Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Clin Endocrinol Metab ; 107(2): e448-e457, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34555150

ABSTRACT

Radioactive iodine has been considered a safe and effective therapeutic option for hyperthyroidism secondary to Graves disease and autonomously functioning thyroid nodules since the mid-20th century. The question of whether I-131 at the doses used for hyperthyroidism might increase the risk of cancer has been investigated in a number of observational cohort studies over the years, with the preponderance of evidence being reassuring as to its safety. In particular, the 1998 Cooperative Thyrotoxicosis Therapy Follow-up Study (CTTFUS) has been widely cited as compelling evidence that I-131 is safe in hyperthyroidism therapy with respect to carcinogenesis. However, in 2019, a study by Kitahara and colleagues re-analyzed the CTTFUS cohort, extending the follow-up time and applying a novel dosimetric model for estimating tissue absorbed doses of radiation. This new analysis concluded that radioactive iodine was associated with an increased risk for mortality from overall cancer, breast cancer, and non-breast solid cancers. Reaction to this study was vociferous and particularly negative in the nuclear medicine literature. This mini-review was inspired by the 2019 CTTFUS controversy, and it is intended to provide the necessary context for clinicians to provide nuanced advice to their patients on the subject. To that end, the pre-2019 literature is surveyed, the 2019 CTTFUS study and a 2020 follow-up are discussed, and lessons from the literature and critical commentaries are considered.


Subject(s)
Hyperthyroidism/therapy , Iodine Radioisotopes/adverse effects , Neoplasms, Radiation-Induced/epidemiology , Thyroid Gland/radiation effects , Thyroid Neoplasms/epidemiology , Dose-Response Relationship, Radiation , Humans , Incidence , Neoplasms, Radiation-Induced/pathology , Observational Studies as Topic , Radiometry , Risk Assessment/statistics & numerical data , Thyroid Gland/pathology , Thyroid Neoplasms/etiology , Thyroid Neoplasms/pathology
3.
Thyroid ; 26(6): 759-64, 2016 06.
Article in English | MEDLINE | ID: mdl-27141822

ABSTRACT

BACKGROUND: The American Thyroid Association (ATA) has recently revised its guidance pertaining to thyroid nodules and follicular cell-derived thyroid cancer. The 2015 guidelines are massive in both scope and scale, with changes in the organizational approach to risk stratification of nodules and cancer, as well as multiple sections covering new material. This review highlights the major structural and organizational changes, focusing attention on the most dramatically changed recommendations, that is, those recommendations that clinicians will find striking because they call for significant divergence from prior clinical practice. SUMMARY: The revised approach to thyroid nodule risk stratification is based on sonographic pattern, with an emphasis on pattern rather than growth in the long-term surveillance of nodules. Accumulating data have also been incorporated into an updated risk stratification scheme for thyroid cancer that increases the size of the low-risk pool, in part because low-volume lymph nodal metastases are now considered low risk. The most fundamentally altered recommendation is that lobectomy might be considered as the initial surgical approach for follicular cell-derived thyroid cancers from 1 to 4 cm in size. CONCLUSIONS: The underlying theme of the 2015 ATA guidelines is that "less is more." As these new recommendations are adopted, fewer fine-needle aspiration biopsies will need to be done, less extensive surgeries will become more common, less radioactive iodine will be used either for treatment or for diagnostics, and less stimulated thyroglobulin testing will be done. Mastery of these guidelines will help clinicians know when it is reasonable to do less, thus providing responsibly individualized therapy for their patients.


Subject(s)
Practice Guidelines as Topic , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Thyroid Nodule/pathology , Humans , Risk Assessment , Thyroid Gland/surgery , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/therapy , Thyroid Nodule/diagnosis , Thyroid Nodule/therapy , United States
4.
Endocrinology ; 156(11): 4325-35, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26287403

ABSTRACT

Thyroid hormone (T3) has been known to regulate the basal metabolic rate for more than a century, but mechanistic understanding is lacking both at the level of the intact organism and in terms of how T3 alters energy expenditure in individual tissues. The current studies investigate the question of which metabolically relevant genes respond acutely as T3 concentrations increase through the physiologic range in liver cells. Because this has been technically unfeasible historically, we developed a modified protocol for extracellular flux analysis using a 96-well Extracellular Flux Analyzer (Seahorse Bioscience). Using a modified extracellular flux protocol and LH86 human hepatoma cells, we established an experimental system where small but significant changes in O2 consumption could be reproducibly quantified as hypothyroid cells were exposed to near-physiologic final concentrations of T3 approximately 2 orders of magnitude lower than most studies (0.04 nM free T3), in only 6-7 hours. Taking advantage of the nondestructive nature of 96-well Extracellular Flux Analyzer measurements, the acute, direct, transcriptional changes that occur were measured in the exact same cells demonstrating increased O2 consumption. An unbiased, genome-wide microarray analysis identified potential candidate genes related to fatty acid oxidation, angiogenesis, nucleotide metabolism, immune signaling, mitochondrial respiration, and cell proliferation. The identified transcriptome is likely enriched in the genes most important for mediating the energetic effects of T3 in hepatoma cells.


Subject(s)
Liver/drug effects , Mitochondria/drug effects , Oxygen Consumption/drug effects , Triiodothyronine/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Homeostasis/drug effects , Humans , Hypothyroidism/metabolism , Liver/metabolism , Liver Neoplasms/metabolism , Mitochondria/metabolism , Oxidation-Reduction
5.
Blood Rev ; 29(2): 71-80, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25433571

ABSTRACT

Metabolic syndrome constitutes a constellation of findings including central obesity, insulin resistance/type 2 diabetes mellitus (DM), dyslipidemia and hypertension. Metabolic syndrome affects 1 in 4 adults in the United States and is rapidly rising in prevalence, largely driven by the dramatic rise in obesity and insulin resistance/DM. Being central to the development of metabolic syndrome and its other related diseases, much focus has been placed on identifying the mitogenic effects of obesity and insulin resistance/DM as mechanistic clues of the link between metabolic syndrome and cancer. Pertinent mechanisms identified include altered lipid signaling, adipokine and inflammatory cytokine effects, and activation of PI3K/Akt/mTOR and RAS/RAF/MAPK/ERK pathways via dysregulated insulin/insulin-like growth factor-1 (IGF-1) signaling. Through variable activation of these multiple pathways, obesity and insulin resistance/DM pre-dispose to hematologic malignancies, imposing the aggressive and chemo-resistant phenotypes typically seen in cancer patients with underlying metabolic syndrome. Growing understanding of these pathways has identified druggable cancer targets, rationalizing the development and testing of agents like PI3K inhibitor idelalisib, mTOR inhibitors everolimus and temsirolimus, and IGF-1 receptor inhibitor linsitinib. It has also led to exploration of obesity and diabetes-directed therapies including statins and oral hypoglycemic for the management of metabolic syndrome-related hematologic neoplasms.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Hematologic Neoplasms/epidemiology , Metabolic Syndrome/epidemiology , Obesity/epidemiology , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Hematologic Neoplasms/complications , Hematologic Neoplasms/metabolism , Humans , Insulin Resistance , Lipid Metabolism , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Obesity/complications , Obesity/metabolism , Signal Transduction
6.
Thyroid ; 24(12): 1670-751, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25266247

ABSTRACT

BACKGROUND: A number of recent advances in our understanding of thyroid physiology may shed light on why some patients feel unwell while taking levothyroxine monotherapy. The purpose of this task force was to review the goals of levothyroxine therapy, the optimal prescription of conventional levothyroxine therapy, the sources of dissatisfaction with levothyroxine therapy, the evidence on treatment alternatives, and the relevant knowledge gaps. We wished to determine whether there are sufficient new data generated by well-designed studies to provide reason to pursue such therapies and change the current standard of care. This document is intended to inform clinical decision-making on thyroid hormone replacement therapy; it is not a replacement for individualized clinical judgment. METHODS: Task force members identified 24 questions relevant to the treatment of hypothyroidism. The clinical literature relating to each question was then reviewed. Clinical reviews were supplemented, when relevant, with related mechanistic and bench research literature reviews, performed by our team of translational scientists. Ethics reviews were provided, when relevant, by a bioethicist. The responses to questions were formatted, when possible, in the form of a formal clinical recommendation statement. When responses were not suitable for a formal clinical recommendation, a summary response statement without a formal clinical recommendation was developed. For clinical recommendations, the supporting evidence was appraised, and the strength of each clinical recommendation was assessed, using the American College of Physicians system. The final document was organized so that each topic is introduced with a question, followed by a formal clinical recommendation. Stakeholder input was received at a national meeting, with some subsequent refinement of the clinical questions addressed in the document. Consensus was achieved for all recommendations by the task force. RESULTS: We reviewed the following therapeutic categories: (i) levothyroxine therapy, (ii) non-levothyroxine-based thyroid hormone therapies, and (iii) use of thyroid hormone analogs. The second category included thyroid extracts, synthetic combination therapy, triiodothyronine therapy, and compounded thyroid hormones. CONCLUSIONS: We concluded that levothyroxine should remain the standard of care for treating hypothyroidism. We found no consistently strong evidence for the superiority of alternative preparations (e.g., levothyroxine-liothyronine combination therapy, or thyroid extract therapy, or others) over monotherapy with levothyroxine, in improving health outcomes. Some examples of future research needs include the development of superior biomarkers of euthyroidism to supplement thyrotropin measurements, mechanistic research on serum triiodothyronine levels (including effects of age and disease status, relationship with tissue concentrations, as well as potential therapeutic targeting), and long-term outcome clinical trials testing combination therapy or thyroid extracts (including subgroup effects). Additional research is also needed to develop thyroid hormone analogs with a favorable benefit to risk profile.


Subject(s)
Hormone Replacement Therapy , Hypothyroidism/drug therapy , Thyroid Gland/physiopathology , Thyroxine/therapeutic use , Humans , Hypothyroidism/physiopathology , Thyroid Function Tests
7.
J Clin Endocrinol Metab ; 99(5): 1537-42, 2014 May.
Article in English | MEDLINE | ID: mdl-24606101

ABSTRACT

CONTEXT: Acute infectious thyroiditis, particularly fungal thyroiditis, is rare and typically presents in immunocompromised individuals. Here we report the first case of coccidiomycosis thyroiditis occurring in an organ recipient as a consequence of likely allograft contamination and discuss the management strategies for thyroid masses in the setting of disseminated infection. EVIDENCE ACQUISITION AND SYNTHESIS: In this clinical case seminar, we summarize the previously published cases of coccidiomycosis thyroiditis based on a MEDLINE search of all peer-reviewed publications (original articles and reviews) on this topic. We identified six other cases, five of which also occurred in immunocompromised hosts, although none occurred in organ recipients. CONCLUSION: A case of coccidiomycosis thyroiditis occurring in a post-liver transplant immunocompromised host is reported. Analysis of donor serum revealed the liver allograft as the likely infectious source, resulting in hematological spread to the thyroid. Although our patient's thyroid gland was lacking gross structural abnormalities at presentation, new-onset thyroid masses developed after relative immune restoration and initiation of antifungal therapies. The differential diagnosis of new-onset thyroid masses in immunocompromised hosts is discussed, with a focus on immune reconstitution inflammatory syndrome. The role of thyroidectomy in the management of fungal thyroiditis is also discussed.


Subject(s)
Coccidioidomycosis/diagnosis , Immunocompromised Host , Liver Transplantation , Thyroiditis, Suppurative/diagnosis , Adult , Female , Humans
8.
Thyroid ; 24(1): 88-168, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24001133

ABSTRACT

BACKGROUND: An in-depth understanding of the fundamental principles that regulate thyroid hormone homeostasis is critical for the development of new diagnostic and treatment approaches for patients with thyroid disease. SUMMARY: Important clinical practices in use today for the treatment of patients with hypothyroidism, hyperthyroidism, or thyroid cancer are the result of laboratory discoveries made by scientists investigating the most basic aspects of thyroid structure and molecular biology. In this document, a panel of experts commissioned by the American Thyroid Association makes a series of recommendations related to the study of thyroid hormone economy and action. These recommendations are intended to promote standardization of study design, which should in turn increase the comparability and reproducibility of experimental findings. CONCLUSIONS: It is expected that adherence to these recommendations by investigators in the field will facilitate progress towards a better understanding of the thyroid gland and thyroid hormone dependent processes.


Subject(s)
Research Design/standards , Thyroid Gland/physiology , Animals , Behavior, Animal , Cells, Cultured , Female , Humans , Hyperthyroidism/drug therapy , Hypothyroidism/drug therapy , Iodine/deficiency , Iodine/metabolism , Iodine Radioisotopes , Models, Animal , Pregnancy , Reproducibility of Results , Thyroid Diseases/drug therapy , Thyroid Hormones/physiology
9.
Endocrinology ; 154(10): 3937-46, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23861374

ABSTRACT

Altered glucose metabolism in the heart is an important characteristic of cardiovascular and metabolic disease. Because thyroid hormones have major effects on peripheral metabolism, we examined the metabolic effects of heart-selective increase in T3 using transgenic mice expressing human type 2 iodothyronine deiodinase (D2) under the control of the α-myosin heavy chain promoter (MHC-D2). Hyperinsulinemic-euglycemic clamps showed normal whole-body glucose disposal but increased hepatic insulin action in MHC-D2 mice as compared to wild-type (WT) littermates. Insulin-stimulated glucose uptake in heart was not altered, but basal myocardial glucose metabolism was increased by more than two-fold in MHC-D2 mice. Myocardial lipid levels were also elevated in MHC-D2 mice, suggesting an overall up-regulation of cardiac metabolism in these mice. The effects of doxorubicin (DOX) treatment on cardiac function and structure were examined using M-mode echocardiography. DOX treatment caused a significant reduction in ventricular fractional shortening and resulted in more than 50% death in WT mice. In contrast, MHC-D2 mice showed increased survival rate after DOX treatment, and this was associated with a six-fold increase in myocardial glucose metabolism and improved cardiac function. Myocardial activity and expression of AMPK, GLUT1, and Akt were also elevated in MHC-D2 and WT mice following DOX treatment. Thus, our findings indicate an important role of thyroid hormone in cardiac metabolism and further suggest a protective role of glucose utilization in DOX-mediated cardiac dysfunction.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Doxorubicin/adverse effects , Glucose/metabolism , Heart Ventricles/drug effects , Insulin Resistance , Iodide Peroxidase/biosynthesis , Ventricular Dysfunction/chemically induced , AMP-Activated Protein Kinases/biosynthesis , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Glucose Clamp Technique , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Heart Ventricles/diagnostic imaging , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Survival Analysis , Triiodothyronine/metabolism , Ultrasonography , Ventricular Dysfunction/diagnostic imaging , Ventricular Dysfunction/metabolism , Ventricular Dysfunction/physiopathology , Iodothyronine Deiodinase Type II
10.
Endocrinology ; 153(8): 4039-48, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22719054

ABSTRACT

The type 2 iodothyronine selenodeiodinase (D2) is a critical determinant of local thyroid signaling, converting T(4) to the active form T(3) at the cytoplasmic face of the endoplasmic reticulum, thus supplying the nucleus with T(3) without immediately affecting circulating thyroid hormone levels. Although inhibitors of the cholesterol synthesis/isoprenylation pathway, such as hydroxy-methyl-glutaryl-coenzyme A reductase inhibitors (statins) have been to shown to down-regulate selenoproteins via interruption of normal selenocysteine incorporation, little is known about the effect of statins on D2. Here, we report that statins and prenyl transferase inhibitors actually increase D2 activity in cells with endogenous D2 expression. Although we confirmed that lovastatin (LVS) decreases the activity of transiently expressed D2 in HEK-293 cells, the prenyl transferase inhibitors increase activity in this system as well. LVS treatment increases endogenous Dio2 mRNA in MSTO-211H cells but does not alter transiently expressed Dio2 mRNA in HEK-293 cells. The prenyl transferase inhibitors do not increase Dio2 mRNA in either system, indicating that a posttranscriptional mechanism must exist. Cotreatment with LVS or the prenyl transferase inhibitors with the proteasome inhibitor MG-132 did not lead to additive increases in D2 activity, indirectly implicating the ubiquitin-proteasomal system in the mechanism. Finally, C57BL/6J mice treated with LVS or farnesyl transferase inhibitor-277 for 24 h exhibited increased D2 activity in their brown adipose tissue. These data indicate that statins and downstream inhibitors of the isoprenylation pathway may increase thyroid signaling via stimulation of D2 activity.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Iodide Peroxidase/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Cell Line , Cell Line, Tumor , Enzyme Activation/drug effects , HEK293 Cells , Humans , In Vitro Techniques , Iodide Peroxidase/genetics , Leupeptins/pharmacology , Lovastatin/pharmacology , Male , Mice , Mice, Inbred C57BL , Iodothyronine Deiodinase Type II
11.
J Lipid Res ; 50(11): 2212-21, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19502644

ABSTRACT

Pctp(-/-) mice that lack phosphatidylcholine transfer protein (Pctp) exhibit a marked shift toward utilization of fatty acids for oxidative phosphorylation, suggesting that Pctp may regulate the entry of fatty acyl-CoAs into mitochondria. Here, we examined the influence of Pctp expression on the function and structure of brown adipose tissue (BAT), a mitochondrial-rich, oxidative tissue that mediates nonshivering thermogenesis. Consistent with increased thermogenesis, Pctp(-/-) mice exhibited higher core body temperatures than wild-type controls at room temperature. During a 24 h cold challenge, Pctp(-/-) mice defended core body temperature efficiently enough that acute, full activation of BAT thermogenic genes did not occur. Brown adipocytes lacking Pctp harbored enlarged and elongated mitochondria. Consistent with increased fatty acid utilization, brown adipocytes cultured from Pctp(-/-) mice exhibited higher oxygen consumption rates in response to norepinephrine. The absence of Pctp expression during brown adipogenesis in vitro altered the expression of key transcription factors, which could be corrected by adenovirus-mediated overexpression of Pctp early but not late during the differentiation. Collectively, these findings support a key role for Pctp in limiting mitochondrial oxidation of fatty acids and thus regulating adaptive thermogenesis in BAT.


Subject(s)
Adipose Tissue, Brown/metabolism , Mitochondria/metabolism , Phospholipid Transfer Proteins/metabolism , Thermogenesis , Adaptation, Physiological , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/ultrastructure , Animals , Blotting, Western , Body Temperature , Cells, Cultured , Gene Expression Profiling , Lipid Metabolism/drug effects , Mice , Mice, Knockout , Microscopy, Electron , Microscopy, Fluorescence , Mitochondria/ultrastructure , Norepinephrine/pharmacology , Oxygen Consumption , Phospholipid Transfer Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism
12.
J Clin Endocrinol Metab ; 94(9): 3521-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19549744

ABSTRACT

CONTEXT: Fatty liver is an important complication of obesity; however, regulatory mechanisms mediating altered gene expression patterns have not been identified. OBJECTIVE: The aim of the study was to identify novel transcriptional changes in human liver that could contribute to hepatic lipid accumulation and associated insulin resistance, type 2 diabetes, and nonalcoholic steatohepatitis. DESIGN: We evaluated gene expression in surgical liver biopsies from 13 obese (nine with type 2 diabetes) and five control subjects using Affymetrix U133A microarrays. PCR validation was performed in liver biopsies using an additional 16 subjects. We also tested thyroid hormone responses in mice fed chow or high-fat diet. SETTING: Recruitment was performed in an academic medical center. PARTICIPANTS: Individuals undergoing elective surgery for obesity or gallstones participated in the study. RESULTS: The top-ranking gene set, down-regulated in obese subjects, was comprised of genes previously demonstrated to be positively regulated by T(3) in human skeletal muscle (n = 399; P < 0.001; false discovery rate = 0.07). This gene set included genes related to RNA metabolism (SNRPE, HNRPH3, TIA1, and SFRS2), protein catabolism (PSMA1, PSMD12, USP9X, IBE2B, USP16, and PCMT1), and energy metabolism (ATP5C1, COX7C, UQCRB). We verified thyroid hormone regulation of these genes in the liver after injection of C57BL/6J mice with T(3) (100 microg/100 g body weight); furthermore, T(3)-induced increases in expression of these genes were abolished by high-fat diet. In agreement, expression of these genes inversely correlated with liver fat content in humans. CONCLUSIONS: These data suggest that impaired thyroid hormone action may contribute to altered patterns of gene expression in fatty liver.


Subject(s)
Fatty Liver/metabolism , Gene Expression Regulation , Triiodothyronine/pharmacology , Adult , Animals , Diabetes Mellitus, Type 2/metabolism , Female , Heat-Shock Proteins/genetics , Humans , Insulin Resistance , Iodide Peroxidase/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Receptors, Leptin/genetics , Transcription Factors/genetics
14.
Endocrinology ; 150(4): 1976-83, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19036883

ABSTRACT

The thyroid hormone activating type 2 deiodinase (D2) is known to play a role in brown adipose tissue-mediated adaptive thermogenesis in rodents, but the finding of D2 in skeletal muscle raises the possibility of a broader metabolic role. In the current study, we examined the regulation of the D2 pathway in primary skeletal muscle myoblasts taken from both humans and mice. We found that pioglitazone treatment led to a 1.6- to 1.9-fold increase in primary human skeletal myocyte D2 activity; this effect was seen with other peroxisomal proliferator-activated receptor-gamma agonists. D2 activity in primary murine skeletal myotubes increased 2.8-fold in response to 5 microM pioglitazone and 1.6-fold in response to 5 nM insulin and increased in a dose-dependent manner in response to lithocholic acid (maximum response at 25 microM was approximately 3.8-fold). We compared Akt phosphorylation in primary myotubes derived from wild-type and D2 knockout (D2KO) mice: phospho-Akt was reduced by 50% in the D2KO muscle after 1 nM insulin exposure. Expression of T(3)-responsive muscle genes via quantitative RT-PCR suggests that D2KO cells have decreased thyroid hormone signaling, which could contribute to the abnormalities in insulin signaling. D2 activity in skeletal muscle fragments from both murine and human sources was low, on the order of about 0.01 fmol/min . mg of muscle protein. The phenotypic changes seen with D2KO cells support a metabolic role for D2 in muscle, hinting at a D2-mediated linkage between thyroid hormone and insulin signaling, but the low activity calls into question whether skeletal muscle D2 is a major source of plasma T(3).


Subject(s)
Iodide Peroxidase/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/enzymology , PPAR gamma/agonists , Animals , Cells, Cultured , Chromans/pharmacology , Enzyme Activation/drug effects , Immunohistochemistry , Insulin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Pioglitazone , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thiazolidinediones/pharmacology , Troglitazone
15.
Endocr Rev ; 29(7): 898-938, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18815314

ABSTRACT

The iodothyronine deiodinases initiate or terminate thyroid hormone action and therefore are critical for the biological effects mediated by thyroid hormone. Over the years, research has focused on their role in preserving serum levels of the biologically active molecule T(3) during iodine deficiency. More recently, a fascinating new role of these enzymes has been unveiled. The activating deiodinase (D2) and the inactivating deiodinase (D3) can locally increase or decrease thyroid hormone signaling in a tissue- and temporal-specific fashion, independent of changes in thyroid hormone serum concentrations. This mechanism is particularly relevant because deiodinase expression can be modulated by a wide variety of endogenous signaling molecules such as sonic hedgehog, nuclear factor-kappaB, growth factors, bile acids, hypoxia-inducible factor-1alpha, as well as a growing number of xenobiotic substances. In light of these findings, it seems clear that deiodinases play a much broader role than once thought, with great ramifications for the control of thyroid hormone signaling during vertebrate development and metamorphosis, as well as injury response, tissue repair, hypothalamic function, and energy homeostasis in adults.


Subject(s)
Iodide Peroxidase/metabolism , Thyroid Gland/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism , Animals , Humans , Signal Transduction , Thyroid Diseases/metabolism , Thyroid Gland/enzymology
16.
Cancer ; 111(6): 508-16, 2007 Dec 25.
Article in English | MEDLINE | ID: mdl-17999413

ABSTRACT

BACKGROUND: The diagnostic evaluation of patients with thyroid nodules is imprecise. Despite the benefits of fine-needle aspiration (FNA), most patients who are referred for surgery because of abnormal cytology prove to have benign disease. Recent technologic and procedural advances suggest that this shortcoming can be mitigated, although few data confirm this benefit in unselected patients. METHODS: A total of 2587 sequential patients were evaluated by thyroid ultrasound and were offered ultrasound-guided FNA (UG-FNA) of all thyroid nodules that measured > or =1 cm during a 10-year period. Results of aspiration cytology were correlated with histologic findings. The prevalence of thyroid cancer in all patients and in those who underwent surgery was determined. Surgical risk was calculated. RESULTS: Tumors that measured > or =1 cm were present in 14% of patients: Forty-three percent of patients had tumors that measured <2 cm in greatest dimension, and 93% had American Joint Committee on Cancer stage I or II disease. The cytologic diagnoses 'positive for malignancy' and 'no malignant cells' were 97% predictive and 99.7% predictive, respectively. Repeat FNA of initial insufficient aspirates, as well as more detailed classification of inconclusive aspirates, improved preoperative assessment of cancer risk and reduced surgical intervention. Fifty-six percent of patients who were referred for surgery because of abnormal cytology had cancer compared with from 10% to 45% of patients historically. An analysis of operative complications from a subset of 296 patients demonstrated a 1% risk of permanent surgical complications. CONCLUSIONS: The current findings demonstrated the benefits of UG-FNA and of a more detailed classification of inconclusive aspirates in the preoperative risk assessment of thyroid nodules, supporting adherence to recently published guidelines.


Subject(s)
Biopsy, Fine-Needle , Thyroid Nodule/diagnostic imaging , Thyroid Nodule/pathology , Adolescent , Adult , Aged , Cytodiagnosis , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Sensitivity and Specificity , Thyroid Nodule/surgery , Thyroidectomy , Ultrasonography
18.
Diabetes ; 56(3): 767-76, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17327447

ABSTRACT

Disturbances in energy homeostasis can result in obesity and other metabolic diseases. Here we report a metabolic pathway present in normal human skeletal muscle myoblasts that is activated by the small polyphenolic molecule kaempferol (KPF). Treatment with KPF leads to an approximately 30% increase in skeletal myocyte oxygen consumption. The mechanism involves a several-fold increase in cyclic AMP (cAMP) generation and protein kinase A activation, and the effect of KPF can be mimicked via treatment with dibutyryl cAMP. Microarray and real-time PCR studies identified a set of metabolically relevant genes influenced by KPF including peroxisome proliferator-activated receptor gamma coactivator-1alpha, carnitine palmitoyl transferase-1, mitochondrial transcription factor 1, citrate synthase, and uncoupling protein-3, although KPF itself is not a direct mitochondrial uncoupler. The cAMP-responsive gene for type 2 iodothyronine deiodinase (D2), an intracellular enzyme that activates thyroid hormone (T3) for the nucleus, is approximately threefold upregulated by KPF; furthermore, the activity half-life for D2 is dramatically and selectively increased as well. The net effect is an approximately 10-fold stimulation of D2 activity as measured in cell sonicates, with a concurrent increase of approximately 2.6-fold in the rate of T3 production, which persists even 24 h after KPF has been removed from the system. The effects of KPF on D2 are independent of sirtuin activation and only weakly reproduced by other small polyphenolic molecules such as quercetin and fisetin. These data document a novel mechanism by which a xenobiotic-activated pathway can regulate metabolically important genes as well as thyroid hormone activation and thus may influence metabolic control in humans.


Subject(s)
Energy Metabolism/drug effects , Kaempferols/pharmacology , Triiodothyronine/metabolism , Animals , Cell Line , Chalcones/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Myoblasts/drug effects , Oxygen Consumption/drug effects , RNA Interference , Rats , Resveratrol , Stilbenes/pharmacology , Iodothyronine Deiodinase Type II
19.
J Clin Invest ; 116(10): 2571-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17016550

ABSTRACT

The deiodinases activate or inactivate thyroid hormone, and their importance in thyroid hormone homeostasis has become increasingly clear with the availability of deiodinase-deficient animals. At the same time, heightened interest in the field has been generated following the discovery that the type 2 deiodinase can be an important component in both the Hedgehog signaling pathway and the G protein-coupled bile acid receptor 1-mediated (GPBAR1-mediated) signaling cascade. The discovery of these new roles for the deiodinases indicates that tissue-specific deiodination plays a much broader role than once thought, extending into the realms of developmental biology and metabolism.


Subject(s)
Iodide Peroxidase/metabolism , Thyroid Hormones/metabolism , Animals , Energy Metabolism/physiology , Homeostasis/physiology , Humans , Iodide Peroxidase/chemistry , Iodide Peroxidase/physiology , Models, Biological , Models, Molecular , Protein Conformation , Signal Transduction/physiology
20.
Nature ; 439(7075): 484-9, 2006 Jan 26.
Article in English | MEDLINE | ID: mdl-16400329

ABSTRACT

While bile acids (BAs) have long been known to be essential in dietary lipid absorption and cholesterol catabolism, in recent years an important role for BAs as signalling molecules has emerged. BAs activate mitogen-activated protein kinase pathways, are ligands for the G-protein-coupled receptor (GPCR) TGR5 and activate nuclear hormone receptors such as farnesoid X receptor alpha (FXR-alpha; NR1H4). FXR-alpha regulates the enterohepatic recycling and biosynthesis of BAs by controlling the expression of genes such as the short heterodimer partner (SHP; NR0B2) that inhibits the activity of other nuclear receptors. The FXR-alpha-mediated SHP induction also underlies the downregulation of the hepatic fatty acid and triglyceride biosynthesis and very-low-density lipoprotein production mediated by sterol-regulatory-element-binding protein 1c. This indicates that BAs might be able to function beyond the control of BA homeostasis as general metabolic integrators. Here we show that the administration of BAs to mice increases energy expenditure in brown adipose tissue, preventing obesity and resistance to insulin. This novel metabolic effect of BAs is critically dependent on induction of the cyclic-AMP-dependent thyroid hormone activating enzyme type 2 iodothyronine deiodinase (D2) because it is lost in D2-/- mice. Treatment of brown adipocytes and human skeletal myocytes with BA increases D2 activity and oxygen consumption. These effects are independent of FXR-alpha, and instead are mediated by increased cAMP production that stems from the binding of BAs with the G-protein-coupled receptor TGR5. In both rodents and humans, the most thermogenically important tissues are specifically targeted by this mechanism because they coexpress D2 and TGR5. The BA-TGR5-cAMP-D2 signalling pathway is therefore a crucial mechanism for fine-tuning energy homeostasis that can be targeted to improve metabolic control.


Subject(s)
Bile Acids and Salts/pharmacology , Energy Metabolism/drug effects , Thyroid Hormones/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/metabolism , Adiposity/drug effects , Animals , Body Weight/drug effects , Carbon Dioxide/metabolism , Cholic Acid/pharmacology , Cyclic AMP/biosynthesis , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Gene Deletion , Homeostasis/drug effects , Humans , Iodide Peroxidase/deficiency , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Muscle Cells/drug effects , Muscle Cells/enzymology , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Oxygen Consumption/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Iodothyronine Deiodinase Type II
SELECTION OF CITATIONS
SEARCH DETAIL
...