were also correlated with the number of double strand DNA breaks determined by 53BP1 staining. Lower ROS levels appear to be a result of decreased mitochondrial oxidative phosphorylation and elevated GSH contents. Given the importance of CSC-targeted therapy for achieving long-term disease eradication by exhausting self-renewal and growth potential of cancer tissues, ROS may be a good candidate for CSC-targeted therapy in thyroid cancer.


Assuntos
Células-Tronco Neoplásicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Aldeído Desidrogenase/metabolismo , Linhagem Celular Tumoral , Citoplasma/metabolismo , Humanos , Espaço Intracelular/metabolismo
5.
Horm Metab Res ; 50(12): 922-931, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30081426

RESUMO

The association between thyroid cancer and thyroid autoimmunity has long been suggested, but remains to be elucidated for several decades. Here the data on this issue are updated by summarizing relevant papers published between 2012 and early 2018. Although numerous papers demonstrated the significant increase in the prevalence of thyroid autoimmunity (positive intrathyroidal lymphocyte infiltration and/or anti-thyroglobulin/thyroid peroxidase antibodies) in patients with thyroid cancers as compared to those with benign nodules, and also the significant increase in the prevalence of papillary thyroid cancer (PTC) in patients with thyroid autoimmunity as compared to those without, there are some crucial biases that should be taken into account for their interpretation. However, a difference in the incidence of thyroid autoimmunity in patients with PTCs and those with other types of thyroid cancers appears to support the significant association of two conditions. Thyroid autoimmunity is, at least partly, likely to be elicited against antigens shared by normal and cancerous thyroid tissues, thereby inducing autoimmunity. At the same time, elevated TSH levels (even within the normal reference ranges), which often accompany Hashimoto's patients are a risk factor for thyroid cancer. However, it is still unclear whether or not the co-existence of thyroid autoimmunity impacts on cancer characteristics and prognosis. This issue needs to be further investigated with large-scale prospective studies.


Assuntos
Autoimunidade , Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/imunologia , Animais , Humanos , Inflamação/patologia , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/cirurgia
6.
Radiat Environ Biophys ; 56(4): 405-412, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28871381

RESUMO

We evaluated the effect of the antioxidant N-acetyl-L-cysteine (NAC) on the levels of reactive oxygen species (ROS), DNA double strand breaks (DSB) and micronuclei (MN) induced by internal and external irradiation using a rat thyroid cell line PCCL3. In internal irradiation experiments, ROS and DSB levels increased immediately after 131I addition and then gradually declined, resulting in very high levels of MN at 24 and 48 h. NAC administration both pre- and also post-131I addition suppressed ROS, DSB and MN. In external irradiation experiments with a low dose (0.5 Gy), ROS and DSB increased shortly and could be prevented by NAC administration pre-, but not post-irradiation. In contrast, external irradiation with a high dose (5 Gy) increased ROS and DSB in a bimodal way: ROS and DSB levels increased immediately after irradiation, quickly returned to the basal levels and gradually rose again after >24 h. The second phase was in parallel with an increase in 4-hydroxy-2-nonenal. The number of MN induced by the second wave of ROS/DSB elevations was much higher than that by the first peak. In this situation, NAC administered pre- and post-irradiation comparably suppressed MN induced by a delayed ROS elevation. In conclusion, a prolonged ROS increase during internal irradiation and a delayed ROS increase after external irradiation with a high dose caused serious DNA damage, which were efficiently prevented by NAC. Thus, NAC administration even both after internal or external irradiation prevents ROS increase and eventual DNA damage.


Assuntos
Acetilcisteína/farmacologia , Dano ao DNA , Protetores contra Radiação/farmacologia , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/efeitos da radiação , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos da radiação , Linhagem Celular , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Relação Dose-Resposta à Radiação , Radioisótopos do Iodo/efeitos adversos , Radioisótopos do Iodo/metabolismo , Testes para Micronúcleos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo
7.
Am J Hum Genet ; 92(5): 807-19, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23623389

RESUMO

Cockayne syndrome (CS) is a genetic disorder characterized by developmental abnormalities and photodermatosis resulting from the lack of transcription-coupled nucleotide excision repair, which is responsible for the removal of photodamage from actively transcribed genes. To date, all identified causative mutations for CS have been in the two known CS-associated genes, ERCC8 (CSA) and ERCC6 (CSB). For the rare combined xeroderma pigmentosum (XP) and CS phenotype, all identified mutations are in three of the XP-associated genes, ERCC3 (XPB), ERCC2 (XPD), and ERCC5 (XPG). In a previous report, we identified several CS cases who did not have mutations in any of these genes. In this paper, we describe three CS individuals deficient in ERCC1 or ERCC4 (XPF). Remarkably, one of these individuals with XP complementation group F (XP-F) had clinical features of three different DNA-repair disorders--CS, XP, and Fanconi anemia (FA). Our results, together with those from Bogliolo et al., who describe XPF alterations resulting in FA alone, indicate a multifunctional role for XPF.


Assuntos
Síndrome de Cockayne/genética , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Anemia de Fanconi/genética , Predisposição Genética para Doença/genética , Fenótipo , Xeroderma Pigmentoso/genética , Sequência de Aminoácidos , Sequência de Bases , Síndrome de Cockayne/enzimologia , Síndrome de Cockayne/patologia , Primers do DNA/genética , Anemia de Fanconi/enzimologia , Anemia de Fanconi/patologia , Evolução Fatal , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Análise de Sequência de DNA , Xeroderma Pigmentoso/enzimologia , Xeroderma Pigmentoso/patologia
8.
J Allergy Clin Immunol ; 136(4): 1007-17, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26255102

RESUMO

BACKGROUND: Nonhomologous end-joining (NHEJ) is the major DNA double-strand break (DSB) repair mechanism in human cells. The final rejoining step requires DNA ligase IV (LIG4) together with the partner proteins X-ray repair cross-complementing protein 4 (XRCC4) and XRCC4-like factor. Patients with mutations in genes encoding LIG4, XRCC4-like factor, or the other NHEJ proteins DNA-dependent protein kinase catalytic subunit and Artemis are DSB repair defective and immunodeficient because of the requirement for NHEJ during V(D)J recombination. OBJECTIVE: We found a patient displaying microcephaly and progressive ataxia but a normal immune response. We sought to determine pathogenic mutations and to describe the molecular pathogenesis of the patient. METHODS: We performed next-generation exome sequencing. We evaluated the DSB repair activities and V(D)J recombination capacity of the patient's cells, as well as performing a standard blood immunologic characterization. RESULTS: We identified causal mutations in the XRCC4 gene. The patient's cells are radiosensitive and display the most severe DSB repair defect we have encountered using patient-derived cell lines. In marked contrast, a V(D)J recombination plasmid assay revealed that the patient's cells did not display the junction abnormalities that are characteristic of other NHEJ-defective cell lines. The mutant protein can interact efficiently with LIG4 and functions normally in in vitro assays and when transiently expressed in vivo. However, the mutation makes the protein unstable, and it undergoes proteasome-mediated degradation. CONCLUSION: Our findings reveal a novel separation of impact phenotype: there is a pronounced DSB repair defect and marked clinical neurological manifestation but no clinical immunodeficiency.


Assuntos
Ataxia/genética , Proteínas de Ligação a DNA/genética , Síndromes de Imunodeficiência/genética , Microcefalia/genética , Estabilidade Proteica , Ataxia/imunologia , DNA Ligase Dependente de ATP , DNA Ligases/metabolismo , Análise Mutacional de DNA , Reparo do DNA/genética , Feminino , Células HEK293 , Humanos , Síndromes de Imunodeficiência/imunologia , Microcefalia/imunologia , Mutação/genética , Ligação Proteica/genética , Tolerância a Radiação/genética , Recombinação V(D)J/genética , Adulto Jovem
9.
Diabetologia ; 58(11): 2606-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26271342

RESUMO

AIMS/HYPOTHESIS: Interferon regulatory factor (IRF)4 plays a critical role in lymphoid development and the regulation of immune responses. Genetic deletion of IRF4 has been shown to suppress autoimmune disease in several mouse models, but its role in autoimmune diabetes in NOD mice remains unknown. METHODS: To address the role of IRF4 in the pathogenesis of autoimmune diabetes in NOD mice, we generated IRF4-knockout NOD mice and investigated the impact of the genetic deletion of IRF4 on diabetes, insulitis and insulin autoantibody; the effector function of T cells in vivo and in vitro; and the proportion of dendritic cell subsets. RESULTS: Heterozygous IRF4-deficient NOD mice maintained the number and phenotype of T cells at levels similar to NOD mice. However, diabetes and autoantibody production were completely suppressed in both heterozygous and homozygous IRF4-deficient NOD mice. The level of insulitis was strongly suppressed in both heterozygous and homozygous IRF4-deficient mice, with minimal insulitis observed in heterozygous mice. An adoptive transfer study revealed that IRF4 deficiency conferred disease resistance in a gene-dose-dependent manner in recipient NOD/severe combined immunodeficiency mice. Furthermore, the proportion of migratory dendritic cells in lymph nodes was reduced in heterozygous and homozygous IRF4-deficient NOD mice in an IRF4 dose-dependent manner. These results suggest that the levels of IRF4 in T cells and dendritic cells are important for the pathogenesis of diabetes in NOD mice. CONCLUSIONS/INTERPRETATION: Haploinsufficiency of IRF4 halted disease development in NOD mice. Our findings suggest that an IRF4-targeted strategy might be useful for modulating autoimmunity in type 1 diabetes.


Assuntos
Autoanticorpos/sangue , Diabetes Mellitus Tipo 1/genética , Haploinsuficiência , Insulina/imunologia , Fatores Reguladores de Interferon/genética , Animais , Células Dendríticas/imunologia , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/imunologia , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Fatores de Proteção , Linfócitos T/imunologia
10.
Endocr J ; 61(3): 297-302, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24335009

RESUMO

Transforming growth factor ß (TGF-ß) members, pleiotropic cytokines, play a critical role for carcinogenesis generally as a tumor suppressor in the early cancer development, but as a tumor promoter in the late stage of cancer progression. The present study was designed to clarify the role for TGF-ß signaling in early thyroid carcinogenesis using the conditional Tgfbr2(floxE2/floxE2) knock-in mice, having 2 loxP sites at introns 1 and 2 of Tgfb2r gene. When these mice were crossed with thyroid peroxidase (TPO)-Cre or fibroblast-specific protein-1 (FSP1)-Cre, the resultant mice, Tgfbr2(tpoKO) and Tgfbr2(fspKO), lost TGF-ß II receptor expression (thereby TGF-ß signaling) specifically in the thyroid follicular epithelial cells or fibroblasts, respectively. The thyroid morphology was monitored up to 52 weeks in these mice, showing no tumor development, except one Tgfbr2(tpoKO) mouse developing follicular adenoma like-lesion. Our data suggest that TGF-ß signaling in mesenchymal or follicular epithelial cells of the thyroid does not appear to function as a tumor suppressive barrier at the early stage of thyroid carcinogenesis.


Assuntos
Transdução de Sinais , Glândula Tireoide/citologia , Neoplasias da Glândula Tireoide/etiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Carcinogênese , Células Epiteliais/fisiologia , Feminino , Fibroblastos/metabolismo , Masculino , Camundongos
11.
Endocr J ; 61(5): 481-90, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24531915

RESUMO

Cancer stem-like cells (CSCs) play important roles in cancer initiation and progression. CSCs have been isolated using several markers, but those for thyroid CSCs remain to be confirmed. We therefore conducted a comprehensive search for thyroid CSC markers. Expression of nine cell surface markers (CD13, CD15, CD24, CD44, CD90, CD117, CD133, CD166, and CD326) and aldehyde dehydrogenase (ALDH) activity, which are CSC markers in various solid cancers, and the ability to form spheres in vitro and tumors in vivo were investigated using eight thyroid cancer cell lines (FRO, KTC1/2/3, TPC1, WRO, ACT1, and 8505C). Among these, four cell lines (FRO, KTC3, ACT1, and 8505C) possessed the both abilities; however, common markers indicative of CSCs were not observed. The pattern of ability to form spheres was completely matched to that of tumor formation, suggesting that our sphere assay is valuable for assessment of tumor-forming ability. Next, the cells were sorted using these markers and subjected to the sphere assay. In three cell lines (FRO, KTC3, and ACT1), ALDH(pos) cells showed higher sphere forming ability than ALDH(neg) cells but not in other cells. CD326(hi) also appeared to be a candidate marker only in FRO cells. However, these subpopulations did not follow a classical hierarchical model because ALDH(neg) and CD326(low) fractions also generated ALDH(pos) and CD326(hi) cells, respectively. These data suggest that ALDH activity is probably a major candidate marker to enrich thyroid CSCs but not universal; other markers such as CD326 that regulate different CSC properties may exist.


Assuntos
Antígenos de Superfície/metabolismo , Biomarcadores Tumorais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Animais , Antígenos de Superfície/análise , Biomarcadores Tumorais/análise , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/patologia , Esferoides Celulares/patologia , Neoplasias da Glândula Tireoide/patologia
12.
Cureus ; 16(6): e63338, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-39070443

RESUMO

OBJECTIVE: Thyrotoxicosis causes excess energy expenditure, resulting in weight loss, despite increased appetite, and changes in body composition, which are typically reversible with the normalization of thyroid hormone levels. However, patients with hyperthyroidism due to Graves' disease are sometimes hesitant to undergo treatment because of the perceived morbidity associated with weight gain. Therefore, obtaining data to explain the details of such weight gain to these patients is important. This study aimed to investigate changes in body weight and composition in patients with Graves' disease after total thyroidectomy. METHODS: In total, 21 patients with Graves' disease who underwent total thyroidectomy were enrolled. Among them, nine patients were hyperthyroid (group A) and 12 were euthyroid (group B, control) immediately before surgery. Body weight, height, and body composition using bioelectrical impedance were measured preoperatively and five months postoperatively. RESULTS: In all patients, body weight, body mass index, and skeletal muscle mass, but not fat mass, significantly increased postoperatively. In individual groups, a significant increase in skeletal muscle and fat masses was observed solely in groups A and B, respectively. Furthermore, a significant positive correlation between preoperative thyroid function and differences in skeletal muscle mass preoperatively and postoperatively was found. CONCLUSION: Our study shows that the normalization of thyroid function using thyroidectomy in patients with Graves' disease is accompanied by weight gain, mainly due to an increase in skeletal muscle mass. These data are clinically significant because they enable physicians to explain to patients that weight gain after surgical treatment for Graves' disease is favorable and reassure them of their concern.

13.
Sci Rep ; 13(1): 149, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36599897

RESUMO

Oncocytic thyroid cancer is characterized by the aberrant accumulation of abnormal mitochondria in the cytoplasm and a defect in oxidative phosphorylation. We performed metabolomics analysis to compare metabolic reprogramming among the oncocytic and non-oncocytic thyroid cancer cell lines XTC.UC1 and TPC1, respectively, and a normal thyroid cell line Nthy-ori 3-1. We found that although XTC.UC1 cells exhibit higher glucose uptake than TPC1 cells, the glycolytic intermediates are not only utilized to generate end-products of glycolysis, but also diverted to branching pathways such as lipid metabolism and the serine synthesis pathway. Glutamine is preferentially used to produce glutathione to reduce oxidative stress in XTC.UC1 cells, rather than to generate α-ketoglutarate for anaplerotic flux into the TCA cycle. Thus, growth, survival and redox homeostasis of XTC.UC1 cells rely more on both glucose and glutamine than do TPC1 cells. Furthermore, XTC.UC1 cells contained higher amounts of intracellular amino acids which is due to higher expression of the amino acid transporter ASCT2 and enhanced autophagy, thus providing the building blocks for macromolecules and energy production. These metabolic alterations are required for oncocytic cancer cells to compensate their defective mitochondrial function and to alleviate excess oxidative stress.


Assuntos
Glutamina , Neoplasias da Glândula Tireoide , Humanos , Linhagem Celular Tumoral , Glutamina/metabolismo , Glicólise , Metabolômica/métodos , Mitocôndrias/metabolismo , Neoplasias da Glândula Tireoide/metabolismo
14.
Thyroid ; 33(3): 330-337, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36565031

RESUMO

Background: The main molecular mechanism underlying acute suppression of iodine organification in normal thyroids after an excessive iodine load, that is, the Wolff-Chaikoff effect, is assumed to be suppression of iodine oxidation and iodothyronine synthesis. However, the mechanism underlying chronic antithyroid action of inorganic iodine in Graves' disease is not fully understood. Using a mouse model of Graves' hyperthyroidism, we examined changes in iodothyronine content and gene expression profiles in the thyroid glands after inorganic iodine loading. Materials and Methods: Graves' hyperthyroidism was induced and maintained in BALB/c mice by repeated immunizations of recombinant adenovirus expressing the human thyrotropin (TSH) receptor A-subunit. Hyperthyroid mice were left untreated (GD-C; n = 8) or treated with inorganic iodine for 12 weeks (GD-NaI; n = 8). We used unimmunized BALB/c mice as a control group (n = 10). In each mouse, serum thyroxine (T4) levels were measured with enzyme-linked immunosorbent assay (ELISA) at 4-week intervals. The intrathyroidal iodothyronine content and gene expression levels were, respectively, evaluated by mass spectrometry and RNA sequencing (RNA-seq) at the end of the experimental period. Results: Serum T4 levels in the GD-C group remained higher than in the control group, whereas those in the GD-NaI group declined to normal levels during the experimental period. Intrathyroidal triiodothyronine (T3), reverse T3 (rT3), and T4 contents in the GD-C group were higher than the control group, and rT3 and T4 were further increased in the GD-NaI group. The observed alterations in iodothyronine levels in the thyroid and sera may be explained by altered expression levels of genes for iodothyronine biosynthetic molecules, their transporter, and deiodinases. Conclusion: In this mouse model of hyperthyroidism, higher intrathyroidal accumulation of T4 and reduced gene expression data of iodothyronine transporters in the GD-NaI group suggest that chronic antithyroid action of iodine in Graves' disease involves suppression of hormone secretion.


Assuntos
Doença de Graves , Hipertireoidismo , Iodo , Humanos , Tiroxina , Hipertireoidismo/genética , Tri-Iodotironina , Doença de Graves/genética , Doença de Graves/metabolismo , Iodo/metabolismo , Receptores da Tireotropina , Tri-Iodotironina Reversa , Expressão Gênica
15.
Nihon Rinsho ; 70(3): 436-40, 2012 Mar.
Artigo em Japonês | MEDLINE | ID: mdl-22514922

RESUMO

The thyroid glands are a vulnerable organ to ionizing radiation. Indeed the epidemiological studies have revealed an increase in the incidences of thyroid cancer among atomic bomb survivors in Hiroshima and Nagasaki and radiation casualties in Chernobyl. The carcinogenic risk for the thyroids is dependent on radiation dose, and higher in younger people. Recent advances in molecular biology contribute to clarify the mechanisms for thyroid carcinogenesis at genetic and molecular levels. Here radiation-induced thyroid carcinogenesis is reviewed from epidemiological data to basic research.


Assuntos
Neoplasias Induzidas por Radiação , Neoplasias da Glândula Tireoide/etiologia , Acidente Nuclear de Chernobyl , Humanos , Japão , Neoplasias Induzidas por Radiação/epidemiologia , Armas Nucleares , Neoplasias da Glândula Tireoide/epidemiologia
16.
Metabolites ; 12(12)2022 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-36557253

RESUMO

Metabolism is a series of life-sustaining chemical reactions in organisms, providing energy required for cellular processes and building blocks for cellular constituents of proteins, lipids, carbohydrates and nucleic acids. Cancer cells frequently reprogram their metabolic behaviors to adapt their rapid proliferation and altered tumor microenvironments. Not only aerobic glycolysis (also termed the Warburg effect) but also altered mitochondrial metabolism, amino acid metabolism and lipid metabolism play important roles for cancer growth and aggressiveness. Thus, the mechanistic elucidation of these metabolic changes is invaluable for understanding the pathogenesis of cancers and developing novel metabolism-targeted therapies. In this review article, we first provide an overview of essential metabolic mechanisms, and then summarize the recent findings of metabolic reprogramming and the recent reports of metabolism-targeted therapies for thyroid cancer.

17.
Front Endocrinol (Lausanne) ; 13: 932754, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36187114

RESUMO

Mitochondria-eating protein (MIEAP) is a molecule important for non-canonical mitophagy and thought to be a tumor suppressor. Our previous study found that MIEAP expression is defective in thyroid oncocytomas, irrespective of being benign or malignant, and also in non-oncocytic thyroid cancers. Thyroid oncocytomas are composed of large polygonal cells with eosinophilic cytoplasm that is rich in abnormal mitochondria. Thus, our data indicate that, together with increased mitochondrial biogenesis that compensates for the dysfunction of the mitochondria, MIEAP plays a critical role in the accumulation of mitochondria in thyroid oncocytic tumors, whereas a defective MIEAP expression alone is not sufficient for mitochondrial accumulation in non-oncocytic cancers with normal mitochondria. To clarify whether MIEAP is a tumor suppressor in the thyroids and whether MIEAP knockout (KO) alone is sufficient for the oncocytic phenotype and also to extend our effort toward canonical mitophagy (a selective autophagy), we here conducted mouse studies using genetically engineered mice. BrafCA/wt mice developed thyroid cancers 1 year after intrathyroidal injection of adenovirus expressing Cre, while cancer development was observed at 6 months in adenovirus-Cre-injected BrafCA/wt;MieapKO/KO and BrafCA/wt;Atg5flox/flox mice [where autophagy-related 5 (ATG5) is a component of autophagic machinery], although KO of either molecule alone was not sufficient for cancer development. These data demonstrate that MIEAP or ATG5 KO accelerated thyroid cancer development. However, cancers in adenovirus-Cre-injected BrafCA/wt ;MieapKO/KO and BrafCA/wt ;Atg5flox/flox mice were not oncocytic. In conclusion, we here show that MIEAP and ATG5 are both tumor suppressors in thyroid carcinogenesis, but as we have anticipated from our previous data, KO of either molecule does not confer the oncocytic phenotype to BRAFV600E-positive thyroid cancers. The combination of disruptive mitochondrial function and impaired mitochondrial quality control may be necessary to establish a mouse model of thyroid oncocytoma.


Assuntos
Adenoma Oxífilo , Proteínas Mitocondriais , Neoplasias da Glândula Tireoide , Animais , Camundongos , Adenoma Oxífilo/patologia , Autofagia/genética , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Mitofagia/genética , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias da Glândula Tireoide/patologia
18.
Sci Rep ; 11(1): 19096, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34580369

RESUMO

Childhood radiation exposure is a known thyroid cancer risk factor. This study evaluated the effects of age on radiation-induced thyroid carcinogenesis in rats irradiated with 8 Gy X-rays. We analyzed cell proliferation, cell death, DNA damage response, and autophagy-related markers in 4-week-old (4W) and 7-month-old (7M) rats and the incidence of thyroid tumors in 4W, 4-month-old (4M), and 7M rats 18 months after irradiation. Cell death and DNA damage response were increased in 4W rats compared to those in controls at 1 month post-irradiation. More Ki-67-positive cells were observed in 4W rats at 12 months post-irradiation. Thyroid tumors were confirmed in 61.9% (13/21), 63.6% (7/11), and 33.3% (2/6) of irradiated 4W, 4M, and 7M rats, respectively, compared to 0%, 14.3% (1/7), and 16.7% (1/6) in the respective nonirradiated controls. There were 29, 9, and 2 tumors in irradiated 4W, 4M, and 7M rats, respectively. The expression of several autophagy components was downregulated in the area surrounding radiation-induced thyroid carcinomas in 4W and 7M rats. LC3 and p62 expression levels decreased in radiation-induced follicular carcinoma in 4W rats. Radiosensitive cells causing thyroid tumors may be more prevalent in young rats, and abrogation of autophagy may be associated with radiation-induced thyroid carcinogenesis.


Assuntos
Carcinogênese/efeitos da radiação , Neoplasias Induzidas por Radiação/epidemiologia , Lesões Experimentais por Radiação/epidemiologia , Neoplasias da Glândula Tireoide/epidemiologia , Adulto , Fatores Etários , Animais , Criança , Relação Dose-Resposta à Radiação , Humanos , Incidência , Masculino , Neoplasias Induzidas por Radiação/etiologia , Neoplasias Induzidas por Radiação/patologia , Lesões Experimentais por Radiação/etiologia , Lesões Experimentais por Radiação/patologia , Tolerância a Radiação , Ratos , Fatores de Risco , Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/etiologia , Neoplasias da Glândula Tireoide/patologia , Raios X/efeitos adversos
19.
J Endocr Soc ; 4(11): bvaa146, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-33123658

RESUMO

The appropriate amount of iodine is critical for normal function of thyroid cells synthesizing thyroid hormones. Although normal thyroid cell lines such as rat PCCL3 and FRTL5 and human Nthy-ori 3-1 have been widely used for in vitro studies on physiological and pathophysiological effects of iodine on thyroid cells, we have recently pointed out the critical differences between FRTL5/PCCL3 cells and Nthy-ori 3-1 cells. Therefore, we here directly compared some of the cellular characteristics-iodine uptake, differentiated status, iodine-induced cytotoxicity, and iodine-regulation of autophagy-between PCCL3 and Nthy-ori 3-1 cells. PCCL3 cells express messenger RNAs for thyrotropin receptor and sodium/iodine symporter and incorporate iodine in a thyrotropin-dependent manner, whereas Nthy-ori 3-1 cells do not either. Nevertheless, both cells were comparably resistant to iodine cytotoxicity: Only far excess iodine (5 × 10-2 M) killed 20% to 40% cells in 24 hours with perchlorate exhibiting no effect, suggesting this cytotoxic effect is due to extracellular iodine. In contrast, a wide range of iodine (5 × 10-9 to 5 × 10-2 M) induced autophagy in PCCL3 cells, which was abolished by perchlorate, indicating intracellular iodine-induction of autophagy, but this effect was not observed in Nthy-ori 3-1 cells. In conclusion, it is critical to discriminate the effect of iodine incorporated into cells from that of extracellular iodine on thyroid cells. Iodine-uptake competent thyroid cells such as PCCL3 and FRTL5 cells, not Nthy-ori 3-1 cells, should be used for studies on iodine effect on thyroid cells.

20.
Endocrine ; 69(3): 571-577, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32281047

RESUMO

PURPOSE: Transforming growth factor-ß (TGFß) has pleiotropic actions, including both anti- and pro-tumorigenic abilities. We have previously shown no tumor development in the thyroid-specific TGFß receptor type II knockout (Tgfßr2 KO) mice, indicating the insufficiency of defective TGFß signal itself for thyroid cancer initiation. In the current study, we evaluated whether defective TGFß signal accelerates BRAFV600E-mediated thyroid carcinogenesis in our mouse model, in which intrathyroidal injection of adenovirus expressing Cre under thyroglobulin (TG) promoter (Ad-TgP-Cre) into thyroid lobes of conditional BrafV600E knock-in mice (BrafCA) induces thyroid cancers 12 months later. METHODS: BrafCA/wt;Tgfbr2floxE2/floxE2 mice were generated by crossing Tgfbr2floxE2/floxE2 and BrafCA mice, and Ad-TgP-Cre was injected into the left lobes of 4-6-week-old mice. Mice were sacrificed at 6 and 12 months, and the thyroid tissues were subjected to H&E and immune-histochemistry and -fluorecence. RESULTS: Thyroid tumors were observed in 8 of 10 mice at 6 months and 4 of 7 mice at 12 months. These tumors were judged to be malignant by H&E staining, because of the presence of papillary growth of atypical follicular cells, intranuclear cytoplasmic inclusions and so on. Immunohistochemical analyses using thyroid cancer tissues obtained at 6 months demonstrated variable levels of TG but steady levels of Paired Box-8 expression and higher Ki67 positivity. The degree of epithelial-to-mesenchymal transition could not be evaluated because normal thyroid tissues and thyroid cancers developed in BrafCA and BrafCA/wt;Tgfbr2floxE2/floxE2 mice were all E-cadherin+/vimentin-, that is, epithelial type. CONCLUSION: In a mouse model, defective TGFß signaling pathway accelerates BRAFV600E-induced thyroid cancer development, which is occasionally accompanied by reduced TG expression implying dedifferentiation. The former finding is consistent with anti-tumorigenic ability of TGFß in early tumorigenic process, but the latter is contradictory to generally accepted concept for TGFß-induction of dedifferentiation.


Assuntos
Carcinoma Papilar , Neoplasias da Glândula Tireoide , Aceleração , Animais , Carcinogênese/genética , Camundongos , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Glândula Tireoide , Neoplasias da Glândula Tireoide/genética , Fator de Crescimento Transformador beta/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa