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










Publication year range
1.
Clin Cancer Res ; 30(7): 1352-1366, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37921808

ABSTRACT

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


Subject(s)
Symporters , Thyroid Neoplasms , Mice , Animals , Humans , Vorinostat/pharmacology , Sodium Pertechnetate Tc 99m/metabolism , Iodine Radioisotopes/therapeutic use , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics , Symporters/genetics , Symporters/metabolism , Histone Deacetylase Inhibitors , Cell Line, Tumor
2.
Cell Chem Biol ; 29(3): 502-516.e7, 2022 03 17.
Article in English | MEDLINE | ID: mdl-34520744

ABSTRACT

The sodium iodide symporter (NIS) functions to transport iodide and is critical for successful radioiodide ablation of cancer cells. Approaches to bolster NIS function and diminish recurrence post-radioiodide therapy are impeded by oncogenic pathways that suppress NIS, as well as the inherent complexity of NIS regulation. Here, we utilize NIS in high-throughput drug screening and undertake rigorous evaluation of lead compounds to identify and target key processes underpinning NIS function. We find that multiple proteostasis pathways, including proteasomal degradation and autophagy, are central to the cellular processing of NIS. Utilizing inhibitors targeting distinct molecular processes, we pinpoint combinatorial drug strategies giving robust >5-fold increases in radioiodide uptake. We also reveal significant dysregulation of core proteostasis genes in human tumors, identifying a 13-gene risk score classifier as an independent predictor of recurrence in radioiodide-treated patients. We thus propose and discuss a model for targetable steps of intracellular processing of NIS function.


Subject(s)
Neoplasms , Symporters , Biological Transport , Humans , Symporters/genetics , Symporters/metabolism
3.
J Clin Endocrinol Metab ; 107(5): 1392-1406, 2022 04 19.
Article in English | MEDLINE | ID: mdl-34791326

ABSTRACT

CONTEXT: Thyroid cancer recurrence is associated with increased mortality and adverse outcomes. Recurrence risk is currently predicted using clinical tools, often restaging patients after treatment. Detailed understanding of recurrence risk at disease onset could lead to personalized and improved patient care. OBJECTIVE: We aimed to perform a comprehensive bioinformatic and experimental analysis of 3 levels of genetic change (mRNA, microRNA, and somatic mutation) apparent in recurrent tumors and construct a new combinatorial prognostic risk model. METHODS: We analyzed The Cancer Genome Atlas data (TCGA) to identify differentially expressed genes (mRNA/microRNA) in 46 recurrent vs 455 nonrecurrent thyroid tumors. Two exonic mutational pipelines were used to identify somatic mutations. Functional gene analysis was performed in cell-based assays in multiple thyroid cell lines. The prognostic value of genes was evaluated with TCGA datasets. RESULTS: We identified 128 new potential biomarkers associated with recurrence, including 40 mRNAs, 39 miRNAs, and 59 genetic variants. Among differentially expressed genes, modulation of FN1, ITGα3, and MET had a significant impact on thyroid cancer cell migration. Similarly, ablation of miR-486 and miR-1179 significantly increased migration of TPC-1 and SW1736 cells. We further utilized genes with a validated functional role and identified a 5-gene risk score classifier as an independent predictor of thyroid cancer recurrence. CONCLUSION: Our newly proposed risk model based on combinatorial mRNA and microRNA expression has potential clinical utility as a prognostic indicator of recurrence. These findings should facilitate earlier prediction of recurrence with implications for improving patient outcome by tailoring treatment to disease risk and increasing posttreatment surveillance.


Subject(s)
MicroRNAs , Thyroid Neoplasms , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , RNA, Messenger/genetics , Risk Factors , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
4.
Cancer Res ; 80(1): 102-115, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31672844

ABSTRACT

The sodium iodide symporter (NIS) is required for iodide uptake, which facilitates thyroid hormone biosynthesis. NIS has been exploited for over 75 years in ablative radioiodine (RAI) treatment of thyroid cancer, where its ability to transport radioisotopes depends on its localization to the plasma membrane. The advent of NIS-based in vivo imaging and theranostic strategies in other malignancies and disease modalities has recently increased the clinical importance of NIS. However, NIS trafficking remains ill-defined. Here, we used tandem mass spectrometry followed by coimmunoprecipitation and proximity ligation assays to identify and validate two key nodes-ADP-ribosylation factor 4 (ARF4) and valosin-containing protein (VCP)-controlling NIS trafficking. Using cell-surface biotinylation assays and highly inclined and laminated optical sheet microscopy, we demonstrated that ARF4 enhanced NIS vesicular trafficking from the Golgi to the plasma membrane, whereas VCP-a principal component of endoplasmic reticulum (ER)-associated degradation-governed NIS proteolysis. Gene expression analysis indicated VCP expression was particularly induced in aggressive thyroid cancers and in patients who had poorer outcomes following RAI treatment. Two repurposed FDA-approved VCP inhibitors abrogated VCP-mediated repression of NIS function, resulting in significantly increased NIS at the cell-surface and markedly increased RAI uptake in mouse and human thyroid models. Collectively, these discoveries delineate NIS trafficking and highlight the new possibility of systemically enhancing RAI therapy in patients using FDA-approved drugs. SIGNIFICANCE: These findings show that ARF4 and VCP are involved in NIS trafficking to the plasma membrane and highlight the possible therapeutic role of VCP inhibitors in enhancing radioiodine effectiveness in radioiodine-refractory thyroid cancer.


Subject(s)
ADP-Ribosylation Factors/metabolism , Golgi Apparatus/metabolism , Iodine Radioisotopes/pharmacology , Symporters/metabolism , Thyroid Cancer, Papillary/therapy , Thyroid Neoplasms/therapy , Valosin Containing Protein/metabolism , Adult , Animals , Breast/pathology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Membrane/metabolism , Chemoradiotherapy/methods , Female , Gene Expression Profiling , Humans , Iodine Radioisotopes/therapeutic use , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Primary Cell Culture , Prognosis , Progression-Free Survival , Proteolysis , Thyroid Cancer, Papillary/mortality , Thyroid Cancer, Papillary/pathology , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/pathology , Thyroid Gland/radiation effects , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Tissue Distribution , Valosin Containing Protein/antagonists & inhibitors
5.
Thyroid ; 29(10): 1485-1498, 2019 10.
Article in English | MEDLINE | ID: mdl-31310151

ABSTRACT

Background: The ability of thyroid follicular epithelial cells to accumulate iodide via the sodium/iodide symporter (NIS) is exploited to successfully treat most thyroid cancers, although a subset of patients lose functional NIS activity and become unresponsive to radioiodide therapy, with poor clinical outcome. Our knowledge of NIS regulation remains limited, however. While numerous membrane proteins are functionally regulated via dimerization, there is little definitive evidence of NIS dimerization, and whether this might impact upon radioiodide uptake and treatment success is entirely unknown. We hypothesized that NIS dimerizes and that dimerization is a prerequisite for iodide uptake. Methods: Coimmunoprecipitation, proximity ligation, and Förster resonance energy transfer (FRET) assays were used to assess NIS:NIS interaction. To identify residues involved in dimerization, a homology model of NIS structure was built based on the crystal structure of the dimeric bacterial protein vSGLT. Results: Abundant cellular NIS dimerization was confirmed in vitro via three discrete methodologies. FRET and proximity ligation assays demonstrated that while NIS can exist as a dimer at the plasma membrane (PM), it is also apparent in other cellular compartments. Homology modeling revealed one key potential site of dimeric interaction, with six residues <3Å apart. In particular, NIS residues Y242, T243, and Q471 were identified as critical to dimerization. Individual mutation of residues Y242 and T243 rendered NIS nonfunctional, while abrogation of Q471 did not impact radioiodide uptake. FRET data show that the putative dimerization interface can tolerate the loss of one, but not two, of these three clustered residues. Conclusions: We show for the first time that NIS dimerizes in vitro, and we identify the key residues via which this happens. We hypothesize that dimerization of NIS is critical to its trafficking to the PM and may therefore represent a new mechanism that would need to be considered in overcoming therapeutic failure in patients with thyroid cancer.


Subject(s)
Iodine Radioisotopes/metabolism , Protein Multimerization , Symporters/metabolism , Thyroid Neoplasms/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Immunoprecipitation , In Vitro Techniques , Protein Conformation , Protein Structure, Quaternary , Symporters/ultrastructure , Thyroid Neoplasms/radiotherapy
6.
Cancer Res ; 78(20): 5863-5876, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30154144

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is the 6th most common cancer worldwide and poses a significant health burden due to its rising incidence. Although the proto-oncogene pituitary tumor-transforming gene 1 (PTTG) predicts poor patient outcome, its mechanisms of action are incompletely understood. We show here that the protein PBF modulates PTTG function, is overexpressed in HNSCC tumors, and correlates with significantly reduced survival. Lentiviral shRNA attenuation of PTTG or PBF expression in HNSCC cells with either wild-type or mutant p53, and with and without HPV infection, led to dysregulated expression of p53 target genes involved in DNA repair and apoptosis. Mechanistically, PTTG and PBF affected each other's interaction with p53 and cooperated to reduce p53 protein stability in HNSCC cells independently of HPV. Depletion of either PTTG or PBF significantly repressed cellular migration and invasion and impaired colony formation in HNSCC cells, implicating both proto-oncogenes in basic mechanisms of tumorigenesis. Patients with HNSCC with high tumoral PBF and PTTG had the poorest overall survival, which reflects a marked impairment of p53-dependent signaling.Significance: These findings reveal a complex and novel interrelationship between the expression and function of PTTG, PBF, and p53 in human HNSCC that significantly influences patient outcome. Cancer Res; 78(20); 5863-76. ©2018 AACR.


Subject(s)
Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/metabolism , Membrane Proteins/metabolism , Securin/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , DNA Repair , Female , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins , Kaplan-Meier Estimate , Lentivirus/metabolism , Male , Middle Aged , Mutation , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Papillomavirus Infections/complications , Proto-Oncogene Mas , RNA, Small Interfering/metabolism , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Tissue Array Analysis , Treatment Outcome
7.
J Clin Endocrinol Metab ; 101(12): 4551-4563, 2016 12.
Article in English | MEDLINE | ID: mdl-27603901

ABSTRACT

CONTEXT: Metastatic disease is responsible for the majority of endocrine cancer deaths. New therapeutic targets are urgently needed to improve patient survival rates. OBJECTIVE: The proto-oncogene PTTG1-binding factor (PBF/PTTG1IP) is overexpressed in multiple endocrine cancers and circumstantially associated with tumor aggressiveness. This study aimed to understand the role of PBF in tumor cell invasion and identify possible routes to inhibit its action. Design, Setting, Patients, and Interventions: Thyroid, breast, and colorectal cells were transfected with PBF and cultured for in vitro analysis. PBF and cortactin (CTTN) expression was determined in differentiated thyroid cancer and The Cancer Genome Atlas RNA-seq data. PRIMARY OUTCOME MEASURE: Pro-invasive effects of PBF were evaluated by 2D Boyden chamber, 3D organotypic, and proximity ligation assays. RESULTS: Our study identified that PBF and CTTN physically interact and co-localize, and that this occurs at the cell periphery, particularly at the leading edge of migrating cancer cells. Critically, PBF induces potent cellular invasion and migration in thyroid and breast cancer cells, which is entirely abrogated in the absence of CTTN. Importantly, we found that CTTN is over-expressed in differentiated thyroid cancer, particularly in patients with regional lymph node metastasis, which significantly correlates with elevated PBF expression. Mutation of PBF (Y174A) or pharmacological intervention modulates the PBF: CTTN interaction and attenuates the invasive properties of cancer cells. CONCLUSION: Our results demonstrate a unique role for PBF in regulating CTTN function to promote endocrine cell invasion and migration, as well as identify a new targetable interaction to block tumor cell movement.


Subject(s)
Breast Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Cortactin/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Neoplasm Invasiveness , Cell Line, Tumor , Female , Humans , Intracellular Signaling Peptides and Proteins , Proto-Oncogene Mas , Thyroid Neoplasms/metabolism
8.
Hum Mol Genet ; 25(9): 1836-45, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26945007

ABSTRACT

Familial medullary thyroid cancer (MTC) and its precursor, C cell hyperplasia (CCH), is associated with germline RET mutations causing multiple endocrine neoplasia type 2. However, some rare families with apparent MTC/CCH predisposition do not have a detectable RET mutation. To identify novel MTC/CCH predisposition genes we undertook exome resequencing studies in a family with apparent predisposition to MTC/CCH and no identifiable RET mutation. We identified a novel ESR2 frameshift mutation, c.948delT, which segregated with histological diagnosis following thyroid surgery in family members and demonstrated loss of ESR2-encoded ERß expression in the MTC tumour. ERα and ERß form heterodimers binding DNA at specific oestrogen-responsive elements (EREs) to regulate gene transcription. ERß represses ERα-mediated activation of the ERE and the RET promoter contains three EREs. In vitro, we showed that ESR2 c.948delT results in unopposed ERα mediated increased cellular proliferation, activation of the ERE and increased RET expression. In vivo, immunostaining of CCH and MTC using an anti-RET antibody demonstrated increased RET expression. Together these findings identify germline ESR2 mutation as a novel cause of familial MTC/CCH and provide important insights into a novel mechanism causing increased RET expression in tumourigenesis.


Subject(s)
Carcinoma, Medullary/congenital , Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic , Germ-Line Mutation/genetics , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Adult , Carcinoma, Medullary/genetics , Carcinoma, Medullary/metabolism , Carcinoma, Medullary/pathology , Cell Proliferation , Disease Susceptibility , Genotype , Humans , Male , Multiple Endocrine Neoplasia Type 2a/pathology , Pedigree , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/pathology , Tumor Cells, Cultured , Up-Regulation , Young Adult
9.
Mol Carcinog ; 55(1): 15-26, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25408419

ABSTRACT

The PTTG1-binding factor (PBF) is a transforming gene capable of eliciting tumor formation in xenograft models. However, the precise role of PBF in tumorigenesis and its prognostic value as a cancer biomarker remain largely uncharacterised, particularly in malignancies outside the thyroid. Here, we provide the first evidence that PBF represents a promising prognostic marker in colorectal cancer. Examination of a total of 39 patients demonstrated higher PBF expression at both the mRNA (P = 0.009) and protein (P < 0.0001) level in colorectal tumors compared to matched normal tissue. Critically, PBF was most abundant in colorectal tumors associated with Extramural Vascular Invasion (EMVI), increased genetic instability (GI) and somatic TP53 mutations, all features linked with recurrence and poorer patient survival. We further demonstrate by glutathione-S-transferase (GST) pull-down and coimmunoprecipitation that PBF binds to the tumor suppressor protein p53, as well as to p53 mutants (Δ126-132, M133K, V197E, G245D, I255F and R273C) identified in the colorectal tumors. Importantly, overexpression of PBF in colorectal HCT116 cells interfered with the transcriptional activity of p53-responsive genes such as mdm2, p21 and sfn. Diminished p53 stability (> 90%; P < 0.01) was also evident with a concurrent increase in ubiquitinated p53. Human colorectal tumors with wild-type TP53 and high PBF expression also had low p53 protein levels (P < 0.05), further emphasizing a putative interaction between these genes in vivo. Overall, these results demonstrate an emerging role for PBF in colorectal tumorigenesis through regulating p53 activity, with implications for PBF as a prognostic indicator for invasive tumors.


Subject(s)
Colorectal Neoplasms/metabolism , Membrane Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Neoplasm Invasiveness , Prognosis , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Mas , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Stem Cell Assay , Ubiquitination
10.
Cancer Discov ; 5(11): 1178-93, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26359368

ABSTRACT

UNLABELLED: Ch22q LOH is preferentially associated with RAS mutations in papillary and in poorly differentiated thyroid cancer (PDTC). The 22q tumor suppressor NF2, encoding merlin, is implicated in this interaction because of its frequent loss of function in human thyroid cancer cell lines. Nf2 deletion or Hras mutation is insufficient for transformation, whereas their combined disruption leads to murine PDTC with increased MAPK signaling. Merlin loss induces RAS signaling in part through inactivation of Hippo, which activates a YAP-TEAD transcriptional program. We find that the three RAS genes are themselves YAP-TEAD1 transcriptional targets, providing a novel mechanism of promotion of RAS-induced tumorigenesis. Moreover, pharmacologic disruption of YAP-TEAD with verteporfin blocks RAS transcription and signaling and inhibits cell growth. The increased MAPK output generated by NF2 loss in RAS-mutant cancers may inform therapeutic strategies, as it generates greater dependency on the MAPK pathway for viability. SIGNIFICANCE: Intensification of mutant RAS signaling through copy-number imbalances is commonly associated with transformation. We show that NF2/merlin inactivation augments mutant RAS signaling by promoting YAP/TEAD-driven transcription of oncogenic and wild-type RAS, resulting in greater MAPK output and increased sensitivity to MEK inhibitors.


Subject(s)
Gene Deletion , Genes, ras , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurofibromin 2/genetics , Nuclear Proteins/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Cell Cycle Proteins , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromosome Deletion , Chromosomes, Human, Pair 22 , DNA Copy Number Variations , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Order , Gene Targeting , Humans , Mice , Mice, Transgenic , Models, Biological , Neoplasm Staging , Nucleotide Motifs , Position-Specific Scoring Matrices , Promoter Regions, Genetic , Protein Binding , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Transcriptional Activation
11.
Endocrinology ; 155(4): 1222-34, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24506068

ABSTRACT

The PTTG1-binding factor (PBF/PTTG1IP) has an emerging repertoire of roles, especially in thyroid biology, and functions as a protooncogene. High PBF expression is independently associated with poor prognosis and lower disease-specific survival in human thyroid cancer. However, the precise role of PBF in thyroid tumorigenesis is unclear. Here, we present extensive evidence demonstrating that PBF is a novel regulator of p53, a tumor suppressor protein with a key role in maintaining genetic stability, which is infrequently mutated in differentiated thyroid cancer. By coimmunoprecipitation and proximity-ligation assays, we show that PBF binds specifically to p53 in thyroid cells and significantly represses transactivation of responsive promoters. Further, we identify that PBF decreases p53 stability by enhancing ubiquitination, which appears dependent on the E3 ligase activity of Mdm2. Impaired p53 function was evident in a transgenic mouse model with thyroid-specific PBF overexpression (transgenic PBF mice), which had significantly increased genetic instability as indicated by fluorescent inter simple sequence repeat-PCR analysis. Consistent with this, approximately 40% of all DNA repair genes examined were repressed in transgenic PBF primary cultures, including genes with critical roles in maintaining genomic integrity such as Mgmt, Rad51, and Xrcc3. Our data also revealed that PBF induction resulted in up-regulation of the E2 enzyme Rad6 in murine thyrocytes and was associated with Rad6 expression in human thyroid tumors. Overall, this work provides novel insights into the role of the protooncogene PBF as a negative regulator of p53 function in thyroid tumorigenesis, in which PBF is generally overexpressed and p53 mutations are rare compared with other tumor types.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Thyroid Gland/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Cell Transformation, Neoplastic/genetics , Cells, Cultured , DNA Repair , Female , Genes, Reporter , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Transgenic , Protein Binding , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Ubiquitin/chemistry
12.
Endocrinology ; 154(11): 4408-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23867215

ABSTRACT

Human pituitary tumor transforming gene (hPTTG) is a multifunctional proto-oncogene implicated in the initiation and progression of several tumors. Phosphorylation of hPTTG is mediated by cyclin-dependent kinase 2 (CDC2), whereas cellular expression is regulated by specificity protein 1 (SP1). The mechanisms underlying hPTTG propagation of aberrant thyroid cell growth have not been fully defined. We set out to investigate the interplay between hPTTG and growth factors, as well as the effects of phosphorylation and SP1 regulation on hPTTG expression and function. In our study, epidermal growth factor (EGF), TGFα, and IGF-1 induced hPTTG expression and phosphorylation in thyroid cells, which was associated with activation of MAPK and phosphoinositide 3-kinase. Growth factors induced hPTTG independently of CDC2 and SP1 in thyroid carcinoma cells. Strikingly, CDC2 depletion in TPC-1 cells resulted in enhanced expression and phosphorylation of hPTTG and reduced cellular proliferation. In reciprocal experiments, hPTTG overexpression induced EGF, IGF-1, and TGFα mRNAs in primary human thyrocytes. Treatment of primary human thyrocytes with conditioned media derived from hPTTG-transfected cells resulted in autocrine upregulation of hPTTG protein, which was ameliorated by growth factor depletion or growth factor receptor tyrosine kinase inhibitors. A transgenic murine model of thyroid targeted hPTTG overexpression (hPTTG-Tg) (FVB/N strain, both sexes) demonstrated smaller thyroids with reduced cellular proliferation and enhanced secretion of Egf. In contrast, Pttg(-/-) knockout mice (c57BL6 strain, both sexes) showed reduced thyroidal Egf mRNA expression. These results define hPTTG as having a central role in thyroid autocrine signaling mechanisms via growth factors, with profound implications for promotion of transformed cell growth.


Subject(s)
Securin/metabolism , Thyroid Gland/cytology , Animals , Autocrine Communication , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Line , Cell Proliferation , Cricetinae , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Gene Expression Regulation/physiology , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Mice , Mice, Transgenic , Paracrine Communication , Phosphorylation , Proto-Oncogene Mas , Securin/genetics
13.
Cancer Res ; 71(19): 6153-64, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21844185

ABSTRACT

Pituitary tumor transforming gene (PTTG)-binding factor (PBF or PTTG1IP) is a little characterized proto-oncogene that has been implicated in the etiology of breast and thyroid tumors. In this study, we created a murine transgenic model to target PBF expression to the thyroid gland (PBF-Tg mice) and found that these mice exhibited normal thyroid function, but a striking enlargement of the thyroid gland associated with hyperplastic and macrofollicular lesions. Expression of the sodium iodide symporter (NIS), a gene essential to the radioiodine ablation of thyroid hyperplasia, neoplasia, and metastasis, was also potently inhibited in PBF-Tg mice. Critically, iodide uptake was repressed in primary thyroid cultures from PBF-Tg mice, which could be rescued by PBF depletion. PBF-Tg thyroids exhibited upregulation of Akt and the TSH receptor (TSHR), each known regulators of thyrocyte proliferation, along with upregulation of the downstream proliferative marker cyclin D1. We extended and confirmed findings from the mouse model by examining PBF expression in human multinodular goiters (MNG), a hyperproliferative thyroid disorder, where PBF and TSHR was strongly upregulated relative to normal thyroid tissue. Furthermore, we showed that depleting PBF in human primary thyrocytes was sufficient to increase radioiodine uptake. Together, our findings indicate that overexpression of PBF causes thyroid cell proliferation, macrofollicular lesions, and hyperplasia, as well as repression of the critical therapeutic route for radioiodide uptake.


Subject(s)
Membrane Proteins/metabolism , Symporters/metabolism , Thyroid Gland , Animals , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Regulation , Goiter, Nodular/metabolism , Goiter, Nodular/pathology , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Intracellular Signaling Peptides and Proteins , Iodine/metabolism , Iodine Radioisotopes , Membrane Proteins/genetics , Mice , Mice, Transgenic , Proto-Oncogene Mas , Symporters/genetics , Thyroid Gland/metabolism , Thyroid Gland/pathology
14.
J Endocrinol ; 210(2): 157-63, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21450804

ABSTRACT

Pituitary tumor-transforming gene (PTTG)-binding factor (PBF; PTTG1IP) was initially identified through its interaction with the human securin, PTTG. Like PTTG, PBF is upregulated in multiple endocrine tumours including thyroid cancer. PBF is believed to induce the translocation of PTTG into the cell nucleus where it can drive tumourigenesis via a number of different mechanisms. However, an independent transforming ability has been demonstrated both in vitro and in vivo, suggesting that PBF is itself a proto-oncogene. Studied in only a limited number of publications to date, PBF is emerging as a protein with a growing repertoire of roles. Recent data suggest that PBF possesses a complex multifunctionality in an increasing number of tumour settings. For example, PBF is upregulated by oestrogen and mediates oestrogen-stimulated cell invasion in breast cancer cells. In addition to a possible role in the induction of thyroid tumourigenesis, PBF overexpression in thyroid cancers inhibits iodide uptake. PBF has been shown to repress sodium iodide symporter (NIS) activity by transcriptional regulation of NIS expression through the human NIS upstream enhancer and further inhibits iodide uptake via a post-translational mechanism of NIS governing subcellular localisation. This review discusses the current data describing PBF expression and function in thyroid cancer and highlights PBF as a novel target for improving radioiodine uptake and thus prognosis in thyroid cancer.


Subject(s)
Neoplasm Proteins/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/radiotherapy , Humans , Neoplasm Proteins/genetics , Prognosis , Proto-Oncogene Mas , Securin , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism
15.
Expert Rev Mol Med ; 12: e38, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21129230

ABSTRACT

The pituitary tumor-transforming gene (PTTG1) encodes a multifunctional protein (PTTG) that is overexpressed in numerous tumours, including pituitary, thyroid, breast and ovarian carcinomas. PTTG induces cellular transformation in vitro and tumourigenesis in vivo, and several mechanisms by which PTTG contributes to tumourigenesis have been investigated. Also known as the human securin, PTTG is involved in cell cycle regulation, controlling the segregation of sister chromatids during mitosis. This review outlines current information regarding PTTG structure, expression, regulation and function in the pathogenesis of neoplasia. Recent progress concerning the use of PTTG as a prognostic marker or therapeutic target will be considered. In addition, the PTTG binding factor (PBF), identified through its interaction with PTTG, has also been established as a proto-oncogene that is upregulated in several cancers. Current knowledge regarding PBF is outlined and its role both independently and alongside PTTG in endocrine and related cancers is discussed.


Subject(s)
Endocrine Gland Neoplasms/genetics , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Amino Acid Sequence , Animals , Cell Line , Endocrine Gland Neoplasms/metabolism , Female , Gene Expression Regulation , Genomic Instability , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Proto-Oncogene Mas , Rats , Securin , Tumor Suppressor Protein p53/metabolism
16.
Cancer Res ; 70(9): 3739-49, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20406982

ABSTRACT

Pituitary tumor transforming gene (PTTG) binding factor (PBF; PTTG1IP) is a relatively uncharacterized oncoprotein whose function remains obscure. Because of the presence of putative estrogen response elements (ERE) in its promoter, we assessed PBF regulation by estrogen. PBF mRNA and protein expression were induced by both diethylstilbestrol and 17beta-estradiol in estrogen receptor alpha (ERalpha)-positive MCF-7 cells. Detailed analysis of the PBF promoter showed that the region -399 to -291 relative to the translational start site contains variable repeats of an 18-bp sequence housing a putative ERE half-site (gcccctcGGTCAcgcctc). Sequencing the PBF promoter from 122 normal subjects revealed that subjects may be homozygous or heterozygous for between 1 and 6 repeats of the ERE. Chromatin immunoprecipitation and oligonucleotide pull-down assays revealed ERalpha binding to the PBF promoter. PBF expression was low or absent in normal breast tissue but was highly expressed in breast cancers. Subjects with greater numbers of ERE repeats showed higher PBF mRNA expression, and PBF protein expression positively correlated with ERalpha status. Cell invasion assays revealed that PBF induces invasion through Matrigel, an action that could be abrogated both by siRNA treatment and specific mutation. Furthermore, PBF is a secreted protein, and loss of secretion prevents PBF inducing cell invasion. Given that PBF is a potent transforming gene, we propose that estrogen treatment in postmenopausal women may upregulate PBF expression, leading to PBF secretion and increased cell invasion. Furthermore, the number of ERE half-sites in the PBF promoter may significantly alter the response to estrogen treatment in individual subjects.


Subject(s)
Breast Neoplasms/genetics , Neoplasm Proteins/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Diethylstilbestrol/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/biosynthesis , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Response Elements , Securin
17.
J Cell Sci ; 122(Pt 18): 3393-402, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19706688

ABSTRACT

Differentiated thyroid cancers and their metastases frequently exhibit reduced iodide uptake, impacting on the efficacy of radioiodine ablation therapy. PTTG binding factor (PBF) is a proto-oncogene implicated in the pathogenesis of thyroid cancer. We recently reported that PBF inhibits iodide uptake, and have now elucidated a mechanism by which PBF directly modulates sodium iodide symporter (NIS) activity in vitro. In subcellular localisation studies, PBF overexpression resulted in the redistribution of NIS from the plasma membrane into intracellular vesicles, where it colocalised with the tetraspanin CD63. Cell-surface biotinylation assays confirmed a reduction in plasma membrane NIS expression following PBF transfection compared with vector-only treatment. Coimmunoprecipitation and GST-pull-down experiments demonstrated a direct interaction between NIS and PBF, the functional consequence of which was assessed using iodide-uptake studies in rat thyroid FRTL-5 cells. PBF repressed iodide uptake, whereas three deletion mutants, which did not localise within intracellular vesicles, lost the ability to inhibit NIS activity. In summary, we present an entirely novel mechanism by which the proto-oncogene PBF binds NIS and alters its subcellular localisation, thereby regulating its ability to uptake iodide. Given that PBF is overexpressed in thyroid cancer, these findings have profound implications for thyroid cancer ablation using radioiodine.


Subject(s)
Cell Differentiation , Repressor Proteins/metabolism , Symporters/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Animals , Antigens, CD/metabolism , Caveolins/metabolism , Cell Line , Gene Deletion , Humans , Intracellular Signaling Peptides and Proteins , Iodides/metabolism , Membrane Proteins/metabolism , Platelet Membrane Glycoproteins/metabolism , Protein Binding , Protein Transport , Proto-Oncogene Mas , Rats , Subcellular Fractions/metabolism , Tetraspanin 30
18.
Diabetes ; 56(12): 3020-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17804763

ABSTRACT

OBJECTIVE: To identify the type 2 diabetes gene located at chromosome 18p11. RESEARCH DESIGN AND METHODS: We investigated the region in a young genetically isolated population by genotyping 34 single nucleotide polymorphisms (SNPs) in 78 case subjects and 101 control subjects. Two SNPs were selected and followed up in two cohorts. The first cohort came from a general Dutch population. In this cohort, association with type 2 diabetes was investigated using 616 type 2 diabetic case subjects and 2,890 control subjects; association with oral glucose tolerance test data was performed in 361 normoglycemic people. Association with fat distribution was studied in the second replication cohort, consisting of 836 people from the genetically isolated population. RESULTS: At the initial step, we found that the common C allele of SNP rs3745012 was associated with type 2 diabetes (odds ratio 2.01, P = 0.03). This SNP is located at the 3' untranslated region of the LPIN2 gene, which is a plausible candidate for type 2 diabetes and obesity. In the cohort from the general Dutch population, we demonstrated that rs3745012 interacts with BMI in determination of type 2 diabetes: whereas in subjects with high BMI, the common C allele is associated with type 2 diabetes, the same allele exhibits a neutral or protective effect in lean subjects (P = 0.05 overall effect, P = 0.02 interaction). Most remarkably, rs3745012 strongly affected composite insulin sensitivity index (P = 0.006 for overall effect, P = 0.004 for interaction). In the second replication cohort, we found that the allele C of rs3745012 increases trunk-to-legs fat mass ratio (P = 0.001) and may affect other fat-related measurements. CONCLUSIONS: rs3745012 SNP of the LPIN2 gene is associated with type 2 diabetes and fat distribution.


Subject(s)
Adipose Tissue/anatomy & histology , Blood Glucose/metabolism , Body Composition/genetics , Diabetes Mellitus, Type 2/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Body Mass Index , Chromosome Mapping , Chromosomes, Human, Pair 18 , Diabetes Mellitus, Type 2/physiopathology , Female , Genetic Predisposition to Disease , Genotype , Humans , Linkage Disequilibrium , Male , Reference Values
19.
J Infect Dis ; 194(12): 1729-36, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17109346

ABSTRACT

The G1P1A[8] rotavirus vaccine candidate 89-12, the precursor to Rotarix, stimulated high titers of neutralizing antibodies to non-G1/P1A[8] serotypes of human rotavirus in naturally infected subjects before attenuation by cell-culture passages. These responses were greatly diminished in young infants (median age, 11 weeks) administered the attenuated vaccine. Because of the possibility of improved responses in older infants, the immunogenicity of the 89-12 vaccine candidate was evaluated after administration of 2 doses beginning at either 4 or 6 months of age. As was found in young infants, neutralizing antibody responses to non-G1/P1A[8] rotaviruses were considerably lower than those observed after natural infection. The reasons identified were overall (P<.0001) lower neutralizing antibody responses stimulated by the attenuated 89-12 strain, compared with those stimulated by its virulent precursor, and 5 mutations selected in the gene encoding the immunodominant VP4 (P) neutralization protein. Even so, the Rotarix vaccine developed from attenuated 89-12 was found to elicit excellent protection against non-G1 rotaviruses.


Subject(s)
Antibodies, Viral/immunology , Rotavirus Infections/immunology , Rotavirus Vaccines/immunology , Rotavirus/immunology , Vaccination , Administration, Oral , Antibodies, Viral/blood , Antigens, Viral/genetics , Capsid Proteins/genetics , Humans , Immunization Schedule , Infant , Mutation , Neutralization Tests , Rotavirus Vaccines/administration & dosage , Rotavirus Vaccines/genetics , Serial Passage , United States
20.
J Infect Dis ; 189(12): 2290-3, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15181577

ABSTRACT

A quadrivalent precursor to the pentavalent rotavirus vaccine candidate RotaTeq was evaluated in a 3-dose, 439-subject study. To determine immunogenicity, the quantity of rotavirus immunoglobulin A (IgA) in stool specimens obtained, at 1 of 10 study sites, from 37 placebo and 37 vaccine recipients was measured. None of the placebo recipients showed a clinically important (>/=3-fold) increase in stool rotavirus IgA, whereas 31 vaccine recipients showed an increase after at least 1 dose of vaccine. In total, 16, 19, and 15 vaccine recipients had increases after 1, 2, and 3 doses, respectively, indicating that a 3-dose regimen increased the immune response elicited by this vaccine.


Subject(s)
Feces/chemistry , Immunoglobulin A/analysis , Reassortant Viruses/immunology , Rotavirus Vaccines/administration & dosage , Rotavirus/immunology , Vaccines, Attenuated/administration & dosage , Animals , Antibodies, Viral/analysis , Antibodies, Viral/blood , Cattle , Dose-Response Relationship, Immunologic , Humans , Immunoglobulin A/blood , Infant , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Vaccines/immunology , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...