Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Virol ; 93(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30996091

ABSTRACT

Here, we show that the cellular DNA replication protein and ATR substrate SMARCAL1 is recruited to viral replication centers early during adenovirus infection and is then targeted in an E1B-55K/E4orf6- and cullin RING ligase-dependent manner for proteasomal degradation. In this regard, we have determined that SMARCAL1 is phosphorylated at S123, S129, and S173 early during infection in an ATR- and CDK-dependent manner, and that pharmacological inhibition of ATR and CDK activities attenuates SMARCAL1 degradation. SMARCAL1 recruitment to viral replication centers was shown to be largely dependent upon SMARCAL1 association with the RPA complex, while Ad-induced SMARCAL1 phosphorylation also contributed to SMARCAL1 recruitment to viral replication centers, albeit to a limited extent. SMARCAL1 was found associated with E1B-55K in adenovirus E1-transformed cells. Consistent with its ability to target SMARCAL1, we determined that E1B-55K modulates cellular DNA replication. As such, E1B-55K expression initially enhances cellular DNA replication fork speed but ultimately leads to increased replication fork stalling and the attenuation of cellular DNA replication. Therefore, we propose that adenovirus targets SMARCAL1 for degradation during infection to inhibit cellular DNA replication and promote viral replication.IMPORTANCE Viruses have evolved to inhibit cellular DNA damage response pathways that possess antiviral activities and utilize DNA damage response pathways that possess proviral activities. Adenovirus has evolved, primarily, to inhibit DNA damage response pathways by engaging with the ubiquitin-proteasome system and promoting the degradation of key cellular proteins. Adenovirus differentially regulates ATR DNA damage response signaling pathways during infection. The cellular adenovirus E1B-55K binding protein E1B-AP5 participates in ATR signaling pathways activated during infection, while adenovirus 12 E4orf6 negates Chk1 activation by promoting the proteasome-dependent degradation of the ATR activator TOPBP1. The studies detailed here indicate that adenovirus utilizes ATR kinase and CDKs during infection to promote the degradation of SMARCAL1 to attenuate normal cellular DNA replication. These studies further our understanding of the relationship between adenovirus and DNA damage and cell cycle signaling pathways during infection and establish new roles for E1B-55K in the modulation of cellular DNA replication.


Subject(s)
Adenoviridae Infections/metabolism , Adenovirus E1B Proteins/metabolism , Adenoviruses, Human/physiology , DNA Helicases/metabolism , DNA Replication , Virus Replication , A549 Cells , Adenoviridae Infections/virology , Ataxia Telangiectasia Mutated Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin/metabolism
2.
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
3.
Value Health ; 26(4): 614-616, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36764516
4.
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
5.
Nature ; 465(7299): 813-7, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20535211

ABSTRACT

Down's syndrome (DS) is a genetic disorder caused by full or partial trisomy of human chromosome 21 and presents with many clinical phenotypes including a reduced incidence of solid tumours. Recent work with the Ts65Dn model of DS, which has orthologues of about 50% of the genes on chromosome 21 (Hsa21), has indicated that three copies of the ETS2 (ref. 3) or DS candidate region 1 (DSCR1) genes (a previously known suppressor of angiogenesis) is sufficient to inhibit tumour growth. Here we use the Tc1 transchromosomic mouse model of DS to dissect the contribution of extra copies of genes on Hsa21 to tumour angiogenesis. This mouse expresses roughly 81% of Hsa21 genes but not the human DSCR1 region. We transplanted B16F0 and Lewis lung carcinoma tumour cells into Tc1 mice and showed that growth of these tumours was substantially reduced compared with wild-type littermate controls. Furthermore, tumour angiogenesis was significantly repressed in Tc1 mice. In particular, in vitro and in vivo angiogenic responses to vascular endothelial growth factor (VEGF) were inhibited. Examination of the genes on the segment of Hsa21 in Tc1 mice identified putative anti-angiogenic genes (ADAMTS1and ERG) and novel endothelial cell-specific genes, never previously shown to be involved in angiogenesis (JAM-B and PTTG1IP), that, when overexpressed, are responsible for inhibiting angiogenic responses to VEGF. Three copies of these genes within the stromal compartment reduced tumour angiogenesis, explaining the reduced tumour growth in DS. Furthermore, we expect that, in addition to the candidate genes that we show to be involved in the repression of angiogenesis, the Tc1 mouse model of DS will permit the identification of other endothelium-specific anti-angiogenic targets relevant to a broad spectrum of cancer patients.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Disease Models, Animal , Down Syndrome/genetics , Gene Dosage/genetics , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS1 Protein , Animals , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chromosomes, Mammalian/genetics , Down Syndrome/complications , Down Syndrome/physiopathology , Female , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Intracellular Signaling Peptides and Proteins , Male , Melanoma, Experimental/complications , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Transcription Factors , Transcriptional Regulator ERG , Trisomy/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
bioRxiv ; 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38168185

ABSTRACT

The current study in prostate cancer (PCa) focused on the genomic mechanisms at the cross-roads of pro-differentiation signals and the emergence of lineage plasticity. We explored an understudied cistromic mechanism involving RARγ's ability to govern AR cistrome-transcriptome relationships, including those associated with more aggressive PCa features. The RARγ complex in PCa cell models was enriched for canonical cofactors, as well as proteins involved in RNA processing and bookmarking. Identifying the repertoire of miR-96 bound and regulated gene targets, including those recognition elements marked by m6A, revealed their significant enrichment in the RARγ complex. RARγ significantly enhanced the AR cistrome, particularly in active enhancers and super-enhancers, and overlapped with the binding of bookmarking factors. Furthermore, RARγ expression led to nucleosome-free chromatin enriched with H3K27ac, and significantly enhanced the AR cistrome in G2/M cells. RARγ functions also antagonized the transcriptional actions of the lineage master regulator ONECUT2. Similarly, gene programs regulated by either miR-96 or antagonized by RARγ were enriched in alternative lineages and more aggressive PCa phenotypes. Together these findings reveal an under-investigated role for RARγ, modulated by miR-96, to bookmark enhancer sites during mitosis. These sites are required by the AR to promote transcriptional competence, and emphasize luminal differentiation, while antagonizing ONECUT2.

7.
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
8.
Carcinogenesis ; 34(2): 248-56, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23087083

ABSTRACT

The current study investigated transcriptional distortion in prostate cancer cells using the vitamin D receptor (VDR) as a tool to examine how epigenetic events driven by corepressor binding and CpG methylation lead to aberrant gene expression. These relationships were investigated in the non-malignant RWPE-1 cells that were 1α,25(OH)(2)D(3) responsive (RWPE-1) and malignant cell lines that were 1α,25(OH)(2)D(3) partially responsive (RWPE-2) and resistant (PC-3). These studies revealed that selective attenuation and repression of VDR transcriptional responses in the cancer cell lines reflected their loss of antiproliferative sensitivity. This was evident in VDR target genes including VDR, CDKN1A (encodes p21( (waf1/cip1) )) and GADD45A; NCOR1 knockdown alleviated this malignant transrepression. ChIP assays in RWPE-1 and PC-3 cells revealed that transrepression of CDKN1A was associated with increased NCOR1 enrichment in response to 1α,25(OH)(2)D(3) treatment. These findings supported the concept that retained and increased NCOR1 binding, associated with loss of H3K9ac and increased H3K9me2, may act as a beacon for the initiation and recruitment of DNA methylation. Overexpressed histone methyltransferases (KMTs) were detectable in a wide panel of prostate cancer cell lines compared with RWPE-1 and suggested that generation of H3K9me2 states would be favored. Cotreatment of cells with the KMT inhibitor, chaetocin, increased 1α,25(OH)(2)D(3)-mediated induction of CDKN1A expression supporting a role for this event to disrupt CDKN1A regulation. Parallel surveys in PC-3 cells of CpG methylation around the VDR binding regions on CDKN1A revealed altered basal and VDR-regulated DNA methylation patterns that overlapped with VDR-induced recruitment of NCOR1 and gene transrepression. Taken together, these findings suggest that sustained corepressor interactions with nuclear-resident transcription factors may inappropriately transform transient-repressive histone states into more stable and repressive DNA methylation events.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Methylation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Nuclear Receptor Co-Repressor 1/metabolism , Prostatic Neoplasms/genetics , Receptors, Calcitriol/metabolism , Apoptosis/drug effects , Bone Density Conservation Agents/pharmacology , Calcitriol/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Epigenesis, Genetic , Humans , Male , Nuclear Receptor Co-Repressor 1/genetics , Promoter Regions, Genetic/genetics , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
9.
J Virol ; 86(6): 3167-79, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22205733

ABSTRACT

The ability of adenovirus early region proteins, E1B-55K and E4orf6, to usurp control of cellular ubiquitin ligases and target proteins for proteasome-dependent degradation during infection is well established. Here we show that the E4 gene product, E4orf3 can, independently of E1B-55K and E4orf6, target the transcriptional corepressor transcriptional intermediary factor 1γ (TIF1γ) for proteasome-mediated degradation during infection. Initial mass spectrometric studies identified TIF1 family members-TIF1α, TIF1ß, and TIF1γ-as E1B-55K-binding proteins in both transformed and infected cells, but analyses revealed that, akin to TIF1α, TIF1γ is reorganized in an E4orf3-dependent manner to promyelocytic leukemia protein-containing nuclear tracks during infection. The use of a number of different adenovirus early region mutants identified the specific and sole requirement for E4orf3 in mediating TIF1γ degradation. Further analyses revealed that TIF1γ is targeted for degradation by a number of divergent human adenoviruses, suggesting that the ability of E4orf3 to regulate TIF1γ expression is evolutionarily conserved. We also determined that E4orf3 does not utilize the Cullin-based ubiquitin ligases, CRL2 and CRL5, or the TIF1α ubiquitin ligase in order to promote TIF1γ degradation. Further studies suggested that TIF1γ possesses antiviral activity and limits adenovirus early and late gene product expression during infection. Indeed, TIF1γ knockdown accelerates the adenovirus-mediated degradation of MRE11, while TIF1γ overexpression delays the adenovirus-mediated degradation of MRE11. Taken together, these studies have identified novel adenovirus targets and have established a new role for the E4orf3 protein during infection.


Subject(s)
Adenoviridae Infections/metabolism , Adenovirus E4 Proteins/metabolism , Adenoviruses, Human/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Adenoviridae Infections/genetics , Adenoviridae Infections/virology , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Adenovirus E4 Proteins/genetics , Adenoviruses, Human/genetics , Cell Line , Humans , Protein Binding , Transcription Factors/genetics
10.
Nucleic Acids Res ; 39(6): 2045-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21088000

ABSTRACT

In non-malignant RWPE-1 prostate epithelial cells signaling by the nuclear receptor Vitamin D Receptor (VDR, NR1I1) induces cell cycle arrest through targets including CDKN1A (encodes p21((waf1/cip1))). VDR dynamically induced individual histone modification patterns at three VDR binding sites (R1, 2, 3) on the CDKN1A promoter. The magnitude of these modifications was specific to each phase of the cell cycle. For example, H3K9ac enrichment occurred rapidly only at R2, whereas parallel accumulation of H3K27me3 occurred at R1; these events were significantly enriched in G(1) and S phase cells, respectively. The epigenetic events appeared to allow VDR actions to combine with p53 to enhance p21((waf1/cip1)) activation further. In parallel, VDR binding to the MCM7 gene induced H3K9ac enrichment associated with rapid mRNA up-regulation to generate miR-106b and consequently regulate p21((waf1/cip1)) expression. We conclude that VDR binding site- and promoter-specific patterns of histone modifications combine with miRNA co-regulation to form a VDR-regulated feed-forward loop to control p21((waf1/cip1)) expression and cell cycle arrest. Dissection of this feed-forward loop in a non-malignant prostate cell system illuminates mechanisms of sensitivity and therefore possible resistance in prostate and other VDR responsive cancers.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Epigenesis, Genetic , Prostate/metabolism , Receptors, Calcitriol/metabolism , Animals , Calcitriol/pharmacology , Cell Cycle/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Feedback, Physiological , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Promoter Regions, Genetic , Prostate/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
11.
Sci Rep ; 12(1): 7706, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35562181

ABSTRACT

Thyroid cancer is the most common primary endocrine malignancy in adults and its incidence is rapidly increasing. Long non-coding RNAs (lncRNAs), generally defined as RNA molecules longer than 200 nucleotides with no protein-encoding capacity, are highly tissue-specific molecules that serve important roles in gene regulation through a variety of different mechanisms, including acting as competing endogenous RNAs (ceRNAs) that 'sponge' microRNAs (miRNAs). In the present study, using an integrated approach through RNA-sequencing of paired thyroid tumor and non-tumor samples, we have identified an interactome network between lncRNAs and miRNAs and examined the functional consequences in vitro and in vivo of one of such interactions. We have identified a likely operative post-transcriptional regulatory network in which the downregulated lncRNA, SPTY2D1-AS1, is predicted to target the most abundant and upregulated miRNAs in thyroid cancer, particularly miR-221, a well-known oncomiRNA in cancer. Indeed, SPTY2D1-AS1 functions as a potent tumor suppressor in vitro and in vivo, it is downregulated in the most advanced stages of human thyroid cancer, and it seems to block the processing of the primary form of miR-221. Overall, our results link SPTY2D1-AS1 to thyroid cancer progression and highlight the potential use of this lncRNA as a therapeutic target of thyroid cancer.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Thyroid Neoplasms , Adult , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Thyroid Neoplasms/genetics
12.
Front Endocrinol (Lausanne) ; 13: 1061555, 2022.
Article in English | MEDLINE | ID: mdl-36686426

ABSTRACT

Introduction: Radioactive iodine (RAI) therapy is a critical component in the post-surgical management of thyroid cancer patients, as well as being a central therapeutic option in the treatment of hyperthyroidism. Previous work suggests that antithyroid drugs hinder the efficacy of RAI therapy in patients. However, the effects of other background medications on RAI treatment efficacy have not been evaluated. Therefore, we performed a systematic review and meta-analysis investigating the potential off-target effects of medication on RAI therapy in patients with thyroid cancer and hyperthyroidism. Methods: Systematic review and meta-analysis according to the 2020 PRISMA guidelines. Databases searched: MEDLINE, EMBASE and Cochrane Library for studies published between 2001 and 2021. Results: Sixty-nine unique studies were identified. After screening, 17 studies with 3313 participants were included. One study investigated thyroid cancer, with the rest targeted to hyperthyroidism. The majority of studies evaluated the effects of antithyroid drugs; the other drugs studied included lithium, prednisone and glycididazole sodium. Antithyroid drugs were associated with negative impacts on post-RAI outcomes (n = 5 studies, RR = 0.81, p = 0.02). However, meta-analysis found moderate heterogeneity between studies (I2 = 51%, τ2 = 0.0199, p = 0.08). Interestingly, lithium (n = 3 studies), prednisone (n = 1 study) and glycididazole (n = 1 study) appeared to have positive impacts on post-RAI outcomes upon qualitative analysis. Conclusion: Our systematic review strengthens previous work on antithyroid medication effects on RAI, and highlights that this field remains under researched especially for background medications unrelated to thyroid disease, with very few papers on non-thyroid medications published. Systematic review registration: https://www.crd.york.ac.uk/prospero/display_record.php, identifier CRD42021274026.


Subject(s)
Hyperthyroidism , Thyroid Neoplasms , Humans , Antithyroid Agents/therapeutic use , Iodine Radioisotopes/therapeutic use , Lithium/therapeutic use , Prednisone/therapeutic use , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/radiotherapy , Hyperthyroidism/radiotherapy , Hyperthyroidism/drug therapy , Treatment Outcome
13.
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
14.
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
15.
Hum Mol Genet ; 18(9): 1704-13, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19244275

ABSTRACT

Graves' disease (GD) is a common autoimmune disease (AID) that shares many of its susceptibility loci with other AIDs. The thyroid stimulating hormone receptor (TSHR) represents the primary autoantigen in GD, in which autoantibodies bind to the receptor and mimic its ligand, thyroid stimulating hormone, causing the characteristic clinical phenotype. Although early studies investigating the TSHR and GD proved inconclusive, more recently we provided convincing evidence for association of the TSHR region with disease. In the current study, we investigated a combined panel of 98 SNPs, including 70 tag SNPs, across an extended 800 kb region of the TSHR to refine association in a cohort of 768 GD subjects and 768 matched controls. In total, 28 SNPs revealed association with GD (P < 0.05), with strongest SNP associations at rs179247 (chi(2) = 32.45, P = 8.90 x 10(-8), OR = 1.53, 95% CI = 1.32-1.78) and rs12101255 (chi(2) = 30.91, P = 1.95 x 10(-7), OR = 1.55, 95% CI = 1.33-1.81), both located in intron 1 of the TSHR. Association of the most associated SNP, rs179247, was replicated in 303 GD families (P = 7.8 x 10(-4)). In addition, we provide preliminary evidence that the disease-associated genotypes of rs179247 (AA) and rs12101255 (TT) show reduced mRNA expression ratios of flTSHR relative to two alternate TSHR mRNA splice variants.


Subject(s)
Graves Disease/genetics , Receptors, Thyrotropin/genetics , Case-Control Studies , Cohort Studies , Gene Expression , Graves Disease/metabolism , Haplotypes , Humans , Introns , Linkage Disequilibrium , Polymorphism, Single Nucleotide , Receptors, Thyrotropin/metabolism , White People/genetics
16.
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
17.
Carcinogenesis ; 31(9): 1650-60, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20466759

ABSTRACT

The loss of anti-proliferative responsiveness in prostate cancer cell lines toward ligands for vitamin D receptor, retinoic acid receptors/retinoid X receptors and peroxisome proliferator activated receptor (PPAR)alpha/gamma may entail underlying epigenetic events, as ligand insensitivity reflects significantly altered messenger RNA expression of corepressors and histone-modifying enzymes. Expression patterns were dependent on phases of the cell cycle and associated with repressed basal gene expression of vitamin D receptor and PPARalpha/gamma target genes, for example CDKN1A [encodes p21((waf1/cip1))]. Elevated nuclear corepressor 1 (NCOR1) and nuclear corepressor 2/silencing mediator of retinoic acid and thyroid hormone receptor protein levels were detected in prostate cancer cell lines compared with non-malignant counterparts. Knockdown of the corepressor NCOR1 significantly elevated basal expression of a cohort of target genes, including CDKN1A. Both chemical [histone deacetylases inhibitor (HDACi)] and NCOR1 knockdown targeting enhanced anti-proliferative sensitivity toward PPARalpha/gamma ligands in prostate cancer cell lines. Pursuing PPARalpha/gamma signaling, microarray approaches were undertaken to identify pathways and genes regulated uniquely by a combination of PPARalpha/gamma activation and HDAC inhibition. Again, HDACi and knockdown approaches demonstrated that elevated NCOR1 expression and activity distorted PPARalpha/gamma gene targets centered on, for example cell cycle control, including CDKN1A and TGFBRAP1. Quantitative real time polymerase chain reaction validation and chromatin immunoprecipitation assays both confirmed that elevated NCOR1 disrupted the ability of PPARalpha/gamma to regulate key target genes (CDKN1A and TGFBRAP1). Interrogation of these relationships in prostate cancer samples using principal component and partial correlation analyses established significant interdependent relationships between NCOR1-PPARalpha/gamma and representative target genes, independently of androgen receptor expression. Therefore, we conclude that elevated NCOR1 distorts the actions of PPARalpha/gamma selectively and generates a potential epigenetic lesion with diagnostic and prognostic significance.


Subject(s)
Biomarkers, Tumor/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Epigenesis, Genetic , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Receptor Co-Repressor 1/metabolism , PPAR alpha/metabolism , PPAR gamma/metabolism , Prostatic Neoplasms/metabolism , Apoptosis , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Cycle , Cell Proliferation , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Profiling , Histone Deacetylase Inhibitors/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Nuclear Receptor Co-Repressor 1/antagonists & inhibitors , Nuclear Receptor Co-Repressor 1/genetics , Oligonucleotide Array Sequence Analysis , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL