Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
2.
J Biol Chem ; 295(26): 8834-8845, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32398261

ABSTRACT

Anaplastic thyroid cancer (ATC) is one of the most aggressive human malignancies, with an average life expectancy of ∼6 months from the time of diagnosis. The genetic and epigenetic changes that underlie this malignancy are incompletely understood. We found that ASH1-like histone lysine methyltransferase (ASH1L) is overexpressed in ATC relative to the much less aggressive and more common differentiated thyroid cancer. This increased expression was due at least in part to reduced levels of microRNA-200b-3p (miR-200b-3p), which represses ASH1L expression, in ATC. Genetic knockout of ASH1L protein expression in ATC cell lines decreased cell growth both in culture and in mouse xenografts. RNA-Seq analysis of ASH1L knockout versus WT ATC cell lines revealed that ASH1L is involved in the regulation of numerous cancer-related genes and gene sets. The pro-oncogenic long noncoding RNA colon cancer-associated transcript 1 (CCAT1) was one of the most highly (approximately 68-fold) down-regulated transcripts in ASH1L knockout cells. Therefore, we investigated CCAT1 as a potential mediator of the growth-inducing activity of ASH1L. Supporting this hypothesis, CCAT1 knockdown in ATC cells decreased their growth rate, and ChIP-Seq data indicated that CCAT1 is likely a direct target of ASH1L's histone methyltransferase activity. These results indicate that ASH1L contributes to the aggressiveness of ATC and suggest that ASH1L, along with its upstream regulator miR-200b-3p and its downstream mediator CCAT1, represents a potential therapeutic target in ATC.


Subject(s)
DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase/genetics , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Neoplasms/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Mice, SCID , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/pathology
3.
J Biol Chem ; 294(48): 18408-18420, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31615896

ABSTRACT

Although adipogenesis is mainly controlled by a small number of master transcription factors, including CCAAT/enhancer-binding protein family members and peroxisome proliferator-activated receptor γ (PPARγ), other transcription factors also are involved in this process. Thyroid cancer cells expressing a paired box 8 (PAX8)-PPARγ fusion oncogene trans-differentiate into adipocyte-like cells in the presence of the PPARγ ligand pioglitazone, but this trans-differentiation is inhibited by the transcription factor NK2 homeobox 1 (NKX2-1). Here, we tested whether NKX family members may play a role also in normal adipogenesis. Using quantitative RT-PCR (RT-qPCR), we examined the expression of all 14 NKX family members during 3T3-L1 adipocyte differentiation. We found that most NKX members, including NKX2-1, are expressed at very low levels throughout differentiation. However, mRNA and protein expression of a related family member, NKX1-2, was induced during adipocyte differentiation. NKX1-2 also was up-regulated in cultured murine ear mesenchymal stem cells (EMSCs) during adipogenesis. Importantly, shRNA-mediated NKX1-2 knockdown in 3T3-L1 preadipocytes or EMSCs almost completely blocked adipocyte differentiation. Furthermore, NKX1-2 overexpression promoted differentiation of the ST2 bone marrow-derived mesenchymal precursor cell line into adipocytes. Additional findings suggested that NKX1-2 promotes adipogenesis by inhibiting expression of the antiadipogenic protein COUP transcription factor II. Bone marrow mesenchymal precursor cells can differentiate into adipocytes or osteoblasts, and we found that NKX1-2 both promotes ST2 cell adipogenesis and inhibits their osteoblastogenic differentiation. These results support a role for NKX1-2 in promoting adipogenesis and possibly in regulating the balance between adipocyte and osteoblast differentiation of bone marrow mesenchymal precursor cells.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation/genetics , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Osteoblasts/metabolism , Transcription Factors/genetics , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cell Line , Cells, Cultured , Gene Expression Regulation , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Osteoblasts/cytology , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , RNA Interference , Transcription Factors/metabolism
4.
Article in English | MEDLINE | ID: mdl-29946481

ABSTRACT

BACKGROUND: Malignant struma ovarii is an ovarian teratoma containing at least 50% thyroid tissue which has the potential to metastasize and produce thyroid hormone. Given its rarity, management strategies are not well-established. We report a case of metastatic malignant struma ovarii discovered during pregnancy with lessons for evaluation and management. CASE PRESENTATION: A 30-year-old woman who was two months pregnant was discovered to have struma ovarii with over half of the struma comprised of papillary thyroid cancer. Following tumor resection, delivery, and thyroidectomy, she underwent evaluation with stimulated thyroglobulin testing and diagnostic staging sodium iodide-131 scan (I-131), which revealed the presence of skeletal metastases. Following administration of 320 mCi I-131, post-therapy scan also showed miliary pulmonary metastases with improved ability to localize the bony and pulmonary metastases with concurrent SPECT/CT imaging. A second dosimetry-guided I-131 therapy resulted in complete resolution of pulmonary metastases; however, small foci of residual bone disease persisted. Post-therapy scans demonstrated additional findings not shown on diagnostic I-131 scans obtained prior to both her initial and second I-131 therapy. CONCLUSIONS: SPECT/CT provides accurate anatomic correlation and localization of metastatic foci and can serve as a baseline study to assess interval response to treatment. Post-therapy scans should always be obtained when I-131 treatment is administered, as additional findings may be revealed versus low dose I-131 activity diagnostic scans. This patient had a high metastatic burden that would not have been discovered in a timely fashion with the conservative approach advocated by others. Thyroidectomy followed by a diagnostic staging radioiodine scan and a stimulated thyroglobulin level should be considered in patients with malignant struma ovarii for guiding therapeutic I-131 administration as metastatic risk is difficult to predict based on histopathologic examination.

5.
J Endocr Soc ; 2(5): 437-443, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29696242

ABSTRACT

Fibroblast growth factor 23 (FGF23)-induced hypophosphatemia is a rare paraneoplastic syndrome of phosphate wasting that, if unrecognized, may cause tumor-induced osteomalacia. It is classically associated with benign mesenchymal tumors but occasionally has been found in patients with other malignancies. Hypophosphatemia has been associated with acute leukemia but has not previously been reported to be due to inappropriate FGF23 secretion. Here, we describe FGF23-induced severe hypophosphatemia and renal phosphate wasting associated with a mixed-phenotype Philadelphia chromosome-like acute leukemia in a previously healthy 22-year-old man. He was found to have low serum 1,25-dihydroxyvitamin D and extremely high FGF23 levels, as well as inappropriate urinary phosphorus excretion. The hypophosphatemia improved with calcitriol and oral phosphate treatment but normalized only during chemotherapy-induced ablation of the blasts. FGF23 levels declined with a reduction in peripheral blast counts. Using real-time reverse transcription polymerase chain reaction, we found that the leukemia cells were the source of FGF23. To our knowledge, this is the first description of FGF23-induced hypophosphatemia associated with acute leukemia. We recommend that the FGF23 paraneoplastic syndrome be considered as a possible etiology of hypophosphatemia in patients with acute leukemia.

6.
Endocrinology ; 159(3): 1463-1468, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29420754

ABSTRACT

Peroxisome proliferator-activated receptor γ (PPARγ) is widely expressed at low levels and regulates many physiological processes. In mice and humans, there is evidence that PPARγ can function as a tumor suppressor. A PAX8-PPARγ fusion protein (PPFP) is oncogenic in a subset of thyroid cancers, suggesting that inhibition of endogenous PPARγ function by the fusion protein could contribute to thyroid oncogenesis. However, the function of PPARγ within thyrocytes has never been directly tested. Therefore, we have created a thyroid-specific genetic knockout of murine Pparg and have studied thyroid biology in these mice. Thyroid size and histology, the expression of thyroid-specific genes, and serum T4 levels all are unaffected by loss of thyroidal PPARγ expression. PPFP thyroid cancers have increased activation of AKT, and mice with thyroid-specific expression of PPFP combined with thyroid-specific loss of PTEN (a negative regulator of AKT) develop thyroid cancer. Therefore we created mice with combined thyroid-specific deletions of Pparg and Pten to test if there is oncogenic synergy between these deletions. Pten deletion alone results in benign thyroid hyperplasia, and this is unchanged when combined with deletion of Pparg. We conclude that, at least in the contexts studied, thyrocyte PPARγ does not play a significant role in the development or function of the thyroid and does not function as a tumor suppressor.


Subject(s)
Gene Deletion , PPAR gamma/genetics , Thyroid Gland/metabolism , Animals , Female , Humans , Male , Mice , Mice, Knockout , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Organ Specificity , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , PPAR gamma/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Thyroid Gland/pathology , Thyroid Hormones/blood , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
7.
J Clin Endocrinol Metab ; 103(4): 1277-1281, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29373711

ABSTRACT

Context: A subset of thyroid carcinomas expresses an oncogenic paired box 8 (PAX8) and peroxisome proliferator activated receptor γ (PPARγ) fusion protein (PPFP). The PPARγ/PPFP ligand pioglitazone is highly therapeutic in a transgenic mouse model of PPFP thyroid carcinoma, but whether pioglitazone is therapeutic in patients with PPFP thyroid carcinoma is unknown. Case Description: Tumor blocks from 40 patients with progressive thyroid cancer despite standard-of-care therapy were screened for PPFP, and the tumor from only one patient (2.5%) was positive. The patient had a 6.0-cm acetabular soft tissue metastasis from Hürthle cell carcinoma that caused severe pain on weight bearing and had a serum thyroglobulin level of 1974 ng/mL. After 24 weeks of therapy with pioglitazone, the metastatic lesion was 3.9 cm, the thyroglobulin level was 49.4 ng/mL, and the patient was pain-free. Thirteen months after discontinuation of pioglitazone, the metastatic lesion was 3.6 cm, the thyroglobulin level was 4.7 ng/mL, and the patient remained pain-free. Conclusions: Pioglitazone may be therapeutic in patients with PPFP thyroid cancer. However, thyroid cancers that are progressive despite standard-of-care therapy appear to only rarely express PPFP.


Subject(s)
Antineoplastic Agents/therapeutic use , Oncogene Proteins, Fusion/analysis , Thiazolidinediones/therapeutic use , Thyroid Neoplasms/drug therapy , Adenoma, Oxyphilic/chemistry , Adenoma, Oxyphilic/diagnostic imaging , Adenoma, Oxyphilic/drug therapy , Adenoma, Oxyphilic/secondary , Aged, 80 and over , Humans , Male , Mutation , Pioglitazone , Soft Tissue Neoplasms/chemistry , Soft Tissue Neoplasms/diagnostic imaging , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/secondary , Thyroid Neoplasms/chemistry , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/genetics , Thyroid Neoplasms/secondary , Tomography, X-Ray Computed
8.
Oncotarget ; 8(4): 5761-5773, 2017 Jan 24.
Article in English | MEDLINE | ID: mdl-28008156

ABSTRACT

PAX8-PPARG fusion protein (PPFP) results from a t(2;3)(q13;p25) chromosomal translocation, is found in 30% of follicular thyroid carcinomas, and demonstrates oncogenic capacity in transgenic mice. A PPARG ligand, pioglitazone, is highly therapeutic in mice with PPFP thyroid cancer. However, only limited data exist to characterize the binding sites and oncogenic function of PPFP, or to explain the observed therapeutic effect of pioglitazone. Here we used our previously characterized transgenic mouse model of PPFP follicular thyroid carcinoma to identify PPFP binding sites in vivo using ChIP-seq, and to distinguish genes and pathways regulated directly or indirectly by PPFP with and without pioglitazone treatment via integration with RNA-seq data. PPFP bound to DNA regions containing the PAX8 and/or the PPARG motif, near genes involved in lipid metabolism, the cell cycle, apoptosis, and cell motility; the binding site distribution was highly concordant with our previous study in a rat PCCL3 cell line. Most strikingly, pioglitazone induced an immune cell infiltration including macrophages and T cells only in the presence of PPFP, which may be central to its therapeutic effect.


Subject(s)
Adenocarcinoma, Follicular/genetics , Gene Regulatory Networks , PAX8 Transcription Factor/metabolism , PPAR gamma/metabolism , Thyroid Neoplasms/genetics , Adenocarcinoma, Follicular/drug therapy , Adenocarcinoma, Follicular/metabolism , Animals , Binding Sites , Chromatin Immunoprecipitation , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Mice , Mice, Transgenic , PAX8 Transcription Factor/genetics , PPAR gamma/genetics , Pioglitazone , Rats , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Sequence Analysis, RNA , Signal Transduction , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Translocation, Genetic
9.
Sci Rep ; 6: 35531, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759039

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), the most common form of chronic liver disease, manifests as an over-accumulation of hepatic fat. We have recently shown that mice with genetic knockout of a long non-coding RNA (lncRNA) steroid receptor RNA activator (SRA) (SRAKO) are resistant to high fat diet-induced obesity with a phenotype that includes improved glucose tolerance and attenuated hepatic steatosis. The underlying mechanism was investigated in the present study. We found that hepatic levels of SRA and adipose triglyceride lipase (ATGL), a major hepatic triacylglycerol (TAG) hydrolase, were inversely regulated by fasting in mice, and the expression of liver ATGL was induced by SRAKO under normal and high fat diet (HFD) feeding. Loss of SRA in primary hepatocytes or a hepatocyte cell line upregulates, but forced expression of SRA inhibits ATGL expression and free fatty acids (FFA) ß-oxidation. SRA inhibits ATGL promoter activity, primarily by inhibiting the otherwise-inductive effects of the transcription factor, forkhead box protein O1 (FoxO1). Our data reveal a novel function of SRA in promoting hepatic steatosis through repression of ATGL expression.


Subject(s)
Carrier Proteins/genetics , Fatty Liver/metabolism , Hepatocytes/physiology , Liver/pathology , Non-alcoholic Fatty Liver Disease/genetics , RNA, Long Noncoding/genetics , Animals , Diet, High-Fat , Fasting , Fatty Liver/genetics , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation , Hep G2 Cells , Humans , Lipase/genetics , Lipase/metabolism , Lipid Peroxidation , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/genetics , RNA, Small Interfering/genetics , Up-Regulation
10.
J Biol Chem ; 291(37): 19274-86, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27435678

ABSTRACT

A subset of thyroid carcinomas contains a t(2;3)(q13;p25) chromosomal translocation that fuses paired box gene 8 (PAX8) with the peroxisome proliferator-activated receptor γ gene (PPARG), resulting in expression of a PAX8-PPARγ fusion protein, PPFP. We previously generated a transgenic mouse model of PPFP thyroid carcinoma and showed that feeding the PPARγ agonist pioglitazone greatly decreased the size of the primary tumor and prevented metastatic disease in vivo The antitumor effect correlates with the fact that pioglitazone turns PPFP into a strongly PPARγ-like molecule, resulting in trans-differentiation of the thyroid cancer cells into adipocyte-like cells that lose malignant character as they become more differentiated. To further study this process, we performed cell culture experiments with thyrocytes from the PPFP mouse thyroid cancers. Our data show that pioglitazone induced cellular lipid accumulation and the expression of adipocyte marker genes in the cultured cells, and shRNA knockdown of PPFP eliminated this pioglitazone effect. In addition, we found that PPFP and thyroid transcription factor 1 (TTF-1) physically interact, and that these transcription factors bind near each other on numerous target genes. TTF-1 knockdown and overexpression studies showed that TTF-1 inhibits PPFP target gene expression and impairs adipogenic trans-differentiation. Surprisingly, pioglitazone repressed TTF-1 expression in PPFP-expressing thyrocytes. Our data indicate that TTF-1 interacts with PPFP to inhibit the pro-adipogenic response to pioglitazone, and that the ability of pioglitazone to decrease TTF-1 expression contributes to its pro-adipogenic action.


Subject(s)
Adipogenesis , Cell Differentiation , Oncogene Proteins, Fusion/metabolism , PAX8 Transcription Factor/metabolism , PPAR gamma/metabolism , Thyroid Neoplasms/metabolism , Animals , Cell Line, Tumor , Mice , Nuclear Proteins , Oncogene Proteins, Fusion/genetics , PAX8 Transcription Factor/genetics , PPAR gamma/genetics , Rats , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Nuclear Factor 1 , Transcription Factors
11.
Oncotarget ; 6(38): 40418-32, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26595524

ABSTRACT

A chromosomal translocation results in production of an oncogenic PAX8-PPARG fusion protein (PPFP) in thyroid carcinomas. PAX8 is a thyroid transcription factor, and PPARG is a transcription factor that plays important roles in adipocytes and macrophages. PPFP retains the DNA binding domains of both proteins; however, the genomic binding sites of PPFP have not been identified, and only limited data exist to characterize gene expression in PPFP thyroid carcinomas. Therefore, the oncogenic function of PPFP is poorly understood. We expressed PPFP in PCCL3 rat thyroid cells and used ChIP-seq to identify PPFP genomic binding sites (PPFP peaks) and RNA-seq to characterize PPFP-dependent gene expression. PPFP peaks (~20,000) include known PAX8 and PPARG binding sites and are enriched with both motifs, indicating that both DNA binding domains are functional. PPFP binds to and regulates many genes involved in cancer-related processes. In PCCL3 thyroid cells, PPFP binds to adipocyte PPARG target genes in preference to macrophage PPARG target genes, consistent with the pro-adipogenic nature of PPFP and its ligand pioglitazone in thyroid cells. PPFP induces oxidative stress in thyroid cells, and pioglitazone increases susceptibility to further oxidative stress. Our data highlight the complexity of PPFP as a transcription factor and the numerous ways that it regulates thyroid oncogenesis.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Genome/genetics , Oncogene Proteins, Fusion/genetics , PPAR gamma/genetics , Paired Box Transcription Factors/genetics , Thyroid Neoplasms/genetics , Animals , Chromatin Immunoprecipitation , Flow Cytometry , High-Throughput Nucleotide Sequencing/methods , Hypoglycemic Agents/pharmacology , PAX8 Transcription Factor , Pioglitazone , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thiazolidinediones/pharmacology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Tumor Cells, Cultured
12.
Thyroid ; 25(10): 1162-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26179797

ABSTRACT

BACKGROUND: It can be useful to know the transgene insertion site in transgenic mice for a variety of reasons, but determining the insertion site generally is a time consuming, expensive, and laborious task. METHODS: A simple method is presented to determine transgene insertion sites that combines the enrichment of a sequencing library by polymerase chain reaction (PCR) for sequences containing the transgene, followed by next-generation sequencing of the enriched library. This method was applied to determine the site of integration of the thyroid peroxidase promoter-Cre recombinase mouse transgene that is commonly used to create thyroid-specific gene deletions. RESULTS: The insertion site was found to be between bp 12,372,316 and 12,372,324 on mouse chromosome 9, with the nearest characterized genes being Cntn5 and Jrkl, ∼1.5 and 0.9 Mbp from the transgene, respectively. One advantage of knowing a transgene insertion site is that it facilitates distinguishing hemizygous from homozygous transgenic mice. Although this can be accomplished by real-time quantitative PCR, the expected Ct difference is only one cycle, which is challenging to assess accurately. Therefore, the transgene insertion site information was used to develop a 3-primer qualitative PCR assay that readily distinguishes wild type, hemizygous, and homozygous TPO-Cre mice based upon size differences of the wild type and transgenic allele PCR products. CONCLUSIONS: Identification of the genomic insertion site of the thyroid peroxidase promoter-Cre mouse transgene should facilitate the use of these mice in studies of thyroid biology.


Subject(s)
Iodide Peroxidase/genetics , Promoter Regions, Genetic , Transgenes , Animals , High-Throughput Nucleotide Sequencing , Integrases/genetics , Mice , Mice, Transgenic
13.
Article in English | MEDLINE | ID: mdl-28702232

ABSTRACT

A 53 year old woman was referred to us because of large goiter, enlarged pituitary and grossly elevated TSH and free T4. The differential diagnosis included a TSH producing adenoma vs. artifactual laboratory tests. A careful step-by step analysis of different possibilities allowed correct diagnosis and treatment.

14.
Nat Rev Endocrinol ; 10(10): 616-23, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25069464

ABSTRACT

Thyroid carcinoma is the most common endocrine malignancy, and its incidence is continuing to increase. Most thyroid carcinomas contain one of several known driver mutations, such as the Val600Glu substitution in B-Raf, Ras mutations, RET gene fusions, or PAX8-PPARG gene fusions. The PAX8-PPARG gene fusion results in the production of a Pax-8-PPAR-γ fusion protein (PPFP), which is found in approximately one-third of follicular thyroid carcinomas, as well as some follicular-variant papillary thyroid carcinomas. In vitro and in vivo evidence indicates that PPFP is an oncoprotein. Although specific mechanisms of action remain to be defined, PPFP is considered to act as a dominant-negative inhibitor of wild-type PPAR-γ and/or as a unique transcriptional activator of subsets of PPAR-γ-responsive and Pax-8-responsive genes. Detection of the fusion transcript in thyroid nodule biopsy specimens can aid clinical decision-making when cytological findings are indeterminate. The PPAR-γ agonist pioglitazone is highly therapeutic in a transgenic mouse model of PPFP-positive thyroid carcinoma, suggesting that PPAR-γ agonists might be beneficial in patients with PPFP-positive thyroid carcinomas.


Subject(s)
Biomarkers, Tumor/genetics , Gene Fusion/genetics , PPAR gamma/genetics , Paired Box Transcription Factors/genetics , Thyroid Neoplasms/genetics , Animals , Humans , PAX8 Transcription Factor , Thyroid Neoplasms/diagnosis
15.
PLoS One ; 9(4): e95416, 2014.
Article in English | MEDLINE | ID: mdl-24743795

ABSTRACT

The Steroid Receptor RNA Activator (SRA) enhances adipogenesis and increases both glucose uptake and phosphorylation of Akt and FOXO1 in response to insulin. To assess the mechanism, we differentiated ST2 mesenchymal precursor cells that did or did not overexpress SRA into adipocytes using combinations of methylisobutylxanthine, dexamethasone and insulin. These studies showed that SRA overexpression promotes full adipogenesis in part by stimulation of insulin/insulin-like growth factor-1 (IGF-1) signaling. SRA overexpression inhibited phosphorylation of p38 mitogen activated protein kinase (MAPK) and c-Jun NH2-terminal kinase (JNK) in the early differentiation of ST2 cells. Conversely, knockdown of endogenous SRA in 3T3-L1 cells increased phosphorylation of JNK. Knockdown of SRA in mature 3T3-L1 adipocytes reduced insulin receptor (IR) mRNA and protein levels, which led to decreased autophosphorylation of IRß and decreased phosphorylation of insulin receptor substrate-1 (IRS-1) and Akt. This likely reflects a stimulatory role of SRA on IR transcription, as transfection studies showed that SRA increased expression of an IR promoter-luciferase reporter construct.


Subject(s)
Adipogenesis/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , RNA, Long Noncoding/metabolism , Receptor, Insulin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , 3T3 Cells , Adipogenesis/genetics , Animals , Cell Line , Humans , Immunoblotting , Mice , Phosphorylation , RNA, Long Noncoding/genetics , Receptor, Insulin/genetics , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/genetics
16.
J Biol Chem ; 289(19): 13000-9, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24675075

ABSTRACT

We have recently shown that the non-coding RNA, steroid receptor RNA activator (SRA), functions as a transcriptional coactivator of PPARγ and promotes adipocyte differentiation in vitro. To assess SRA function in vivo, we have generated a whole mouse Sra1 gene knock-out (SRA(-/-)). Here, we show that the Sra1 gene is an important regulator of adipose tissue mass and function. SRA is expressed at a higher level in adipose tissue than other organs in wild type mice. SRA(-/-) mice are resistant to high fat diet-induced obesity, with decreased fat mass and increased lean content. This lean phenotype of SRA(-/-) mice is associated with decreased expression of a subset of adipocyte marker genes and reduced plasma TNFα levels. The SRA(-/-) mice are more insulin sensitive, as evidenced by reduced fasting insulin, and lower blood glucoses in response to IP glucose and insulin. In addition, the livers of SRA(-/-) mice have fewer lipid droplets after high fat diet feeding, and the expression of lipogenesis-associated genes is decreased. To our knowledge, these data are the first to indicate a functional role for SRA in adipose tissue biology and glucose homeostasis in vivo.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Blood Glucose/metabolism , Dietary Fats/adverse effects , Obesity/metabolism , RNA, Long Noncoding/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Blood Glucose/genetics , Dietary Fats/pharmacology , Homeostasis/genetics , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , RNA, Long Noncoding/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
Mol Cell Endocrinol ; 386(1-2): 55-66, 2014 Apr 05.
Article in English | MEDLINE | ID: mdl-23933154

ABSTRACT

Thyroid carcinoma is the most common endocrine malignancy, and although the disease generally has an excellent prognosis, therapeutic options are limited for patients not cured by surgery and radioiodine. Thyroid carcinomas commonly contain one of a small number of recurrent genetic mutations. The identification and study of these mutations has led to a deeper understanding of the pathophysiology of this disease and is providing new approaches to diagnosis and therapy. Papillary thyroid carcinomas usually contain an activating mutation in the RAS cascade, most commonly in BRAF and less commonly in RAS itself or through gene fusions that activate RET. A chromosomal translocation that results in production of a PAX8-PPARG fusion protein is found in follicular carcinomas. Anaplastic carcinomas may contain some of the above changes as well as additional mutations. Therapies that are targeted to these mutations are being used in patient care and clinical trials.


Subject(s)
Epigenesis, Genetic , Thyroid Neoplasms/genetics , Animals , Humans , Mice , Mutation/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
18.
Endocr Relat Cancer ; 20(5): 725-39, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24025583

ABSTRACT

A chromosomal translocation results in the production of a paired box 8-peroxisome proliferator-activated receptor gamma (PAX8-PPARG) fusion protein (PPFP) in ∼35% of follicular thyroid carcinomas. To examine the role of PPFP in thyroid oncogenesis, the fusion protein was stably expressed in the non-transformed rat thyroid cell line PCCL3. PPFP conferred on PCCL3 cells the ability to invade through Matrigel and to form colonies in anchorage-independent conditions. PPFP also increased the fraction of cells with Wnt/TCF-responsive green fluorescent protein reporter gene expression. This Wnt/TCF-activated population was enriched for colony-forming and invading cells. These actions of PPFP required a functional PPARG DNA binding domain (DBD) within PPFP and were further stimulated by PPARG agonists. These data indicate that PPFP, through its PPARG DBD, induces Wnt/TCF pathway activation in a subpopulation of cells, and these cells have properties of cellular transformation including increased invasiveness and anchorage-independent growth.


Subject(s)
Oncogene Proteins, Fusion/metabolism , PPAR gamma/metabolism , Paired Box Transcription Factors/metabolism , TCF Transcription Factors/metabolism , Wnt Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Humans , Mice , Mice, Transgenic , PPAR gamma/agonists , Phenotype , Pioglitazone , Rats , Thiazolidinediones/pharmacology , Thyroid Neoplasms/metabolism
20.
Ann Surg ; 258(2): 354-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23567930

ABSTRACT

UNLABELLED: By linking surgeon surveys to the National Cancer Database, we found that surgeons' tendency to perform more extensive thyroid resection is associated with greater use of radioactive iodine for stage I thyroid cancer. OBJECTIVE: To determine the relationships between surgeon recommendations for extent of resection and radioactive iodine use in low-risk thyroid cancer. BACKGROUND: There has been an increase in thyroid cancer treatment intensity; the relationship between extent of resection and medical treatment with radioactive iodine remains unknown. METHODS: We randomly surveyed thyroid surgeons affiliated with 368 hospitals with Commission on Cancer-accredited cancer programs. Survey responses were linked to the National Cancer Database. The relationship between extent of resection and the proportion of the American Joint Committee on Cancer stage I well-differentiated thyroid cancer patients treated with radioactive iodine after total thyroidectomy was assessed with multivariable weighted regression, controlling for hospital and surgeon characteristics. RESULTS: The survey response rate was 70% (560/804). Surgeons who recommend total thyroidectomy over lobectomy for subcentimeter unifocal thyroid cancer were significantly more likely to recommend prophylactic central lymph node dissection for thyroid cancer regardless of tumor size (P < 0.001). They were also more likely to favor radioactive iodine in patients with intrathyroidal unifocal cancer ≤1 cm (P = 0.001), 1.1-2 cm (P = 0.004), as well as intrathyroidal multifocal cancer ≤1 cm (P = 0.004). In multivariable analysis, high hospital case volume, fewer surgeon years of experience, general surgery specialty, and preference for more extensive resection were independently associated with greater hospital-level use of radioactive iodine for stage I disease. CONCLUSIONS: In addition to surgeon experience and specialty, surgeons' tendency to perform more extensive thyroid resection is associated with greater use of radioactive iodine for stage I thyroid cancer.


Subject(s)
Iodine Radioisotopes/therapeutic use , Practice Patterns, Physicians'/statistics & numerical data , Thyroid Neoplasms/radiotherapy , Thyroid Neoplasms/surgery , Thyroidectomy/methods , Adult , Databases, Factual , Female , Health Care Surveys , Humans , Male , Middle Aged , Multivariate Analysis , Neck Dissection/statistics & numerical data , Radiotherapy, Adjuvant/statistics & numerical data , Regression Analysis , Thyroidectomy/statistics & numerical data , United States
SELECTION OF CITATIONS
SEARCH DETAIL